• Open Access
    Review

    The role of microbiome in uveitis

    Hind Amin 1
    Samir Shoughy 2,3*

    Explor Med. 2023;4:1001–1013 DOI: https://doi.org/10.37349/emed.2023.00190

    Received: July 07, 2023 Accepted: September 28, 2023 Published: December 25, 2023

    Academic Editor: Dimitra Skondra, University of Chicago, USA

    Abstract

    The gut microbiota comprises a complex bacterial community that resides in the intestine. Imbalances in the gut microbiota can disrupt immune homeostasis, triggering autoimmune diseases including non-infectious uveitis. Despite recent advances, the underlying mechanisms linking the microbiome and uveitis are not fully understood. This review offers a comprehensive analysis of the literature addressing microbiome’s relationship with ocular inflammation. Additionally, it explores the potential of modulating the gut microbiota as a novel therapeutic target. A literature search of published articles related to the role of ocular microbiome in non-infectious uveitis in PubMed and Scopus databases was conducted. The following keywords were used: microbiome, uveitis, and immune-mediate diseases.

    Keywords

    Microbiome, uveitis, autoimmune

    Introduction

    While uveitis is a diverse and complex disease, the prevailing belief is that a combination of environmental and genetic factors, coupled with immune-mediated inflammation, plays a crucial role in its onset and progression. The condition is caused by a disturbed balance between the regulatory and effector components of the immune system, a phenomenon marked by a complex interplay of pro- and anti-inflammatory factors leading to the destruction of the immune-privileged tissues, specifically those of the eye [1].

    The process of colonization by bacteria following birth plays a fundamental role in shaping the human immune system whereby the body’s surfaces become covered by innumerable bacteria by the time of adulthood. The term “microbiome” is used to refer to these wide varieties of microorganisms [2].

    The etiology of uveitis is diverse; it could be infectious, immune-mediated, iatrogenic, or undetermined etiologies. This literature review compiles existing research on the correlation between microbiota and non-infectious uveitis (NIU). There are contradictory findings suggesting that the relationship between microbiota and uveitis may be more complex than initially thought. While further research is needed to fully understand this relationship, the existing literature highlights the various mechanisms through which intestinal dysbiosis may contribute to the pathogenesis of NIU as well as the potential value of microbiome-based interventions for the prevention and treatment of uveitis.

    Methodology

    A search of published articles related to the role of microbiome in uveitis was conducted. The literature review of published articles was done using PubMed and Scopus databases. The following search terms in various combinations were used: non-infectious uveitis, gut microbiome, dysbiosis, and HLA.

    Gut microbiome

    The microbiome is a complex ecosystem that varies in population and diversity according to different factors such as age, genetics, diet, lifestyle, and environmental factors [2]. Studies have shown that disruptions in the balance of the microbiome, known as dysbiosis, can result in immune dysregulation, thereby contributing to the development of various autoimmune disorders such as inflammatory bowel disease (IBD) [3]. Furthermore, the research has established a link between the intestinal microbiota and extraintestinal disorders like multiple sclerosis (MS), fibromyalgia, arthritis, and mental disorders. However, some experts argue that the current body of evidence is insufficient to definitively establish a causal relationship between gut health and extraintestinal diseases [2]. However, studies on mice have shown that changes in the composition of gut microbiota can modulate cytokine production and T-cell activation, which can influence the development of autoimmune diseases in the eyes [4].

    The human gut microbiome comprises approximately 160 bacterial species, the majority can be classified into three groups based on their physiological functions: fermentative bacteria, bile-tolerant bacteria, and mucin-degrading bacteria [5, 6]. The fermentative bacteria produce a range of organic acids as end products of carbohydrate fermentation. These include short-chain fatty acids (SCFAs) such as butyrate, succinate, and propionate, which play a vital role in the physiology of the gastrointestinal tract and the overall health of the host [7].

    The group of butyrate-producing bacteria (BPB) is considered a significant anti-inflammatory bacterial population [8]. This group, consisting of bacterial species like Faecalibacterium, Blautia, Roseburia, Lachnospira, and Ruminococcus, has potential benefits such as modulation of immune response and production of butyrate, which stimulates the production of immunosuppressive regulatory T (Treg) cells as well as inhibits the expression of proinflammatory mediators. Additionally, it encourages mucin secretion, which strengthens and protects the gastrointestinal epithelial barrier [912].

    A reduction in butyrate production may result in a disruption of the gut microflora equilibrium, thereby causing a decline in the Treg cells and the initiation of T-effector responses. These changes may have long-term consequences for both the composition of the gut microbiota and the efficiency of the immune system [12]. Methanogens, including the Candidatus, Methanomethylophilus, and Methanoculleus bacterial species, constitute an additional category of beneficial bacteria which produce methane that has anti-inflammatory and anti-apoptotic effects on intestinal as well as retinal tissues [12].

    Pathogenic microbes with proinflammatory properties include sulfate-reducing bacteria (SRB) like Bilophila spp. and opportunistic pathogens like Parabacteroides spp., Paraprevotella spp., and Fusobacterium spp. Hydrogen sulfide (H2S), produced during sulphate reduction by SRB, inhibits butyrate metabolism in intestinal cells. Additionally, this gas promotes inflammatory response in the gut by disrupting the integrity of the intestinal barrier and thereby exposing the intestinal epithelium to bacterial products [13, 14]. These SRB have been detected in the microbiota of patients with autoimmune uveitis. Additionally, several studies have compared the bacterial profiles of individuals with uveitis to those of healthy individuals. The results revealed a shift in the microbial community structure which may suggest a disruption in the balance between protective (symbiotic) and harmful (pathogenic) microorganisms. Particularly, uveitis patients exhibited a significant reduction in protective microorganisms, coupled with an increase in gut proinflammatory bacteria. While protective (symbiotic) bacteria with anti-inflammatory effects are more common in healthy individuals [12, 15].

    The mechanism by which this disruption can contribute to the development of NIU is an area of interest to many researchers. The following section will illustrate the various mechanisms suggested in the literature.

    The mechanism underlying dysbiosis-induced uveitis

    In recent years, there has been a growing interest in studying the role of microbiota in the development of NIU [16]. Dysbiosis is involved in the development of uveitis through four complementary mechanisms, which include antigenic or molecular mimicry, increased intestinal permeability, loss of immune intestinal homeostasis, and reduced production of beneficial anti-inflammatory metabolites [17].

    Molecular mimicry

    Antigenic mimicry is a prominent mechanism that contributes to the development of autoimmune diseases. In this process, the production of autoreactive T cells is a direct consequence of the cross-reactivity that exists between self-antigens and microbial peptides. As a result, pro-inflammatory cytokines are produced, causing tissue damage and inflammation [18].

    Studies conducted on mice with experimental autoimmune uveitis (EAU) have demonstrated the role of this pathogenic mechanism since the severity of uveitis can be reduced and intestinal T helper 17 cells (Th17, effector T cells) activation can be minimized by administering oral broad-spectrum antibiotics, which eliminate the microbial community. In contrast, the transfer of T cells from a microbiota-grown transgenic mouse can induce uveitis in wild mice [4].

    Loss of intestinal immune homeostasis

    Dysbiosis results in the disruption of intestinal homeostasis, leading to an imbalance between effector T cells (Th1 and Th17) and Treg cells, which in turn triggers immune activation by upregulating Th17 [and interleukin 17 (IL-17)] and downregulating Treg (and IL-10) [19, 20]. For example, Klebsiella triggers the activation of Th (Th1) cells in the intestinal region through antigen-presenting cells (APCs), resulting in the secretion of inflammatory mediators like tumor necrosis factor alpha (TNFα) and interferon gamma (IFNγ). Bacteroides fragilis is known to generate polysaccharide A, which stimulates Treg cells. Bifidobacterium has been observed to stimulate Treg cells as well, which are known to mitigate inflammation by regulating the activity of Th1, Th2, and Th17 cells [21]. In addition, it has been observed that mice models with EAU showed the presence of T cells originating from the intestine within their ocular tissues, thereby providing evidence in favor of the migration of immune cells from the gut to the eye (gut-eye axis) [1, 22].

    Increased intestinal permeability

    Although the intestinal barrier is naturally permeable to some degree, an increase in permeability resulting from dysbiosis-induced mucosal inflammation, facilitates the translocation of microbiota or its products to the circulation [23, 24]. These products, namely lipopolysaccharides (LPS) and β-glucan can disseminate throughout various tissues via the vascular system. This can result in the direct induction of inflammation within targeted organs such as the uveal tissue. The study conducted by Janowitz et al. [25] focused on examining intestinal alterations in mice with EAU induced by immunization with inter-photoreceptor retinoid-binding protein (IRBP) along with killed Mycobacterium tuberculosis (MTB) antigen as an adjuvant. The control group consisted of mice that were immunized with MTB, which did not exhibit any ocular inflammation. The research findings indicate that there was a noticeable increase in intestinal permeability in mice that were immunized with IRBP, as compared to those that were not immunized. This increase in permeability was found to be consistent with the progression of uveitis along with alterations in the composition of the intestinal microbiota [25].

    Reduction of anti-inflammatory microbial metabolites

    Numerous metabolites produced by gut microbiota have the potential to modulate immune responses. SCFAs have been previously mentioned, and are produced via the process of dietary fiber fermentation which offers anti-inflammatory effects by stimulating and enhancing Treg cells in the intestinal lamina propria, while also inhibiting effector T cells and reducing their migration between the intestine and spleen [22, 26]. It was found that adding SCFAs to the diet from outside sources reduced the severity of uveitis in mice with EAU [22, 27].

    From an immunological perspective, it is necessary to present retinal antigens to the immune system to develop autoimmune uveitis. The following section will delve into the hypothesis of the gut-eye axis.

    The gut-eye axis

    The ocular and brain tissues have been recognized as areas of immunological privilege. The sequestration of target antigens behind the blood-retinal barrier and blood-brain barrier leads to their inaccessibility to the immune system, thereby impeding the activation of self-reactive T cells that have bypassed thymic negative selection and are present in the bloodstream [28, 29]. There is a strong correlation between IBD and ocular inflammatory conditions, as approximately 10% of patients with IBD can develop episcleritis, uveitis, and conjunctivitis [4]. Multiple studies have demonstrated the presence of a gut-eye axis, wherein gut microbiota can impact the immune response in the eye. Furthermore, as previously stated, numerous studies have highlighted the significance of the intestinal microbiome and its metabolites, specifically SCFAs, in the modulation of essential immune cell functions [30, 31]. NIU is an example of many ocular conditions that have been linked to abnormalities in the gut microbiome; others include glaucoma, diabetic retinopathy, and age-related macular degeneration [30, 31].

    The study conducted by Nakamura et al. [1] showed a significant reduction in uveitis severity in the EAU model following oral administration of specific antibiotics. However, when the antibiotics were administered intraperitoneally, no effect was observed. These findings underscore the crucial role of gut microbiota in influencing uveitis in this model. Moreover, the improvement of uveitis seems to be linked with significant changes in bacterial species caused by the administration of oral antibiotics. This leads to the differential buildup of effector (Th1 and Th17) and Treg-cell populations in different lymphoid tissues, including the intestinal lamina propria and cervical lymph nodes located near the eye. Additionally, Horai et al. [28] demonstrated in their research that the microbiota present in the gastrointestinal contents of immunized mice exhibited modifications in comparison to non-immunized mice. As well, antibiotic-treated mice in the EAU study revealed a diverse microbial composition, which conferred a protective effect against uveitis [32].

    However, in contrast to the experimental disease, most cases of human autoimmune uveitis cannot be attributed directly to an immune system exposure to ocular antigens since these antigens are typically sequestered behind a tight blood-retinal barrier in a healthy eye. This presents a paradoxical situation as retinal antigens are not typically expressed in the periphery and the activation of retina-specific T cells circulating in the periphery is a crucial step to be able to breach the blood-retinal barrier and trigger inflammation. This scenario prompts fundamental research regarding the location and manner in which autoreactive T cells, capable of recognizing retinal antigens and initiating uveitis, are initially stimulated [28].

    To study this scenario, the spontaneous uveitis model in R161H mice was used to elaborate the role of microbiota in activating the retina-specific T cells and triggering autoimmune uveitis. The administration of oral broad-spectrum antibiotics to R161H mice prior to birth led to a depletion of commensal microbiota. This resulted in a significant reduction of spontaneous uveitis, which was also observed in R161H mice raised under germ-free conditions. The development of uveitis was found to be correlated with an elevated population of Th17 cells in the intestinal lamina propria. In mice that were either treated with antibiotics or were germ-free, these Th17 cells were observed to be significantly diminished. These findings provide robust evidence in support of the hypothesis that commensal microbiota plays a significant role in the pathogenesis of spontaneous uveitis [4].

    Clinical studies

    Preclinical studies conducted on EAU models have demonstrated the involvement of the gut microbiome in the pathogenesis of various immune or inflammatory diseases, such as Behcet’s disease (BD) [33], Vogt-Koyanagi-Harada disease (VKH), rheumatoid arthritis (RA) [34], psoriatic arthritis (PsA), IBD [35], MS [36], spondylarthritis (SpA) [37], and systemic lupus erythematosus (SLE), as shown in Table 1 [38, 39]. While clinical studies are necessary to characterize dysbiosis of the gut microbiome and its role in modulating immune homeostasis, it is challenging to draw causal conclusions from these studies.

    Characterization of gut dysbiosis in different autoimmune conditions compared to healthy or disease controls

    DiseaseAuthorStudy objectStudy findings
    Increased microbiotaDecreased microbiota
    BDYasar et al., 2020 [40]27 BD vs. 10 HCsLachnospiraceae NK4A136, Actinomyces, Libanicoccus, Collinsella, Eggerthella, Enetrohabdus, Catenibacterium, EnterobacterBacteroides, Cricetibacter, Alistipes, Lachnospira, Dielma, Akkermansia, Sutterella, Anaerofilum, Ruminococcease-UCG007, Acetanaerobacterium, Copropaacter
    Shimizu et al., 2019 [41]13 BD vs. 27 HCsEggerthella lenta, Acidaminococcus bifidum, Lactobacillus iners, Streptococcus species, Lactobacillus salivariusMegamonas hypermegale, Butyrivibrio, Streptococcus infantis, Filifacto
    Oezguen et al., 2019 [42]13 BD vs. 14 HCsParabacteroides, Clostridiales, Geminger, Butyricimonas, Actinobacteria, ErysipelotrichaceaeVampirovibrio, unclassified Lachnospiraceae, Prevotella
    Ye et al., 2018 [33]32 BD vs. 74 HCsBilophila spp., Parabacteroides spp., Paraprevotella spp., Stenotrophomonas spp., Actinomyces spp., Corynebacterium spp.Clostridium spp. (BPB), Methanoculleus spp., Methanomethylophilus spp.
    VKHYe et al., 2020 [39]55 VKH vs. 52 HCsRamularia, Alternaria, RhizophagusMethanoculleus, Candidatus Methanomethylophilus, Azospirillum
    Li et al., 2022 [43]11 VKH vs. 11 HCs vs. 20 NIASStomatobaculum, Pseudomonas, LachnoanaerobaculumGordonibacter, Slackia
    ASZhang et al., 2020 [44]20 AS vs. 19 HCsPrevotellaceae, Actinomycetaceae, Dialister, Escherichia-Shigella, KlebsiellaLachnospiraceae, Bacteroides, Parasutterella, Bifidobacterium
    Yin et al., 2020 [45]127 AS vs. 123 HCsClostridiales bacterium 1_7_47FAA, Clostridium hatheway, Clostridium bolteaeBifidobacterium adolescentis, Coprococcus comes, Lachnospiraceae bacterium 5_1_63FAA, Roseburia inulinivorans
    Klingberg et al., 2019 [46]150 AS vs. 17 HCsProteobacteria, Enterobacteriaceae, Bacilli, Streptococcus species, ActinobacteriaBacteroides, Lachnospiraceae
    IBDTakahashi et al., 2016 [47]10 IBD vs. 10 HCsActinomyces, BifidobacteriumBacteroides, Eubacterium, Faecalibacterium, Ruminococcus
    Franzosa et al., 2019 [48]121 IBD vs. 34 HCsBifidobacterium breve, Clostridium symbiosum, Ruminococcus gnavusa, Escherichia coli, Clostridium clostridioformeRoseburia hominis, Dorea formicigenerans, Ruminococcus obeum
    Display full size

    HCs: healthy controls; AS: ankylosing spondylitis; NIAS: non-infectious anterior scleritis

    Numerous investigations conducted on particular disease populations, including BD and VKH disease, have established a robust correlation primarily by comparing the microbiome of patients with the disease to that of healthy individuals [33, 39].

    Clinical investigations conducted on patients with BD have revealed a modification in the microbiota composition, along with notable variations in the gut microbiome composition between BD patients with and without uveitis [33]. This implies a correlation between intestinal dysbiosis and the pathophysiology of the disease. The observed alterations primarily consisted of a rise in SRB, Stenotrophomonas species, Actinomyces species, and Paraprevotella species, accompanied by a decline in BPB and methanogens. The lack of equilibrium leads to impairment of the intestinal epithelial barrier, thereby promoting the infiltration of effector molecules into the intestinal epithelial cells [4951]. A study conducted by Emmi et al. [52] wherein patients with BD were randomly assigned to receive diets enriched with butyrate. The findings of the study indicate a decrease in disease activity and a reduction in the use of immunosuppressive medication, despite the lack of significant changes in blood inflammatory markers [52].

    The microbial composition of the intestinal microbiome in patients diagnosed with active VKH disease exhibits differences in comparison to that of healthy individuals, which is akin to the observed differences in patients with BD. Specifically, Paraprevotella spp. were enriched in active VKH patients, whereas BPB like Clostridium spp. and methanogens like Methanoculleus spp. were depleted [39, 43].

    Regarding the SpA disease spectrum, the initial evidence linking SpA and intestinal inflammation was derived from histological analysis of tissue obtained during colonoscopy as part of a research study involving patients with SpA and no apparent intestinal symptoms. The study revealed that over 50% of patients exhibited subclinical or unrecognized inflammation associated with heightened levels of intestinal permeability. Moreover, reactive arthritis and acute anterior uveitis (AAU) can also be triggered by a range of enteric infections such as Salmonella, Yersinia, Shigella, and Campylobacter. Furthermore, the efficacy of sulfasalazine in addressing arthritis associated with SpA may be ascribed to its established influence on the intestinal microbiota and its ability to reduce intestinal permeability [23].

    A comparative analysis was conducted to examine the composition of gut microbiota in patients with SpA in relation to healthy controls. The results revealed a higher level of microbial richness in SpA patients. Furthermore, an observed positive correlation has been identified between the abundance of the bacterial genus Dialister and the level of inflammatory activity in SpA. This suggests that Dialister has the potential to serve as an indicator of disease activity [53].

    The role of human leukocyte antigen

    The human leukocyte antigen (HLA), also known as the major histocompatibility complex (MHC) is responsible for antigen presentation. However, the precise mechanism through which HLA molecules confer susceptibility to diseases is frequently not understood. HLA molecules have been observed to exert an impact on the susceptibility of certain immune-mediated diseases, including ankylosing spondylitis [54], birdshot chorioretinopathy (BSRC) [55], and Crohn’s disease [56], wherein the presence of autoantibodies is not an essential aspect of the condition. This implies that in the aforementioned illnesses, the susceptibility produced by the HLA allele may not be working through an autoimmune reaction. Given that the gut microbiome plays a role in educating the immune response and exhibits a wide range of antigenic diversity, it is plausible to consider an alternative hypothesis that HLA molecules may, in certain cases, contribute to disease susceptibility through their impact on the gut microbiome [23]. To test this hypothesis that HLA molecules would influence the intestinal bacterial composition, the microbiome of HLA-B27-transgenic mice that express HLA-B27, was compared with the microbiome of controls [57, 58]. Although these studies lend weight to the idea that human MHC expression affects the composition of the gut microbiota, they cannot be held solely responsible for the onset of disease. Because of the low incidence of BSRC and SpA among individuals who express the HLA-A29 and HLA-B27 alleles, respectively. This points to the importance of additional factors, either genetic or environmental [5961].

    Therapies targeting the gut microbiome

    Due to the significant impact of gut microbiome on immunity and metabolism in uveitis, there has been a growing trend toward therapeutic interventions that target the gut microbiome in order to modify disease outcomes. At present, the primary therapeutic approaches comprise antibiotics, probiotics, prebiotics, dietary modifications, and fecal microbiota transplantation (FMT) [62].

    Antibiotics

    The use of antibiotics in the treatment of gut microbiome dysbiosis is well-established. As previously reported in the literature [4], administration of oral antibiotics demonstrated a reduction in the severity of EAU in mice. This was achieved through modulation of the gut microbiome composition, resulting in an increase in the frequency of Treg cells in the intestinal lamina propria and extraintestinal lymphoid tissues, as well as a decrease in the number of Th1 and Th17 cells, and the level of inflammatory cytokines. The oral administration of broad-spectrum antibiotics starting one week prior to the immune induction of EAU model has the potential to safeguard mice from severe uveitis. This effect is comparable to that observed in germ-free mice, with a reduction in retinal T-cell infiltration and inflammatory cytokine levels in comparison to EAU mice [1, 63]. Furthermore, the oral administration of minocycline a broad-spectrum tetracycline antibiotic has demonstrated anti-inflammatory and immunomodulatory effects. In experimental studies, it has been observed that minocycline has the potential to alter the gut microenvironment of mice with EAU. After treatment with minocycline, the relative abundances of pro-inflammatory bacteria such as Desulfovibrio, Ruminococcus bromii, Streptococcus hyointestinalis, and pathogenic Spirochaeta were significantly reduced. In addition, minocycline supplementation can increase the growth of Parabacteroides goldsteinii, which has been demonstrated to decrease IL-1β and TNFα levels and maintain intestinal permeability thereby significantly reducing the severity of EAU in mice model [64].

    Probiotics

    Probiotics are defined as live microorganisms that, when administered in sufficient quantities, can provide a health benefit to the host. The mechanism of action of probiotics is associated with their capacity to compete with pathogenic microorganisms for adhesion sites, exhibit antagonistic effects against these pathogens, or regulate the host’s immune response by enhancing Treg cell differentiation. Lactic acid bacteria are one of the most commonly used probiotic strains to improve the intestinal barrier and immune function [65, 66]. The impact of probiotics on the EAU mouse model was studied in which the experimental design involved administering antibiotics to the mice prior to providing them with IRT-5 probiotics, which consisted of a combination of mixture of five strains of lactic acid bacteria that includes Bifidobacterium bifidum, Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus reuteri, and Streptococcus thermophilus [10]. The findings indicate that administering IRT-5 probiotics can potentially modulate the clinical manifestations of ocular autoimmunity in comparison to the control group, thereby serving as a viable preventive measure against the onset of uveitis. Therefore, the administration of antibiotics can lead to an immediate reduction of the gut microbiota, which can be followed by the introduction of beneficial oral probiotics, such as IRT-5, to restore the intestinal flora [10, 17]. The achievement of optimal colonization of probiotics in the intestinal tract poses a substantial challenge that necessitates careful consideration [17].

    Prebiotic

    The prebiotics concept was introduced for the first time in 1995 by Glenn Gibson and Marcel Roberfroid [67]. Prebiotic was described as “a non-digestible food ingredient that beneficially affects the host by selectively stimulating the growth and/or activity of one or a limited number of bacteria in the colon, and thus improves host health”. Fructooligosaccharides (FOS), inulin, and galactooligosaccharides (GOS) are recognized as conventional prebiotics that have been shown to have a significant positive impact on human health as a result of their ability to enhance the presence of lactobacilli and bifidobacteria by generating advantageous metabolites, facilitating calcium absorption, mitigating protein fermentation and pathogenic bacteria, reducing intestinal permeability, and augmenting the immune system [68].

    Dietary modifications

    Various lifestyle factors, including dietary habits, socioeconomic status, physical inactivity, smoking, and environmental factors, have the potential to disrupt the equilibrium of the microbiome ecosystem [69]. Among all these factors, diet is possibly the most crucial, as it may influence both the richness and diversity of microbiota which may as a consequence impact the immune system. For example, a high-fiber diet such as unrefined cereals, fruits, vegetables, and legumes is believed to enhance the intestinal microecological environment by increasing the gut microbiome’s capacity to produce endogenous SCFAs [70]. Moreover, the consumption of plant-based protein has been reported to increase gut-commensal Bifidobacterium and Lactobacillus which increase the intestinal SCFA levels, while additionally decreasing the pathogenic Bacteroides fragilis and Clostridium perfringens. Consumption of animal-based protein was found, on the other hand, to increase the number of bile-tolerant anaerobes like Bacteroides, Alistipes, and Bilophila [71, 72]. Research studies in both animals and humans have demonstrated a correlation between high fat diets and reduced richness and diversity of the intestinal microbiota [73]. Additionally, studies have demonstrated that exogenous administration of propionate and butyrate can modulate the immune system and potentially serve as treatment strategies by reducing the severity of EAU in animal models [22, 27].

    FMT

    FMT, namely stool transplantation, involves the replacement of a patient’s original gut microbiome with the entire fecal microbiome of a healthy donor. This procedure aims to modify the microorganism composition within the gut. The utilization of FMT is currently being studied for a diverse range of medical conditions. It has received authorization for the treatment of recurrent colitis induced by Clostridium difficile and has demonstrated significant effectiveness in this respect [74]. EAU mouse models developed more severe uveitis after receiving FMT from BD or VKH patients, accompanied by increased IL-17 and IFNγ production. However, there is still not enough clinical evidence to support its use in other conditions like IBD, ankylosing spondylitis, or uveitis because of its substantial interindividual variability [75].

    Conclusions

    The current literature supports the possible causality between intestinal dysbiosis and immune mediated uveitis. There is still much to learn about the microbiome, and many questions remain unanswered, such as how the microbiome is established in early life, how it is affected by environmental factors, and how it can be manipulated to promote health and prevent disease. The complexity of the microbiome presents challenges in studying and understanding its role in human health and disease, as well as developing effective treatments or interventions. The gut-eye axis, which has been proposed as a potential explanation for the microbiome effect on ocular inflammation and diseases, is not yet well-understood and requires further research. By exploring this area, researchers can expand the current knowledge base and potentially uncover new therapeutic targets for these conditions. In the future, the ability to modulate the composition of the intestinal microbiome through dietary supplementation or the use of certain medications may pave the road for new therapeutic modalities.

    Abbreviations

    BD:

    Behcet’s disease

    BPB:

    butyrate-producing bacteria

    EAU:

    experimental autoimmune uveitis

    FMT:

    fecal microbiota transplantation

    HLA:

    human leukocyte antigen

    IBD:

    inflammatory bowel disease

    IL:

    interleukin

    IRT-5:

    probiotics consisted of a combination of mixture of five strains of lactic acid bacteria

    NIU:

    non-infectious uveitis

    SCFA:

    short-chain fatty acid

    SpA:

    spondylarthritis

    SRB:

    sulfate-reducing bacteria

    Th:

    T helper

    Treg:

    regulatory T

    VKH:

    Vogt-Koyanagi-Harada disease

    Declarations

    Author contributions

    HA and SS equally contributed to: Conceptualization, Writing—original draft, Writing—review & editing.

    Conflicts of interest

    The authors declare that they have no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2023.

    References

    Nakamura YK, Metea C, Karstens L, Asquith M, Gruner H, Moscibrocki C, et al. Gut microbial alterations associated with protection from autoimmune uveitis. Invest Ophthalmol Vis Sci. 2016;57:374758. [DOI] [PubMed] [PMC]
    Shivaji S. We are not alone: a case for the human microbiome in extra intestinal diseases. Gut Pathog. 2017;9:13. [DOI] [PubMed] [PMC]
    Wendling D, Guillot X, Prati C, Miceli-Richard C, Molto A, Lories R, et al. Effect of gut involvement in patients with high probability of early spondyloarthritis: data from the DESIR cohort. J Rheumatol. 2020;47:34953. [DOI] [PubMed]
    Horai R, Zárate-Bladés CR, Dillenburg-Pilla P, Chen J, Kielczewski JL, Silver PB, et al. Microbiota-dependent activation of an autoreactive T cell receptor provokes autoimmunity in an immunologically privileged site. Immunity. 2015;43:34353. [DOI] [PubMed] [PMC]
    McLean MH, Dieguez D Jr, Miller LM, Young HA. Does the microbiota play a role in the pathogenesis of autoimmune diseases? Gut. 2015;64:33241. [DOI] [PubMed] [PMC]
    Glover JS, Ticer TD, Engevik MA. Characterizing the mucin-degrading capacity of the human gut microbiota. Sci Rep. 2022;12:8456. [DOI] [PubMed] [PMC]
    Hooper LV, Gordon JI. Commensal host-bacterial relationships in the gut. Science. 2001;292:11158. [DOI] [PubMed]
    Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504:4515. [DOI] [PubMed] [PMC]
    Wu J, Wang R, Ye Z, Sun X, Chen Z, Xia F, et al. Protective effects of methane-rich saline on diabetic retinopathy via anti-inflammation in a streptozotocin-induced diabetic rat model. Biochem Biophys Res Commun. 2015;466:15561. [DOI] [PubMed]
    Kim J, Choi SH, Kim YJ, Jeong HJ, Ryu JS, Lee HJ, et al. Clinical effect of IRT-5 probiotics on immune modulation of autoimmunity or alloimmunity in the eye. Nutrients. 2017;9:1166. [DOI] [PubMed] [PMC]
    Richards JL, Yap YA, McLeod KH, Mackay CR, Mariño E. Dietary metabolites and the gut microbiota: an alternative approach to control inflammatory and autoimmune diseases. Clin Transl Immunology. 2016;5:e82. [DOI] [PubMed] [PMC]
    Kalyana Chakravarthy S, Jayasudha R, Sai Prashanthi G, Ali MH, Sharma S, Tyagi M, et al. Dysbiosis in the gut bacterial microbiome of patients with uveitis, an inflammatory disease of the eye. Indian J Microbiol. 2018;58:45769. [DOI] [PubMed] [PMC]
    Ijssennagger N, van der Meer R, van Mil SWC. Sulfide as a mucus barrier-breaker in inflammatory bowel disease? Trends Mol Med. 2016;22:1909. [DOI] [PubMed]
    Ijssennagger N, Belzer C, Hooiveld GJ, Dekker J, van Mil SWC, Müller M, et al. Gut microbiota facilitates dietary heme-induced epithelial hyperproliferation by opening the mucus barrier in colon. Proc Natl Acad Sci U S A. 2015;112:1003843. [DOI] [PubMed] [PMC]
    de Paiva CS, Jones DB, Stern ME, Bian F, Moore QL, Corbiere S, et al. Altered mucosal microbiome diversity and disease severity in Sjögren syndrome. Sci Rep. 2016;6:23561. [DOI] [PubMed] [PMC]
    Nishida A, Inoue R, Inatomi O, Bamba S, Naito Y, Andoh A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin J Gastroenterol. 2018;11:110. [DOI] [PubMed]
    Fu X, Chen Y, Chen D. The role of gut microbiome in autoimmune uveitis. Ophthalmic Res. 2021;64:16877. [DOI] [PubMed]
    Rojas M, Restrepo-Jiménez P, Monsalve DM, Pacheco Y, Acosta-Ampudia Y, Ramírez-Santana C, et al. Molecular mimicry and autoimmunity. J Autoimmun. 2018;95:10023. [DOI] [PubMed] [PMC]
    Zhuang Z, Wang Y, Zhu G, Gu Y, Mao L, Hong M, et al. Imbalance of Th17/Treg cells in pathogenesis of patients with human leukocyte antigen B27 associated acute anterior uveitis. Sci Rep. 2017;7:40414. [DOI] [PubMed] [PMC]
    Asquith MJ, Stauffer P, Davin S, Mitchell C, Lin P, Rosenbaum JT. Perturbed mucosal immunity and dysbiosis accompany clinical disease in a rat model of spondyloarthritis. Arthritis Rheumatol. 2016;68:215162. [DOI] [PubMed] [PMC]
    Shim JA, Ryu JH, Jo Y, Hong C. The role of gut microbiota in T cell immunity and immune mediated disorders. Int J Biol Sci. 2023;19:117891. [DOI] [PubMed] [PMC]
    Nakamura YK, Janowitz C, Metea C, Asquith M, Karstens L, Rosenbaum JT, et al. Short chain fatty acids ameliorate immune-mediated uveitis partially by altering migration of lymphocytes from the intestine. Sci Rep. 2017;7:11745. [DOI] [PubMed] [PMC]
    Rosenbaum JT, Asquith M. The microbiome and HLA-B27-associated acute anterior uveitis. Nat Rev Rheumatol. 2018;14:70413. [DOI] [PubMed] [PMC]
    Ciccia F, Guggino G, Rizzo A, Alessandro R, Luchetti MM, Milling S, et al. Dysbiosis and zonulin upregulation alter gut epithelial and vascular barriers in patients with ankylosing spondylitis. Ann Rheum Dis. 2017;76:112332. [DOI] [PubMed] [PMC]
    Janowitz C, Nakamura YK, Metea C, Gligor A, Yu W, Karstens L, et al. Disruption of intestinal homeostasis and intestinal microbiota during experimental autoimmune uveitis. Invest Ophthalmol Vis Sci. 2019;60:4209. [DOI] [PubMed] [PMC]
    Asquith M, Davin S, Stauffer P, Michell C, Janowitz C, Lin P, et al. Intestinal metabolites are profoundly altered in the context of HLA-B27 expression and functionally modulate disease in a rat model of spondyloarthritis. Arthritis Rheumatol. 2017;69:198495. [DOI] [PubMed] [PMC]
    Chen X, Su W, Wan T, Yu J, Zhu W, Tang F, et al. Sodium butyrate regulates Th17/Treg cell balance to ameliorate uveitis via the Nrf2/HO-1 pathway. Biochem Pharmacol. 2017;142:1119. [DOI] [PubMed]
    Horai R, Caspi RR. Microbiome and autoimmune uveitis. Front Immunol. 2019;10:232. [DOI] [PubMed] [PMC]
    Caspi RR. A look at autoimmunity and inflammation in the eye. J Clin Invest. 2010;120:307383. [DOI] [PubMed] [PMC]
    Fernandes R, Viana SD, Nunes S, Reis F. Diabetic gut microbiota dysbiosis as an inflammaging and immunosenescence condition that fosters progression of retinopathy and nephropathy. Biochim Biophys Acta Mol Basis Dis. 2019;1865:187697. [DOI] [PubMed]
    Cavuoto KM, Banerjee S, Galor A. Relationship between the microbiome and ocular health. Ocul Surf. 2019;17:38492. [DOI] [PubMed]
    Horai R, Silver PB, Chen J, Agarwal RK, Chong WP, Jittayasothorn Y, et al. Breakdown of immune privilege and spontaneous autoimmunity in mice expressing a transgenic T cell receptor specific for a retinal autoantigen. J Autoimmun. 2013;44:2133. [DOI] [PubMed] [PMC]
    Ye Z, Zhang N, Wu C, Zhang X, Wang Q, Huang X, et al. A metagenomic study of the gut microbiome in Behcet’s disease. Microbiome. 2018;6:135. [DOI] [PubMed] [PMC]
    Scher JU, Sczesnak A, Longman RS, Segata N, Ubeda C, Bielski C, et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife. 2013;2:e01202. [DOI] [PubMed] [PMC]
    Naftali T, Reshef L, Kovacs A, Porat R, Amir I, Konikoff FM, et al. Distinct microbiotas are associated with ileum-restricted and colon-involving Crohn’s disease. Inflamm Bowel Dis. 2016;22:293302. [DOI] [PubMed]
    Cantarel BL, Waubant E, Chehoud C, Kuczynski J, DeSantis TZ, Warrington J, et al. Gut microbiota in multiple sclerosis: possible influence of immunomodulators. J Investig Med. 2015;63:72934. [DOI] [PubMed] [PMC]
    Costello ME, Ciccia F, Willner D, Warrington N, Robinson PC, Gardiner B, et al. Brief report: intestinal dysbiosis in ankylosing spondylitis. Arthritis Rheumatol. 2015;67:68691. [DOI] [PubMed]
    López P, Sánchez B, Margolles A, Suárez A. Intestinal dysbiosis in systemic lupus erythematosus: cause or consequence? Curr Opin Rheumatol. 2016;28:51522. [DOI] [PubMed]
    Ye Z, Wu C, Zhang N, Du L, Cao Q, Huang X, et al. Altered gut microbiome composition in patients with Vogt-Koyanagi-Harada disease. Gut Microbes. 2020;11:53955. [DOI] [PubMed] [PMC]
    Yasar Bilge NS, Pérez Brocal V, Kasifoglu T, Bilge U, Kasifoglu N, Moya A, et al. Intestinal microbiota composition of patients with Behçet’s disease: differences between eye, mucocutaneous and vascular involvement. The Rheuma-BIOTA study. Clin Exp Rheumatol. 2020;38:608. [PubMed]
    Shimizu J, Kubota T, Takada E, Takai K, Fujiwara N, Arimitsu N, et al. Relative abundance of Megamonas hypermegale and Butyrivibrio species decreased in the intestine and its possible association with the T cell aberration by metabolite alteration in patients with Behcet’s disease (210 characters). Clin Rheumatol. 2019;38:143745. [DOI] [PubMed]
    Oezguen N, Yalcinkaya N, Kücükali CI, Dahdouli M, Hollister EB, Luna RA, et al. Microbiota stratification identifies disease-specific alterations in neuro-Behçet’s disease and multiple sclerosis. Clin Exp Rheumatol. 2019;37:5866. [PubMed]
    Li M, Yang L, Cao J, Liu T, Liu X. Enriched and decreased intestinal microbes in active VKH patients. Invest Ophthalmol Vis Sci. 2022;63:21. [DOI] [PubMed] [PMC]
    Zhang F, Ma C, Zhang B. Dynamic variations in gut microbiota in ankylosing spondylitis patients treated with anti-TNF-α for six months. Ann Clin Lab Sci. 2020;50:99106. [PubMed]
    Yin J, Sternes PR, Wang M, Song J, Morrison M, Li T, et al. Shotgun metagenomics reveals an enrichment of potentially cross-reactive bacterial epitopes in ankylosing spondylitis patients, as well as the effects of TNFi therapy upon microbiome composition. Ann Rheum Dis. 2020;79:13240. [DOI] [PubMed]
    Klingberg E, Magnusson MK, Strid H, Deminger A, Ståhl A, Sundin J, et al. A distinct gut microbiota composition in patients with ankylosing spondylitis is associated with increased levels of fecal calprotectin. Arthritis Res Ther. 2019;21:248. [DOI] [PubMed] [PMC]
    Takahashi K, Nishida A, Fujimoto T, Fujii M, Shioya M, Imaeda H, et al. Reduced abundance of butyrate-producing bacteria species in the fecal microbial community in Crohn’s disease. Digestion. 2016;93:5965. [DOI] [PubMed]
    Franzosa EA, Sirota-Madi A, Avila-Pacheco J, Fornelos N, Haiser HJ, Reinker S, et al. Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nat Microbiol. 2019;4:293305. [DOI] [PubMed] [PMC]
    Wang Q, Yi S, Su G, Du Z, Pan S, Huang X, et al. Changes in the gut microbiome contribute to the development of Behcet’s disease via adjuvant effects. Front Cell Dev Biol. 2021;9:716760. [DOI] [PubMed] [PMC]
    van der Houwen TB, van Laar JAM, Kappen JH, van Hagen PM, de Zoete MR, van Muijlwijk GH, et al. Behçet’s disease under microbiotic surveillance? A combined analysis of two cohorts of Behçet’s disease patients. Front Immunol. 2020;11:1192. [DOI] [PubMed] [PMC]
    Islam SMS, Ryu HM, Sayeed HM, Byun HO, Jung JY, Kim HA, et al. Eubacterium rectale attenuates HSV-1 induced systemic inflammation in mice by inhibiting CD83. Front Immunol. 2021;12:712312. [DOI] [PubMed] [PMC]
    Emmi G, Bettiol A, Niccolai E, Ramazzotti M, Amedei A, Pagliai G, et al. Butyrate-rich diets improve redox status and fibrin lysis in Behçet’s syndrome. Circ Res. 2021;128:27880. [DOI] [PubMed]
    Tito RY, Cypers H, Joossens M, Varkas G, Van Praet L, Glorieus E, et al. Brief report: dialister as a microbial marker of disease activity in spondyloarthritis. Arthritis Rheumatol. 2017;69:11421. [DOI] [PubMed]
    Asquith M, Sternes PR, Costello ME, Karstens L, Diamond S, Martin TM, et al. HLA alleles associated with risk of ankylosing spondylitis and rheumatoid arthritis influence the gut microbiome. Arthritis Rheumatol. 2019;71:164250. [DOI] [PubMed]
    Brézin AP, Monnet D, Cohen JH, Levinson RD. HLA-A29 and birdshot chorioretinopathy. Ocul Immunol Inflamm. 2011;19:397400. [DOI] [PubMed]
    Ellinghaus D, Bethune J, Petersen BS, Franke A. The genetics of Crohn’s disease and ulcerative colitis – status quo and beyond. Scand J Gastroenterol. 2015;50:1323. [DOI] [PubMed]
    Lin P, Bach M, Asquith M, Lee AY, Akileswaran L, Stauffer P, et al. HLA-B27 and human β2-microglobulin affect the gut microbiota of transgenic rats. PLoS One. 2014;9:e105684. [DOI] [PubMed] [PMC]
    Gill T, Asquith M, Brooks SR, Rosenbaum JT, Colbert RA. Effects of HLA-B27 on gut microbiota in experimental spondyloarthritis implicate an ecological model of dysbiosis. Arthritis Rheumatol. 2018;70:55565. [DOI] [PubMed] [PMC]
    Zamecki KJ, Jabs DA. HLA typing in uveitis: use and misuse. Am J Ophthalmol. 2010;149:18993.e2. [DOI] [PubMed]
    Kuiper JJ, Van Setten J, Ripke S, Van ‘T Slot R, Mulder F, Missotten T, et al. A genome-wide association study identifies a functional ERAP2 haplotype associated with birdshot chorioretinopathy. Hum Mol Genet. 2014;23:60817. [DOI] [PubMed] [PMC]
    Kuiper JJW, Setten JV, Devall M, Cretu-Stancu M, Hiddingh S, Ophoff RA, et al. Functionally distinct ERAP1 and ERAP2 are a hallmark of HLA-A29-(Birdshot) uveitis. Hum Mol Genet. 2018;27:433343. [DOI] [PubMed] [PMC]
    Cheng YW, Phelps E, Ganapini V, Khan N, Ouyang F, Xu H, et al. Fecal microbiota transplantation for the treatment of recurrent and severe Clostridium difficile infection in solid organ transplant recipients: a multicenter experience. Am J Transplant. 2019;19:50111. [DOI] [PubMed] [PMC]
    Heissigerova J, Seidler Stangova P, Klimova A, Svozilkova P, Hrncir T, Stepankova R, et al. The microbiota determines susceptibility to experimental autoimmune uveoretinitis. J Immunol Res. 2016;2016:5065703. [DOI] [PubMed] [PMC]
    Zhou J, Yang J, Dai M, Lin D, Zhang R, Liu H, et al. A combination of inhibiting microglia activity and remodeling gut microenvironment suppresses the development and progression of experimental autoimmune uveitis. Biochem Pharmacol. 2020;180:114108. [DOI] [PubMed]
    Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol. 2014;11:50614. [DOI] [PubMed]
    Tsai YL, Lin TL, Chang CJ, Wu TR, Lai WF, Lu CC, et al. Probiotics, prebiotics and amelioration of diseases. J Biomed Sci. 2019;26:3. [DOI] [PubMed] [PMC]
    Gibson GR, Roberfroid MB. Dietary modulation of the human colonic microbiota: introducing the concept of prebiotics. J Nutr. 1995;125:140112. [DOI] [PubMed]
    Gibson GR, Hutkins R, Sanders ME, Prescott SL, Reimer RA, Salminen SJ, et al. Expert consensus document: the International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat Rev Gastroenterol Hepatol. 2017;14:491502. [DOI] [PubMed]
    Martinez JE, Kahana DD, Ghuman S, Wilson HP, Wilson J, Kim SCJ, et al. Unhealthy lifestyle and gut dysbiosis: a better understanding of the effects of poor diet and nicotine on the intestinal microbiome. Front Endocrinol (Lausanne). 2021;12:667066. [DOI] [PubMed] [PMC]
    Zmora N, Suez J, Elinav E. You are what you eat: diet, health and the gut microbiota. Nat Rev Gastroenterol Hepatol. 2019;16:3556. [DOI] [PubMed]
    Świątecka D, Narbad A, Ridgway KP, Kostyra H. The study on the impact of glycated pea proteins on human intestinal bacteria. Int J Food Microbiol. 2011;145:26772. [DOI] [PubMed]
    Cotillard A, Kennedy SP, Kong LC, Prifti E, Pons N, Le Chatelier E, et al. Dietary intervention impact on gut microbial gene richness. Nature. 2013;500:5858. [DOI] [PubMed]
    Agus A, Denizot J, Thévenot J, Martinez-Medina M, Massier S, Sauvanet P, et al. Western diet induces a shift in microbiota composition enhancing susceptibility to Adherent-Invasive E. coli infection and intestinal inflammation. Sci Rep. 2016;6:19032. [DOI] [PubMed] [PMC]
    van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, Zoetendal EG, de Vos WM, et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med. 2013;368:40715. [DOI] [PubMed]
    Rodríguez-Fernández CA, Iglesias MB, de Domingo B, Conde-Pérez K, Vallejo JA, Rodríguez-Martínez L, et al. Microbiome in immune-mediated uveitis. Int J Mol Sci. 2022;23:7020. [DOI] [PubMed] [PMC]