Next Article in Journal
Advances in Biomedical Applications of Solution Blow Spinning
Next Article in Special Issue
Multiple Sclerosis-Associated Gut Microbiome in the Israeli Diverse Populations: Associations with Ethnicity, Gender, Disability Status, Vitamin D Levels, and Mediterranean Diet
Previous Article in Journal
Zinc Finger 521 Modulates the Nrf2-Notch Signaling Pathway in Human Ovarian Carcinoma
Previous Article in Special Issue
An Animal Model for Chronic Meningeal Inflammation and Inflammatory Demyelination of the Cerebral Cortex
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Interaction of the Gut Microbiome and Immunity in Multiple Sclerosis: Impact of Diet and Immune Therapy

by
Sudhir Kumar Yadav
1,
Kouichi Ito
1 and
Suhayl Dhib-Jalbut
1,2,*
1
Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
2
Rutgers New Jersey Medical School, Newark, NJ 07101, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(19), 14756; https://doi.org/10.3390/ijms241914756
Submission received: 31 August 2023 / Revised: 24 September 2023 / Accepted: 27 September 2023 / Published: 29 September 2023
(This article belongs to the Special Issue Molecular Mechanism in Multiple Sclerosis and Related Disorders)

Abstract

:
The bidirectional communication between the gut and central nervous system (CNS) through microbiota is known as the microbiota–gut–brain axis. The brain, through the enteric neural innervation and the vagus nerve, influences the gut physiological activities (motility, mucin, and peptide secretion), as well as the development of the mucosal immune system. Conversely, the gut can influence the CNS via intestinal microbiota, its metabolites, and gut-homing immune cells. Growing evidence suggests that gut immunity is critically involved in gut–brain communication during health and diseases, including multiple sclerosis (MS). The gut microbiota can influence the development and function of gut immunity, and conversely, the innate and adaptive mucosal immunity can influence microbiota composition. Gut and systemic immunity, along with gut microbiota, are perturbed in MS. Diet and disease-modifying therapies (DMTs) can affect the composition of the gut microbial community, leading to changes in gut and peripheral immunity, which ultimately affects MS. A high-fat diet is highly associated with gut dysbiosis-mediated inflammation and intestinal permeability, while a high-fiber diet/short-chain fatty acids (SCFAs) can promote the development of Foxp3 Tregs and improvement in intestinal barrier function, which subsequently suppress CNS autoimmunity in the animal model of MS (experimental autoimmune encephalomyelitis or EAE). This review will address the role of gut immunity and its modulation by diet and DMTs via gut microbiota during MS pathophysiology.

1. Introduction

Multiple sclerosis (MS) is a chronic autoimmune inflammatory disease of the central nervous system (CNS). MS is characterized by peripheral immune dysregulation and immune cell infiltration into the CNS, leading to demyelination and axonal damage, and, ultimately, neurodegeneration. It is suggested that CD4+Th1/Th17, B cells, and CD8+T cells play an important role during the initial inflammatory phase of MS [1]. In addition, glial cells such as microglia and astrocytes play critical roles during the neurodegenerative phase of MS. Both genetic and environmental factors contribute to immune dysregulation during MS onset and progression [2]. Earlier, we reported that the gut microbiota interacts with MS susceptibility genes to break immune tolerance to myelin antigens and leads to the development of experimental autoimmune encephalomyelitis (EAE) in 3A6 TCR/DR2a Tg mice [3]. So far, nine classes of disease-modifying therapies (DMTs), including interferons, glatiramer acetate, teriflunomide, sphingosine 1-phosphate receptor modulators, fumarates, cladribine, and monoclonal antibodies, have been approved by regulatory authorities to treat MS. These DMTs have been helpful in reducing clinical relapses and gadolinium-enhancing lesions on brain magnetic resonance imaging (MRI) [4].
The CNS influences gut physiological (gut motility and secretion) and immunological (development and functioning of the mucosal immune system) functions, while the gut influences CNS development and function via the intestinal microbiota and its metabolites, gut-homing immune cells, gut hormones, and the vagus nerve [5,6]. Such bidirectional communication between the gut and the CNS is commonly known as the gut–brain axis and could be a potential target for therapeutic intervention. Accumulating evidence indicates that brain-resident and gut-resident immune cells are critically involved in orchestrating gut–brain axis communication. The gut microbiota can influence the development and function of the intestinal immune system, and, conversely, the innate and adaptive immune system can influence microbiota composition [7]. Recently, we demonstrated in an MS animal model that perturbed gut immune homeostasis is associated with EAE development [8]. Similarly, immune dysregulation has been demonstrated in MS with excessive Th17 cell expansion in the intestine [9]. These reports suggest that gut immunity has a significant role in the immunopathogenesis of MS. Since diet and DMTs can influence gut immunity by affecting the gut microbial community [10], we hereby review their roles in modulating MS pathophysiology via gut immunity.

2. Role of Gut–Immune Axis in MS Pathogenesis

In a spontaneous EAE mouse model, we recently showed that CNS autoimmunity is triggered by altered gut and peripheral immunity due to gut dysbiosis [3,8]. Similarly, altered gut microbiota and gut immunity were found to be correlated with high disease activity in MS patients [9]. In addition, the transplantation of stool samples from MS patients into germ-free mice can increase the severity and incidence of EAE in the transplanted mice [11,12]. These reports suggest that gut dysbiosis may trigger the initiation and progression of CNS autoimmunity by promoting immune dysregulation in the gut (Figure 1).
Humanized gnotobiotic EAE mouse models contributed significantly to the understanding of the role of gut microbiota in MS pathogenesis [11,12]. However, the lack of standardized protocols, interspecies variation between mouse and human, and the type of fecal sample used (RRMS vs. SPMS vs. PPMS) in creating humanized gnotobiotic mouse models can lead to translatability issues. Interspecies parameters that can affect fecal transplantation are differences in human and mouse gut anatomy, digestion process and metabolic rate (quicker in mouse), and mouse genetic and immunological background [13]. In addition, another important limitation is the lack of a clear understanding of the cause–effect relationship between MS and intestinal microbiota dysbiosis.
Although they appear similar at the phyla level, human and murine gut floras have key discrepancies in the microbial composition and abundance. For example, a higher Firmicutes/Bacterioidetes ratio is observed in humans compared to mice. Further, the phylum Bacteroidetes mainly consists of the S24-7 family, and Firmicutes consists of Clostridiales in mice. But Bacteroidetes mainly consists of Bacteroidaceae, Prevotellaceae, and Firmicutes of the Ruminococcaceae family in humans [14]. Therefore, the results from EAE models should be interpreted with caution while studying the role of the gut microbiota in MS.

2.1. MS-Associated Gut Microbiota

The gut microbiome of treatment-naïve early-stage MS patients of different ethnicities (Caucasian, Hispanic, and African American) have increased relative abundances of Clostridia species compared to ethnicity-matched controls. However, other taxa showed significant differences among different ethnicities [15]. In both progressive MS and RRMS, Clostridium bolteae, Ruthenibacterium lactatiformans, and Akkermansia muciniphila were increased, while Blautia wexlerae, Dorea formicigenerans, and Erysipelotrichaceae CCMM were decreased. However, increased Enterobacteriaceae and Clostridium g24 FCEY and decreased Blautia and Agathobaculum were unique to progressive MS. Interestingly, several Clostridium species were associated with higher EDSS and fatigue scores [16]. A certain gut microbiota composition may be associated with subsequent MS relapse, especially in pediatric MS. Notably, Fusobacteria depletion was associated with pediatric MS relapse [17]. Future studies should focus on identifying more bacterial species involved in MS relapse. Except for increased A. muciniphila, the bacterial taxa involved in the development of gut dysbiosis among individuals with MS varied among studies [11,12,16,18]. These variations may be due to interindividual differences in the baseline microbial composition caused by host genetic factors, long-term dietary habits, environmental exposures related to race/ethnicity, and/or geographical location [15,19,20]. It is unknown whether increased A. muciniphila contribute to MS pathogenesis or are a consequence of the disease. Interestingly, a recent report suggests a link between increased Akkermansia and lower disability, suggesting that Akkermansia may have a beneficial role [16]. It is important that future studies explore the disease-specific roles and mode of action of MS-associated gut bacteria.

2.2. MS-Associated Dysregulation of Gut Immunity

Dysregulated gut and peripheral immunity are consequences of gut dysbiosis (change in gut microbiota). Many reports suggest a link between MS pathogenicity and gut immunity. For example, Th17 cells are now widely accepted to be key players in MS pathogenesis, and an increased frequency of intestinal Th17 cells correlates with high disease activity and altered gut microbiota in MS patients [9]. Also, disruption of the intestinal barrier and increased permeability, often referred to as “leaky gut”, are involved in the pathogenesis of autoimmune diseases [21]. Interestingly, intestinal barrier dysfunction develops at the onset of EAE, which is associated with Th17 cell infiltration in the small intestine as well as an increase in intestinal permeability [22]. Distinct signals from gut microorganisms coordinately activate myelin oligodendrocyte glycoprotein (MOG)-specific Th17 cells in the small intestine. Germ-free mice colonized with two bacteria from the small intestine, a strain from the Erysipelotrichaceae family and Lactobacillus reuteri, develop more severe EAE compared to germ-free or monocolonized mice. The strain from Erysipelotrichaceae acts as an adjuvant to enhance the Th17 cells’ response, while Lactobacillus reuteri possesses peptides that potentially mimic MOG. Therefore, the synergistic effects of these microorganisms may be involved in the pathogenicity of MS [23]. MOG-specific Th17 cells infiltrate the colonic lamina propria prior to the development of neurological symptoms in active and adaptive transfer EAE models and alter gut microbiota composition. Disrupting Th17 cell trafficking to the large intestine significantly attenuates EAE [24]. Likewise, we have demonstrated the infiltration of myelin basic protein (MBP)-specific Th17 cells as well as the recruitment of neutrophils in the colon of a spontaneous EAE mouse model [8]. Of note, neutrophils can promote Th17 cell differentiation by neutrophil extracellular traps (NET) and their histones via the Toll-like receptor (TLR) pathway [25]. These reports suggest that the gut may be a location for the differentiation of encephalitogenic Th17 cells in the periphery, with the microbiota playing an important role in their differentiation, activation, and migration to the CNS. It was hypothesized that this could be the result of cross-reactivity between bacterial antigens and endogenous CNS antigens (molecular mimicry) or bystander activation [26]. Recently, we discovered a variant of surface layer protein A (SLPA) in a subtype of Clostridioides difficile (strain DJNS06-36), which can activate MBP89-98-reactive T cells. SLPA contains an amino acid sequence that resembles immunodominant myelin basic protein 89–98. Importantly, active immunization with SLPA activates MBP-specific T cells and induces EAE in MBP-TCR/DR2a Tg mice. This study suggests that the encephalitogenic mimotope of MBP of gut bacteria can activate autoreactive myelin-specific T cells and trigger CNS autoimmunity [27]. In summary, MS-associated gut bacterial species have functional effects on the immune system that can potentially modulate MS pathogenesis (Table 1).
In addition, in severe cases of MS, commensal-specific gut IgA responses are drastically reduced, with a simultaneous increase in serum IgG responses against IgA-unbound bacteria compared to controls [31]. Further, the mobilization of IgA+ Plasma blast and/or plasma cells from the gut to the CNS can significantly suppress neuroinflammation [32]. In healthy individuals, C-C chemokine receptor type 9 (CCR9)+ memory T cells exhibited a regulatory profile characterized by both the expression of C-MAF and the production of IL-4 and IL-10. However, in CCR9+ memory T cells, the expression of RORγt was specifically upregulated, and the production of IL-17A and IFN-γ was high in patients with secondary progressive MS (SPMS) compared to healthy controls, indicating the loss of regulatory function [33]. These animal and clinical studies suggest that an imbalance in the gut microbiome between anti-inflammatory and pro-inflammatory bacteria may promote immune dysregulation and increase the risk of MS.

2.3. Dysregulated Gut Immunity May Promote MS Relapses

Gut dysbiosis-mediated intestinal inflammation could be a risk factor for disease exacerbation in MS. Recently, we showed that CNS autoimmunity is associated with gut inflammation, which is probably triggered by a reduction in the enteric bacteria involved in the development of regulatory immune cells. Also, we observed the gut infiltration of Th1 and Th17 cells as well as the recruitment of neutrophils during the development of spontaneous EAE [8]. Inflammatory cytokines like TNF-α, IFN-γ, and IL-1β produced by immune cells during gut inflammation disrupt tight junction proteins, resulting in increased intestinal permeability [34,35]. Notably, intestinal permeability is increased in patients with MS compared with healthy donors [36]. This could be one of the risk factors involved in disease exacerbation. Intestinal permeability induces the translocation of microbial components into the systemic circulation, which could break peripheral immune tolerance [3]. Animal studies also show that increased intestinal permeability exacerbates EAE and promotes disease progression [22]. Further, microbial dysbiosis can affect the production of bacterial metabolites that promote or suppress CNS autoimmunity. For example, microbial dysbiosis reduces the production of short-chain fatty acids (SCFAs), which are important metabolites for the development of forkhead box P3 (Foxp3) Tregs and for the maintenance of immune homeostasis [37,38]. Therefore, the gut dysbiosis-mediated reduction of SCFAs could be a risk factor in MS. Finally, intestinal regulatory and pathogenic immune cells can migrate from the intestine to the CNS [39,40]. Therefore, gut dysbiosis may reduce the migration of regulatory cells and increase the migration of pathogenic immune cells to the CNS. Collectively, these studies suggest that dysregulated gut immunity may promote MS disease activity.

2.4. Contribution of Gut Microbiota to CNS Pathology in MS

MS pathology involves inflammatory and neurodegenerative processes. Neuroinflammation is predominant in the early stages of MS, which is mediated by immune cells, whereas neurodegeneration is dominant in the later stages of MS and is mainly driven by microglia and astrocytes, whose activity can be modulated by the gut microbiota [41]. An immature microglia phenotype with diminished immune function and enhanced proliferation and survival observed in germ free mice compared to specific-pathogen-free (SPF) mice, point to the role of microbiota in microglia development and function [42]. Further, a subtype of reactive astrocytes (A1 astrocytes) is induced by activated neuroinflammatory microglia by secreting IL-1α, TNF-α, and C1q [43]. In addition, transforming growth factor-α (TGF-α) and vascular endothelial growth factor-B (VEGF-B), produced by microglia, regulate the pathogenic activities of astrocytes in EAE. Microglia-derived TGF-α limits pathogenic activities of astrocytes and acts via the ErbB1 receptor during EAE. Conversely, microglial-derived VEGF-B activates fms-related receptor tyrosine kinase-1 (FLT-1) signaling in astrocytes to worsen EAE. Interestingly, the metabolites produced by the commensal flora from dietary tryptophan control microglial activation and TGF-α and VEGF-B production by the aryl hydrocarbon receptor [44]. Similarly, neurotoxicity in MS is induced via the microbially derived metabolites, phenol and indole, produced by the tryptophan and phenylalanine catabolism [45]. During homeostatic conditions, a subset of astrocytes expresses the lysosomal-associated membrane protein 1 (LAMP1) and the TNF-related apoptosis-inducing ligand (TRAIL). The TRAIL expression in the astrocytes is promoted by interferon-γ (IFN-γ) produced by meningeal natural killer (NK) cells. Notably, IFN-γ expression in the NK cells are modulated by the gut microbiome. These LAMP1+TRAIL+ astrocytes limit neuroinflammation in the CNS by inducing T cell apoptosis through TRAIL–DR5 signaling. However, during inflammation, TRAIL expression in astrocytes is suppressed by molecules produced by T cells and microglia [46]. In addition, the gut microbiota may affect CNS pathology by producing toxins. For example, Clostridium perfringens, which can produce epsilon toxin (ETX), were found in the majority of RRMS patients. Importantly, ETX can disrupt the blood–brain barrier to promote multifocal lesions in the brain and spinal cord of the EAE model, resembling MS lesion pathology [47]. These reports suggest that the gut microbiota can modulate glial cell phenotypes and the blood–brain barrier to promote CNS pathology in MS.

3. Mechanisms of the Gut–Immune-Axis-Mediated Effect in the CNS

The gut–immune axis affects the CNS during health and disease by several mechanisms (Figure 2). First, microbial metabolites and endogenous components from the gut microbiota can pass to the circulation and affect peripheral immunity during health and disease. The most notable microbial metabolites are SCFAs like acetic acid, butyric acid, and propionic acid. SCFAs are produced by the fermentation of dietary fiber in the colon by anaerobic gut bacteria [48,49,50]. In RRMS patients, the fecal levels of acetate, propionate, and butyrate are significantly lower compared to HC [51]. Interestingly, female RRMS patients showed significantly reduced fecal SCFA concentrations compared to male RRMS patients, possibly contributing to the higher female susceptibility to MS [52]. SCFAs are known to suppresses autoimmunity through immunomodulatory effects, which have been attributed partially to the epigenetic modulation of immune cells via the inhibition of the histone deacetylase (HDAC) enzyme. One of the downstream effects is enhanced regulatory T cells (Tregs), which suppress autoimmunity [53]. Indeed, the number of CD4+ CD25+ FOXP3+ Tregs are decreased in MS patients and have impaired suppressive capacity [54,55]. The SCFA pentanoate suppresses autoimmunity by inducing IL-10 production in lymphocytes and reprogramming their metabolic activity towards elevated glucose oxidation. In addition, pentanoate-induced regulatory B cells suppress autoimmune pathology in colitis and MS animal models [50]. Also, tryptophan and its metabolites are emerging as important modulators of mucosal and CNS immunity via the aryl hydrocarbon receptor (AHR). Tryptophan metabolites like kynurenine, kynurenic acid, anthranilic acid, quinolinate, indole-3-acetic acid, indoxyl-3-sulfate, indole-3-propionic acid, and indole-3-aldehyde are produced by the action of gut microbiota on dietary tryptophan [56]. Tryptophan metabolites protect against increased gut permeability through the aryl hydrocarbon receptor by maintaining the apical junctional complex and its regulatory proteins (myosin IIA and ezrin) [57]. In addition, tryptophan metabolites also signal through the AHR in astrocytes and reduce CNS autoimmunity via the Suppressor of Cytokine Signaling 2 (SOCS2)-mediated inhibition of NF-κB-driven inflammation [58]. Interestingly, the tryptophan metabolites kynurenine, kynurenic acid, anthranilic acid, and quinolinate are low in MS serum, indicating a possible role in MS pathogenesis [58]. Further, lower serum tryptophan and indole lactate (tryptophan metabolite) are associated with pediatric MS risk and disease course [59]. Contrary to previous reports, kynurenic acid has been shown to promote the accumulation of Th17-inducing GPR35+ Ly6C+ macrophages in the small intestine of EAE mice before disease induction. Sporosarcina pasteurii, Staphylococcus lentus, Pseudoxanthomonas mexicana, and Sphingomonas were identified as potential species involved in kynurenic acid production [60]. Therefore, tryptophan-metabolizing gut bacteria and the metabolic end-product will modulate the effect on CNS autoimmunity. Among bacterial components, polysaccharide-A (PSA) and LPS are well investigated. PSA is a capsular polysaccharide produced by a Gram-negative symbiont, Bacteroides fragilis, in the colon. PSA from human gut bacteria Bacteroides fragilis protects against EAE by inducing IL-10-producing FoxP3+ Treg cells [61]. Further, PSA promotes human CD39+Foxp3+Treg cells and Treg function [62]. PSA suppresses EAE by the expansion of CD4+Treg. Further, CD39, which is an ectonucleotidase, promotes the accumulation of CD39+CD4+ Tregs in the CNS to suppress autoimmunity [63]. Interestingly, Bacteroides fragilis is significantly depleted in pediatric MS patients, suggesting its possible role in MS pathogenesis [64]. In addition, gut dysbiosis during CNS autoimmunity increases the serum level of lipopolysaccharide (LPS), which promotes the loss of peripheral immune tolerance [3,37]. LPS injection has been shown to exacerbate EAE [65]. Further, systemic LPS can activate microglia to increase pro-inflammatory factors in the CNS, consequently contributing to neurodegeneration [66]. In summary, it can be concluded that microbiota-derived metabolites and components will either promote or suppress CNS autoimmunity.
Second, in homeostatic and disease conditions, CNS function and behavior can be affected by resident innate and adaptive immune cell populations [67,68,69]. Interestingly, many of these immune cells are derived from the periphery and their development can be modulated by gut microbiota [70,71]. Depending on the composition of the gut microbiota, the latter can promote or suppress neuroinflammation and/or demyelination via the development of Th1/Th17 or Treg cells, respectively [72,73]. In fact, the dysregulation of TGF-beta/Smad7 signaling has been reported in the intestine of MS patients. This favors an inflammatory phenotype in intestinal CD4+ T cells and leads to migration to the CNS to cause autoimmunity [40]. Further, the gut-homing of myelin-specific Th17 cells is required for disease induction in the adoptive transfer EAE model, suggesting a role for gut microbiota/gut environment in the differentiation of encephalitogenic Th17 cells [24]. Conversely, gut-microbiota-specific IgA+ B cells can traffic to the CNS in active MS and dampen excessive inflammation [32,74]. Therefore, the gut microbiota can affect the CNS during health and disease by affecting immune cell homing and migration pathways.
Third, the vagus nerve and enteric nervous system (ENS) innervate the intestinal wall and play a significant role in bidirectional communication between the gut and the CNS. The ENS can function independently of the vagus nerve to control gut physiology like motility and secretion. The ENS is made of myenteric (Auerbach’s) and submucosal (Meissner’s) plexuses, which are connected to CNS via the vagus nerve [75]. The gut wall is enervated with both afferent and efferent fibers of the vagus nerve and its activity can be modulated by the diffusion of bacterial components (like LPS) and metabolites (like SCFAs) or hormonal signals from specialized enteroendocrine cells (EECs), which are capable of sensing luminal bacterial content [76]. Only a few studies have explored the role of the ENS in CNS autoimmunity. For example, in a B-cell- and antibody-dependent mouse EAE model, the degeneration of the myenteric plexus with gliosis and axonal loss caused a decrease in intestinal motility before the onset of EAE had been reported. Interestingly, gliosis and ENS degeneration were also detected in resected colon from MS patients. Further, both EAE mice and MS patients have serum autoantibodies against antigens derived from enteric neurons and/or glia [77]. Also, altered gastrointestinal motility in EAE models was reported due to autoantibodies targeting the ENS [78]. These reports suggest that the ENS may be involved in the pathogenesis of MS, which warrants further studies.
Lastly, gut hormones can also affect the CNS. It is important to note that gut-microbiota-derived LPS and SCFAs modulate the production and release of gut hormones. Cholecystokinin (CCK), ghrelin, peptide YY, glucagon-like peptide-1 (GLP-1), and 5-hydroxytryptamine (5-HT) or serotonin are some of the important gut hormones produced by EEC in the gastrointestinal tract [79]. GLP-1 potentially suppresses neuroinflammation since it attenuates LPS-induced inflammatory responses in microglia [80]. In fact, Semaglutide, a novel glucagon-like peptide-1 agonist, has been shown to suppress EAE in rats [80]. Most of the body’s serotonin is secreted by EEC. Fluoxetine, a serotonin reuptake inhibitor, and 5-HT suppress Th17-immune responses in multiple sclerosis (MS). Such an effect is facilitated by the activation of 5-HT2B receptors, which reduces IL-17, IFN-γ, and GM-CSF production in MS [81]. Therefore, gut hormones may play an important role in MS pathogenesis.

4. Effect of Diet on the Gut–Immune Axis in MS

4.1. High-Fiber Diet

Dietary fiber is an important food component involved in the maintenance of health, which can be mediated by microbiota-dependent or independent mechanisms [82]. Dietary fiber is an edible plant carbohydrate polymer that is resistant to digestion and absorption in the small intestine, with complete or partial fermentation in the large intestine. Dietary fiber includes polysaccharides (cellulose, hemicellulose, and pectin), oligosaccharides, lignin, and related plant molecules [83]. Bacterial species from Bifidobacterium, Prevotella, and Bacteroides are the main degraders of dietary fiber to SCFAs such as butyrate, propionate, and acetate [84]. Dietary fiber and SCFAs can suppress an immune response through G protein-coupled receptors or acting as histone deacetylase inhibitors [85]. SCFAs are critical to maintain intestinal homeostasis and immunity by inducing IL-22 production in CD4+ T cells and innate lymphoid cells 3 (ILC3) [86]. Further, SCFAs drive monocyte-to-macrophage differentiation via histone deacetylase 3 (HDAC3) inhibition. Interestingly, these macrophages have enhanced antimicrobial activity due to a shift in metabolism, a reduction in mammalian target of rapamycin (mTOR) kinase activity, and increased LC3-associated host defense [87]. Therefore, SCFAs can prevent gut dysbiosis by inhibiting the overgrowth of pathogenic bacteria. Importantly, dietary fiber and SCFAs have been shown to inhibit CNS autoimmunity in vivo through the differentiation of Tregs and the inhibition of pathogenic Th cells [88,89]. Tregs differentiation is mediated by the induction of tolerogenic dendritic cells (DC) and/or the enhanced acetylation of the Foxp3 gene by SCFAs through the inhibition of histone deacetylase activity [38,90]. In addition, SCFAs can affect immune cell metabolism and T helper cell differentiation through mTOR signaling [91].
Besides immune modulation, SCFAs are the preferred energy source for intestinal epithelial cells and maintain a healthy intestinal epithelial layer by promoting epithelial cell proliferation and turnover [92]. Further, dietary fiber and SCFAs can prevent autoimmunity by promoting intestinal barrier function and preventing the migration of intestinal lymphocytes to extraintestinal tissues [93,94]. The SCFAs produced by the microbiota are important to suppress inflammatory microglia, which partially depend on epigenetic modifications [95]. In addition, butyrate treatment has been shown to suppress demyelination and enhance remyelination by modulating oligodendrocytes directly [96]. SCFAs maintain the blood–brain barrier (BBB) and blood–cerebrospinal fluid (CSF) barrier through the increased expression of tight junction (TJ) proteins as well as cytoskeleton rearrangement in endothelial cells [97,98,99]. SCFA diets alleviate cognitive and spatial memory deficits by enhanced astrocyte–neuron metabolic coupling, leading to reduced oxidative damage [100]. Therefore, dietary fiber and its metabolite SCFAs are important for immune and nervous system homeostasis, thereby helpful in suppressing autoimmunity and neurodegeneration.
MS is highly associated with gut dysbiosis, characterized by the loss of bacterial taxa involved in the fermentation of dietary fiber and the production of SCFAs [8,101]. Since SCFAs can promote the differentiation of Tregs, a decrease in SCFA-producing bacteria in the gut would be a risk factor for MS. Indeed, the oral intake of propionic acid promotes a sustained increase in functionally competent Treg cells, while Th1 and Th17 cells are significantly decreased in MS patients [102]. Similarly, a high-vegetable/low-protein diet (HV/LP diet), which is rich in dietary fiber, is associated with reduced relapse rate and less disability due to microbial composition change and the induction of IL10-producing monocytes and Tregs [103]. Also, animal studies have indicated that feeding a high-fiber diet or diet rich in SCFAs ameliorated EAE through the increased differentiation of Tregs and the downregulation of Th1 cells [50,89]. Important ongoing or completed studies addressing the effects of different diets on MS are summarized in Table 2. Dietary fiber and SCFAs also affect neurodegenerative diseases. For example, fecal SCFA levels are lower in Alzheimer’s disease (AD) patients, and its supplementation can potentially provide therapeutic benefits for AD since they can prevent the formation of neurotoxic Aβ aggregates and Aβ-induced microglia activation [104,105,106]. Thus, an increase in the consumption of a high-fiber diet could be beneficial for neurodegenerative diseases including MS. Recently, a high-fiber supplement was shown to efficiently reduce gut dysbiosis and promote the growth of SCFA-producing gut bacteria in humans [107]. The effect of the high-fiber supplement in MS gut dysbiosis and disease induction in EAE is currently being investigated in our laboratory.

4.2. Western Diet/High-Fat Diet (HFD)

Western diets/HFDs can impact gut–brain communications by altering the gut microbiota and intestinal permeability. Commonly, the consumption of a HFD leads to an increase in Firmicutes and a decrease in Bacteroidetes, which are associated with obesity and chronic diseases [110]. Interestingly, obesity is associated with higher disease activity and poorer outcome in newly diagnosed MS patients [111]. The consumption of a HFD led to unfavorable changes in fecal and plasma metabolite and plasma factors, resulting in poor long-term health outcomes [112]. In addition, HFDs have unfavorable effects due to the dysregulation of gut immunity and barrier function [113,114]. HFDs have been shown to induce neuroinflammation, oxidative stress, and neuronal death in the brain cortex and hippocampus of mice [115]. Further, a HFD exacerbated neuroinflammation in an animal model of MS by promoting microglial activation and T cell infiltration [116]. As the neurotrophic factor BDNF plays a key role in brain function, HFD-induced neuroinflammation suppresses brain-derived neurotrophic factor (BDNF)-related pathways [117]. Similarly, saturated fat increases the risk of relapse in pediatric MS [118]. However, polyunsaturated fatty acids (PUFAs) such as omega-3 fatty acids have been shown to reduce disease severity in an MS mouse model. This beneficial effect is due to the reduced polarization of naïve T cells toward proinflammatory Th1 and Th17 phenotypes [119]. In fact, a higher intake of omega-3 supplementation reduces the risk of MS in patients with clinically isolated syndrome [120]. In addition, omega-3 supplementation has been associated with decreased relapse rate and inflammatory markers and improved quality of life in MS patients [121]. Similarly, omega-6 supplementation decreases pro-inflammatory monocyte number and function, with a simultaneous increase in anti-inflammatory monocyte subsets and functions in MS patients [122]. A HFD increases the risk of neurodegenerative diseases like Alzheimer’s and Parkinson’s disease [123,124], perhaps in connection with HFD-induced gut dysbiosis [125].

5. Effect of MS Therapeutics on the Gut–Immune Axis in MS

Disease-modifying therapy (DMT)-induced changes in gut microbiota may contribute toward their therapeutic effects (Table 3). For example, by promoting anti-inflammatory gut bacteria, DMTs can enhance Tregs or the production of regulatory cytokines in the gut and the circulation. Recently, the gut microbiomes of 576 MS patients and 1152 genetically unrelated household healthy controls (HHC) were studied by the International Multiple Sclerosis Microbiome Study (iMSMS). In this study, differences in β-diversity were observed when patients within each treatment group were compared to their corresponding HHC. However, β-diversity did not differ between the treated and untreated RRMS patients except for the IFN-β treated group [10]. Similarly, in another study, the overall composition of the microbiota did not differ significantly between treated and untreated MS patients. However, specific bacteria were found to be linked to each DMT [16]. Overall, DMT-induced changes in microbial populations may impact biological and metabolic pathways. For example, glatiramer acetate (GA) and dimethyl fumarate (DMF) mainly upregulate retinol (vitamin A) metabolism, which is known to promote the differentiation of Tregs and suppress the reprogramming of Treg to Th17 cells during intestinal inflammation [126,127]. In addition, both therapies decreased methane metabolism. An increased relative abundance of proinflammatory methane-producing bacteria, Methanobrevibacter, and exhaled methane was reported in MS patients compared with healthy controls [18,126]. Therefore, by decreasing methane-producing bacteria, GA and DMF have beneficial therapeutic effects. Although certain DMT-induced changes in the gut microbiota have been identified, no specific microbiota DMT signatures are currently established. This can be explained by two factors: interindividual differences in base microbiota and use of previous DMTs. First, the baseline microbial compositions have interindividual differences caused by host genetic factors, dietary habits, environmental exposures, and/or geographical location [15,19,128]. Therefore, different study cohorts might have different microbiota compositions after DMT. The effects of individual DMTs on bacterial composition are summarized in Table 3. Second, treatment guidelines do not provide guidance on how to select or sequence DMTs, which leads to switches or discontinuations of initial DMTs (~70% of patients) [129]. Therefore, it will be hard to predict a gut microbiota signature for a specific DMT. It is also unknown how the gut microbiota modulates the response to DMT. We hypothesize that the gut microbiota can affect DMT actions by affecting its metabolism via the production of enzymes that degrade or activate drugs [130]. In addition, the gut microbiota can produce some metabolites that could compete with DMTs or affect the level of metabolite transporters, which may affect its bioavailability. Therefore, gut microbiota composition may contribute to the efficacy of DMTs.

6. Conclusions

Emerging evidence suggests important roles for the gut microbiome and gut immunity in human health and disease. MS is associated with perturbed gut microbiomes and immunity, supporting the role of the gut microbiome in the initiation and progression of MS. However, comprehensive knowledge about the functional effects of MS-associated bacteria is lacking at present. It is important that future studies explore the functional effects and mode of action of MS-associated gut bacteria, which would be helpful in designing therapeutic strategies. A high-fiber diet and disease-modifying therapies (DMTs) can have beneficial effects for MS by shifting the composition of the gut microbial community to an anti-inflammatory phenotype, leading to the differentiation of regulatory immune cells and an improvement in intestinal barrier function. However, it is not known how the gut microbiota modulates DMTs; the role played by baseline microbiomes in determining the response to DMTs is also unknown. Studies in this direction could help personalize the choice of DMTs, since the microbiome is varied among different MS populations and disease states.

Author Contributions

S.K.Y. wrote the manuscript. K.I. and S.D.-J. provided content, reviewed, and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant from the USA National Multiple Sclerosis Society (RG-1901-33077) and the Ruth Dunietz Kushner and Michael Jay Serwitz endowed Chair in Multiple Sclerosis.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rodriguez Murua, S.; Farez, M.F.; Quintana, F.J. The Immune Response in Multiple Sclerosis. Annu. Rev. Pathol. 2022, 17, 121–139. [Google Scholar] [CrossRef] [PubMed]
  2. Waubant, E.; Lucas, R.; Mowry, E.; Graves, J.; Olsson, T.; Alfredsson, L.; Langer-Gould, A. Environmental and genetic risk factors for MS: An integrated review. Ann. Clin. Transl. Neurol. 2019, 6, 1905–1922. [Google Scholar] [CrossRef] [PubMed]
  3. Yadav, S.K.; Boppana, S.; Ito, N.; Mindur, J.E.; Mathay, M.T.; Patel, A.; Dhib-Jalbut, S.; Ito, K. Gut dysbiosis breaks immunological tolerance toward the central nervous system during young adulthood. Proc. Natl. Acad. Sci. USA 2017, 114, E9318–E9327. [Google Scholar] [CrossRef] [PubMed]
  4. McGinley, M.P.; Goldschmidt, C.H.; Rae-Grant, A.D. Diagnosis and Treatment of Multiple Sclerosis: A Review. JAMA 2021, 325, 765–779. [Google Scholar] [CrossRef]
  5. Rutsch, A.; Kantsjo, J.B.; Ronchi, F. The Gut-Brain Axis: How Microbiota and Host Inflammasome Influence Brain Physiology and Pathology. Front. Immunol. 2020, 11, 604179. [Google Scholar] [CrossRef] [PubMed]
  6. Kasarello, K.; Cudnoch-Jedrzejewska, A.; Czarzasta, K. Communication of gut microbiota and brain via immune and neuroendocrine signaling. Front. Microbiol. 2023, 14, 1118529. [Google Scholar] [CrossRef] [PubMed]
  7. Sittipo, P.; Choi, J.; Lee, S.; Lee, Y.K. The function of gut microbiota in immune-related neurological disorders: A review. J. Neuroinflamm. 2022, 19, 154. [Google Scholar] [CrossRef]
  8. Yadav, S.K.; Ito, N.; Mindur, J.E.; Kumar, H.; Youssef, M.; Suresh, S.; Kulkarni, R.; Rosario, Y.; Balashov, K.E.; Dhib-Jalbut, S.; et al. Fecal Lcn-2 level is a sensitive biological indicator for gut dysbiosis and intestinal inflammation in multiple sclerosis. Front. Immunol. 2022, 13, 1015372. [Google Scholar] [CrossRef]
  9. Cosorich, I.; Dalla-Costa, G.; Sorini, C.; Ferrarese, R.; Messina, M.J.; Dolpady, J.; Radice, E.; Mariani, A.; Testoni, P.A.; Canducci, F.; et al. High frequency of intestinal T(H)17 cells correlates with microbiota alterations and disease activity in multiple sclerosis. Sci. Adv. 2017, 3, e1700492. [Google Scholar] [CrossRef]
  10. iMSMS Consortium. Gut microbiome of multiple sclerosis patients and paired household healthy controls reveal associations with disease risk and course. Cell 2022, 185, 3467–3486.e16. [Google Scholar] [CrossRef]
  11. Berer, K.; Gerdes, L.A.; Cekanaviciute, E.; Jia, X.; Xiao, L.; Xia, Z.; Liu, C.; Klotz, L.; Stauffer, U.; Baranzini, S.E.; et al. Gut microbiota from multiple sclerosis patients enables spontaneous autoimmune encephalomyelitis in mice. Proc. Natl. Acad. Sci. USA 2017, 114, 10719–10724. [Google Scholar] [CrossRef] [PubMed]
  12. 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.; et al. 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] [PubMed]
  13. Park, J.C.; Im, S.H. Of men in mice: The development and application of a humanized gnotobiotic mouse model for microbiome therapeutics. Exp. Mol. Med. 2020, 52, 1383–1396. [Google Scholar] [CrossRef] [PubMed]
  14. Nagpal, R.; Wang, S.; Solberg Woods, L.C.; Seshie, O.; Chung, S.T.; Shively, C.A.; Register, T.C.; Craft, S.; McClain, D.A.; Yadav, H. Comparative Microbiome Signatures and Short-Chain Fatty Acids in Mouse, Rat, Non-human Primate, and Human Feces. Front. Microbiol. 2018, 9, 2897. [Google Scholar] [CrossRef]
  15. Ventura, R.E.; Iizumi, T.; Battaglia, T.; Liu, M.; Perez-Perez, G.I.; Herbert, J.; Blaser, M.J. Gut microbiome of treatment-naive MS patients of different ethnicities early in disease course. Sci. Rep. 2019, 9, 16396. [Google Scholar] [CrossRef] [PubMed]
  16. Cox, L.M.; Maghzi, A.H.; Liu, S.; Tankou, S.K.; Dhang, F.H.; Willocq, V.; Song, A.; Wasen, C.; Tauhid, S.; Chu, R.; et al. Gut Microbiome in Progressive Multiple Sclerosis. Ann. Neurol. 2021, 89, 1195–1211. [Google Scholar] [CrossRef]
  17. Tremlett, H.; Fadrosh, D.W.; Faruqi, A.A.; Hart, J.; Roalstad, S.; Graves, J.; Lynch, S.; Waubant, E.; US Network of Pediatric MS Centers. Gut microbiota composition and relapse risk in pediatric MS: A pilot study. J. Neurol. Sci. 2016, 363, 153–157. [Google Scholar] [CrossRef]
  18. Jangi, S.; Gandhi, R.; Cox, L.M.; Li, N.; von Glehn, F.; Yan, R.; Patel, B.; Mazzola, M.A.; Liu, S.; Glanz, B.L.; et al. Alterations of the human gut microbiome in multiple sclerosis. Nat. Commun. 2016, 7, 12015. [Google Scholar] [CrossRef]
  19. Kurilshikov, A.; Medina-Gomez, C.; Bacigalupe, R.; Radjabzadeh, D.; Wang, J.; Demirkan, A.; Le Roy, C.I.; Raygoza Garay, J.A.; Finnicum, C.T.; Liu, X.; et al. Large-scale association analyses identify host factors influencing human gut microbiome composition. Nat. Genet. 2021, 53, 156–165. [Google Scholar] [CrossRef]
  20. Bourdeau-Julien, I.; Castonguay-Paradis, S.; Rochefort, G.; Perron, J.; Lamarche, B.; Flamand, N.; Di Marzo, V.; Veilleux, A.; Raymond, F. The diet rapidly and differentially affects the gut microbiota and host lipid mediators in a healthy population. Microbiome 2023, 11, 26. [Google Scholar] [CrossRef]
  21. An, J.; Liu, Y.; Wang, Y.; Fan, R.; Hu, X.; Zhang, F.; Yang, J.; Chen, J. The Role of Intestinal Mucosal Barrier in Autoimmune Disease: A Potential Target. Front. Immunol. 2022, 13, 871713. [Google Scholar] [CrossRef] [PubMed]
  22. 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]
  23. Miyauchi, E.; Kim, S.W.; Suda, W.; Kawasumi, M.; Onawa, S.; Taguchi-Atarashi, N.; Morita, H.; Taylor, T.D.; Hattori, M.; Ohno, H. Gut microorganisms act together to exacerbate inflammation in spinal cords. Nature 2020, 585, 102–106.e4. [Google Scholar] [CrossRef]
  24. Duc, D.; Vigne, S.; Bernier-Latmani, J.; Yersin, Y.; Ruiz, F.; Gaïa, N.; Leo, S.; Lazarevic, V.; Schrenzel, J.; Petrova, T.V.; et al. Disrupting Myelin-Specific Th17 Cell Gut Homing Confers Protection in an Adoptive Transfer Experimental Autoimmune Encephalomyelitis. Cell Rep. 2019, 29, 378–390.e4. [Google Scholar] [CrossRef] [PubMed]
  25. Wilson, A.S.; Randall, K.L.; Pettitt, J.A.; Ellyard, J.I.; Blumenthal, A.; Enders, A.; Quah, B.J.; Bopp, T.; Parish, C.R.; Brustle, A. Neutrophil extracellular traps and their histones promote Th17 cell differentiation directly via TLR2. Nat. Commun. 2022, 13, 528. [Google Scholar] [CrossRef]
  26. Berer, K.; Krishnamoorthy, G. Microbial view of central nervous system autoimmunity. FEBS Lett. 2014, 588, 4207–4213. [Google Scholar] [CrossRef]
  27. Mindur, J.E.; Yadav, S.K.; Ito, N.; Senoh, M.; Kato, H.; Dhib-Jalbut, S.; Ito, K. Surface Layer Protein A Expressed in Clostridioides difficile DJNS06-36 Possesses an Encephalitogenic Mimotope of Myelin Basic Protein. Microorganisms 2020, 9, 34. [Google Scholar] [CrossRef]
  28. Miyake, S.; Kim, S.; Suda, W.; Oshima, K.; Nakamura, M.; Matsuoka, T.; Chihara, N.; Tomita, A.; Sato, W.; Kim, S.W.; et al. Dysbiosis in the Gut Microbiota of Patients with Multiple Sclerosis, with a Striking Depletion of Species Belonging to Clostridia XIVa and IV Clusters. PLoS ONE 2015, 10, e0137429. [Google Scholar] [CrossRef]
  29. Thirion, F.; Sellebjerg, F.; Fan, Y.; Lyu, L.; Hansen, T.H.; Pons, N.; Levenez, F.; Quinquis, B.; Stankevic, E.; Sondergaard, H.B.; et al. The gut microbiota in multiple sclerosis varies with disease activity. Genome Med. 2023, 15, 1. [Google Scholar] [CrossRef]
  30. 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]
  31. Sterlin, D.; Larsen, M.; Fadlallah, J.; Parizot, C.; Vignes, M.; Autaa, G.; Dorgham, K.; Juste, C.; Lepage, P.; Aboab, J.; et al. Perturbed Microbiota/Immune Homeostasis in Multiple Sclerosis. Neurol. Neuroimmunol. Neuroinflamm. 2021, 8, e997. [Google Scholar] [CrossRef]
  32. Rojas, O.L.; Probstel, A.K.; Porfilio, E.A.; Wang, A.A.; Charabati, M.; Sun, T.; Lee, D.S.W.; Galicia, G.; Ramaglia, V.; Ward, L.A.; et al. Recirculating Intestinal IgA-Producing Cells Regulate Neuroinflammation via IL-10. Cell 2019, 176, 610–624.e18. [Google Scholar] [CrossRef]
  33. Kadowaki, A.; Saga, R.; Lin, Y.; Sato, W.; Yamamura, T. Gut microbiota-dependent CCR9+CD4+ T cells are altered in secondary progressive multiple sclerosis. Brain 2019, 142, 916–931. [Google Scholar] [CrossRef]
  34. Woznicki, J.A.; Saini, N.; Flood, P.; Rajaram, S.; Lee, C.M.; Stamou, P.; Skowyra, A.; Bustamante-Garrido, M.; Regazzoni, K.; Crawford, N.; et al. TNF-alpha synergises with IFN-gamma to induce caspase-8-JAK1/2-STAT1-dependent death of intestinal epithelial cells. Cell Death Dis. 2021, 12, 864. [Google Scholar] [CrossRef] [PubMed]
  35. Meyer, F.; Wendling, D.; Demougeot, C.; Prati, C.; Verhoeven, F. Cytokines and intestinal epithelial permeability: A systematic review. Autoimmun. Rev. 2023, 22, 103331. [Google Scholar] [CrossRef] [PubMed]
  36. Buscarinu, M.C.; Gargano, F.; Lionetto, L.; Capi, M.; Morena, E.; Fornasiero, A.; Renie, R.; Landi, A.C.; Pellicciari, G.; Romano, C.; et al. Intestinal Permeability and Circulating CD161+CCR6+CD8+T Cells in Patients With Relapsing-Remitting Multiple Sclerosis Treated With Dimethylfumarate. Front. Neurol. 2021, 12, 683398. [Google Scholar] [CrossRef] [PubMed]
  37. Saresella, M.; Marventano, I.; Barone, M.; La Rosa, F.; Piancone, F.; Mendozzi, L.; d’Arma, A.; Rossi, V.; Pugnetti, L.; Roda, G.; et al. Alterations in Circulating Fatty Acid Are Associated With Gut Microbiota Dysbiosis and Inflammation in Multiple Sclerosis. Front. Immunol. 2020, 11, 1390. [Google Scholar] [CrossRef] [PubMed]
  38. Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly, Y.M.; Glickman, J.N.; Garrett, W.S. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569–573. [Google Scholar] [CrossRef]
  39. Fitzpatrick, Z.; Frazer, G.; Ferro, A.; Clare, S.; Bouladoux, N.; Ferdinand, J.; Tuong, Z.K.; Negro-Demontel, M.L.; Kumar, N.; Suchanek, O.; et al. Gut-educated IgA plasma cells defend the meningeal venous sinuses. Nature 2020, 587, 472–476. [Google Scholar] [CrossRef]
  40. Haupeltshofer, S.; Leichsenring, T.; Berg, S.; Pedreiturria, X.; Joachim, S.C.; Tischoff, I.; Otte, J.M.; Bopp, T.; Fantini, M.C.; Esser, C.; et al. Smad7 in intestinal CD4(+) T cells determines autoimmunity in a spontaneous model of multiple sclerosis. Proc. Natl. Acad. Sci. USA 2019, 116, 25860–25869. [Google Scholar] [CrossRef]
  41. Mey, G.M.; Mahajan, K.R.; DeSilva, T.M. Neurodegeneration in multiple sclerosis. WIREs Mech. Dis. 2023, 15, e1583. [Google Scholar] [CrossRef] [PubMed]
  42. Erny, D.; Hrabe de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef] [PubMed]
  43. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Munch, A.E.; Chung, W.S.; Peterson, T.C.; et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef] [PubMed]
  44. Rothhammer, V.; Borucki, D.M.; Tjon, E.C.; Takenaka, M.C.; Chao, C.-C.; Ardura-Fabregat, A.; De Lima, K.A.; Gutiérrez-Vázquez, C.; Hewson, P.; Staszewski, O.; et al. Microglial control of astrocytes in response to microbial metabolites. Nature 2018, 557, 724–728. [Google Scholar] [CrossRef] [PubMed]
  45. Ntranos, A.; Park, H.J.; Wentling, M.; Tolstikov, V.; Amatruda, M.; Inbar, B.; Kim-Schulze, S.; Frazier, C.; Button, J.; Kiebish, M.A.; et al. Bacterial neurotoxic metabolites in multiple sclerosis cerebrospinal fluid and plasma. Brain 2022, 145, 569–583. [Google Scholar] [CrossRef]
  46. Sanmarco, L.M.; Wheeler, M.A.; Gutierrez-Vazquez, C.; Polonio, C.M.; Linnerbauer, M.; Pinho-Ribeiro, F.A.; Li, Z.; Giovannoni, F.; Batterman, K.V.; Scalisi, G.; et al. Gut-licensed IFNgamma(+) NK cells drive LAMP1(+)TRAIL(+) anti-inflammatory astrocytes. Nature 2021, 590, 473–479. [Google Scholar] [CrossRef]
  47. Reder, A.T. Clostridium epsilon toxin is excessive in multiple sclerosis and provokes multifocal lesions in mouse models. J. Clin. Investig. 2023, 133, e169643. [Google Scholar] [CrossRef]
  48. West, D.B.; Diaz, J.; Roddy, S.; Woods, S.C. Long-term effects on adiposity after preweaning nutritional manipulations in the gastrostomy-reared rat. J. Nutr. 1987, 117, 1259–1264. [Google Scholar] [CrossRef]
  49. Fusco, W.; Lorenzo, M.B.; Cintoni, M.; Porcari, S.; Rinninella, E.; Kaitsas, F.; Lener, E.; Mele, M.C.; Gasbarrini, A.; Collado, M.C.; et al. Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota. Nutrients 2023, 15, 2211. [Google Scholar] [CrossRef]
  50. Luu, M.; Pautz, S.; Kohl, V.; Singh, R.; Romero, R.; Lucas, S.; Hofmann, J.; Raifer, H.; Vachharajani, N.; Carrascosa, L.C.; et al. The short-chain fatty acid pentanoate suppresses autoimmunity by modulating the metabolic-epigenetic crosstalk in lymphocytes. Nat. Commun. 2019, 10, 760. [Google Scholar] [CrossRef]
  51. Takewaki, D.; Suda, W.; Sato, W.; Takayasu, L.; Kumar, N.; Kimura, K.; Kaga, N.; Mizuno, T.; Miyake, S.; Hattori, M.; et al. 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]
  52. Becker, A.; Abuazab, M.; Schwiertz, A.; Walter, S.; Fassbender, K.C.; Fousse, M.; Unger, M.M. Short-chain fatty acids and intestinal inflammation in multiple sclerosis: Modulation of female susceptibility by microbial products? Auto. Immun. Highlights 2021, 12, 7. [Google Scholar] [CrossRef]
  53. McBride, D.A.; Dorn, N.C.; Yao, M.; Johnson, W.T.; Wang, W.; Bottini, N.; Shah, N.J. Short-chain fatty acid-mediated epigenetic modulation of inflammatory T cells in vitro. Drug Deliv. Transl. Res. 2023, 13, 1912–1924. [Google Scholar] [CrossRef] [PubMed]
  54. Li, Y.F.; Zhang, S.X.; Ma, X.W.; Xue, Y.L.; Gao, C.; Li, X.Y.; Xu, A.D. The proportion of peripheral regulatory T cells in patients with Multiple Sclerosis: A meta-analysis. Mult. Scler. Relat. Disord. 2019, 28, 75–80. [Google Scholar] [CrossRef] [PubMed]
  55. Viglietta, V.; Baecher-Allan, C.; Weiner, H.L.; Hafler, D.A. Loss of functional suppression by CD4+CD25+ regulatory T cells in patients with multiple sclerosis. J. Exp. Med. 2004, 199, 971–979. [Google Scholar] [CrossRef]
  56. Ma, N.; He, T.; Johnston, L.J.; Ma, X. Host-microbiome interactions: The aryl hydrocarbon receptor as a critical node in tryptophan metabolites to brain signaling. Gut Microbes 2020, 11, 1203–1219. [Google Scholar] [CrossRef]
  57. Scott, S.A.; Fu, J.; Chang, P.V. Microbial tryptophan metabolites regulate gut barrier function via the aryl hydrocarbon receptor. Proc. Natl. Acad. Sci. USA 2020, 117, 19376–19387. [Google Scholar] [CrossRef]
  58. Rothhammer, V.; Mascanfroni, I.D.; Bunse, L.; Takenaka, M.C.; Kenison, J.E.; Mayo, L.; Chao, C.C.; Patel, B.; Yan, R.; Blain, M.; et al. Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor. Nat. Med. 2016, 22, 586–597. [Google Scholar] [CrossRef]
  59. Nourbakhsh, B.; Bhargava, P.; Tremlett, H.; Hart, J.; Graves, J.; Waubant, E. Altered tryptophan metabolism is associated with pediatric multiple sclerosis risk and course. Ann. Clin. Transl. Neurol. 2018, 5, 1211–1221. [Google Scholar] [CrossRef]
  60. Miyamoto, K.; Sujino, T.; Harada, Y.; Ashida, H.; Yoshimatsu, Y.; Yonemoto, Y.; Nemoto, Y.; Tomura, M.; Melhem, H.; Niess, J.H.; et al. The gut microbiota-induced kynurenic acid recruits GPR35-positive macrophages to promote experimental encephalitis. Cell Rep. 2023, 42, 113005. [Google Scholar] [CrossRef]
  61. Ochoa-Reparaz, J.; Mielcarz, D.W.; Wang, Y.; Begum-Haque, S.; Dasgupta, S.; Kasper, D.L.; Kasper, L.H. A polysaccharide from the human commensal Bacteroides fragilis protects against CNS demyelinating disease. Mucosal Immunol. 2010, 3, 487–495. [Google Scholar] [CrossRef] [PubMed]
  62. Telesford, K.M.; Yan, W.; Ochoa-Reparaz, J.; Pant, A.; Kircher, C.; Christy, M.A.; Begum-Haque, S.; Kasper, D.L.; Kasper, L.H. A commensal symbiotic factor derived from Bacteroides fragilis promotes human CD39(+)Foxp3(+) T cells and Treg function. Gut Microbes 2015, 6, 234–242. [Google Scholar] [CrossRef] [PubMed]
  63. Wang, Y.; Begum-Haque, S.; Telesford, K.M.; Ochoa-Reparaz, J.; Christy, M.; Kasper, E.J.; Kasper, D.L.; Robson, S.C.; Kasper, L.H. A commensal bacterial product elicits and modulates migratory capacity of CD39(+) CD4 T regulatory subsets in the suppression of neuroinflammation. Gut Microbes 2014, 5, 552–561. [Google Scholar] [CrossRef] [PubMed]
  64. Mirza, A.; Forbes, J.D.; Zhu, F.; Bernstein, C.N.; Van Domselaar, G.; Graham, M.; Waubant, E.; Tremlett, H. The multiple sclerosis gut microbiota: A systematic review. Mult. Scler. Relat. Disord. 2020, 37, 101427. [Google Scholar] [CrossRef] [PubMed]
  65. Mardiguian, S.; Ladds, E.; Turner, R.; Shepherd, H.; Campbell, S.J.; Anthony, D.C. The contribution of the acute phase response to the pathogenesis of relapse in chronic-relapsing experimental autoimmune encephalitis models of multiple sclerosis. J. Neuroinflamm. 2017, 14, 196. [Google Scholar] [CrossRef]
  66. Qin, L.; Wu, X.; Block, M.L.; Liu, Y.; Breese, G.R.; Hong, J.S.; Knapp, D.J.; Crews, F.T. Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration. Glia 2007, 55, 453–462. [Google Scholar] [CrossRef]
  67. Croese, T.; Castellani, G.; Schwartz, M. Immune cell compartmentalization for brain surveillance and protection. Nat. Immunol. 2021, 22, 1083–1092. [Google Scholar] [CrossRef]
  68. Rustenhoven, J.; Drieu, A.; Mamuladze, T.; de Lima, K.A.; Dykstra, T.; Wall, M.; Papadopoulos, Z.; Kanamori, M.; Salvador, A.F.; Baker, W.; et al. Functional characterization of the dural sinuses as a neuroimmune interface. Cell 2021, 184, 1000–1016.e27. [Google Scholar] [CrossRef]
  69. Alves de Lima, K.; Rustenhoven, J.; Da Mesquita, S.; Wall, M.; Salvador, A.F.; Smirnov, I.; Martelossi Cebinelli, G.; Mamuladze, T.; Baker, W.; Papadopoulos, Z.; et al. Meningeal gammadelta T cells regulate anxiety-like behavior via IL-17a signaling in neurons. Nat. Immunol. 2020, 21, 1421–1429. [Google Scholar] [CrossRef]
  70. Choileain, S.N.; Kleinewietfeld, M.; Raddassi, K.; Hafler, D.A.; Ruff, W.E.; Longbrake, E.E. CXCR3+ T cells in multiple sclerosis correlate with reduced diversity of the gut microbiome. J. Transl. Autoimmun. 2020, 3, 100032. [Google Scholar] [CrossRef]
  71. Sonner, J.K.; Keil, M.; Falk-Paulsen, M.; Mishra, N.; Rehman, A.; Kramer, M.; Deumelandt, K.; Rowe, J.; Sanghvi, K.; Wolf, L.; et al. Dietary tryptophan links encephalogenicity of autoreactive T cells with gut microbial ecology. Nat. Commun. 2019, 10, 4877. [Google Scholar] [CrossRef] [PubMed]
  72. Mangalam, A.; Shahi, S.K.; Luckey, D.; Karau, M.; Marietta, E.; Luo, N.; Choung, R.S.; Ju, J.; Sompallae, R.; Gibson-Corley, K.; et al. Human Gut-Derived Commensal Bacteria Suppress CNS Inflammatory and Demyelinating Disease. Cell Rep. 2017, 20, 1269–1277. [Google Scholar] [CrossRef] [PubMed]
  73. 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] [PubMed]
  74. Probstel, A.K.; Zhou, X.; Baumann, R.; Wischnewski, S.; Kutza, M.; Rojas, O.L.; Sellrie, K.; Bischof, A.; Kim, K.; Ramesh, A.; et al. Gut microbiota-specific IgA(+) B cells traffic to the CNS in active multiple sclerosis. Sci. Immunol. 2020, 5, eabc7191. [Google Scholar] [CrossRef] [PubMed]
  75. Fung, C.; Vanden Berghe, P. Functional circuits and signal processing in the enteric nervous system. Cell Mol. Life Sci. 2020, 77, 4505–4522. [Google Scholar] [CrossRef]
  76. Han, Y.; Wang, B.; Gao, H.; He, C.; Hua, R.; Liang, C.; Zhang, S.; Wang, Y.; Xin, S.; Xu, J. Vagus Nerve and Underlying Impact on the Gut Microbiota-Brain Axis in Behavior and Neurodegenerative Diseases. J. Inflamm. Res. 2022, 15, 6213–6230. [Google Scholar] [CrossRef]
  77. Wunsch, M.; Jabari, S.; Voussen, B.; Enders, M.; Srinivasan, S.; Cossais, F.; Wedel, T.; Boettner, M.; Schwarz, A.; Weyer, L.; et al. The enteric nervous system is a potential autoimmune target in multiple sclerosis. Acta Neuropathol. 2017, 134, 281–295. [Google Scholar] [CrossRef]
  78. Spear, E.T.; Holt, E.A.; Joyce, E.J.; Haag, M.M.; Mawe, S.M.; Hennig, G.W.; Lavoie, B.; Applebee, A.M.; Teuscher, C.; Mawe, G.M. Altered gastrointestinal motility involving autoantibodies in the experimental autoimmune encephalomyelitis model of multiple sclerosis. Neurogastroenterol. Motil. 2018, 30, e13349. [Google Scholar] [CrossRef]
  79. Martin, A.M.; Sun, E.W.; Rogers, G.B.; Keating, D.J. The Influence of the Gut Microbiome on Host Metabolism Through the Regulation of Gut Hormone Release. Front. Physiol. 2019, 10, 428. [Google Scholar] [CrossRef]
  80. Yoon, G.; Kim, Y.K.; Song, J. Glucagon-like peptide-1 suppresses neuroinflammation and improves neural structure. Pharmacol. Res. 2020, 152, 104615. [Google Scholar] [CrossRef]
  81. Sviridova, A.; Rogovskii, V.; Kudrin, V.; Pashenkov, M.; Boyko, A.; Melnikov, M. The role of 5-HT(2B)-receptors in fluoxetine-mediated modulation of Th17- and Th1-cells in multiple sclerosis. J. Neuroimmunol. 2021, 356, 577608. [Google Scholar] [CrossRef] [PubMed]
  82. Cai, Y.; Folkerts, J.; Folkerts, G.; Maurer, M.; Braber, S. Microbiota-dependent and -independent effects of dietary fibre on human health. Br. J. Pharmacol. 2020, 177, 1363–1381. [Google Scholar] [CrossRef] [PubMed]
  83. Berding, K.; Carbia, C.; Cryan, J.F. Going with the grain: Fiber, cognition, and the microbiota-gut-brain-axis. Exp. Biol. Med. 2021, 246, 796–811. [Google Scholar] [CrossRef] [PubMed]
  84. Tan, J.K.; Macia, L.; Mackay, C.R. Dietary fiber and SCFAs in the regulation of mucosal immunity. J. Allergy Clin. Immunol. 2023, 151, 361–370. [Google Scholar] [CrossRef]
  85. Van der Hee, B.; Wells, J.M. Microbial Regulation of Host Physiology by Short-chain Fatty Acids. Trends Microbiol. 2021, 29, 700–712. [Google Scholar] [CrossRef]
  86. Yang, W.; Yu, T.; Huang, X.; Bilotta, A.J.; Xu, L.; Lu, Y.; Sun, J.; Pan, F.; Zhou, J.; Zhang, W.; et al. Intestinal microbiota-derived short-chain fatty acids regulation of immune cell IL-22 production and gut immunity. Nat. Commun. 2020, 11, 4457. [Google Scholar] [CrossRef]
  87. Schulthess, J.; Pandey, S.; Capitani, M.; Rue-Albrecht, K.C.; Arnold, I.; Franchini, F.; Chomka, A.; Ilott, N.E.; Johnston, D.G.W.; Pires, E.; et al. The Short Chain Fatty Acid Butyrate Imprints an Antimicrobial Program in Macrophages. Immunity 2019, 50, 432–445.e7. [Google Scholar] [CrossRef]
  88. Park, J.; Wang, Q.; Wu, Q.; Mao-Draayer, Y.; Kim, C.H. Bidirectional regulatory potentials of short-chain fatty acids and their G-protein-coupled receptors in autoimmune neuroinflammation. Sci. Rep. 2019, 9, 8837. [Google Scholar] [CrossRef]
  89. Fettig, N.M.; Robinson, H.G.; Allanach, J.R.; Davis, K.M.; Simister, R.L.; Wang, E.J.; Sharon, A.J.; Ye, J.; Popple, S.J.; Seo, J.H.; et al. Inhibition of Th1 activation and differentiation by dietary guar gum ameliorates experimental autoimmune encephalomyelitis. Cell Rep. 2022, 40, 111328. [Google Scholar] [CrossRef]
  90. Goverse, G.; Molenaar, R.; Macia, L.; Tan, J.; Erkelens, M.N.; Konijn, T.; Knippenberg, M.; Cook, E.C.; Hanekamp, D.; Veldhoen, M.; et al. Diet-Derived Short Chain Fatty Acids Stimulate Intestinal Epithelial Cells To Induce Mucosal Tolerogenic Dendritic Cells. J. Immunol. 2017, 198, 2172–2181. [Google Scholar] [CrossRef]
  91. Kim, C.H.; Park, J.; Kim, M. Gut microbiota-derived short-chain Fatty acids, T cells, and inflammation. Immune Netw. 2014, 14, 277–288. [Google Scholar] [CrossRef] [PubMed]
  92. Bilotta, A.J.; Ma, C.; Yang, W.; Yu, Y.; Yu, Y.; Zhao, X.; Zhou, Z.; Yao, S.; Dann, S.M.; Cong, Y. Propionate Enhances Cell Speed and Persistence to Promote Intestinal Epithelial Turnover and Repair. Cell Mol. Gastroenterol. Hepatol. 2021, 11, 1023–1044. [Google Scholar] [CrossRef] [PubMed]
  93. Feng, Y.; Wang, Y.; Wang, P.; Huang, Y.; Wang, F. Short-Chain Fatty Acids Manifest Stimulative and Protective Effects on Intestinal Barrier Function Through the Inhibition of NLRP3 Inflammasome and Autophagy. Cell Physiol. Biochem. 2018, 49, 190–205. [Google Scholar] [CrossRef] [PubMed]
  94. Nakamura, Y.K.; Janowitz, C.; Metea, C.; Asquith, M.; Karstens, L.; Rosenbaum, J.T.; Lin, P. Short chain fatty acids ameliorate immune-mediated uveitis partially by altering migration of lymphocytes from the intestine. Sci. Rep. 2017, 7, 11745. [Google Scholar] [CrossRef] [PubMed]
  95. Caetano-Silva, M.E.; Rund, L.; Hutchinson, N.T.; Woods, J.A.; Steelman, A.J.; Johnson, R.W. Inhibition of inflammatory microglia by dietary fiber and short-chain fatty acids. Sci. Rep. 2023, 13, 2819. [Google Scholar] [CrossRef] [PubMed]
  96. Chen, T.; Noto, D.; Hoshino, Y.; Mizuno, M.; Miyake, S. Butyrate suppresses demyelination and enhances remyelination. J. Neuroinflamm. 2019, 16, 165. [Google Scholar] [CrossRef]
  97. Knox, E.G.; Aburto, M.R.; Tessier, C.; Nagpal, J.; Clarke, G.; O’Driscoll, C.M.; Cryan, J.F. Microbial-derived metabolites induce actin cytoskeletal rearrangement and protect blood-brain barrier function. iScience 2022, 25, 105648. [Google Scholar] [CrossRef]
  98. Xie, J.; Bruggeman, A.; De Nolf, C.; Vandendriessche, C.; Van Imschoot, G.; Van Wonterghem, E.; Vereecke, L.; Vandenbroucke, R.E. Gut microbiota regulates blood-cerebrospinal fluid barrier function and Abeta pathology. EMBO J. 2023, 42, e111515. [Google Scholar] [CrossRef]
  99. Tang, C.F.; Wang, C.Y.; Wang, J.H.; Wang, Q.N.; Li, S.J.; Wang, H.O.; Zhou, F.; Li, J.M. Short-Chain Fatty Acids Ameliorate Depressive-like Behaviors of High Fructose-Fed Mice by Rescuing Hippocampal Neurogenesis Decline and Blood-Brain Barrier Damage. Nutrients 2022, 14, 1882. [Google Scholar] [CrossRef]
  100. Sun, Y.; Zhang, H.; Zhang, X.; Wang, W.; Chen, Y.; Cai, Z.; Wang, Q.; Wang, J.; Shi, Y. Promotion of astrocyte-neuron glutamate-glutamine shuttle by SCFA contributes to the alleviation of Alzheimer’s disease. Redox Biol. 2023, 62, 102690. [Google Scholar] [CrossRef]
  101. Ordonez-Rodriguez, A.; Roman, P.; Rueda-Ruzafa, L.; Campos-Rios, A.; Cardona, D. Changes in Gut Microbiota and Multiple Sclerosis: A Systematic Review. Int. J. Environ. Res. Public. Health 2023, 20, 4624. [Google Scholar] [CrossRef] [PubMed]
  102. Duscha, A.; Gisevius, B.; Hirschberg, S.; Yissachar, N.; Stangl, G.I.; Dawin, E.; Bader, V.; Haase, S.; Kaisler, J.; David, C.; et al. Propionic Acid Shapes the Multiple Sclerosis Disease Course by an Immunomodulatory Mechanism. Cell 2020, 180, 1067–1080.e16. [Google Scholar] [CrossRef]
  103. Saresella, M.; Mendozzi, L.; Rossi, V.; Mazzali, F.; Piancone, F.; LaRosa, F.; Marventano, I.; Caputo, D.; Felis, G.E.; Clerici, M. Immunological and Clinical Effect of Diet Modulation of the Gut Microbiome in Multiple Sclerosis Patients: A Pilot Study. Front. Immunol. 2017, 8, 1391. [Google Scholar] [CrossRef] [PubMed]
  104. Ho, L.; Ono, K.; Tsuji, M.; Mazzola, P.; Singh, R.; Pasinetti, G.M. Protective roles of intestinal microbiota derived short chain fatty acids in Alzheimer’s disease-type beta-amyloid neuropathological mechanisms. Expert. Rev. Neurother. 2018, 18, 83–90. [Google Scholar] [CrossRef] [PubMed]
  105. Wu, L.; Han, Y.; Zheng, Z.; Peng, G.; Liu, P.; Yue, S.; Zhu, S.; Chen, J.; Lv, H.; Shao, L.; et al. Altered Gut Microbial Metabolites in Amnestic Mild Cognitive Impairment and Alzheimer’s Disease: Signals in Host-Microbe Interplay. Nutrients 2021, 13, 228. [Google Scholar] [CrossRef]
  106. Liu, J.; Li, H.; Gong, T.; Chen, W.; Mao, S.; Kong, Y.; Yu, J.; Sun, J. Anti-neuroinflammatory Effect of Short-Chain Fatty Acid Acetate against Alzheimer’s Disease via Upregulating GPR41 and Inhibiting ERK/JNK/NF-kappaB. J. Agric. Food Chem. 2020, 68, 7152–7161. [Google Scholar] [CrossRef] [PubMed]
  107. Zhao, L.; Zhang, F.; Ding, X.; Wu, G.; Lam, Y.Y.; Wang, X.; Fu, H.; Xue, X.; Lu, C.; Ma, J.; et al. Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science 2018, 359, 1151–1156. [Google Scholar] [CrossRef]
  108. Black, L.J.; Rowley, C.; Sherriff, J.; Pereira, G.; Ponsonby, A.L.; Lucas, R.M. A healthy dietary pattern associates with a lower risk of a first clinical diagnosis of central nervous system demyelination. Mult. Scler. 2019, 25, 1514–1525. [Google Scholar] [CrossRef]
  109. Sedaghat, F.; Jessri, M.; Behrooz, M.; Mirghotbi, M.; Rashidkhani, B. Mediterranean diet adherence and risk of multiple sclerosis: A case-control study. Asia Pac. J. Clin. Nutr. 2016, 25, 377–384. [Google Scholar] [CrossRef]
  110. Malesza, I.J.; Malesza, M.; Walkowiak, J.; Mussin, N.; Walkowiak, D.; Aringazina, R.; Bartkowiak-Wieczorek, J.; Madry, E. High-Fat, Western-Style Diet, Systemic Inflammation, and Gut Microbiota: A Narrative Review. Cells 2021, 10, 3164. [Google Scholar] [CrossRef]
  111. Lutfullin, I.; Eveslage, M.; Bittner, S.; Antony, G.; Flaskamp, M.; Luessi, F.; Salmen, A.; Gisevius, B.; Klotz, L.; Korsukewitz, C.; et al. Association of obesity with disease outcome in multiple sclerosis. J. Neurol. Neurosurg. Psychiatry 2023, 94, 57–61. [Google Scholar] [CrossRef] [PubMed]
  112. Wan, Y.; Wang, F.; Yuan, J.; Li, J.; Jiang, D.; Zhang, J.; Li, H.; Wang, R.; Tang, J.; Huang, T.; et al. Effects of dietary fat on gut microbiota and faecal metabolites, and their relationship with cardiometabolic risk factors: A 6-month randomised controlled-feeding trial. Gut 2019, 68, 1417–1429. [Google Scholar] [CrossRef] [PubMed]
  113. Luck, H.; Khan, S.; Kim, J.H.; Copeland, J.K.; Revelo, X.S.; Tsai, S.; Chakraborty, M.; Cheng, K.; Tao Chan, Y.; Nohr, M.K.; et al. Gut-associated IgA(+) immune cells regulate obesity-related insulin resistance. Nat. Commun. 2019, 10, 3650. [Google Scholar] [CrossRef] [PubMed]
  114. Rohr, M.W.; Narasimhulu, C.A.; Rudeski-Rohr, T.A.; Parthasarathy, S. Negative Effects of a High-Fat Diet on Intestinal Permeability: A Review. Adv. Nutr. 2020, 11, 77–91. [Google Scholar] [CrossRef] [PubMed]
  115. Ribeiro, M.F.; Santos, A.A.; Afonso, M.B.; Rodrigues, P.M.; Sa Santos, S.; Castro, R.E.; Rodrigues, C.M.P.; Sola, S. Diet-dependent gut microbiota impacts on adult neurogenesis through mitochondrial stress modulation. Brain Commun. 2020, 2, fcaa165. [Google Scholar] [CrossRef]
  116. 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]
  117. Zhao, Z.; Yao, M.; Wei, L.; Ge, S. Obesity caused by a high-fat diet regulates the Sirt1/PGC-1alpha/FNDC5/BDNF pathway to exacerbate isoflurane-induced postoperative cognitive dysfunction in older mice. Nutr. Neurosci. 2020, 23, 971–982. [Google Scholar] [CrossRef]
  118. Azary, S.; Schreiner, T.; Graves, J.; Waldman, A.; Belman, A.; Guttman, B.W.; Aaen, G.; Tillema, J.M.; Mar, S.; Hart, J.; et al. Contribution of dietary intake to relapse rate in early paediatric multiple sclerosis. J. Neurol. Neurosurg. Psychiatry 2018, 89, 28–33. [Google Scholar] [CrossRef]
  119. Kim, J.S.; Soto-Diaz, K.; Bingham, T.W.; Steelman, A.J.; Das, A. Role of omega-3 endocannabinoids in the modulation of T-cell activity in a multiple sclerosis experimental autoimmune encephalomyelitis (EAE) model. J. Biol. Chem. 2023, 299, 102886. [Google Scholar] [CrossRef]
  120. Hoare, S.; Lithander, F.; van der Mei, I.; Ponsonby, A.L.; Lucas, R.; Ausimmune Investigator, G. 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. 2016, 22, 884–892. [Google Scholar] [CrossRef]
  121. AlAmmar, W.A.; Albeesh, F.H.; Ibrahim, L.M.; Algindan, Y.Y.; Yamani, L.Z.; Khattab, R.Y. Effect of omega-3 fatty acids and fish oil supplementation on multiple sclerosis: A systematic review. Nutr. Neurosci. 2021, 24, 569–579. [Google Scholar] [CrossRef] [PubMed]
  122. Fleck, A.K.; Hucke, S.; Teipel, F.; Eschborn, M.; Janoschka, C.; Liebmann, M.; Wami, H.; Korn, L.; Pickert, G.; Hartwig, M.; et al. Dietary conjugated linoleic acid links reduced intestinal inflammation to amelioration of CNS autoimmunity. Brain 2021, 144, 1152–1166. [Google Scholar] [CrossRef] [PubMed]
  123. Samadi, M.; Moradi, S.; Moradinazar, M.; Mostafai, R.; Pasdar, Y. Dietary pattern in relation to the risk of Alzheimer’s disease: A systematic review. Neurol. Sci. 2019, 40, 2031–2043. [Google Scholar] [CrossRef]
  124. Hantikainen, E.; Roos, E.; Bellocco, R.; D’Antonio, A.; Grotta, A.; Adami, H.O.; Ye, W.; Trolle Lagerros, Y.; Bonn, S. Dietary fat intake and risk of Parkinson disease: Results from the Swedish National March Cohort. Eur. J. Epidemiol. 2022, 37, 603–613. [Google Scholar] [CrossRef]
  125. Saiyasit, N.; Chunchai, T.; Prus, D.; Suparan, K.; Pittayapong, P.; Apaijai, N.; Pratchayasakul, W.; Sripetchwandee, J.; Chattipakorn, M.D.P.D.N.; Chattipakorn, S.C. Gut dysbiosis develops before metabolic disturbance and cognitive decline in high-fat diet-induced obese condition. Nutrition 2020, 69, 110576. [Google Scholar] [CrossRef] [PubMed]
  126. Katz Sand, I.; Zhu, Y.; Ntranos, A.; Clemente, J.C.; Cekanaviciute, E.; Brandstadter, R.; Crabtree-Hartman, E.; Singh, S.; Bencosme, Y.; Debelius, J.; et al. Disease-modifying therapies alter gut microbial composition in MS. Neurol. Neuroimmunol. Neuroinflamm 2019, 6, e517. [Google Scholar] [CrossRef]
  127. Tejon, G.; Manriquez, V.; De Calisto, J.; Flores-Santibanez, F.; Hidalgo, Y.; Crisostomo, N.; Fernandez, D.; Sauma, D.; Mora, J.R.; Bono, M.R.; et al. Vitamin A Impairs the Reprogramming of Tregs into IL-17-Producing Cells during Intestinal Inflammation. Biomed. Res. Int. 2015, 2015, 137893. [Google Scholar] [CrossRef]
  128. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef]
  129. Fox, R.J.; Mehta, R.; Pham, T.; Park, J.; Wilson, K.; Bonafede, M. Real-world disease-modifying therapy pathways from administrative claims data in patients with multiple sclerosis. BMC Neurol. 2022, 22, 211. [Google Scholar] [CrossRef]
  130. Li, X.; Liu, L.; Cao, Z.; Li, W.; Li, H.; Lu, C.; Yang, X.; Liu, Y. Gut microbiota as an “invisible organ” that modulates the function of drugs. Biomed. Pharmacother. 2020, 121, 109653. [Google Scholar] [CrossRef]
  131. Castillo-Alvarez, F.; Perez-Matute, P.; Oteo, J.A.; Marzo-Sola, M.E. The influence of interferon beta-1b on gut microbiota composition in patients with multiple sclerosis. Neurologia 2021, 36, 495–503. [Google Scholar] [CrossRef] [PubMed]
  132. Diebold, M.; Meola, M.; Purushothaman, S.; Siewert, L.K.; Possnecker, E.; Roloff, T.; Lindberg, R.L.; Kuhle, J.; Kappos, L.; Derfuss, T.; et al. Gut microbiota composition as a candidate risk factor for dimethyl fumarate-induced lymphopenia in multiple sclerosis. Gut Microbes 2022, 14, 2147055. [Google Scholar] [CrossRef] [PubMed]
  133. Ferri, C.; Castellazzi, M.; Merli, N.; Laudisi, M.; Baldin, E.; Baldi, E.; Mancabelli, L.; Ventura, M.; Pugliatti, M. Gut Microbiota Changes during Dimethyl Fumarate Treatment in Patients with Multiple Sclerosis. Int. J. Mol. Sci. 2023, 24, 2720. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Role of gut–immune axis in MS pathogenesis. (A) Dysbiotic MS microbiota can promote the differentiation of myelin-specific T cells into Th1/Th17 cells by bystander action and molecular mimicry. (B,C) Myelin-specific Th1/Th17 cells, neutrophiles, and macrophages can cause gut inflammation and increased gut permeability by proinflammatory cytokines. (D) Increased gut permeability leads to the passage of bacterial endotoxins from the gut to periphery including the CNS, which may activate microglia and astrocytes to cause MS onset and/or progression. (EG) MS gut microbiota is unable to produce sufficient levels of SCFAs, which may further promote intestinal permeability and neuroinflammation. (H) Certain bacterial species produce toxins and promote CNS pathology in MS. (I) Mobilization of IgA+ plasma blast and/or plasma cells from the gut to the CNS can significantly suppress neuroinflammation, and gut dysbiosis may suppress the development of IgA+ B cells in the gut. Schematic diagram was created using BioRender.com (accessed on: 13 September 2023).
Figure 1. Role of gut–immune axis in MS pathogenesis. (A) Dysbiotic MS microbiota can promote the differentiation of myelin-specific T cells into Th1/Th17 cells by bystander action and molecular mimicry. (B,C) Myelin-specific Th1/Th17 cells, neutrophiles, and macrophages can cause gut inflammation and increased gut permeability by proinflammatory cytokines. (D) Increased gut permeability leads to the passage of bacterial endotoxins from the gut to periphery including the CNS, which may activate microglia and astrocytes to cause MS onset and/or progression. (EG) MS gut microbiota is unable to produce sufficient levels of SCFAs, which may further promote intestinal permeability and neuroinflammation. (H) Certain bacterial species produce toxins and promote CNS pathology in MS. (I) Mobilization of IgA+ plasma blast and/or plasma cells from the gut to the CNS can significantly suppress neuroinflammation, and gut dysbiosis may suppress the development of IgA+ B cells in the gut. Schematic diagram was created using BioRender.com (accessed on: 13 September 2023).
Ijms 24 14756 g001
Figure 2. Mechanisms of gut–immune-axis-mediated effect on the CNS. (A) Microbial metabolites (SCFAs from dietary fibers and tryptophan metabolites) generated from gut microbiota and bacterial endogenous components (LPS and PSA) pass to the circulation due to leaky gut and affect peripheral immunity and glial cells in the CNS. (B) Migration of gut-resident innate and adaptive immune cell populations to the CNS and their cytokines can modulate MS pathogenesis. (C) The gut wall is innervated with both afferent and efferent fibers of the vagus nerve. Efferent nerve fibers carry impulses from the CNS to the gut and affect its physiology. On the other hand, afferent fibers carry impulses from the gut to the CNS. The activity of these fibers can be modulated by the diffusion of bacterial components (like LPS) and metabolites (like SCFAs) or hormonal signals from specialized EECs that are capable of sensing luminal bacterial content. (D) Gut-microbiota-derived LPS and SCFAs modulate the production and release of gut hormones (CCK, ghrelin, Peptide YY, GLP-1, 5-HT) from EECs. Gut hormones potentially suppress Th17 responses and neuroinflammation by attenuating activated microglia. (E) Disease-modifying therapies (DMTs) can affect gut immune cells and epithelial cells to induce changes in gut microbiota, which may contribute toward their therapeutic effects. By promoting anti-inflammatory gut bacteria, DMTs can enhance the development of Tregs or the production of regulatory cytokines in the gut and their circulation, which can suppress CNS autoimmunity. (F) Gut microbiota may modulate the response to DMTs. Schematic diagram was created using BioRender.com (accessed on: 13 September 2023).
Figure 2. Mechanisms of gut–immune-axis-mediated effect on the CNS. (A) Microbial metabolites (SCFAs from dietary fibers and tryptophan metabolites) generated from gut microbiota and bacterial endogenous components (LPS and PSA) pass to the circulation due to leaky gut and affect peripheral immunity and glial cells in the CNS. (B) Migration of gut-resident innate and adaptive immune cell populations to the CNS and their cytokines can modulate MS pathogenesis. (C) The gut wall is innervated with both afferent and efferent fibers of the vagus nerve. Efferent nerve fibers carry impulses from the CNS to the gut and affect its physiology. On the other hand, afferent fibers carry impulses from the gut to the CNS. The activity of these fibers can be modulated by the diffusion of bacterial components (like LPS) and metabolites (like SCFAs) or hormonal signals from specialized EECs that are capable of sensing luminal bacterial content. (D) Gut-microbiota-derived LPS and SCFAs modulate the production and release of gut hormones (CCK, ghrelin, Peptide YY, GLP-1, 5-HT) from EECs. Gut hormones potentially suppress Th17 responses and neuroinflammation by attenuating activated microglia. (E) Disease-modifying therapies (DMTs) can affect gut immune cells and epithelial cells to induce changes in gut microbiota, which may contribute toward their therapeutic effects. By promoting anti-inflammatory gut bacteria, DMTs can enhance the development of Tregs or the production of regulatory cytokines in the gut and their circulation, which can suppress CNS autoimmunity. (F) Gut microbiota may modulate the response to DMTs. Schematic diagram was created using BioRender.com (accessed on: 13 September 2023).
Ijms 24 14756 g002
Table 1. Effect of various gut bacteria on immune system in MS patients.
Table 1. Effect of various gut bacteria on immune system in MS patients.
Bacterial TaxaLevel in MSFunctional EffectReference
Clostridium (Clostridia cluster XIV and IV)DecreasedDecreased regulatory T cells (Treg) and IL10 production[28]
PrevotellaDecreasedDifferentiation of Th17 cells[9]
Streptococcus mitis (S. mitis) and Streptococcus oralisIncreased
MethanobrevibacterIncreasedActivation of T cells and monocytes[18]
Akkermansia
ButyricimonasDecreased
Parabacteroides distasonisDecreasedDecreased anti-inflammatory IL-10-expressing human CD4+CD25+ T cells and IL-10+FoxP3+ Tregs in mice
Increased differentiation of Th1 type cells and reduced proportion of CD25+FoxP3+ Treg cells
[12]
Acinetobacter calcoaceticusIncreased
Flavonifractor plautiiIncreasedCorrelate positively with increased monocytes and neutrophils, and blood cell gene expression of IL17A and IL6[29]
Clostridium leptumIncreasedCorrelate positively with increased Type 1 IFN-induced blood cell genes: MX1, IFIT1, IFI44L, and IFI27
FlavonifractorIncreasedCorelate positively with increased serum TNF-α[30]
Faecalibacterium and RoseburiaDecreasedCorelate negatively with serum TNF-α
FaecalibacteriumDecreasedCorelate positively with serum IL8 and MIP-1a
Table 2. Important ongoing or completed studies addressing the effects of different diets on MS.
Table 2. Important ongoing or completed studies addressing the effects of different diets on MS.
Diet/InterventionStudy Cohort or DesignOutcomeClinical Trial/Ethical Committee Approval NumberReference
High-fiber supplement (NBT-NM108)RRMSOngoing studyNCT04574024Not applicable
Propionate (PA)RRMSSignificant increase in functionally competent Treg cells and decrease in Th1 and Th17 cells after two weeks.
Reduced annual relapse rate, disability stabilization, and reduced brain atrophy after 3 years of PA intake.
15-5351
4493-12
17-6235
357_17B
[102]
High-vegetable/low-protein diet (HV/LP dietRRMSInduction of IL10-producing monocytes and Tregs.
Increase in abundance of Lachnospiraceae family.
Decrease in relapse rate and Expanded Disability Status Scale score.
Not available[103]
Healthy/Mediterranean dietCase control retrospective dietary recall studiesLower risk of clinically isolated syndrome (CIS).
Lower risk of MS.
Not applicable[108,109]
Table 3. Effect of DMTs on gut microbiota composition in MS.
Table 3. Effect of DMTs on gut microbiota composition in MS.
Type of DMTDMTsMicrobial ChangesReference
IncreaseDecrease
InjectablesInterferons
(IFN)
Bacteroides uniformis
Prevotella genus
Sutterella genus
Ruthenibacterium lactatiformans
Akkermansia muciniphila
Sarcina genus
Prevotella copri
[10,11,16,18,131]
Glatiramer
Acetate (GA)
Prevotella genus
Sutterella genus
Bacteroides uniformis
Lachnospiraceae family
Veillonellaceae family
Akkermansia muciniphila
Sarcina genus
[10,11,18,126]
OralDimethyl fumarate (DMF)Lactobacillus pentosu
Roseburia intestinalis
Ruthenibacterium lactatiformans
Bacteroidetes phylum
Bacteroides stercoris
Clostridium species
Eubacterium species
Coprococcus eutactus
Enterococcus gilvus
Lachnospiraceae families
Veillonellaceae families
Firmicutes phyla
Fusobacteria phyla
Clostridiales order
[10,16,126,132,133]
Streptococcus, Haemophilus, Clostridium, Lachnospira, Blautia, Subdoligranulum, and Tenericutes in MS subjects with side effectsBacteroidetes, Barnesiella, Odoribacter, Akkermansia, and some Proteobacteria families in MS subjects with side effects
FingolimodRuminococcaceae PAC001607
Ruthenibacterium lactatiformans
Bacteroides finegoldii CAG:203
Roseburia faecis
Blautia species
[10,16]
InfusionOcrelizumab (Anti-CD20)Faecalibacterium prausnitzii
Ruthenibacterium lactatiformans
[16]
NatalizumabPhascolarctobacterium sp. CAG:207
Ruminococcaceae PAC001607
Ruthenibacterium lactatiformans
Bacteroides uniformis
Prevotella species
Bifidobacterium longum
Akkermansia muciniphila
[10,11,16]
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

Yadav, S.K.; Ito, K.; Dhib-Jalbut, S. Interaction of the Gut Microbiome and Immunity in Multiple Sclerosis: Impact of Diet and Immune Therapy. Int. J. Mol. Sci. 2023, 24, 14756. https://doi.org/10.3390/ijms241914756

AMA Style

Yadav SK, Ito K, Dhib-Jalbut S. Interaction of the Gut Microbiome and Immunity in Multiple Sclerosis: Impact of Diet and Immune Therapy. International Journal of Molecular Sciences. 2023; 24(19):14756. https://doi.org/10.3390/ijms241914756

Chicago/Turabian Style

Yadav, Sudhir Kumar, Kouichi Ito, and Suhayl Dhib-Jalbut. 2023. "Interaction of the Gut Microbiome and Immunity in Multiple Sclerosis: Impact of Diet and Immune Therapy" International Journal of Molecular Sciences 24, no. 19: 14756. https://doi.org/10.3390/ijms241914756

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