Next Article in Journal
Associations of the CYP7A1 Gene Polymorphisms Located in the Promoter and Enhancer Regions with the Risk of Acute Coronary Syndrome, Plasma Cholesterol, and the Incidence of Diabetes
Previous Article in Journal
Odontogenic Sarcomas of the Mandible
Previous Article in Special Issue
First Report of Rubber Collection Bowls & Plastic and Bamboo Water Containers as the Major Breeding Source of Ae. albopictus with the Indigenous Transmission of Dengue and Chikungunya in Rural Forested Malaria-Endemic Villages of Dhalai District, Tripura, India: The Importance of Molecular Identification
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Impact of Microbiota–Immunity–Hormone Interactions on Autoimmune Diseases and Infection

by
Serena Martinelli
1,
Giulia Nannini
1,
Fabio Cianchi
1,
Francesco Coratti
1 and
Amedeo Amedei
1,2,3,*
1
Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy
2
SOD of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), 50134 Florence, Italy
3
Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 50139 Florence, Italy
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(3), 616; https://doi.org/10.3390/biomedicines12030616
Submission received: 31 December 2023 / Revised: 21 February 2024 / Accepted: 25 February 2024 / Published: 8 March 2024
(This article belongs to the Special Issue Molecular Research in Infectious Diseases)

Abstract

:
Autoimmune diseases are complex multifactorial disorders, and a mixture of genetic and environmental factors play a role in their onset. In recent years, the microbiota has gained attention as it helps to maintain host health and immune homeostasis and is a relevant player in the interaction between our body and the outside world. Alterations (dysbiosis) in its composition or function have been linked to different pathologies, including autoimmune diseases. Among the different microbiota functions, there is the activation/modulation of immune cells that can protect against infections. However, if dysbiosis occurs, it can compromise the host’s ability to protect against pathogens, contributing to the development and progression of autoimmune diseases. In some cases, infections can trigger autoimmune diseases by several mechanisms, including the alteration of gut permeability and the activation of innate immune cells to produce pro-inflammatory cytokines that recruit autoreactive T and B cells. In this complex scenario, we cannot neglect critical hormones’ roles in regulating immune responses. Different hormones, especially estrogens, have been shown to influence the development and progression of autoimmune diseases by modulating the activity and function of the immune system in different ways. In this review, we summarized the main mechanisms of connection between infections, microbiota, immunity, and hormones in autoimmune diseases’ onset and progression given the influence of some infections and hormone levels on their pathogenesis. In detail, we focused on rheumatoid arthritis, multiple sclerosis, and systemic lupus erythematosus.

1. Introduction

The majority of interactions between the immune system and the external environment occur within the gastrointestinal (GI) tract, primarily affecting the community of resident microorganisms known as the intestinal microbiota [1]. These microorganisms present a significant source of antigenic diversity, which the host immune system must carefully manage its responses to. The preservation of tolerance and anti-inflammatory responses requires the engagement of a large range of innate and adaptive immune pathways that work together to control microbiota shaping and reduce systemic inflammation [2].
Additionally, the microbiota plays crucial roles in signaling the correct development, education, and epigenetic capabilities of various immune cells [3,4]. This mutual relationship has evolved over thousands of years. However, the rapid modernization of human communities has led to significant changes in environmental exposures and microbiota composition, leading to an increase in autoimmune diseases [4,5]. Autoimmune diseases require a combination of uncontrolled inflammation and self-antigen-specific T cells. Three essential conditions must be met for T cell-mediated autoimmune disorders to manifest: (a) self-reactive T cells must be present and be activated; (b) these T cells must proliferate; and (d) immune regulation must fail to regulate autoreactive responses. Complementarily, hormones, especially estrogens, not only modulate the reproductive system but also regulate immunity development and function. Innate, adaptive, humoral, and cell-mediated immune responses are impacted by hormones, and dysregulation of these mechanisms can contribute to immune-mediated disorders, including autoimmunity [6,7,8].
The aim of our paper is to decipher the complexities of how the microbiota, hormones, and the immune system interact, aiming to assess their collective impact on the onset of autoimmune diseases.

2. Microbiota–Immune System Interactions

As previously mentioned, the microbiota is essential for the proper maturation of the host immune system from the earliest stages of life [9]. The immune system has to develop to defend against pathogens while simultaneously tolerating the beneficial microorganisms that coexist symbiotically with the host [10]. Moreover, the microbiota in the large intestine plays a significant role in preserving mucosal and systemic homeostasis. The interaction between the large intestine microbiota and local immune cells is crucial for directing specific immune responses and, consequently, for performing immunomodulatory functions [11]. Notably, the interactions between GM and the immune system established in the first year of life can exert long-term effects on immune responses [12]. This, in turn, may play a role in determining the host’s susceptibility to infections and immune-related disorders later in life [13,14]. In addition, throughout life, GM affects immune functions, often with systemic outcomes that can be independent of the GM colonization site. The GM influences multiple aspects of innate and adaptive immunity. Activation of recognition receptors (PRRs), such as nucleotide-binding oligomerization domain-like receptors (NODs) and Toll-like receptors (TLRs), through commensal bacteria, enhances enterocyte regeneration and survival [15]. The commensal bacterium Bacteroides fragilis (B. fragilis) produces polysaccharide A (PSA) that recognizes the TLR2/TLR1 heterodimer, inducing the expression of anti-inflammatory genes through cyclic adenosine monophosphate (cAMP)-response element-binding protein (CREB) [16].
In addition, GM can prevent intestinal inflammation by controlling the differentiation of T regulatory (Treg) cells [17]. Metabolites produced by GM, such as short-chain fatty acids (SCFAs) and trimethylamine N-oxide (TMAO), can influence innate and adaptive immune cells in several ways, while the butyrate, through enhancing histone H3 acetylation, induces monocyte-to-macrophage differentiation [18] and TMAO can drive their polarization [19]. Moreover, these molecules reinforce antimicrobial defenses and induce the differentiation of naïve CD4+ into Treg cells [20].
Myeloid differentiation primary response protein (MyD88) serves as an adaptor for various innate immune receptors that detect microbial signals and mediate signaling pathways activated by IL-1 and IL-18 through their respective receptors [3]. Mice lacking MyD88 show a modified microbial composition [21] and MyD88 plays a crucial role in controlling the epithelial expression of several antimicrobial peptides (AMPs), including RegIIIγ. This regulation limits the presence of surface-associated Gram+ bacteria and constrains the activation of adaptive immunity [22]. Additionally, MyD88 influences T cell differentiation, supports microbiota homeostasis by promoting immunoglobulin A (IgA) stimulation, and regulates the differentiation of Th17 cells by inhibiting the growth of segmented filamentous bacteria (SFB) in mice [23].
Of note, GM can also modulate the T helper 17 (Th17) cells; indeed, Citrobacter can promote their pro-inflammatory capabilities [24]. Fung et al., show that commensal bacteria residing in lymphoid tissues (LRC) colonized germ-free and antibiotic-treated mice and influenced the cytokines’ production of dendritic cells. This colonization led to the induction of various members of the IL-10 cytokine family, such as dendritic cell-derived IL-10 and group 3 innate lymphoid cell (ILC3)-derived IL-22. As previously reported, IL-10 played a crucial role in limiting pro-inflammatory Th17 cell responses, and IL-22 production contributed to enhanced LRC colonization under steady-state conditions. Those results highlight the straight crosstalk between the host and commensal bacteria [25].
GF colonized by human GM exhibited decreased levels of CD4+ and CD8+ T cells, limited proliferation of T cells, low number of dendritic cells, and decreased expression of antimicrobial peptides in the intestinal tract. Conversely, when GF mice were colonized with SFB derived from mice, the Th17 cell number was restored to levels comparable to those observed in conventionally reared mice (CONV-R mice). These data suggest that specific mice GM may be essential for achieving complete immune maturation in these animals [26].
In addition, gut colonization by SFB elicits IL-17A production by RORγt+ Th17. SFB flagellins stimulate the production of more cytokines, such as IL17, IL21, and IL22, and drive immune endothelial cells (IECs) to secrete serum amyloid (SAA3). These cytokines lately can promote Th17 cell production [27]. Th17 lymphocytes have functional plasticity in response to inflammatory signals; indeed, the presence of high amounts of IL-12 enables them to differentiate in Th17/Th1, while IL-1 and IL-6 can stimulate a Treg-Th17 trans-differentiation [28,29]. These lymphocytes are more pathogenic compared to cells that did not undergo these shifts and can assume a pathogenic role, especially in chronic inflammatory conditions, where inflammation is frequently started by unidentified agents and the immune system lacks the ability to suppress the response [30,31]. On the other hand, Treg cells have a suppressive role (mainly secreting the anti-inflammatory cytokine IL-10). Indeed, they recognize commensal-derived antigens [32], maintain tolerance to intestinal microbes [33], and are essential for suppressing the aberrant activation of myeloid cells and Th17 cells [34]. Clostridium species are able to restore the Treg cells’ colonization in germ-free mice through the SCFAs involvement [20,35,36]. Finally, the Lactobacillus reuteri, Lactobacillus murinus, Helicobacter hepaticus, and B. fragilis increase the proportion of IL-10-producing Treg cells in mice [17,37]. In other words, the GM composition plays a relevant role in maintaining the proper balance and regulation of T cell subtypes, which is crucial in determining a person’s health status.

3. Link between Autoimmunity and Infectious Diseases

Autoimmune diseases (ADs) are a group of chronic and clinically heterogeneous pathologies that affect approximately 5% of the world’s population [38] with a constant increase in westernized societies [39]. Although the understanding of several autoimmune diseases’ pathogenesis still faces open questions, it is usually considered as a result of a mix of genetic and environmental factors. In eubiosis, the gut microbiota (GM) can protect the body against infections through competitive exclusion by contending with pathogenic microorganisms for resources, such as nutrients and space, and so preventing their colonization and growth. In addition, GM secrete antimicrobial compounds, such as bacteriocins and organic acids, that can inhibit the growth and survival of pathogenic microorganisms [40]. However, alterations in GM composition and/or function, such as dysbiosis, can compromise the host’s ability to protect against infections, contributing to the development of infectious diseases [41].
It has been proposed that GM dysbiosis may promote disease onset through infectious pathogens. For example, GM dysbiosis has been linked to various infections, such as Clostridium difficile, Salmonella, and Shigella infections [42]. The GM’s effects on the systemic immune response are mediated by the circulation of microbiota-derived soluble factors from the gut to the periphery [41]. Indeed, GM produce specific molecules (like dsRNAs and peptidoglicans) that can induce the production of cytokines such as interleukin 1 and 6 (IL-1 and IL-6) through the activation of TLRs, promoting the recruitment and activation of immune cells.
These cytokines, especially IL-6, can influence inflammation and regulate adaptive immunity through the induction of Th17 and B cell differentiation [43,44]. Round et al., showed that the PSA of B. fragilis can activate Treg cells directly through TLR2 [45].
Moreover, microbiota and its metabolites can induce epigenetic changes. The SCFAs, for example, can inhibit histone deacetylases and stimulate Treg cell differentiation [46]. Notably, the SCFAs play an important role in maintaining a strong gut barrier and in preserving host homeostasis by enhancing the regeneration of epithelial cells, as well as the production of mucus and antimicrobial peptides, preventing infections [47]. Moreover, the SCFAs induce gene expression for B cell differentiation and provide building blocks and energy for antibody production [48].
Anyway, “molecular mimicry” is the leading mechanism through which infectious or chemical substances can trigger autoimmune responses occurring when similarities between foreign and self-peptides lead to the activation of autoreactive T or B cells in susceptible individuals [49,50].
In 1964, Damian used, for the first time, the term “molecular mimicry” to indicate the existence of antigens expressed by infectious agents that were similar to molecules of human hosts that could help microbes avoid the host’s immune response [51]. Two years later, Zabriskie and Freimer observed the similarity between the membrane of Streptococcus pyogenes and mammalian muscle [52]. Since their discovery, several pathogens have been documented to carry structurally similar antigens to self-antigens, which activate B and T cells and lead to a crossreactive response against both self- and non-self-antigens [53,54,55].
Finally, more recently, we demonstrated that Helicobacter pylori (H. pylori)-infected patients with gastric autoimmunity have gastric CD4+ T cells that recognize both H+, K+-adenosine triphosphatase, and H. pylori antigens. In addition, we characterized the submolecular specificity of these T cells, identifying crossreactive epitopes from nine H. pylori proteins. These peptides were able to induce T cell proliferation and expression of Th- functions [56].
Another mechanism that triggers autoimmunity is the “bystander activation”: a nonspecific and hyperactive antiviral immune response that can create a localized pro-inflammatory environment together with the release of the damaged tissue of self-antigens that can be presented by antigen-presenting cells (APCs) to trigger T cells into an autoreactive state [57,58].
A third way to trigger autoimmunity is “epitope spreading”, where a viral infection can induce the release of new self-antigens that are presented by APCs and activate T cells [59].
In Figure 1, we summarize these three mechanisms of autoimmunity induction, but the infections can also stimulate the secretion of crossreactive antibodies that recognize both the pathogen and the host’s own tissues, leading to tissue damage and inflammation [60].

4. The Role of Specific Microorganisms in Some Autoimmune Diseases

As previously mentioned, autoimmune diseases are thought to arise as the result of acquired environmental risk in a genetically susceptible population. Understanding the interaction of environmental factors and genotype is crucial for the development of targeted preventive strategies. These factors can impact the immune system, leading to the aberrant development of plasma cells, the development of autoreactive T cells, and the abnormal production of pro-inflammatory cytokines. The increasing incidence of autoimmune diseases is thought to be a result of substantial GM alterations, influenced by various factors such as dietary shifts and the widespread use of antibiotics.
Among the environmental risks, viruses are the microbial agents that have received the greatest attention for triggering or exacerbating autoimmune diseases.
In detail, Epstein–Barr virus (EBV) has emerged as the virus with the strongest, most consistent, and most biologically plausible association with autoimmunity [61,62]. EBV is a ubiquitous human virus that infects 95% of humans during their lifetime and, after the acute phase, persists for the individual’s whole life. In the latent phase, EBV is prevented from reactivation through efficient cytotoxic cellular immunity, but it can reactivate (lytic phase) under psychological stress conditions, resulting in weakened cellular immunity. EBV chronic activation is a critical mechanism in the pathogenesis of many diseases including autoimmune disorders.
EBV was found to be associated with several autoimmune diseases such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and multiple sclerosis (MS) [63].
However, as previously reported, in addition to infections, GM, as a relevant modulator of immunity and brain function, has emerged as a likely environmental factor contributing to autoimmune diseases. Several mechanisms have been proposed to explain the link between microbiota and autoimmunity; the first of these is the modulation of gut barrier function through the production of various metabolites and signaling molecules, such as lipopolysaccharides (LPSs), SCFAs, and cytokines [64]. Alterations in the gut barrier function can lead to the translocation of microbial antigens and the activation of autoimmune responses. Furthermore, the microbiota can also influence the production of specific immune cell subsets that produce anti-inflammatory cytokines, such as IL-10 and transforming growth factor beta (TGF-β), resulting in the suppression of autoimmune responses. An imbalance in the microbiota composition can lead to the production of pro-inflammatory cytokines, such as tumor necrosis factor alpha (TNF-α) and IL-6, which can promote autoimmune responses [65,66].

4.1. Rheumatoid Arthritis

RA is characterized by inflamed and painful joints, which arise from inflammation and thickening of the synovial membrane, leading to the development of excessive connective tissue (known as pannus) and the erosion of bone, ultimately causing disability. Additionally, RA often comes with systemic complications like vascular disorders, osteoporosis, and various other issues [67]. Globally, RA incidence is approximately 1%, and the prevalence increases with age [68,69]; the disease typically onsets between the ages of 40 and 50, with a prevalence three to five times higher in women than in men. RA diagnosis relies on evaluating the patient’s physical symptoms and manifestations [70,71].
Currently, there is a lack of effective treatment, and patients experience the burden of musculoskeletal defects, leading to a decline in physical function and quality of life. RA can be grouped into two main subtypes, namely seropositive and seronegative, depending on the presence or absence of specific serum antibodies related to RA (rheumatoid factor or anticitrullinated peptide antibodies) [72]. Seronegativity is typically defined by the absence of anticitrullinated protein antibodies (ACPA) and/or IgM rheumatoid factor (RF). However, recently, it has been shown that the presence of ACPA or newly discovered autoantibodies, as well as rediagnosis to other rheumatic diseases, is rendering this group extremely heterogenic, and its place in the classification of musculoskeletal diseases remains to be clarified [73,74,75,76]. On the other hand, rheumatoid factor (RF), the classic autoantibody, can be detected in 70–80% of patients with RA, in particular ACPA. The presence of autoantibodies has enabled the recognition of a somewhat homogenous subgroup of patients with certain genetic and environmental risk factors and also a more severe course of the disease [77].
Among the several factors involved in RA pathogenesis are also genetic elements, which include mainly class II major histocompatibility antigens/human leukocyte antigens (HLA-DR), along with non-HLA genes [78]. Smoking and potentially other environmental and lifestyle-related elements may favor the production of ACPA and contribute to the onset of ACPA seropositive RA [79,80].
Moreover, production of pro-inflammatory cytokines and lymphocyte activation are fundamental in the pathogenesis of the disease [81]. In this scenario, IL-17 has been recognized as an essential mediator of cartilage and bone destruction [81,82]. The number of Th17 cells is increased in the early disease stages and in active RA [83,84].
Although factors promoting Th17 differentiation in RA are not fully understood, periodontal pathogens have been described to be implicated in RA etiology [85,86,87]. In addition to the association of EBV infection in RA patients, several studies reported the presence of highly severe forms of periodontal disease (PD) [86,88,89]. Other studies showed a reduction in RA severity when the accompanying PD was successfully treated [90,91]. Porphyromonas gingivalis has been described as the main etiological PD agent, and increased antibody titers against Porphyromonas gingivalis have been detected in the serum of patients both at high risk of developing RA and in those with RA [92,93]. Notably, the periodontitis induced by P. gingivalis and P. nigrescens can affect the progression of experimental arthritis in mice, increasing the severity of the induced arthritis [94].
In addition to infectious agents, commensal bacteria have been implicated in RA pathogenesis [95]. Ivanov et al., showed that the introduction of segmented filamentous bacteria (SFB) in GF mice resulted in an increase in Th17 cells in the intestinal lamina propria, promoting the development of autoimmune diseases such as experimental RA [96,97]. Introduction of B. fragilis into GF mice, instead, has been shown to induce the correct development of the immune system and induced Treg cells, preventing the occurrence of colitis, meaning that commensal bacteria can reshape the T cell subset and can drive the immune response [45,98,99]. Moreover, in a clinical study, the authors found a strong correlation between the stool presence of Prevotella copri with disease in new-onset untreated RA patients [100].
In order to define a microbial and metabolite profile that could predict disease RA status, Chen et al., found that RA patients showed reduced GM diversity compared to controls that correlated with disease and with the levels of autoantibody. In detail, Collinsella, Eggerthella, and Faecalibacterium genera were segregated with RA, and Collinsella strongly correlated with high levels of IL-17, suggesting a potential role in altering gut permeability [101].
Moreover, Wu et al., found a decrease in microbial diversity in RA patients’ stool samples compared with healthy subjects, including a lower Firmicutes/Bacteroidetes (F/B) ratio [102] and depletion of butyrate-producing taxa (Faecalibacterium, Roseburia, Subdoligranulum, Ruminococcus, and Pseudobutyrivibrio). Intriguingly, the abundance of Roseburia negatively correlated with erythrocyte sedimentation rate (ESR) and with blood levels of rheumatoid factors (IgM) in RA patients [102].
Finally, Wang et al., performed a data-driven analysis of the gut microbiome–immune–joint interactions in RA, documenting that GM metabolites were implicated in RA at genetic, functional, and phenotypic levels [103].
In conclusion, these studies demonstrate that the GM plays a fundamental role in maintaining the balance between pro- and anti-inflammatory T cells, thus preserving intestinal homeostasis and influencing disease progression. Table 1 summarizes the findings on the role of microorganisms in RA. In addition, modifications in the dental, gut, or saliva microbiota can discriminate RA patients from healthy controls, and since these changes were correlated with clinical measures [89], the microbiota signature could be used to stratify RA patients on the basis of their response to therapy.

4.2. Multiple Sclerosis

Multiple sclerosis (MS) is the most common autoimmune inflammatory demyelinating disease of the central nervous system (CNS), with onset usually between the ages of 20 and 50, affecting more than 2 million people worldwide [104,105]. It is characterized by motor and sensory disturbances associated with vision and cognitive impairment. Three clinical courses of the disease are described: relapsing–remitting (alternating episodes of neurological disability and recovery), primary progressive (gradual worsening from onset), and secondary progressive (relapsing–remitting at the onset but gradual worsening over the MS course) [106,107]. MS etiology is complex and involves the interaction between known susceptibility genes and environmental factors, including infectious agents, lack of sun and vitamin D exposure, smoking, and obesity [108]. Regarding genetic factors, in a recent genome-wide association study (GWAS), 233 single-nucleotide polymorphisms (SNPs or loci) were found to be linked to susceptibility to MS onset. Among these, 32 loci were located within the major histocompatibility complex (MHC), and one locus was identified on the X chromosome. Other SNPs are located within or in close proximity to genes implicated in both the adaptive and innate systems [109,110].
Among the supposed causative factors, the leading candidate is EBV whose contributing role is supported by the increased MS risk after infectious mononucleosis [111], by increased antibody titers against EBV nuclear antigens (EBNAs) in the serum [112], and by the occurrence of EBV in demyelinated lesions [113,114,115]. Finally, Bjornevik and colleagues recently revealed a 32-fold increase in MS risk following EBV infection, with no corresponding increase observed after infection with other viruses, including the similarly transmitted cytomegalovirus. In addition, serum neurofilament light chain levels, a biomarker linked with neuroaxonal degeneration, showed an increase only after seroconversion to EBV. These findings suggest that EBV is the primary MS cause [116].
Additionally, Lanz et al., demonstrated that molecular mimicry between the EBV transcription factor Epstein–Barr nuclear antigen (EBNA1) and glial cell adhesion molecule (GlialCAM) may be the missing molecular link [117]. Indeed, the sequence analysis of immunoglobulin chains from cerebrospinal fluid B cells isolated from nine MS patients showed extensive clonality, suggesting an antigen-specific proliferation. Notably, the B cell-encoded antibodies recognized viral proteins and peptides, particularly EBNA1, which was linked to MS on an epidemiological basis. These findings provide a mechanistic link between EBV infection and the pathobiology of MS [117,118].
As a modulator of the immune response, GM is at the center of research on MS development. Zhou et al., studied the GM of 576 pairs of MS patients and genetically unrelated healthy controls, and they found no difference in α-diversity between MS patients and healthy individuals but a significant difference in β-diversity in disease status. They also did not observe differences in β-diversity between untreated MS and treated MS, suggesting that disease can exert a stronger effect on GM than treatment. On the other hand, Faecalibacterium prausnitzii and other beneficial bacteria that secrete metabolites that block nuclear factor kappa B (NF-κB) and IL-8 activation and upregulate Treg cell differentiation [119] were found to be significantly reduced in untreated MS patients. This depletion had a consequential impact on key metabolic pathways, which could potentially worsen MS-associated inflammation [120]. Streptococcus thermophilus, Azospirillum sp. 47_25, and Rhodospirillum sp. UNK.MSG-17 were then associated with disease severity [120]. Vice versa, the Butyrivibrio, Clostridium, and Ruminococcus species, which are SCFA producers, correlated with lower MS severity [120]. Since SCFAs have well-documented anti-inflammatory properties, these data suggest that the above-mentioned bacteria have the potential to confer benefits through the production of anti-inflammatory metabolites.
Cox et al., found that β-diversity was significantly different between MS patients and controls, but these differences were not observed between relapsing–remitting and progressive MS patients [121]. In both progressive and relapsing–remitting forms, they observed an increase in Clostridium bolteae, Ruthenibacterium lactatiformans, and Akkermansia, along with a decrease in Blautia wexlerae, Dorea formicigenerans, and Erysipelotrichaceae CCM. Notably, in progressive MS, there were unique findings of elevated Enterobacteriaceae and Clostridium g24 FCEY, along with a decrease in Blautia and Agathobaculum. Additionally, various Clostridium species were identified [121].
In a matched case and control longitudinal study, Cantoni and colleagues [122] observed a lower presence of specific bacteria such as Faecalibacteria, Prevotella, Lachnospiraceae, and Anaerostipes in MS patients compared to healthy controls, supporting previous research findings [123]. This observation is biologically plausible because these bacteria are known to produce butyrate, which, by activating G protein-coupled receptors and inhibiting histone deacetylase, plays a crucial role in suppressing the demyelination of the CNS, a prominent feature in MS [124]. Previous studies have also revealed reduced levels of SCFAs, including acetate, butyrate, and propionate, in the feces of relapsing–remitting MS patients compared to those without MS [125,126]. A trend towards lower concentrations of butyrate in the stools of MS patients was observed, aligning with the decreased presence of SCFA-producing bacteria in MS. Remarkably, the dietary choices, such as higher meat consumption among MS patients, may contribute to the observed decline in SCFA levels [122]. Castillo-Álvarez et al., at the phylum level, reported statistically significant changes in the abundance of Firmicutes, Proteobacteria, Actinobacteria, and Lentisphaerae between MS patients and controls [127]. The operational taxonomic units (OTUs) analysis revealed that, among these taxa, seven belonged to the phylum of Bacteroidetes, two to Actinobacteria, one to Proteobacteria, and one to Firmicutes. On the contrary, five OTUs (uncultured Bacteroides sp.; Prevotella copri; uncultured alpha Proteobacterium; Eubacterium eligens; and uncultured Pseudomonas sp.). were less abundant among MS patients. Among these, two were classified under Bacteroidetes, two under Proteobacteria, and one under Firmicutes [127]. The Firmicutes phylum plays a significant role in generating SCFAs, notably butyrate, and it contributes to the differentiation of Treg cells [128,129]. Significant differences in the decrease in Bacteroides and increase in Methanobrevibacter, Streptococcus, and Akkermansia abundances were documented in MS patients compared with healthy controls [130,131]. In mice models, some species of Bifidobacterium and Streptococcus can induce Th17 cells, while Streptococcus mitis can induce Th17 cell differentiation in humans [132,133], suggesting that increasing these two species in MS patients could increase the activity of Th17 cells. Although microbiota-driven Th17 cell activation is a putative trigger of MS, aberrant local inflammatory processes in the brain play also a relevant role in disease progression.
On the other hand, microbiota can induce the activation of Treg cells that maintain immune tolerance by producing SCFAs [35,134], which can stimulate the expression of Foxp3, a transcription factor that is essential for Treg cell development, and inhibit the activation of pro-inflammatory immune cells, such as Th17 cells [134,135].
These findings, summarized in Table 1, hold the promise of paving the way for the development of specific probiotics, designed to rejuvenate the natural balance and functionality of the GM, offering potential benefits for MS patients.

4.3. Systemic Lupus Erythematosus

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease affecting multiple systems, characterized by a pattern of relapsing and remitting symptoms.
SLE has a multifactorial origin, involving factors such as genetics, hormones, and environmental exposures [136,137]. Smoking, exposure to silica dust, UV radiation, stress, air pollution, pesticides, and heavy metals are the main environmental risk factors that show some evidence of association with SLE [138,139]. Regarding genetic factors, GWAS have revealed over 100 genetic loci associated with susceptibility to SLE across diverse populations, suggesting that a significant portion of the genetic risk is shared across borders and ethnicities [140,141,142].
It is more prevalent in women of childbearing age, with a female predominance of 9:1. Moreover, women with SLE often show more severe disease manifestations compared to men [143,144]. Characteristic of SLE is the presence of antibodies targeting nuclear and cytoplasmic antigens, along with a range of other autoantibodies. These include anti-Scl-70 antibodies (linked to systemic sclerosis), anti-La and anti-Ro antibodies (detected in Sjogren’s disease), anticardiolipin antibodies, and antiphospholipid antibodies. This antibody profile suggests a comprehensive association between SLE and various other autoimmune diseases.
Dysregulation of innate and adaptive immune cells, other SLE characteristics, can result in excessive activation of T and B cells, increased autoantibodies’ production, and the accumulation of immune complexes in renal tubules, leading to glomerulonephritis and inflammation in several organs [145]. SLE pathophysiology is influenced by a complex interplay of genetic, environmental, hormonal, and other immunoregulatory variables, but the etiology is still not entirely clear [146].
As suggested by recent reports, the GM seems implicated in SLE development and symptom onset. In both SLE animal models and patients, alterations have been identified in various taxa of bacteria, such as Ruminococcus, Lactobacillus, Akkermansia, and B. fragilis [147,148]. In detail, Luo et al., found that the GM changed significantly before and after SLE onset in New Zealand Black/White F1 (NZB/W F1) mice [149], while Zhang and colleagues observed a notable reduction in Lactobacillaceae abundance and a significant increase in Lachnospiraceae in MRL/lpr mice predisposed to SLE [150]. In agreement, another investigation reported a diminished presence of Lactobacillaceae in MRL/lpr mice [151], and elevated levels of intestinal Lactobacillaceae were linked to the amelioration of SLE symptoms, whereas heightened colonization of Lachnospiraceae was correlated with SLE progression [150]. Zegarra-Ruiz et al., reported an increased abundance of Lactobacillus reuteri in TLR7.1 Tg mice, and the colonization of Lactobacillus reuteri exacerbated systemic autoimmunity in both specific pathogen-free and gnotobiotic conditions [152]. The observed reduced F/B ratio in 6-week-old MRL/lpr mice could potentially contribute to the early disease onset [153]. In addition, Valiente et al., found that NZM2410 mice, when colonized with segmented filamentous bacteria, exhibited a deterioration in glomerulonephritis, along with the deposition of immune complexes in both glomerular and tubular regions and interstitial inflammation [154]. Consequently, GM dysbiosis in SLE mouse models is marked by a decline in beneficial bacteria and some increased detrimental bacteria, correlating with SLE.
Finally, also, human clinical trials showed differences in the GM composition between SLE patients and healthy controls. Wang et al., conducted a comparison between SLE patients and their healthy family members, accounting for living conditions and dietary factors. They revealed that the GM of SLE patients still exhibited differences compared to that of healthy controls. Several studies carried out in various countries worldwide have documented a reduced F/B ratio in the GM of SLE patients when compared to healthy individuals [155,156,157]. In their meta-analysis, Xiang et al., reported an increased abundance of Enterobacteriaceae and Enterococcaceae, along with a decreased abundance of Ruminococcaceae in the GM of SLE patients [158] (Table 1).
Table 1. Studies reporting microorganisms’ involvement in autoimmunity and proposed roles. We need remark that there is no single factor responsible for activating autoimmunity, but it seems that infections and imbalance in microorganism composition are parts of the multifactorial processes involved in autoimmune onset, which can be influenced by several variables.
Table 1. Studies reporting microorganisms’ involvement in autoimmunity and proposed roles. We need remark that there is no single factor responsible for activating autoimmunity, but it seems that infections and imbalance in microorganism composition are parts of the multifactorial processes involved in autoimmune onset, which can be influenced by several variables.
Autoimmune DiseasePathogenRoleReference
Rheumatoid arthritis Epstein–Barr virusDisease onset[63]
Rheumatoid arthritis Porphyromonas gingivalisIncrease disease severity[92,93]
Rheumatoid arthritis Prevotella copriCorrelation with disease onset[100]
Rheumatoid arthritis Firmicutes/Bacteroidetes ratio and butyrate-producing taxaDecreased in RA stool samples[102]
Multiple sclerosisEpstein–Barr virusDisease onset[116,118]
Multiple sclerosisStreptococcus thermophilus, Azospirillum sp. 47_25, and Rhodospirillum sp. UNK.MSG-17Increase disease severity[120]
Multiple sclerosisClostridium g24 FCEYPresent in progressive MS[121]
Multiple sclerosisClostridium bolteae, Ruthenibacterium lactatiformans, and AkkermansiaHighly present in relapsing–remitting MS[121]
Multiple sclerosisBacteroidetes, Actinobacteria, Proteobacteria, and FirmicutesSignificantly more abundant in MS patients compared to healthy controls[128]
Systemic lupus erythematosus Epstein–Barr virusDisease onset[63]
Systemic lupus erythematosus LachnospiraceaeSLE progression[150]
Systemic lupus erythematosus Enterobacteriaceae and EnterococcaceaeIncreased in SLE stool samples[158]
Systemic lupus erythematosus Firmicutes/Bacteroidetes ratioDecreased in SLE stool samples[156,157]

5. Sexual Dimorphism in Immunity Modulation

SLE, RA, and MS have a female-to-male disease susceptibility ratio of 9:1, 3:1, and 2:1, respectively [159,160,161]. Although complex and likely multifactorial, this gender dimorphism is partly attributable to differences in the levels and response to sex steroid hormones in males and females. It is demonstrated that castration of males enhanced disease progression in animal models of SLE [162] and type 1 diabetes (T1D) [163]. Administration of androgens to females led to their protection from autoimmune diseases [162,164], and hormone treatment was used in SLE patients’ therapy [165]. Finally, a recent study revealed a metabolic signature of urinary steroids associated with SLE, characterized by a lower level of total androgens observed in patients and a slightly higher level of total estrogens in SLE patients than controls [166].
Regarding MS, both men and women have lower testosterone levels when compared to healthy controls [167], and some studies analyzing testosterone as a therapeutic agent described its neuroprotective effect. Indeed, after 12 months of testosterone treatment, remarkable improvements in auditory tasks and a reduction in cerebral volume loss were recorded [168,169].
Notably, gestation usually protects against autoimmune diseases [170,171] by developing an immune-tolerant condition in which the maternal immune system adapts to the allogeneic tissues of the fetus. Cytokines produced by the fetoplacental unit can modulate maternal immune responses, promoting a strong Th2 and decreasing Th1/Th17-mediated response to reduce the risks of miscarriage [172,173]. Estrogen/estrogen receptor (E2/ER) signaling plays an active role in the development, differentiation, and functionality of both innate and adaptive immune cells [174,175,176]. Indeed, a direct E2 role in regulating the function and differentiation of immune cells has been confirmed both in the healthy immune system and in several diseases [177].
Regarding the effects of E2 on adaptive immunity, it can influence T cell biology throughout their entire life cycle, from right maturation through to the modulation of effector functions since thymocytes and thymic epithelial cells express ER [178,179,180]. Moreover, in mouse models, it has been shown that E2 can trigger thymic atrophy through apoptosis induction in T cells involving Fas–Fas ligand (FasL) interaction [181,182,183]. E2 can also induce an extrathymic pathway of T cell differentiation in the liver (Figure 2). It is thought that these extrathymically produced T cells are more autoreactive and could thus contribute to the higher incidence of autoimmune disease in women [184]. In a mouse model of ER ablation specifically in T lymphocytes, the authors observed an increased T cell activation, proliferation, survival, and Th subset differentiation, demonstrating the ER relevance in regulating T cell functions and suggesting that ER may be a potential therapeutic target for autoimmune disorders [185].
E2 has been demonstrated to increase the abundance of bone marrow progenitor B cells and enhance the survival of splenic B lymphocytes, promoting the development of autoreactive B cells [186].
In the spleen, E2 can promote the expansion of the transient and marginal B zone and follicular B cell pools, losing the criteria for negative selection, thus allowing the development of autoreactive B lymphocytes [187] (Figure 2).
Finally, in a mouse model of SLE, E2 treatment increased autoreactive B cell survival, and cells were likely to be eliminated in the central tolerance process [188]. The molecular features of these cells suggested that E2 treatment enhanced antiapoptotic Bcl2 gene expression, as well as that of genes like Shp2 and Vcam that are associated with autoreactive B cell survival [186,188].
In MS, B cells act as antigen-presenting cells and produce antimyelin antibodies and cytokines that contribute to the pathogenesis [189]. Comparing mRNA and protein expression of male and female thymus revealed that autoimmune regulator (Aire) levels were higher in males than in females, in mice and in humans [190,191], and in an MS mouse model, androgen administration protected against autoimmunity through Aire-dependent mechanisms. In castrated male mice, sex differences in Aire expression compared to females were lost. These results support an androgen-driven mechanism that contributes to gender differences in autoimmunity reinforcing a central tolerance barrier, which limits the release of autoimmune T cells into the periphery [191].
Regarding androgens, they exert their biological functions by binding to and activating the androgen receptor (AR) [192]. More studies suggest the involvement of androgens/ARs in immunomodulation, influencing both innate and adaptive immunity. Cumulatively, these hormones demonstrate various immunosuppressive effects, such as diminishing antibodies’ production, lowering the count and activation potential of T cells, and promoting the secretion of anti-inflammatory cytokines by antigen-presenting cells [193,194,195]. These hormones, whose levels are higher in males, seem to have a protective role against the development of various immune-inflammatory diseases [194,196]. However, the relationship between androgens and disease activity is still unclear. In RA patients, Cutolo et al., found higher E2 levels and lower testosterone and progesterone levels compared with healthy controls. Accordingly, Gupta and colleagues detected low levels of testosterone, dehydroepiandrosterone sulfate (DHEAS), and androgen/E2 ratio in serum and synovial fluid of RA patients [197,198].
The supposed mechanism is that increased levels of TNF-α, IL-6, and IL-1, famous inflammatory cytokines in RA synovitis, could substantially stimulate aromatase activity in peripheral tissues, thereby converting androgen to E2 [199,200,201]. However, higher serum levels of testosterone and DHEAS may predict low disease activity, with likely lower levels of some cytokines, such as IL-1, IL-6, and TNF-α, which could promote minimum peripheral conversion of androgens to E2 and therefore higher androgens levels [198]. Recently, Wu et al., revealed a distinct steroid profile in patients with SLE marked by elevated levels of three estrogens and two sterols, coupled with a decrease in nine androgens, one corticoid, and two progestins. Notably, the most substantial alterations were observed in androgens, revealing the presence of disorders in the process of androgen-to-estrogen conversion [166]. Within the central nervous system (CNS), dihydrotestosterone inhibits the release of pro-inflammatory factors, such as TNF-α, IL-1β, IL-6, iNOS, COX-2, NO, and PGE2, induced by LPS in primary microglia cells. This inhibition occurs through the suppression of the TLR4-mediated NF-κB and MAPK p38 signaling pathways, protecting neurons from inflammatory damage caused by the activated microglia [202]. Similarly, in animal models, DHEA reduces the T cell response and exhibits anti-inflammatory effects on microglia and astrocytes, thereby alleviating the severity of experimental autoimmune encephalomyelitis (EAE) and inflammation [203,204].
In addition to the sex hormones themselves, males and females also differ in the number of X or Y chromosomes contained in each cell. Mary Lyon suggested that for the maintenance of an equivalent expression of X-coded genes between males and females, one of the X chromosomes in each female cell should be inactivated [205]. X chromosome inactivation (XCI) ensures that females, like males, have a functional copy of the X chromosome in each cell of the body. Because X inactivation is random, in normal females, the maternally inherited X chromosome is active in some cells and the paternally inherited X chromosome is active in other cells [206]. However, some females undergo nonrandom X chromosome silencing, resulting in 80% or more cells of paternal or maternal origin, a phenomenon known as skewed XCI. Notably, distorted XCI is associated with autoimmune diseases. Significant XCI distortion was also observed in patients with rheumatoid arthritis [207].

6. Role of Microbiota in Regulating Sex Hormone Levels

It is now clearly recognized that GM is active and functional, exerting effects locally and over long distances with the ability to modulate metabolic and immunological messengers as well as hormone circulating levels, particularly E2 in women [208,209]. The link between GM and E2 was observed as antibiotic assumption has been shown to reduce E2 levels in women [210]. The E2 hydroxylated and conjugated to their metabolites are secreted into the bile and subsequently into the GI tract, where they can be deconjugated into active E2 accordingly by the activity of the β-glucuronidase enzyme. This enzyme is encoded by several GM genera, including Bacteroides, Bifidobacterium, Escherichia, Fecalibacterium, Lactobacillus, and Roseburia [211,212,213], that are also able to modulate systemic E2 and their metabolites’ (hydroxylated species from estrone or estradiol) concentration. Ervin et al., demonstrated that GM β-glucuronidase can reactivate two different estrogen glucuronides, estrone-3-glucuronide and estradiol-17-glucuronide, to estrone and estradiol, respectively, from their inactive glucuronides [212].
In light of these data, the gut can be a reservoir of estrogenic metabolites with a local and distant action capacity affecting both health and disease condition.
In addition to E2 hormones, there are plant compounds, called phytoestrogens, which show structural and functional similarities to E2 [214]. Phytoestrogens include isoflavones, such as genistein and daidzein, mainly abundant in soya, that are activated after being metabolized by the GM through conversion of the isoflavone daidzein to O-desmethylangolensin (ODMA) and equol. Both of them have estrogenic activity and can cause physiological effects by affecting cell signaling and may trigger also epigenetic effects and intracellular signaling cascades [215,216,217,218].
Finally, there are the endocrine disruptors, defined as “exogenous agents that interfere with the synthesis, secretion, transport, metabolism, binding action, or elimination of natural blood-borne hormones that are present in the body and are responsible for homeostasis, reproduction, and developmental process” [219]. By their binding to ER, they can elicit downstream gene activation and trigger intracellular signaling cascades in more tissues, affecting the host metabolism [220]. There is a bidirectional relationship between GM and endocrine disruptors since GM can metabolize the compounds into biologically active or inactive forms; meanwhile, endocrine disruptors can selectively induce the growth of specific GM populations. In detail, Clostridium methoxybenzovorans and Bifidobacterium pseudocatenulatum WC 401 can deglucosylate, respectively, anhydrosecoisolariciresinol and secoisolariciresinol diglucoside [221,222], transforming them into enterodiol and enterolactone, which exert E2 activity. Anhydrosecoisolariciresinol and secoisolariciresinol can be also demethylated by Peptostreptococcus, Eubacterium limosum, and Clostridium methoxybenzovorans [223].
Likewise, it has recently shown that the gut microbiome is implicated in the metabolism and deglucuronidation of dihydrotestosterone (DHT) and testosterone, resulting in exceptionally high DHT levels [224]. Furthermore, a potential GM mechanism to modulate the sex hormones could be the hydroxysteroid dehydrogenase (HSD) enzymes, which are involved in the metabolism of steroid hormones and control steroids binding to their nuclear receptors, causing them to act as activators or inhibitors [225,226].
Finally, the GM can influence the sex hormones’ concentration through SCFA production [156]. SCFAs function by binding to G protein-coupled receptors (free fatty acid receptors (FFARs) 2 and 3) and, through adenylate cyclase, can lead to inhibition of cAMP pathways. G protein activation leads to hydroxylation of phosphatidylinositol 4,5 bisphosphate (PIP2) to 1,2 diacylglycerol (DAG) and inositol 1,4,5 triphosphate (IP3), activating protein kinase C (PKC) and increasing calcium release [227,228].
Overall, the microbiome–hormone interactions play a critical role in modulating the immune system’s activity and function, and alterations in hormones’ levels or signaling can contribute to the development and progression of autoimmune diseases. Further research is needed to elucidate the specific mechanisms involved and to develop novel therapeutic strategies based on hormone modulation by manipulating the microbiota, including fecal microbiota transplantation (FMT) [229].

7. Clinical Trials Examining the Role of FMT in Autoimmune Diseases

FMT consists of the transferring of the entire community of human fecal microbiota from a healthy donor to the GI tract of a recipient patient with the aim of re-establishing microbial diversity and host intestinal health [230]. FMT is currently a consolidated treatment for recurrent Clostridium difficile infection (CDI) that is not responding to standard therapies, and since 2020, the research has expanded to explore FMT’s potential in a plethora of other pathologies such as neurodegenerative diseases [231] and autoimmune diseases [232].
A study transplanting human fecal material into a mouse MS model showed that mice colonized with microbiota derived from MS patients had a higher frequency of spontaneous experimental autoimmune encephalomyelitis (EAE) than mice transplanted with GM from healthy twins [233]. This study suggested that FMT can be an innovative therapy by modulating the immune response in MS. There are three active clinical trials, one completed and two terminated, on MS, as summarized in Table 2.
Regarding RA, one case report described that a patient with refractory RA was successfully treated with FMT, suggesting its good therapeutic effects on RA [234]. Meanwhile, a clinical trial is reported on the evaluation of FMT efficacy and safety in patients with RA refractory to methotrexate (Table 2).
Regarding SLE treatment, recent studies on mice models documented that the GM derived from SLE patients and transplanted into recipient mice induced the production of autoantibodies and upregulated the expression of genes associated with SLE onset. In addition, Choi et al., transplanting the dysbiotic GM from triple congenic lupus-prone mice into germ-free congenic C57BL/6 mice [235], observed that the transplanted GM activated immune cells and triggered the autoantibodies production in the recipient mice. Similarly, Ma et al., after FMT from SLE mice into germ-free mice, observed that the fecal microbiome from SLE mice stimulated the secretion of anti-dsDNA antibodies and increased the expression of susceptibility genes associated with SLE in germ-free mice [236]. Furthermore, germ-free or germ-depleted mice exhibited elevated blood pressure and vascular complications following the transplantation of GM obtained from hypertensive NZBWF1 mice [237]. Finally, the first clinical trial using FMT for the treatment of SLE patients was conducted by Huang et al., using an oral encapsulated microbiome isolated from the feces of healthy donors [238]. The authors reported that FMT treatment led to a significant reduction in the Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2K) score and the levels of serum anti-dsDNA antibodies. Additionally, they observed a significant decrease in bacterial taxa linked to inflammation and an increase in bacteria producing SCFAs. Finally, the peripheral blood levels of IL-6 and the CD4+ memory/naïve ratio decreased after FMT, whereas the synthesis of SCFAs increased [238].

8. Other GM-Modulating Approaches and Future Perspectives

Regarding GM modulation, in addition to FMT, there are other promising approaches involving the administration of probiotics, prebiotics, symbiotics, and postbiotics.
These natural compounds are nontargeted approaches in GM shaping; however, their use in combination with other therapeutic interventions should be taken into account [239,240]. A synthetic bacterial preparation of microorganisms called “Bacterial Consortium” is under development with the aim to provide the administration of specific beneficial bacterial strains to support the growth of a new community, with the goal of achieving beneficial outcomes [241,242].
Some probiotic strains, including those of Lactobacilli, Bifidobacteria, Propionibacterium, E. coli, Saccharomyces, and Bacillus, can positively regulate TLR activation through the decrease in MAPK activation and NF-κB pathways, thus limiting the production of pro-inflammatory cytokines [243]. Moreover, several small molecules have demonstrated efficacy in inhibiting the bacterial β-glucuronidase enzyme, a pivotal player in metabolizing glucuronide drug conjugates produced by host metabolism [244,245]. This intervention strategy could help in regulating the levels of GM producing E2 in autoimmune diseases, contributing to the limitation of E2 circulating levels. The diet regimes, as well as the administration of biotics, drugs limiting the β-glucuronidase enzyme, and “Bacterial Consortium”, could effectively influence the GM composition. This could enhance the presence of beneficial microbes, preventing infections and restoring a functional gut microbiome, thus contributing to improving patients’ responses to therapies.

9. Conclusions

There are biological differences in immunological responses to stimuli and to hormone circulating levels between males and females, and this can contribute to sex differences in the loss of immunological tolerance and autoantibody production. Although estrogens generally protect women from infections, they predispose the same to chronic inflammatory conditions and are a major risk in the development of autoimmunity compared to their male counterparts.
In addition, microbial metabolism may exert protection or promote exacerbation of some disease processes by regulating both sex hormone circulating levels and immune system homeostasis. Given the complexity of the several factors implicated in autoimmune diseases’ onset, a multifaceted approach is needed to treat these pathologies. By employing approaches such as FMT and other treatments to modulate the microbiota, along with methods to regulate hormones, it becomes imperative to advance personalized medicine. We are confident that this progression is crucial for attaining improved therapeutic results in the treatment of autoimmune diseases.

Author Contributions

Conceptualization, A.A.; methodology, S.M.; investigation, S.M. and G.N.; resources, S.M. and G.N.; writing—original draft preparation, S.M. and G.N.; writing—review and editing, S.M. and G.N.; visualization, S.M., G.N., A.A., F.C. (Fabio Cianchi) and F.C. (Francesco Coratti); supervision, A.A.; project administration, A.A.; funding acquisition, A.A. All authors have read and agreed to the published version of the manuscript.

Funding

Funded by the European Union—NextGenerationEU—National Recovery and Resilience Plan, Mission 4 Component 2—Investment 1.5—THE—Tuscany Health Ecosystem—ECS00000017—CUP B83C22003920001.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Hooper, L.V.; Gordon, J.I. Commensal host-bacterial relationships in the gut. Science 2001, 292, 1115–1118. [Google Scholar] [CrossRef] [PubMed]
  2. Jiao, Y.; Wu, L.; Huntington, N.D.; Zhang, X. Crosstalk Between Gut Microbiota and Innate Immunity and Its Implication in Autoimmune Diseases. Front. Immunol. 2020, 11, 282. [Google Scholar] [CrossRef]
  3. Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef] [PubMed]
  4. Brown, E.M.; Kenny, D.J.; Xavier, R.J. Gut Microbiota Regulation of T Cells During Inflammation and Autoimmunity. Annu. Rev. Immunol. 2019, 37, 599–624. [Google Scholar] [CrossRef] [PubMed]
  5. Okada, H.; Kuhn, C.; Feillet, H.; Bach, J.F. The ‘hygiene hypothesis’ for autoimmune and allergic diseases: An update. Clin. Exp. Immunol. 2010, 160, 1–9. [Google Scholar] [CrossRef]
  6. Klein, S.L.; Flanagan, K.L. Sex differences in immune responses. Nat. Rev. Immunol. 2016, 16, 626–638. [Google Scholar] [CrossRef]
  7. Edwards, M.; Dai, R.; Ahmed, S.A. Our Environment Shapes Us: The Importance of Environment and Sex Differences in Regulation of Autoantibody Production. Front. Immunol. 2018, 9, 478. [Google Scholar] [CrossRef]
  8. Ortona, E.; Pierdominici, M.; Maselli, A.; Veroni, C.; Aloisi, F.; Shoenfeld, Y. Sex-based differences in autoimmune diseases. Ann. Ist. Super Sanita 2016, 52, 205–212. [Google Scholar] [CrossRef]
  9. Gensollen, T.; Iyer, S.S.; Kasper, D.L.; Blumberg, R.S. How colonization by microbiota in early life shapes the immune system. Science 2016, 352, 539–544. [Google Scholar] [CrossRef]
  10. Yoo, J.-S.; Oh, S.F. Unconventional immune cells in the gut mucosal barrier: Regulation by symbiotic microbiota. Exp. Mol. Med. 2023, 55, 1905–1912. [Google Scholar] [CrossRef]
  11. Lin, L.; Zhang, J. Role of intestinal microbiota and metabolites on gut homeostasis and human diseases. BMC Immunol. 2017, 18, 2. [Google Scholar] [CrossRef] [PubMed]
  12. Donald, K.; Finlay, B.B. Early-life interactions between the microbiota and immune system: Impact on immune system development and atopic disease. Nat. Rev. Immunol. 2023, 23, 735–748. [Google Scholar] [CrossRef] [PubMed]
  13. Lubin, J.B.; Green, J.; Maddux, S.; Denu, L.; Duranova, T.; Lanza, M.; Wynosky-Dolfi, M.; Flores, J.N.; Grimes, L.P.; Brodsky, I.E.; et al. Arresting microbiome development limits immune system maturation and resistance to infection in mice. Cell Host Microbe 2023, 31, 554–570. [Google Scholar] [CrossRef] [PubMed]
  14. Barone, M.; Ramayo-Caldas, Y.; Estellé, J.; Tambosco, K.; Chadi, S.; Maillard, F.; Gallopin, M.; Planchais, J.; Chain, F.; Kropp, C.; et al. Gut barrier-microbiota imbalances in early life lead to higher sensitivity to inflammation in a murine model of C-section delivery. Microbiome 2023, 11, 140. [Google Scholar] [CrossRef]
  15. Nigro, G.; Rossi, R.; Commere, P.H.; Jay, P.; Sansonetti, P.J. The cytosolic bacterial peptidoglycan sensor Nod2 affords stem cell protection and links microbes to gut epithelial regeneration. Cell Host Microbe 2014, 15, 792–798. [Google Scholar] [CrossRef] [PubMed]
  16. Erturk-Hasdemir, D.; Oh, S.F.; Okan, N.A.; Stefanetti, G.; Gazzaniga, F.S.; Seeberger, P.H.; Plevy, S.E.; Kasper, D.L. Symbionts exploit complex signaling to educate the immune system. Proc. Natl. Acad. Sci. USA 2019, 116, 26157–26166. [Google Scholar] [CrossRef]
  17. Tang, C.; Kamiya, T.; Liu, Y.; Kadoki, M.; Kakuta, S.; Oshima, K.; Hattori, M.; Takeshita, K.; Kanai, T.; Saijo, S.; et al. Inhibition of Dectin-1 Signaling Ameliorates Colitis by Inducing Lactobacillus-Mediated Regulatory T Cell Expansion in the Intestine. Cell Host Microbe 2015, 18, 183–197. [Google Scholar] [CrossRef]
  18. 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. [Google Scholar] [CrossRef]
  19. Wu, K.; Yuan, Y.; Yu, H.; Dai, X.; Wang, S.; Sun, Z.; Wang, F.; Fei, H.; Lin, Q.; Jiang, H.; et al. The gut microbial metabolite trimethylamine N-oxide aggravates GVHD by inducing M1 macrophage polarization in mice. Blood 2020, 136, 501–515. [Google Scholar] [CrossRef] [PubMed]
  20. Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef] [PubMed]
  21. Wen, L.; Ley, R.E.; Volchkov, P.Y.; Stranges, P.B.; Avanesyan, L.; Stonebraker, A.C.; Hu, C.; Wong, F.S.; Szot, G.L.; Bluestone, J.A.; et al. Innate immunity and intestinal microbiota in the development of Type 1 diabetes. Nature 2008, 455, 1109–1113. [Google Scholar] [CrossRef]
  22. Vaishnava, S.; Yamamoto, M.; Severson, K.M.; Ruhn, K.A.; Yu, X.; Koren, O.; Ley, R.; Wakeland, E.K.; Hooper, L.V. The antibacterial lectin RegIIIgamma promotes the spatial segregation of microbiota and host in the intestine. Science 2011, 334, 255–258. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, S.; Charbonnier, L.M.; Noval Rivas, M.; Georgiev, P.; Li, N.; Gerber, G.; Bry, L.; Chatila, T.A. MyD88 Adaptor-Dependent Microbial Sensing by Regulatory T Cells Promotes Mucosal Tolerance and Enforces Commensalism. Immunity 2015, 43, 289–303. [Google Scholar] [CrossRef] [PubMed]
  24. Omenetti, S.; Bussi, C.; Metidji, A.; Iseppon, A.; Lee, S.; Tolaini, M.; Li, Y.; Kelly, G.; Chakravarty, P.; Shoaie, S.; et al. The Intestine Harbors Functionally Distinct Homeostatic Tissue-Resident and Inflammatory Th17 Cells. Immunity 2019, 51, 77–89. [Google Scholar] [CrossRef] [PubMed]
  25. Fung, T.C.; Bessman, N.J.; Hepworth, M.R.; Kumar, N.; Shibata, N.; Kobuley, D.; Wang, K.; Ziegler, C.G.K.; Goc, J.; Shima, T.; et al. Lymphoid-Tissue-Resident Commensal Bacteria Promote Members of the IL-10 Cytokine Family to Establish Mutualism. Immunity 2016, 44, 634–646. [Google Scholar] [CrossRef] [PubMed]
  26. Chung, H.; Pamp, S.J.; Hill, J.A.; Surana, N.K.; Edelman, S.M.; Troy, E.B.; Reading, N.C.; Villablanca, E.J.; Wang, S.; Mora, J.R.; et al. Gut immune maturation depends on colonization with a host-specific microbiota. Cell 2012, 149, 1578–1593. [Google Scholar] [CrossRef] [PubMed]
  27. Wang, Y.; Yin, Y.; Chen, X.; Zhao, Y.; Wu, Y.; Li, Y.; Wang, X.; Chen, H.; Xiang, C. Induction of Intestinal Th17 Cells by Flagellins From Segmented Filamentous Bacteria. Front. Immunol. 2019, 10, 2750. [Google Scholar] [CrossRef]
  28. Cosmi, L.; Santarlasci, V.; Maggi, L.; Liotta, F.; Annunziato, F. Th17 plasticity: Pathophysiology and treatment of chronic inflammatory disorders. Curr. Opin. Pharmacol. 2014, 17, 12–16. [Google Scholar] [CrossRef]
  29. Niccolai, E.; Boem, F.; Emmi, G.; Amedei, A. The link “Cancer and autoimmune diseases” in the light of microbiota: Evidence of a potential culprit. Immunol. Lett. 2020, 222, 12–28. [Google Scholar] [CrossRef]
  30. Cosmi, L.; Maggi, L.; Santarlasci, V.; Liotta, F.; Annunziato, F. T helper cells plasticity in inflammation. Cytom. Part A 2014, 85, 36–42. [Google Scholar] [CrossRef]
  31. Smith, K.J.; Minns, D.; McHugh, B.J.; Holloway, R.K.; O’Connor, R.; Williams, A.; Melrose, L.; McPherson, R.; Miron, V.E.; Davidson, D.J.; et al. The antimicrobial peptide cathelicidin drives development of experimental autoimmune encephalomyelitis in mice by affecting Th17 differentiation. PLoS Biol. 2022, 20, e3001554. [Google Scholar] [CrossRef]
  32. Lathrop, S.K.; Bloom, S.M.; Rao, S.M.; Nutsch, K.; Lio, C.W.; Santacruz, N.; Peterson, D.A.; Stappenbeck, T.S.; Hsieh, C.S. Peripheral education of the immune system by colonic commensal microbiota. Nature 2011, 478, 250–254. [Google Scholar] [CrossRef] [PubMed]
  33. Cebula, A.; Seweryn, M.; Rempala, G.A.; Pabla, S.S.; McIndoe, R.A.; Denning, T.L.; Bry, L.; Kraj, P.; Kisielow, P.; Ignatowicz, L. Thymus-derived regulatory T cells contribute to tolerance to commensal microbiota. Nature 2013, 497, 258–262. [Google Scholar] [CrossRef]
  34. Huber, S.; Gagliani, N.; Esplugues, E.; O’Connor, W., Jr.; Huber, F.J.; Chaudhry, A.; Kamanaka, M.; Kobayashi, Y.; Booth, C.J.; Rudensky, A.Y.; et al. Th17 cells express interleukin-10 receptor and are controlled by Foxp3 and Foxp3+ regulatory CD4+ T cells in an interleukin-10-dependent manner. Immunity 2011, 34, 554–565. [Google Scholar] [CrossRef] [PubMed]
  35. Atarashi, K.; Tanoue, T.; Shima, T.; Imaoka, A.; Kuwahara, T.; Momose, Y.; Cheng, G.; Yamasaki, S.; Saito, T.; Ohba, Y.; et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 2011, 331, 337–341. [Google Scholar] [CrossRef] [PubMed]
  36. Atarashi, K.; Tanoue, T.; Oshima, K.; Suda, W.; Nagano, Y.; Nishikawa, H.; Fukuda, S.; Saito, T.; Narushima, S.; Hase, K.; et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 2013, 500, 232–236. [Google Scholar] [CrossRef]
  37. Karimi, K.; Inman, M.D.; Bienenstock, J.; Forsythe, P. Lactobacillus reuteri-induced regulatory T cells protect against an allergic airway response in mice. Am. J. Respir. Crit. Care Med. 2009, 179, 186–193. [Google Scholar] [CrossRef]
  38. Amador-Patarroyo, M.J.; Rodriguez-Rodriguez, A.; Montoya-Ortiz, G. How does age at onset influence the outcome of autoimmune diseases? Autoimmune Dis. 2012, 2012, 251730. [Google Scholar] [CrossRef]
  39. Lerner, A.; Jeremias, P.; Matthias, T. The World Incidence and Prevalence of Autoimmune Diseases is Increasing. Int. J. Celiac Dis. 2015, 3, 151–155. [Google Scholar] [CrossRef]
  40. Garcia-Gutierrez, E.; Mayer, M.J.; Cotter, P.D.; Narbad, A. Gut microbiota as a source of novel antimicrobials. Gut Microbes 2019, 10, 1–21. [Google Scholar] [CrossRef]
  41. Clarke, T.B.; Davis, K.M.; Lysenko, E.S.; Zhou, A.Y.; Yu, Y.; Weiser, J.N. Recognition of peptidoglycan from the microbiota by Nod1 enhances systemic innate immunity. Nat. Med. 2010, 16, 228–231. [Google Scholar] [CrossRef]
  42. Grinblat, J.; Weiss, A.; Grosman, B.; Dicker, D.; Beloosesky, Y. Diarrhea in elderly patients due to Clostridium difficile associated with Salmonella and Shigella infection. Arch. Gerontol. Geriatr. 2004, 39, 277–282. [Google Scholar] [CrossRef]
  43. Bahman, Y.; Maryam, M.; Aisa, B.; Falalyeyeva, T.; Kobyliak, N.; Majid, E. Immunomodulatory role of Faecalibacterium prausnitzii in obesity and metabolic disorders. Minerva Biotechnol. Biomol. 2021, 33, 76. [Google Scholar] [CrossRef]
  44. Mills, K.H.G. IL-17 and IL-17-producing cells in protection versus pathology. Nat. Rev. Immunol. 2023, 23, 38–54. [Google Scholar] [CrossRef] [PubMed]
  45. Round, J.L.; Lee, S.M.; Li, J.; Tran, G.; Jabri, B.; Chatila, T.A.; Mazmanian, S.K. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science 2011, 332, 974–977. [Google Scholar] [CrossRef] [PubMed]
  46. Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef] [PubMed]
  47. Rivière, A.; Selak, M.; Lantin, D.; Leroy, F.; De Vuyst, L. Bifidobacteria and Butyrate-Producing Colon Bacteria: Importance and Strategies for Their Stimulation in the Human Gut. Front. Microbiol. 2016, 7, 979. [Google Scholar] [CrossRef]
  48. Kim, M.; Qie, Y.; Park, J.; Kim, C.H. Gut Microbial Metabolites Fuel Host Antibody Responses. Cell Host Microbe 2016, 20, 202–214. [Google Scholar] [CrossRef]
  49. Shahrizaila, N.; Yuki, N. Guillain-barré syndrome animal model: The first proof of molecular mimicry in human autoimmune disorder. J. Biomed. Biotechnol. 2011, 2011, 829129. [Google Scholar] [CrossRef]
  50. Johnson, D.; Jiang, W. Infectious diseases, autoantibodies, and autoimmunity. J. Autoimmun. 2023, 137, 102962. [Google Scholar] [CrossRef]
  51. Damian, R.T. Molecular Mimicry: Antigen Sharing by Parasite and Host and Its Consequences. Am. Nat. 1964, 98, 129–149. [Google Scholar] [CrossRef]
  52. Zabriskie, J.B.; Freimer, E.H. An immunological relationship between the group. A streptococcus and mammalian muscle. J. Exp. Med. 1966, 124, 661–678. [Google Scholar] [CrossRef] [PubMed]
  53. Zhao, Z.S.; Granucci, F.; Yeh, L.; Schaffer, P.A.; Cantor, H. Molecular mimicry by herpes simplex virus-type 1: Autoimmune disease after viral infection. Science 1998, 279, 1344–1347. [Google Scholar] [CrossRef] [PubMed]
  54. Coppieters, K.T.; Wiberg, A.; von Herrath, M.G. Viral infections and molecular mimicry in type 1 diabetes. APMIS 2012, 120, 941–949. [Google Scholar] [CrossRef] [PubMed]
  55. Croxford, J.L.; Olson, J.K.; Miller, S.D. Epitope spreading and molecular mimicry as triggers of autoimmunity in the Theiler’s virus-induced demyelinating disease model of multiple sclerosis. Autoimmun. Rev. 2002, 1, 251–260. [Google Scholar] [CrossRef] [PubMed]
  56. Amedei, A.; Bergman, M.P.; Appelmelk, B.J.; Azzurri, A.; Benagiano, M.; Tamburini, C.; van der Zee, R.; Telford, J.L.; Vandenbroucke-Grauls, C.M.; D’Elios, M.M.; et al. Molecular mimicry between Helicobacter pylori antigens and H+, K+—Adenosine triphosphatase in human gastric autoimmunity. J. Exp. Med. 2003, 198, 1147–1156. [Google Scholar] [CrossRef] [PubMed]
  57. Pacheco, Y.; Acosta-Ampudia, Y.; Monsalve, D.M.; Chang, C.; Gershwin, M.E.; Anaya, J.M. Bystander activation and autoimmunity. J. Autoimmun. 2019, 103, 102301. [Google Scholar] [CrossRef] [PubMed]
  58. Okada, M.; Zhang, V.; Loaiza Naranjo, J.D.; Tillett, B.J.; Wong, F.S.; Steptoe, R.J.; Bergot, A.S.; Hamilton-Williams, E.E. Islet-specific CD8. Immunol. Cell Biol. 2023, 101, 36–48. [Google Scholar] [CrossRef] [PubMed]
  59. Smatti, M.K.; Cyprian, F.S.; Nasrallah, G.K.; Al Thani, A.A.; Almishal, R.O.; Yassine, H.M. Viruses and Autoimmunity: A Review on the Potential Interaction and Molecular Mechanisms. Viruses 2019, 11, 762. [Google Scholar] [CrossRef]
  60. Ali, S.; Afzal, S.; Yousaf, M.Z.; Shahid, M.; Amin, I.; Idrees, M.; Aftab, A. Paradoxical Role of Dengue Virus Envelope Protein Domain III Antibodies in Dengue Virus Infection. Crit. Rev. Eukaryot. Gene Expr. 2020, 30, 199–206. [Google Scholar] [CrossRef]
  61. Murray, J. Infection as a cause of multiple sclerosis. BMJ 2002, 325, 1128. [Google Scholar] [CrossRef] [PubMed]
  62. Jacobs, B.M.; Giovannoni, G.; Cuzick, J.; Dobson, R. Systematic review and meta-analysis of the association between Epstein-Barr virus, multiple sclerosis and other risk factors. Mult. Scler. J. 2020, 26, 1281–1297. [Google Scholar] [CrossRef] [PubMed]
  63. Dostál, C.; Newkirk, M.M.; Duffy, K.N.; Palecková, A.; Bosák, V.; Cerná, M.; Zd’arský, E.; Zvárová, J. Herpes viruses in multicase families with rheumatoid arthritis and systemic lupus erythematosus. Ann. N. Y. Acad. Sci. 1997, 815, 334–337. [Google Scholar] [CrossRef] [PubMed]
  64. Singh, R.; Chandrashekharappa, S.; Bodduluri, S.R.; Baby, B.V.; Hegde, B.; Kotla, N.G.; Hiwale, A.A.; Saiyed, T.; Patel, P.; Vijay-Kumar, M.; et al. Enhancement of the gut barrier integrity by a microbial metabolite through the Nrf2 pathway. Nat. Commun. 2019, 10, 89. [Google Scholar] [CrossRef] [PubMed]
  65. Ghosh, S.S.; Wang, J.; Yannie, P.J.; Ghosh, S. Intestinal Barrier Dysfunction, LPS Translocation, and Disease Development. J. Endocr. Soc. 2020, 4, bvz039. [Google Scholar] [CrossRef]
  66. Fasano, A. Leaky Gut and Autoimmune Diseases. Clin. Rev. Allergy Immunol. 2012, 42, 71–78. [Google Scholar] [CrossRef]
  67. Ding, Q.; Hu, W.; Wang, R.; Yang, Q.; Zhu, M.; Li, M.; Cai, J.; Rose, P.; Mao, J.; Zhu, Y.Z. Signaling pathways in rheumatoid arthritis: Implications for targeted therapy. Signal Transduct. Target. Ther. 2023, 8, 68. [Google Scholar] [CrossRef]
  68. Smolen, J.S.; Aletaha, D.; McInnes, I.B. Rheumatoid arthritis. Lancet 2016, 388, 2023–2038. [Google Scholar] [CrossRef] [PubMed]
  69. Radu, A.F.; Bungau, S.G. Management of Rheumatoid Arthritis: An Overview. Cells 2021, 10, 2857. [Google Scholar] [CrossRef]
  70. Littlejohn, E.A.; Monrad, S.U. Early Diagnosis and Treatment of Rheumatoid Arthritis. Prim. Care 2018, 45, 237–255. [Google Scholar] [CrossRef] [PubMed]
  71. Cush, J.J. Rheumatoid Arthritis: Early Diagnosis and Treatment. Rheum. Dis. Clin. N. Am. 2022, 48, 537–547. [Google Scholar] [CrossRef] [PubMed]
  72. Ajeganova, S.; Huizinga, T.W.J. Seronegative and seropositive RA: Alike but different? Nat. Rev. Rheumatol. 2015, 11, 8–9. [Google Scholar] [CrossRef] [PubMed]
  73. Reed, E.; Hedström, A.K.; Hansson, M.; Mathsson-Alm, L.; Brynedal, B.; Saevarsdottir, S.; Cornillet, M.; Jakobsson, P.-J.; Holmdahl, R.; Skriner, K.; et al. Presence of autoantibodies in “seronegative” rheumatoid arthritis associates with classical risk factors and high disease activity. Arthritis Res. Ther. 2020, 22, 170. [Google Scholar] [CrossRef] [PubMed]
  74. Li, K.; Mo, W.; Wu, L.; Wu, X.; Luo, C.; Xiao, X.; Jia, X.; Yang, H.; Fei, Y.; Chen, H.; et al. Novel autoantibodies identified in ACPA-negative rheumatoid arthritis. Ann. Rheum. Dis. 2021, 80, 739–747. [Google Scholar] [CrossRef] [PubMed]
  75. Paalanen, K.; Puolakka, K.; Nikiphorou, E.; Hannonen, P.; Sokka, T. Is seronegative rheumatoid arthritis true rheumatoid arthritis? A nationwide cohort study. Rheumatology 2021, 60, 2391–2395. [Google Scholar] [CrossRef] [PubMed]
  76. Ishigaki, K.; Sakaue, S.; Terao, C.; Luo, Y.; Sonehara, K.; Yamaguchi, K.; Amariuta, T.; Too, C.L.; Laufer, V.A.; Scott, I.C.; et al. Multi-ancestry genome-wide association analyses identify novel genetic mechanisms in rheumatoid arthritis. Nat. Genet. 2022, 54, 1640–1651. [Google Scholar] [CrossRef]
  77. Willemze, A.; Trouw, L.A.; Toes, R.E.; Huizinga, T.W. The influence of ACPA status and characteristics on the course of RA. Nat. Rev. Rheumatol. 2012, 8, 144–152. [Google Scholar] [CrossRef]
  78. Xu, H.; Liu, M.; Cao, J.; Li, X.; Fan, D.; Xia, Y.; Lu, X.; Li, J.; Ju, D.; Zhao, H. The Dynamic Interplay between the Gut Microbiota and Autoimmune Diseases. J. Immunol. Res. 2019, 2019, 7546047. [Google Scholar] [CrossRef]
  79. van der Woude, D.; Alemayehu, W.G.; Verduijn, W.; de Vries, R.R.; Houwing-Duistermaat, J.J.; Huizinga, T.W.; Toes, R.E. Gene-environment interaction influences the reactivity of autoantibodies to citrullinated antigens in rheumatoid arthritis. Nat. Genet. 2010, 42, 814–816. [Google Scholar] [CrossRef]
  80. Lee, H.S.; Irigoyen, P.; Kern, M.; Lee, A.; Batliwalla, F.; Khalili, H.; Wolfe, F.; Lum, R.F.; Massarotti, E.; Weisman, M.; et al. Interaction between smoking, the shared epitope, and anti-cyclic citrullinated peptide: A mixed picture in three large North American rheumatoid arthritis cohorts. Arthritis Rheum. 2007, 56, 1745–1753. [Google Scholar] [CrossRef]
  81. Lundy, S.K.; Sarkar, S.; Tesmer, L.A.; Fox, D.A. Cells of the synovium in rheumatoid arthritis. T lymphocytes. Arthritis Res. Ther. 2007, 9, 202. [Google Scholar] [CrossRef] [PubMed]
  82. van den Berg, W.B.; Miossec, P. IL-17 as a future therapeutic target for rheumatoid arthritis. Nat. Rev. Rheumatol. 2009, 5, 549–553. [Google Scholar] [CrossRef] [PubMed]
  83. Samson, M.; Audia, S.; Janikashvili, N.; Ciudad, M.; Trad, M.; Fraszczak, J.; Ornetti, P.; Maillefert, J.F.; Miossec, P.; Bonnotte, B. Brief report: Inhibition of interleukin-6 function corrects Th17/Treg cell imbalance in patients with rheumatoid arthritis. Arthritis Rheum. 2012, 64, 2499–2503. [Google Scholar] [CrossRef] [PubMed]
  84. van Hamburg, J.P.; Asmawidjaja, P.S.; Davelaar, N.; Mus, A.M.; Colin, E.M.; Hazes, J.M.; Dolhain, R.J.; Lubberts, E. Th17 cells, but not Th1 cells, from patients with early rheumatoid arthritis are potent inducers of matrix metalloproteinases and proinflammatory cytokines upon synovial fibroblast interaction, including autocrine interleukin-17A production. Arthritis Rheum. 2011, 63, 73–83. [Google Scholar] [CrossRef]
  85. de Pablo, P.; Chapple, I.L.; Buckley, C.D.; Dietrich, T. Periodontitis in systemic rheumatic diseases. Nat. Rev. Rheumatol. 2009, 5, 218–224. [Google Scholar] [CrossRef]
  86. Mercado, F.; Marshall, R.I.; Klestov, A.C.; Bartold, P.M. Is there a relationship between rheumatoid arthritis and periodontal disease? J. Clin. Periodontol. 2000, 27, 267–272. [Google Scholar] [CrossRef]
  87. Jung, E.S.; Choi, Y.Y.; Lee, K.H. Relationship between rheumatoid arthritis and periodontal disease in Korean adults: Data from the Sixth Korea National Health and Nutrition Examination Survey, 2013 to 2015. J. Periodontol. 2019, 90, 350–357. [Google Scholar] [CrossRef]
  88. Scher, J.U.; Ubeda, C.; Equinda, M.; Khanin, R.; Buischi, Y.; Viale, A.; Lipuma, L.; Attur, M.; Pillinger, M.H.; Weissmann, G.; et al. Periodontal disease and the oral microbiota in new-onset rheumatoid arthritis. Arthritis Rheum. 2012, 64, 3083–3094. [Google Scholar] [CrossRef]
  89. Zhang, X.; Zhang, D.; Jia, H.; Feng, Q.; Wang, D.; Liang, D.; Wu, X.; Li, J.; Tang, L.; Li, Y.; et al. The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment. Nat. Med. 2015, 21, 895–905. [Google Scholar] [CrossRef] [PubMed]
  90. Al-Katma, M.K.; Bissada, N.F.; Bordeaux, J.M.; Sue, J.; Askari, A.D. Control of periodontal infection reduces the severity of active rheumatoid arthritis. J. Clin. Rheumatol. 2007, 13, 134–137. [Google Scholar] [CrossRef] [PubMed]
  91. Ortiz, P.; Bissada, N.F.; Palomo, L.; Han, Y.W.; Al-Zahrani, M.S.; Panneerselvam, A.; Askari, A. Periodontal therapy reduces the severity of active rheumatoid arthritis in patients treated with or without tumor necrosis factor inhibitors. J. Periodontol. 2009, 80, 535–540. [Google Scholar] [CrossRef]
  92. Hitchon, C.A.; Chandad, F.; Ferucci, E.D.; Willemze, A.; Ioan-Facsinay, A.; van der Woude, D.; Markland, J.; Robinson, D.; Elias, B.; Newkirk, M.; et al. Antibodies to Porphyromonas gingivalis are associated with anticitrullinated protein antibodies in patients with rheumatoid arthritis and their relatives. J. Rheumatol. 2010, 37, 1105–1112. [Google Scholar] [CrossRef] [PubMed]
  93. Mikuls, T.R.; Thiele, G.M.; Deane, K.D.; Payne, J.B.; O’Dell, J.R.; Yu, F.; Sayles, H.; Weisman, M.H.; Gregersen, P.K.; Buckner, J.H.; et al. Porphyromonas gingivalis and disease-related autoantibodies in individuals at increased risk of rheumatoid arthritis. Arthritis Rheum. 2012, 64, 3522–3530. [Google Scholar] [CrossRef] [PubMed]
  94. de Aquino, S.G.; Abdollahi-Roodsaz, S.; Koenders, M.I.; van de Loo, F.A.J.; Pruijn, G.J.M.; Marijnissen, R.J.; Walgreen, B.; Helsen, M.M.; van den Bersselaar, L.A.; de Molon, R.S.; et al. Periodontal pathogens directly promote autoimmune experimental arthritis by inducing a TLR2- and IL-1-driven Th17 response. J. Immunol. 2014, 192, 4103–4111. [Google Scholar] [CrossRef] [PubMed]
  95. Bergot, A.S.; Giri, R.; Thomas, R. The microbiome and rheumatoid arthritis. Best Pract. Res. Clin. Rheumatol. 2019, 33, 101497. [Google Scholar] [CrossRef] [PubMed]
  96. Ivanov, I.I.; Atarashi, K.; Manel, N.; Brodie, E.L.; Shima, T.; Karaoz, U.; Wei, D.; Goldfarb, K.C.; Santee, C.A.; Lynch, S.V.; et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 2009, 139, 485–498. [Google Scholar] [CrossRef] [PubMed]
  97. Wu, H.J.; Ivanov, I.I.; Darce, J.; Hattori, K.; Shima, T.; Umesaki, Y.; Littman, D.R.; Benoist, C.; Mathis, D. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity 2010, 32, 815–827. [Google Scholar] [CrossRef] [PubMed]
  98. Mazmanian, S.K.; Liu, C.H.; Tzianabos, A.O.; Kasper, D.L. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 2005, 122, 107–118. [Google Scholar] [CrossRef] [PubMed]
  99. Round, J.L.; Mazmanian, S.K. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc. Natl. Acad. Sci. USA 2010, 107, 12204–12209. [Google Scholar] [CrossRef]
  100. Scher, J.U.; Sczesnak, A.; Longman, R.S.; Segata, N.; Ubeda, C.; Bielski, C.; Rostron, T.; Cerundolo, V.; Pamer, E.G.; Abramson, S.B.; et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife 2013, 2, e01202. [Google Scholar] [CrossRef]
  101. Chen, J.; Wright, K.; Davis, J.M.; Jeraldo, P.; Marietta, E.V.; Murray, J.; Nelson, H.; Matteson, E.L.; Taneja, V. An expansion of rare lineage intestinal microbes characterizes rheumatoid arthritis. Genome Med. 2016, 8, 43. [Google Scholar] [CrossRef] [PubMed]
  102. Wu, X.; Liu, J.; Xiao, L.; Lu, A.; Zhang, G. Alterations of Gut Microbiome in Rheumatoid Arthritis. Osteoarthr. Cartil. 2017, 25, S287–S288. [Google Scholar] [CrossRef]
  103. Wang, Q.; Xu, R. Data-driven multiple-level analysis of gut-microbiome-immune-joint interactions in rheumatoid arthritis. BMC Genom. 2019, 20, 124. [Google Scholar] [CrossRef] [PubMed]
  104. Barendregt, J.J.; Doi, S.A.; Lee, Y.Y.; Norman, R.E.; Vos, T. Meta-analysis of prevalence. J. Epidemiol. Community Health 2013, 67, 974–978. [Google Scholar] [CrossRef] [PubMed]
  105. Kuhlmann, T.; Moccia, M.; Coetzee, T.; Cohen, J.A.; Correale, J.; Graves, J.; Marrie, R.A.; Montalban, X.; Yong, V.W.; Thompson, A.J.; et al. Multiple sclerosis progression: Time for a new mechanism-driven framework. Lancet Neurol. 2023, 22, 78–88. [Google Scholar] [CrossRef] [PubMed]
  106. Lublin, F.D.; Reingold, S.C.; Cohen, J.A.; Cutter, G.R.; Sørensen, P.S.; Thompson, A.J.; Wolinsky, J.S.; Balcer, L.J.; Banwell, B.; Barkhof, F.; et al. Defining the clinical course of multiple sclerosis: The 2013 revisions. Neurology 2014, 83, 278–286. [Google Scholar] [CrossRef]
  107. Thompson, A.J.; Banwell, B.L.; Barkhof, F.; Carroll, W.M.; Coetzee, T.; Comi, G.; Correale, J.; Fazekas, F.; Filippi, M.; Freedman, M.S.; et al. Diagnosis of multiple sclerosis: 2017 revisions of the McDonald criteria. Lancet Neurol. 2018, 17, 162–173. [Google Scholar] [CrossRef]
  108. Brodin, P.; Jojic, V.; Gao, T.; Bhattacharya, S.; Angel, C.J.; Furman, D.; Shen-Orr, S.; Dekker, C.L.; Swan, G.E.; Butte, A.J.; et al. Variation in the human immune system is largely driven by non-heritable influences. Cell 2015, 160, 37–47. [Google Scholar] [CrossRef]
  109. International Multiple Sclerosis Genetics Consortium. Multiple sclerosis genomic map implicates peripheral immune cells and microglia in susceptibility. Science 2019, 365, eaav7188. [Google Scholar] [CrossRef] [PubMed]
  110. Goodin, D.S.; Khankhanian, P.; Gourraud, P.A.; Vince, N. The nature of genetic and environmental susceptibility to multiple sclerosis. PLoS ONE 2021, 16, e0246157. [Google Scholar] [CrossRef] [PubMed]
  111. Thacker, E.L.; Mirzaei, F.; Ascherio, A. Infectious mononucleosis and risk for multiple sclerosis: A meta-analysis. Ann. Neurol. 2006, 59, 499–503. [Google Scholar] [CrossRef] [PubMed]
  112. Levin, L.I.; Munger, K.L.; Rubertone, M.V.; Peck, C.A.; Lennette, E.T.; Spiegelman, D.; Ascherio, A. Temporal relationship between elevation of epstein-barr virus antibody titers and initial onset of neurological symptoms in multiple sclerosis. JAMA 2005, 293, 2496–2500. [Google Scholar] [CrossRef] [PubMed]
  113. Serafini, B.; Rosicarelli, B.; Franciotta, D.; Magliozzi, R.; Reynolds, R.; Cinque, P.; Andreoni, L.; Trivedi, P.; Salvetti, M.; Faggioni, A.; et al. Dysregulated Epstein-Barr virus infection in the multiple sclerosis brain. J. Exp. Med. 2007, 204, 2899–2912. [Google Scholar] [CrossRef] [PubMed]
  114. Moreno, M.A.; Or-Geva, N.; Aftab, B.T.; Khanna, R.; Croze, E.; Steinman, L.; Han, M.H. Molecular signature of Epstein-Barr virus infection in MS brain lesions. Neurol. Neuroimmunol. Neuroinflamm. 2018, 5, e466. [Google Scholar] [CrossRef]
  115. Angelini, D.F.; Serafini, B.; Piras, E.; Severa, M.; Coccia, E.M.; Rosicarelli, B.; Ruggieri, S.; Gasperini, C.; Buttari, F.; Centonze, D.; et al. Increased CD8+ T cell response to Epstein-Barr virus lytic antigens in the active phase of multiple sclerosis. PLoS Pathog. 2013, 9, e1003220. [Google Scholar] [CrossRef]
  116. Bjornevik, K.; Münz, C.; Cohen, J.I.; Ascherio, A. Epstein–Barr virus as a leading cause of multiple sclerosis: Mechanisms and implications. Nat. Rev. Neurol. 2023, 19, 160–171. [Google Scholar] [CrossRef]
  117. Lanz, T.V.; Brewer, R.C.; Ho, P.P.; Moon, J.S.; Jude, K.M.; Fernandez, D.; Fernandes, R.A.; Gomez, A.M.; Nadj, G.S.; Bartley, C.M.; et al. Clonally expanded B cells in multiple sclerosis bind EBV EBNA1 and GlialCAM. Nature 2022, 603, 321–327. [Google Scholar] [CrossRef]
  118. Bjornevik, K.; Cortese, M.; Healy, B.C.; Kuhle, J.; Mina, M.J.; Leng, Y.; Elledge, S.J.; Niebuhr, D.W.; Scher, A.I.; Munger, K.L.; et al. Longitudinal analysis reveals high prevalence of Epstein-Barr virus associated with multiple sclerosis. Science 2022, 375, 296–301. [Google Scholar] [CrossRef]
  119. Lopez-Siles, M.; Duncan, S.H.; Garcia-Gil, L.J.; Martinez-Medina, M. Faecalibacterium prausnitzii: From microbiology to diagnostics and prognostics. ISME J. 2017, 11, 841–852. [Google Scholar] [CrossRef]
  120. Zhou, X.; Baumann, R.; Gao, X.; Mendoza, M.; Singh, S.; Sand, I.K.; Xia, Z.; Cox, L.M.; Chitnis, T.; Yoon, H.; et al. Gut microbiome of multiple sclerosis patients and paired household healthy controls reveal associations with disease risk and course. Cell 2022, 185, 3467–3486. [Google Scholar] [CrossRef] [PubMed]
  121. Cox, L.M.; Maghzi, A.H.; Liu, S.; Tankou, S.K.; Dhang, F.H.; Willocq, V.; Song, A.; Wasén, C.; Tauhid, S.; Chu, R.; et al. Gut Microbiome in Progressive Multiple Sclerosis. Ann. Neurol. 2021, 89, 1195–1211. [Google Scholar] [CrossRef]
  122. Cantoni, C.; Lin, Q.; Dorsett, Y.; Ghezzi, L.; Liu, Z.; Pan, Y.; Chen, K.; Han, Y.; Li, Z.; Xiao, H.; et al. Alterations of host-gut microbiome interactions in multiple sclerosis. EBioMedicine 2022, 76, 103798. [Google Scholar] [CrossRef] [PubMed]
  123. Chen, J.; Chia, N.; Kalari, K.R.; Yao, J.Z.; Novotna, M.; Paz Soldan, M.M.; Luckey, D.H.; Marietta, E.V.; Jeraldo, P.R.; Chen, X.; et al. Multiple sclerosis patients have a distinct gut microbiota compared to healthy controls. Sci. Rep. 2016, 6, 28484. [Google Scholar] [CrossRef]
  124. Chen, T.; Noto, D.; Hoshino, Y.; Mizuno, M.; Miyake, S. Butyrate suppresses demyelination and enhances remyelination. J. Neuroinflammation 2019, 16, 165. [Google Scholar] [CrossRef] [PubMed]
  125. Zeng, Q.; Junli, G.; Liu, X.; Chen, C.; Sun, X.; Li, H.; Zhou, Y.; Cui, C.; Wang, Y.; Yang, Y.; et al. Gut dysbiosis and lack of short chain fatty acids in a Chinese cohort of patients with multiple sclerosis. Neurochem. Int. 2019, 129, 104468. [Google Scholar] [CrossRef] [PubMed]
  126. Trend, S.; Leffler, J.; Jones, A.P.; Cha, L.; Gorman, S.; Brown, D.A.; Breit, S.N.; Kermode, A.G.; French, M.A.; Ward, N.C.; et al. Associations of serum short-chain fatty acids with circulating immune cells and serum biomarkers in patients with multiple sclerosis. Sci. Rep. 2021, 11, 5244. [Google Scholar] [CrossRef]
  127. Castillo-Álvarez, F.; Pérez-Matute, P.; Oteo, J.A.; Marzo-Sola, M.E. The influence of interferon β-1b on gut microbiota composition in patients with multiple sclerosis. Neurologia 2021, 36, 495–503. [Google Scholar] [CrossRef]
  128. Castillo-Álvarez, F.; Marzo-Sola, M.E. Role of intestinal microbiota in the development of multiple sclerosis. Neurologia 2017, 32, 175–184. [Google Scholar] [CrossRef]
  129. Hollister, E.B.; Gao, C.; Versalovic, J. Compositional and Functional Features of the Gastrointestinal Microbiome and Their Effects on Human Health. Gastroenterology 2014, 146, 1449–1458. [Google Scholar] [CrossRef]
  130. 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]
  131. Berer, K.; Mues, M.; Koutrolos, M.; Rasbi, Z.A.; Boziki, M.; Johner, C.; Wekerle, H.; Krishnamoorthy, G. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature 2011, 479, 538–541. [Google Scholar] [CrossRef]
  132. 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]
  133. Tan, T.G.; Sefik, E.; Geva-Zatorsky, N.; Kua, L.; Naskar, D.; Teng, F.; Pasman, L.; Ortiz-Lopez, A.; Jupp, R.; Wu, H.J.; et al. Identifying species of symbiont bacteria from the human gut that, alone, can induce intestinal Th17 cells in mice. Proc. Natl. Acad. Sci. USA 2016, 113, E8141–E8150. [Google Scholar] [CrossRef] [PubMed]
  134. Wiechers, C.; Zou, M.; Galvez, E.; Beckstette, M.; Ebel, M.; Strowig, T.; Huehn, J.; Pezoldt, J. The microbiota is dispensable for the early stages of peripheral regulatory T cell induction within mesenteric lymph nodes. Cell. Mol. Immunol. 2021, 18, 1211–1221. [Google Scholar] [CrossRef] [PubMed]
  135. Fan, L.; Qi, Y.; Qu, S.; Chen, X.; Li, A.; Hendi, M.; Xu, C.; Wang, L.; Hou, T.; Si, J.; et al. B. adolescentis ameliorates chronic colitis by regulating Treg/Th2 response and gut microbiota remodeling. Gut Microbes 2021, 13, 1826746. [Google Scholar] [CrossRef] [PubMed]
  136. Woo, J.M.P.; Parks, C.G.; Jacobsen, S.; Costenbader, K.H.; Bernatsky, S. The role of environmental exposures and gene-environment interactions in the etiology of systemic lupus erythematous. J. Intern. Med. 2022, 291, 755–778. [Google Scholar] [CrossRef] [PubMed]
  137. Barbhaiya, M.; Costenbader, K.H. Environmental exposures and the development of systemic lupus erythematosus. Curr. Opin. Rheumatol. 2016, 28, 497–505. [Google Scholar] [CrossRef] [PubMed]
  138. Refai, R.H.; Hussein, M.F.; Abdou, M.H.; Abou-Raya, A.N. Environmental risk factors of systemic lupus erythematosus: A case–control study. Sci. Rep. 2023, 13, 10219. [Google Scholar] [CrossRef] [PubMed]
  139. Gulati, G.; Brunner, H.I. Environmental triggers in systemic lupus erythematosus. Semin. Arthritis Rheum. 2018, 47, 710–717. [Google Scholar] [CrossRef]
  140. Chen, L.; Wang, Y.F.; Liu, L.; Bielowka, A.; Ahmed, R.; Zhang, H.; Tombleson, P.; Roberts, A.L.; Odhams, C.A.; Cunninghame Graham, D.S.; et al. Genome-wide assessment of genetic risk for systemic lupus erythematosus and disease severity. Hum. Mol. Genet. 2020, 29, 1745–1756. [Google Scholar] [CrossRef]
  141. Reid, S.; Alexsson, A.; Frodlund, M.; Morris, D.; Sandling, J.K.; Bolin, K.; Svenungsson, E.; Jönsen, A.; Bengtsson, C.; Gunnarsson, I.; et al. High genetic risk score is associated with early disease onset, damage accrual and decreased survival in systemic lupus erythematosus. Ann. Rheum. Dis. 2020, 79, 363–369. [Google Scholar] [CrossRef]
  142. Langefeld, C.D.; Ainsworth, H.C.; Graham, D.S.C.; Kelly, J.A.; Comeau, M.E.; Marion, M.C.; Howard, T.D.; Ramos, P.S.; Croker, J.A.; Morris, D.L.; et al. Transancestral mapping and genetic load in systemic lupus erythematosus. Nat. Commun. 2017, 8, 16021. [Google Scholar] [CrossRef]
  143. Justiz Vaillant, A.A.; Goyal, A.; Varacallo, M. Systemic Lupus Erythematosus. In StatPearls; StatPearls Publishing LLC.: Treasure Island, FL, USA, 2023. [Google Scholar]
  144. Tian, J.; Zhang, D.; Yao, X.; Huang, Y.; Lu, Q. Global epidemiology of systemic lupus erythematosus: A comprehensive systematic analysis and modelling study. Ann. Rheum. Dis. 2023, 82, 351–356. [Google Scholar] [CrossRef]
  145. Wilson, P.C.; Kashgarian, M.; Moeckel, G. Interstitial inflammation and interstitial fibrosis and tubular atrophy predict renal survival in lupus nephritis. Clin. Kidney J. 2018, 11, 207–218. [Google Scholar] [CrossRef]
  146. Moulton, V.R. Sex Hormones in Acquired Immunity and Autoimmune Disease. Front. Immunol. 2018, 9, 2279. [Google Scholar] [CrossRef] [PubMed]
  147. Azzouz, D.; Omarbekova, A.; Heguy, A.; Schwudke, D.; Gisch, N.; Rovin, B.H.; Caricchio, R.; Buyon, J.P.; Alekseyenko, A.V.; Silverman, G.J. Lupus nephritis is linked to disease-activity associated expansions and immunity to a gut commensal. Ann. Rheum. Dis. 2019, 78, 947–956. [Google Scholar] [CrossRef] [PubMed]
  148. Hevia, A.; Milani, C.; López, P.; Cuervo, A.; Arboleya, S.; Duranti, S.; Turroni, F.; González, S.; Suárez, A.; Gueimonde, M.; et al. Intestinal dysbiosis associated with systemic lupus erythematosus. mBio 2014, 5, e01548-14. [Google Scholar] [CrossRef]
  149. Luo, X.M.; Edwards, M.R.; Mu, Q.; Yu, Y.; Vieson, M.D.; Reilly, C.M.; Ahmed, S.A.; Bankole, A.A. Gut Microbiota in Human Systemic Lupus Erythematosus and a Mouse Model of Lupus. Appl. Environ. Microbiol. 2018, 84, e02288-17. [Google Scholar] [CrossRef]
  150. Zhang, H.; Liao, X.; Sparks, J.B.; Luo, X.M. Dynamics of gut microbiota in autoimmune lupus. Appl. Environ. Microbiol. 2014, 80, 7551–7560. [Google Scholar] [CrossRef] [PubMed]
  151. Kim, D.S.; Park, Y.; Choi, J.W.; Park, S.H.; Cho, M.L.; Kwok, S.K. Lactobacillus acidophilus Supplementation Exerts a Synergistic Effect on Tacrolimus Efficacy by Modulating Th17/Treg Balance in Lupus-Prone Mice via the SIGNR3 Pathway. Front. Immunol. 2021, 12, 696074. [Google Scholar] [CrossRef]
  152. Zegarra-Ruiz, D.F.; El Beidaq, A.; Iñiguez, A.J.; Lubrano Di Ricco, M.; Manfredo Vieira, S.; Ruff, W.E.; Mubiru, D.; Fine, R.L.; Sterpka, J.; Greiling, T.M.; et al. A Diet-Sensitive Commensal Lactobacillus Strain Mediates TLR7-Dependent Systemic Autoimmunity. Cell Host Microbe 2019, 25, 113–127. [Google Scholar] [CrossRef] [PubMed]
  153. Wang, H.; Wang, G.; Banerjee, N.; Liang, Y.; Du, X.; Boor, P.J.; Hoffman, K.L.; Khan, M.F. Aberrant Gut Microbiome Contributes to Intestinal Oxidative Stress, Barrier Dysfunction, Inflammation and Systemic Autoimmune Responses in MRL/lpr Mice. Front. Immunol. 2021, 12, 651191. [Google Scholar] [CrossRef]
  154. Valiente, G.R.; Munir, A.; Hart, M.L.; Blough, P.; Wada, T.T.; Dalan, E.E.; Willis, W.L.; Wu, L.C.; Freud, A.G.; Jarjour, W.N. Gut dysbiosis is associated with acceleration of lupus nephritis. Sci. Rep. 2022, 12, 152. [Google Scholar] [CrossRef]
  155. Toumi, E.; Goutorbe, B.; Plauzolles, A.; Bonnet, M.; Mezouar, S.; Militello, M.; Mege, J.L.; Chiche, L.; Halfon, P. Gut microbiota in systemic lupus erythematosus patients and lupus mouse model: A cross species comparative analysis for biomarker discovery. Front. Immunol. 2022, 13, 943241. [Google Scholar] [CrossRef] [PubMed]
  156. He, Z.; Shao, T.; Li, H.; Xie, Z.; Wen, C. Alterations of the gut microbiome in Chinese patients with systemic lupus erythematosus. Gut Pathog. 2016, 8, 64. [Google Scholar] [CrossRef]
  157. Gerges, M.A.; Esmaeel, N.E.; Makram, W.K.; Sharaf, D.M.; Gebriel, M.G. Altered Profile of Fecal Microbiota in Newly Diagnosed Systemic Lupus Erythematosus Egyptian Patients. Int. J. Microbiol. 2021, 2021, 9934533. [Google Scholar] [CrossRef]
  158. Xiang, S.; Qu, Y.; Qian, S.; Wang, R.; Wang, Y.; Jin, Y.; Li, J.; Ding, X. Association between systemic lupus erythematosus and disruption of gut microbiota: A meta-analysis. Lupus Sci. Med. 2022, 9, e000599. [Google Scholar] [CrossRef] [PubMed]
  159. Whitacre, C.C. Sex differences in autoimmune disease. Nat. Immunol. 2001, 2, 777–780. [Google Scholar] [CrossRef]
  160. Conrad, N.; Misra, S.; Verbakel, J.Y.; Verbeke, G.; Molenberghs, G.; Taylor, P.N.; Mason, J.; Sattar, N.; McMurray, J.J.V.; McInnes, I.B.; et al. Incidence, prevalence, and co-occurrence of autoimmune disorders over time and by age, sex, and socioeconomic status: A population-based cohort study of 22 million individuals in the UK. Lancet 2023, 401, 1878–1890. [Google Scholar] [CrossRef]
  161. Orton, S.M.; Herrera, B.M.; Yee, I.M.; Valdar, W.; Ramagopalan, S.V.; Sadovnick, A.D.; Ebers, G.C. Sex ratio of multiple sclerosis in Canada: A longitudinal study. Lancet Neurol. 2006, 5, 932–936. [Google Scholar] [CrossRef]
  162. Roubinian, J.R.; Talal, N.; Greenspan, J.S.; Goodman, J.R.; Siiteri, P.K. Effect of castration and sex hormone treatment on survival, anti-nucleic acid antibodies, and glomerulonephritis in NZB/NZW F1 mice. J. Exp. Med. 1978, 147, 1568–1583. [Google Scholar] [CrossRef]
  163. Makino, S.; Kunimoto, K.; Muraoka, Y.; Katagiri, K. Effect of castration on the appearance of diabetes in NOD mouse. Jikken Dobutsu 1981, 30, 137–140. [Google Scholar] [CrossRef]
  164. Fox, H.S. Androgen treatment prevents diabetes in nonobese diabetic mice. J. Exp. Med. 1992, 175, 1409–1412. [Google Scholar] [CrossRef] [PubMed]
  165. Kanda, N.; Tsuchida, T.; Tamaki, K. Testosterone suppresses anti-DNA antibody production in peripheral blood mononuclear cells from patients with systemic lupus erythematosus. Arthritis Rheum. 1997, 40, 1703–1711. [Google Scholar] [CrossRef]
  166. Wu, D.; Ye, L.; Zhang, X.; Yin, M.; Guo, Y.; Zhou, J. Characteristics of steroid hormones in systemic lupus erythematosus revealed by GC/MS-based metabolic profiling. Front. Endocrinol. 2023, 14, 1164679. [Google Scholar] [CrossRef]
  167. Bove, R.; Musallam, A.; Healy, B.C.; Raghavan, K.; Glanz, B.I.; Bakshi, R.; Weiner, H.; De Jager, P.L.; Miller, K.K.; Chitnis, T. Low testosterone is associated with disability in men with multiple sclerosis. Mult. Scler. J. 2014, 20, 1584–1592. [Google Scholar] [CrossRef] [PubMed]
  168. Kurth, F.; Luders, E.; Sicotte, N.L.; Gaser, C.; Giesser, B.S.; Swerdloff, R.S.; Montag, M.J.; Voskuhl, R.R.; Mackenzie-Graham, A. Neuroprotective effects of testosterone treatment in men with multiple sclerosis. NeuroImage Clin. 2014, 4, 454–460. [Google Scholar] [CrossRef]
  169. Sicotte, N.L.; Giesser, B.S.; Tandon, V.; Klutch, R.; Steiner, B.; Drain, A.E.; Shattuck, D.W.; Hull, L.; Wang, H.-J.; Elashoff, R.M.; et al. Testosterone Treatment in Multiple Sclerosis: A Pilot Study. Arch. Neurol. 2007, 64, 683–688. [Google Scholar] [CrossRef] [PubMed]
  170. Sicotte, N.L.; Liva, S.M.; Klutch, R.; Pfeiffer, P.; Bouvier, S.; Odesa, S.; Wu, T.C.J.; Voskuhl, R.R. Treatment of multiple sclerosis with the pregnancy hormone estriol. Ann. Neurol. 2002, 52, 421–428. [Google Scholar] [CrossRef]
  171. Soldan, S.S.; Retuerto, A.I.A.; Sicotte, N.L.; Voskuhl, R.R. Immune Modulation in Multiple Sclerosis Patients Treated with the Pregnancy Hormone Estriol 1. J. Immunol. 2003, 171, 6267–6274. [Google Scholar] [CrossRef]
  172. Papenfuss, T.L.; Powell, N.D.; McClain, M.A.; Bedarf, A.; Singh, A.; Gienapp, I.E.; Shawler, T.; Whitacre, C.C. Estriol generates tolerogenic dendritic cells in vivo that protect against autoimmunity. J. Immunol. 2011, 186, 3346–3355. [Google Scholar] [CrossRef]
  173. Tang, C.; Hu, W. The role of Th17 and Treg cells in normal pregnancy and unexplained recurrent spontaneous abortion (URSA): New insights into immune mechanisms. Placenta 2023, 142, 18–26. [Google Scholar] [CrossRef]
  174. Khaw, Y.M.; Anwar, S.; Zhou, J.; Kawano, T.; Lin, P.C.; Otero, A.; Barakat, R.; Drnevich, J.; Takahashi, T.; Ko, C.J.; et al. Estrogen receptor alpha signaling in dendritic cells modulates autoimmune disease phenotype in mice. EMBO Rep. 2023, 24, e54228. [Google Scholar] [CrossRef]
  175. Yao, Y.; Cai, X.; Zhang, M.; Zhang, X.; Ren, F.; Zheng, Y.; Fei, W.; Zhao, M.; Zheng, C. PSTPIP2 regulates synovial macrophages polarization and dynamics via ERβ in the joint microenvironment. Arthritis Res. Ther. 2022, 24, 247. [Google Scholar] [CrossRef]
  176. Fernandez Lahore, G.; Förster, M.; Johannesson, M.; Sabatier, P.; Lönnblom, E.; Aoun, M.; He, Y.; Nandakumar, K.S.; Zubarev, R.A.; Holmdahl, R. Polymorphic estrogen receptor binding site causes Cd2-dependent sex bias in the susceptibility to autoimmune diseases. Nat. Commun. 2021, 12, 5565. [Google Scholar] [CrossRef]
  177. Mohammad, I.; Starskaia, I.; Nagy, T.; Guo, J.; Yatkin, E.; Väänänen, K.; Watford, W.T.; Chen, Z. Estrogen receptor α contributes to T cell–mediated autoimmune inflammation by promoting T cell activation and proliferation. Sci. Signal. 2018, 11, eaap9415. [Google Scholar] [CrossRef] [PubMed]
  178. Khan, D.; Ansar Ahmed, S. The Immune System Is a Natural Target for Estrogen Action: Opposing Effects of Estrogen in Two Prototypical Autoimmune Diseases. Front. Immunol. 2015, 6, 635. [Google Scholar] [CrossRef] [PubMed]
  179. Grossman, C.J.; Sholiton, L.J.; Blaha, G.C.; Nathan, P. Rat thymic estrogen receptor—II. Physiological properties. J. Steroid Biochem. 1979, 11, 1241–1246. [Google Scholar] [CrossRef] [PubMed]
  180. Seiki, K.; Sakabe, K. Sex Hormones and the Thymus in Relation to Thymocyte Proliferation and Maturation. Arch. Histol. Cytol. 1997, 60, 29–38. [Google Scholar] [CrossRef] [PubMed]
  181. Ishibashi, H.; Suzuki, T.; Suzuki, S.; Moriya, T.; Kaneko, C.; Takizawa, T.; Sunamori, M.; Handa, M.; Kondo, T.; Sasano, H. Sex steroid hormone receptors in human thymoma. J. Clin. Endocrinol. Metab. 2003, 88, 2309–2317. [Google Scholar] [CrossRef] [PubMed]
  182. Mor, G.I.L.; MuÑOz, A.; Redlinger Jr, R.; Silva, I.; Song, J.; Lim, C.; Kohen, F. The Role of the Fas/Fas Ligand System in Estrogen-Induced Thymic Alteration. Am. J. Reprod. Immunol. 2001, 46, 298–307. [Google Scholar] [CrossRef] [PubMed]
  183. Okasha, S.A.; Ryu, S.; Do, Y.; McKallip, R.J.; Nagarkatti, M.; Nagarkatti, P.S. Evidence for estradiol-induced apoptosis and dysregulated T cell maturation in the thymus. Toxicology 2001, 163, 49–62. [Google Scholar] [CrossRef] [PubMed]
  184. Do, Y.; Ryu, S.; Nagarkatti, M.; Nagarkatti, P.S. Role of death receptor pathway in estradiol-induced T-cell apoptosis in vivo. Toxicol. Sci. 2002, 70, 63–72. [Google Scholar] [CrossRef] [PubMed]
  185. Okuyama, R.; Abo, T.; Seki, S.; Ohteki, T.; Sugiura, K.; Kusumi, A.; Kumagai, K. Estrogen administration activates extrathymic T cell differentiation in the liver. J. Exp. Med. 1992, 175, 661–669. [Google Scholar] [CrossRef] [PubMed]
  186. Grimaldi, C.M.; Cleary, J.; Dagtas, A.S.; Moussai, D.; Diamond, B. Estrogen alters thresholds for B cell apoptosis and activation. J. Clin. Investig. 2002, 109, 1625–1633. [Google Scholar] [CrossRef] [PubMed]
  187. Smithson, G.; Couse, J.F.; Lubahn, D.B.; Korach, K.S.; Kincade, P.W. The role of estrogen receptors and androgen receptors in sex steroid regulation of B lymphopoiesis. J. Immunol. 1998, 161, 27–34. [Google Scholar] [CrossRef] [PubMed]
  188. Eilat, D.; Wabl, M. B cell tolerance and positive selection in lupus. J. Immunol. 2012, 189, 503–509. [Google Scholar] [CrossRef] [PubMed]
  189. Lehmann-Horn, K.; Kinzel, S.; Weber, M.S. Deciphering the Role of B Cells in Multiple Sclerosis—Towards Specific Targeting of Pathogenic Function. Int. J. Mol. Sci. 2017, 18, 2048. [Google Scholar] [CrossRef]
  190. Dragin, N.; Bismuth, J.; Cizeron-Clairac, G.; Biferi, M.G.; Berthault, C.; Serraf, A.; Nottin, R.; Klatzmann, D.; Cumano, A.; Barkats, M.; et al. Estrogen-mediated downregulation of AIRE influences sexual dimorphism in autoimmune diseases. J. Clin. Investig. 2016, 126, 1525–1537. [Google Scholar] [CrossRef]
  191. Zhu, M.-L.; Bakhru, P.; Conley, B.; Nelson, J.S.; Free, M.; Martin, A.; Starmer, J.; Wilson, E.M.; Su, M.A. Sex bias in CNS autoimmune disease mediated by androgen control of autoimmune regulator. Nat. Commun. 2016, 7, 11350. [Google Scholar] [CrossRef]
  192. Imperato-McGinley, J.; Peterson, R.E.; Gautier, T.; Sturla, E. Androgens and the evolution of male-gender identity among male pseudohermaphrodites with 5alpha-reductase deficiency. N. Engl. J. Med. 1979, 300, 1233–1237. [Google Scholar] [CrossRef] [PubMed]
  193. Ben-Batalla, I.; Vargas-Delgado, M.E.; von Amsberg, G.; Janning, M.; Loges, S. Influence of Androgens on Immunity to Self and Foreign: Effects on Immunity and Cancer. Front. Immunol. 2020, 11, 1184. [Google Scholar] [CrossRef] [PubMed]
  194. Gubbels Bupp, M.R.; Jorgensen, T.N. Androgen-Induced Immunosuppression. Front. Immunol. 2018, 9, 794. [Google Scholar] [CrossRef] [PubMed]
  195. Sakiani, S.; Olsen, N.J.; Kovacs, W.J. Gonadal steroids and humoral immunity. Nat. Rev. Endocrinol. 2013, 9, 56–62. [Google Scholar] [CrossRef] [PubMed]
  196. Cutolo, M. Androgens in rheumatoid arthritis: When are they effectors? Arthritis Res. Ther. 2009, 11, 126. [Google Scholar] [CrossRef] [PubMed]
  197. Cutolo, M.; Seriolo, B.; Villaggio, B.; Pizzorni, C.; Craviotto, C.; Sulli, A. Androgens and estrogens modulate the immune and inflammatory responses in rheumatoid arthritis. Ann. N. Y. Acad. Sci. 2002, 966, 131–142. [Google Scholar] [CrossRef] [PubMed]
  198. Gupta, P.K.; Sheoran, A.; Gupta, P.; Mahto, S.K.; Jain, P.; Varshney, A.K.; Sharma, L.K. Association of Sex Hormones and Androgens with Disease Activity in Premenopausal Females with Rheumatoid Arthritis. Mediterr. J. Rheumatol. 2023, 34, 152–158. [Google Scholar] [CrossRef] [PubMed]
  199. Kawasaki, T.; Ushiyama, T.; Inoue, K.; Hukuda, S. Effects of estrogen on interleukin-6 production in rheumatoid fibroblast-like synoviocytes. Clin. Exp. Rheumatol. 2000, 18, 743–745. [Google Scholar]
  200. Macdiarmid, F.; Wang, D.; Duncan, L.J.; Purohit, A.; Ghilchick, M.W.; Reed, M.J. Stimulation of aromatase activity in breast fibroblasts by tumor necrosis factor alpha. Mol. Cell Endocrinol. 1994, 106, 17–21. [Google Scholar] [CrossRef]
  201. Kondo, N.; Kuroda, T.; Kobayashi, D. Cytokine Networks in the Pathogenesis of Rheumatoid Arthritis. Int. J. Mol. Sci. 2021, 22, 10922. [Google Scholar] [CrossRef]
  202. Yang, L.; Zhou, R.; Tong, Y.; Chen, P.; Shen, Y.; Miao, S.; Liu, X. Neuroprotection by dihydrotestosterone in LPS-induced neuroinflammation. Neurobiol. Dis. 2020, 140, 104814. [Google Scholar] [CrossRef]
  203. Yilmaz, C.; Karali, K.; Fodelianaki, G.; Gravanis, A.; Chavakis, T.; Charalampopoulos, I.; Alexaki, V.I. Neurosteroids as regulators of neuroinflammation. Front. Neuroendocrinol. 2019, 55, 100788. [Google Scholar] [CrossRef]
  204. Boghozian, R.; McKenzie, B.A.; Saito, L.B.; Mehta, N.; Branton, W.G.; Lu, J.; Baker, G.B.; Noorbakhsh, F.; Power, C. Suppressed oligodendrocyte steroidogenesis in multiple sclerosis: Implications for regulation of neuroinflammation. Glia 2017, 65, 1590–1606. [Google Scholar] [CrossRef]
  205. Lyon, M.F. Gene action in the X-chromosome of the mouse (Mus musculus L.). Nature 1961, 190, 372–373. [Google Scholar] [CrossRef] [PubMed]
  206. Prchal, J.T.; Prchal, J.F.; Belickova, M.; Chen, S.; Guan, Y.; Gartland, G.L.; Cooper, M.D. Clonal stability of blood cell lineages indicated by X-chromosomal transcriptional polymorphism. J. Exp. Med. 1996, 183, 561–567. [Google Scholar] [CrossRef] [PubMed]
  207. Ozcelik, T.; Uz, E.; Akyerli, C.B.; Bagislar, S.; Mustafa, C.A.; Gursoy, A.; Akarsu, N.; Toruner, G.; Kamel, N.; Gullu, S. Evidence from autoimmune thyroiditis of skewed X-chromosome inactivation in female predisposition to autoimmunity. Eur. J. Hum. Genet. 2006, 14, 791–797. [Google Scholar] [CrossRef]
  208. Flores, R.; Shi, J.; Fuhrman, B.; Xu, X.; Veenstra, T.D.; Gail, M.H.; Gajer, P.; Ravel, J.; Goedert, J.J. Fecal microbial determinants of fecal and systemic estrogens and estrogen metabolites: A cross-sectional study. J. Transl. Med. 2012, 10, 253. [Google Scholar] [CrossRef]
  209. Maynard, C.L.; Elson, C.O.; Hatton, R.D.; Weaver, C.T. Reciprocal interactions of the intestinal microbiota and immune system. Nature 2012, 489, 231–241. [Google Scholar] [CrossRef]
  210. Adlercreutz, H.; Pulkkinen, M.O.; Hämäläinen, E.K.; Korpela, J.T. Studies on the role of intestinal bacteria in metabolism of synthetic and natural steroid hormones. J. Steroid Biochem. 1984, 20, 217–229. [Google Scholar] [CrossRef]
  211. Candeliere, F.; Raimondi, S.; Ranieri, R.; Musmeci, E.; Zambon, A.; Amaretti, A.; Rossi, M. β-Glucuronidase Pattern Predicted From Gut Metagenomes Indicates Potentially Diversified Pharmacomicrobiomics. Front. Microbiol. 2022, 13, 826994. [Google Scholar] [CrossRef]
  212. Ervin, S.M.; Li, H.; Lim, L.; Roberts, L.R.; Liang, X.; Mani, S.; Redinbo, M.R. Gut microbial β-glucuronidases reactivate estrogens as components of the estrobolome that reactivate estrogens. J. Biol. Chem. 2019, 294, 18586–18599. [Google Scholar] [CrossRef]
  213. Biernat, K.A.; Pellock, S.J.; Bhatt, A.P.; Bivins, M.M.; Walton, W.G.; Tran, B.N.T.; Wei, L.; Snider, M.C.; Cesmat, A.P.; Tripathy, A.; et al. Structure, function, and inhibition of drug reactivating human gut microbial β-glucuronidases. Sci. Rep. 2019, 9, 825. [Google Scholar] [CrossRef]
  214. Gruber, C.J.; Tschugguel, W.; Schneeberger, C.; Huber, J.C. Production and actions of estrogens. N. Engl. J. Med. 2002, 346, 340–352. [Google Scholar] [CrossRef] [PubMed]
  215. Domínguez, R.; Zhang, L.; Rocchetti, G.; Lucini, L.; Pateiro, M.; Munekata, P.E.S.; Lorenzo, J.M. Elderberry (Sambucus nigra L.) as potential source of antioxidants: Characterization, optimization of extraction parameters and bioactive properties. Food Chem. 2020, 330, 127266. [Google Scholar] [CrossRef]
  216. Nakatsu, C.H.; Armstrong, A.; Clavijo, A.P.; Martin, B.R.; Barnes, S.; Weaver, C.M. Fecal bacterial community changes associated with isoflavone metabolites in postmenopausal women after soy bar consumption. PLoS One 2014, 9, e108924. [Google Scholar] [CrossRef]
  217. Soukup, S.T.; Stoll, D.A.; Danylec, N.; Schoepf, A.; Kulling, S.E.; Huch, M. Metabolism of Daidzein and Genistein by Gut Bacteria of the Class Coriobacteriia. Foods 2021, 10, 2741. [Google Scholar] [CrossRef]
  218. Rietjens, I.; Louisse, J.; Beekmann, K. The potential health effects of dietary phytoestrogens. Br. J. Pharmacol. 2017, 174, 1263–1280. [Google Scholar] [CrossRef] [PubMed]
  219. Kavlock, R.J.; Daston, G.P.; DeRosa, C.; Fenner-Crisp, P.; Gray, L.E.; Kaattari, S.; Lucier, G.; Luster, M.; Mac, M.J.; Maczka, C.; et al. Research needs for the risk assessment of health and environmental effects of endocrine disruptors: A report of the U.S. EPA-sponsored workshop. Environ. Health Perspect. 1996, 104 (Suppl. 4), 715–740. [Google Scholar] [CrossRef]
  220. Kabir, E.R.; Rahman, M.S.; Rahman, I. A review on endocrine disruptors and their possible impacts on human health. Environ. Toxicol. Pharmacol. 2015, 40, 241–258. [Google Scholar] [CrossRef]
  221. Senizza, A.; Rocchetti, G.; Mosele, J.I.; Patrone, V.; Callegari, M.L.; Morelli, L.; Lucini, L. Lignans and Gut Microbiota: An Interplay Revealing Potential Health Implications. Molecules 2020, 25, 5709. [Google Scholar] [CrossRef]
  222. Roncaglia, L.; Amaretti, A.; Raimondi, S.; Leonardi, A.; Rossi, M. Role of bifidobacteria in the activation of the lignan secoisolariciresinol diglucoside. Appl. Microbiol. Biotechnol. 2011, 92, 159–168. [Google Scholar] [CrossRef]
  223. Struijs, K.; Vincken, J.P.; Gruppen, H. Bacterial conversion of secoisolariciresinol and anhydrosecoisolariciresinol. J. Appl. Microbiol. 2009, 107, 308–317. [Google Scholar] [CrossRef]
  224. Colldén, H.; Landin, A.; Wallenius, V.; Elebring, E.; Fändriks, L.; Nilsson, M.E.; Ryberg, H.; Poutanen, M.; Sjögren, K.; Vandenput, L.; et al. The gut microbiota is a major regulator of androgen metabolism in intestinal contents. Am. J. Physiol. Endocrinol. Metab. 2019, 317, E1182–E1192. [Google Scholar] [CrossRef]
  225. Doden, H.L.; Ridlon, J.M. Microbial Hydroxysteroid Dehydrogenases: From Alpha to Omega. Microorganisms 2021, 9, 469. [Google Scholar] [CrossRef]
  226. Kisiela, M.; Skarka, A.; Ebert, B.; Maser, E. Hydroxysteroid dehydrogenases (HSDs) in bacteria—A bioinformatic perspective. J. Steroid Biochem. Mol. Biol. 2012, 129, 31–46. [Google Scholar] [CrossRef]
  227. Ang, Z.; Xiong, D.; Wu, M.; Ding, J.L. FFAR2-FFAR3 receptor heteromerization modulates short-chain fatty acid sensing. FASEB J. 2018, 32, 289–303. [Google Scholar] [CrossRef]
  228. Kimura, I.; Ichimura, A.; Ohue-Kitano, R.; Igarashi, M. Free Fatty Acid Receptors in Health and Disease. Physiol. Rev. 2020, 100, 171–210. [Google Scholar] [CrossRef]
  229. Martinelli, S.; Nannini, G.; Cianchi, F.; Staderini, F.; Coratti, F.; Amedei, A. Microbiota Transplant and Gynecological Disorders: The Bridge between Present and Future Treatments. Microorganisms 2023, 11, 2407. [Google Scholar] [CrossRef]
  230. Wallace, B.D.; Wang, H.; Lane, K.T.; Scott, J.E.; Orans, J.; Koo, J.S.; Venkatesh, M.; Jobin, C.; Yeh, L.A.; Mani, S.; et al. Alleviating cancer drug toxicity by inhibiting a bacterial enzyme. Science 2010, 330, 831–835. [Google Scholar] [CrossRef]
  231. Wallace, B.D.; Redinbo, M.R. The human microbiome is a source of therapeutic drug targets. Curr. Opin. Chem. Biol. 2013, 17, 379–384. [Google Scholar] [CrossRef]
  232. Gulati, A.S.; Nicholson, M.R.; Khoruts, A.; Kahn, S.A. Fecal Microbiota Transplantation Across the Lifespan: Balancing Efficacy, Safety, and Innovation. Am. J. Gastroenterol. 2023, 118, 435–439. [Google Scholar] [CrossRef] [PubMed]
  233. Mandrioli, J.; Amedei, A.; Cammarota, G.; Niccolai, E.; Zucchi, E.; D’Amico, R.; Ricci, F.; Quaranta, G.; Spanu, T.; Masucci, L. FETR-ALS Study Protocol: A Randomized Clinical Trial of Fecal Microbiota Transplantation in Amyotrophic Lateral Sclerosis. Front. Neurol. 2019, 10, 1021. [Google Scholar] [CrossRef]
  234. Available online: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/enforcement-policy-regarding-investigational-new-drug-requirements-use-fecal-microbiota (accessed on 1 January 2024).
  235. 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]
  236. Zeng, J.; Peng, L.; Zheng, W.; Huang, F.; Zhang, N.; Wu, D.; Yang, Y. Fecal microbiota transplantation for rheumatoid arthritis: A case report. In Clinical Case Report; John Wiley & Sons Ltd.: Chichester, UK, 2021; Volume 9, pp. 906–909. [Google Scholar]
  237. Choi, S.C.; Brown, J.; Gong, M.; Ge, Y.; Zadeh, M.; Li, W.; Croker, B.P.; Michailidis, G.; Garrett, T.J.; Mohamadzadeh, M.; et al. Gut microbiota dysbiosis and altered tryptophan catabolism contribute to autoimmunity in lupus-susceptible mice. Sci. Transl. Med. 2020, 12, eaax2220. [Google Scholar] [CrossRef] [PubMed]
  238. Ma, Y.; Xu, X.; Li, M.; Cai, J.; Wei, Q.; Niu, H. Gut microbiota promote the inflammatory response in the pathogenesis of systemic lupus erythematosus. Mol. Med. 2019, 25, 35. [Google Scholar] [CrossRef] [PubMed]
  239. de la Visitación, N.; Robles-Vera, I.; Toral, M.; Gómez-Guzmán, M.; Sánchez, M.; Moleón, J.; González-Correa, C.; Martín-Morales, N.; O’Valle, F.; Jiménez, R.; et al. Gut microbiota contributes to the development of hypertension in a genetic mouse model of systemic lupus erythematosus. Br. J. Pharmacol. 2021, 178, 3708–3729. [Google Scholar] [CrossRef] [PubMed]
  240. Huang, C.; Yi, P.; Zhu, M.; Zhou, W.; Zhang, B.; Yi, X.; Long, H.; Zhang, G.; Wu, H.; Tsokos, G.C.; et al. Safety and efficacy of fecal microbiota transplantation for treatment of systemic lupus erythematosus: An EXPLORER trial. J. Autoimmun. 2022, 130, 102844. [Google Scholar] [CrossRef]
  241. Ying, Z.H.; Mao, C.L.; Xie, W.; Yu, C.H. Postbiotics in rheumatoid arthritis: Emerging mechanisms and intervention perspectives. Front. Microbiol. 2023, 14, 1290015. [Google Scholar] [CrossRef]
  242. Ravi, A.K.; Muthukrishnan, S.K. Combination of Probiotics and Natural Compounds to Treat Multiple Sclerosis via Warburg Effect. Adv. Pharm. Bull. 2022, 12, 515–523. [Google Scholar] [CrossRef]
  243. Quaranta, G.; Ianiro, G.; De Maio, F.; Guarnaccia, A.; Fancello, G.; Agrillo, C.; Iannarelli, F.; Bibbo, S.; Amedei, A.; Sanguinetti, M.; et al. “Bacterial Consortium”: A Potential Evolution of Fecal Microbiota Transplantation for the Treatment of. BioMed Res. Int. 2022, 2022, 5787373. [Google Scholar] [CrossRef]
  244. Louie, T.; Golan, Y.; Khanna, S.; Bobilev, D.; Erpelding, N.; Fratazzi, C.; Carini, M.; Menon, R.; Ruisi, M.; Norman, J.M.; et al. VE303, a Defined Bacterial Consortium, for Prevention of Recurrent Clostridioides difficile Infection: A Randomized Clinical Trial. JAMA 2023, 329, 1356–1366. [Google Scholar] [CrossRef] [PubMed]
  245. Plaza-Diaz, J.; Gomez-Llorente, C.; Fontana, L.; Gil, A. Modulation of immunity and inflammatory gene expression in the gut, in inflammatory diseases of the gut and in the liver by probiotics. World J. Gastroenterol. 2014, 20, 15632–15649. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Infections can trigger or exacerbate autoimmune diseases through several mechanisms, leading to autoimmunity induction. (A) Molecular mimicry is the mechanism by which infectious antigens similar to self-molecules and presented by APCs can trigger T autoreactive cells, leading to the development of autoimmune diseases. (B) Bystander activation refers to the way in which over-reactive antiviral immune responses lead to the release of self-antigens and inflammatory cytokines from damaged tissue. Autoreactive T cells are then activated by APCs. (C) The epitope spreading model predicts that a persistent infection induces tissue damage and release of new self-antigens that are presented by APCs. Nonspecific triggering of several autoreactive T cells can lead to autoimmunity. APC = antigen-presenting cell.
Figure 1. Infections can trigger or exacerbate autoimmune diseases through several mechanisms, leading to autoimmunity induction. (A) Molecular mimicry is the mechanism by which infectious antigens similar to self-molecules and presented by APCs can trigger T autoreactive cells, leading to the development of autoimmune diseases. (B) Bystander activation refers to the way in which over-reactive antiviral immune responses lead to the release of self-antigens and inflammatory cytokines from damaged tissue. Autoreactive T cells are then activated by APCs. (C) The epitope spreading model predicts that a persistent infection induces tissue damage and release of new self-antigens that are presented by APCs. Nonspecific triggering of several autoreactive T cells can lead to autoimmunity. APC = antigen-presenting cell.
Biomedicines 12 00616 g001
Figure 2. Schematization of estrogen (E2) actions on B (green) and T (red) cells. There are several actions of estrogens on B cells such as the increase in cell number progenitors in the bone marrow, the enhanced survival in the spleen, and the induction of antibody production. Regarding E2’s effects on T cells, the promotion of cell activation, proliferation, survival, and differentiation have been described in Th-1 subtype. Extrathymic cell differentiation in the liver was observed. All these features could lead to the predisposition to autoimmunity and disease development when an imbalance occurs.
Figure 2. Schematization of estrogen (E2) actions on B (green) and T (red) cells. There are several actions of estrogens on B cells such as the increase in cell number progenitors in the bone marrow, the enhanced survival in the spleen, and the induction of antibody production. Regarding E2’s effects on T cells, the promotion of cell activation, proliferation, survival, and differentiation have been described in Th-1 subtype. Extrathymic cell differentiation in the liver was observed. All these features could lead to the predisposition to autoimmunity and disease development when an imbalance occurs.
Biomedicines 12 00616 g002
Table 2. Clinical trials involving FMT in multiple sclerosis, rheumatoid arthritis, and systemic lupus erythematosus.
Table 2. Clinical trials involving FMT in multiple sclerosis, rheumatoid arthritis, and systemic lupus erythematosus.
DiseaseStudy TitleClinical Trial IDStatus of the StudyStudy Start
Multiple SclerosisFecal Microbial Transplantation in Relapsing Multiple Sclerosis PatientsNCT03183869Terminated with results24 August 2017
Multiple SclerosisFecal Microbiota Transplantation After Autologous HSCT in Patients with Multiple SclerosisNCT04203017Terminated because of corrupted biological samples 6 December 2023
Multiple SclerosisFecal Microbiota Transplantation (FMT) in Multiple SclerosisNCT03975413Completed8 October 2020
Multiple SclerosisSafety and Efficacy of Fecal Microbiota TransplantationNCT04014413Active, recruiting30 May 2023
Multiple SclerosisFecal Microbiota Transplantation (FMT) of FMP30 in Relapsing–Remitting Multiple Sclerosis (MS-BIOME)NCT03594487Active, not recruiting3 July 2023
Rheumatoid Arthritis Efficacy and Safety of Fecal Microbiota Transplantation in Patients With Rheumatoid Arthritis Refractory to Methotrexate (FARM)NCT03944096Unknown status30 April 2019
Systemic Lupus ErythematosusSafety and Efficacy of Fecal Microbiota Transplantation for Treatment of Systemic Lupus Erythematosus: An EXPLORER TrialChiCTR2000036352Completed22 August 2020
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

Martinelli, S.; Nannini, G.; Cianchi, F.; Coratti, F.; Amedei, A. The Impact of Microbiota–Immunity–Hormone Interactions on Autoimmune Diseases and Infection. Biomedicines 2024, 12, 616. https://doi.org/10.3390/biomedicines12030616

AMA Style

Martinelli S, Nannini G, Cianchi F, Coratti F, Amedei A. The Impact of Microbiota–Immunity–Hormone Interactions on Autoimmune Diseases and Infection. Biomedicines. 2024; 12(3):616. https://doi.org/10.3390/biomedicines12030616

Chicago/Turabian Style

Martinelli, Serena, Giulia Nannini, Fabio Cianchi, Francesco Coratti, and Amedeo Amedei. 2024. "The Impact of Microbiota–Immunity–Hormone Interactions on Autoimmune Diseases and Infection" Biomedicines 12, no. 3: 616. https://doi.org/10.3390/biomedicines12030616

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