Next Article in Journal
Marginal Bone Loss Compared in Internal and External Implant Connections: Retrospective Clinical Study at 6-Years Follow-Up
Next Article in Special Issue
Cardiac Resynchronization Therapy and Left Atrial Remodeling: A Novel Insight?
Previous Article in Journal
Exploring the Complex Relationship between Diabetes and Cardiovascular Complications: Understanding Diabetic Cardiomyopathy and Promising Therapies
Previous Article in Special Issue
Measurement of Aβ Amyloid Plaques and Tau Protein in Postmortem Human Alzheimer’s Disease Brain by Autoradiography Using [18F]Flotaza, [125I]IBETA, [124/125I]IPPI and Immunohistochemistry Analysis Using QuPath
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genes and Microbiota Interaction in Monogenic Autoimmune Disorders

1
Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy
2
Maggiore della Carità University Hospital, 28100 Novara, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2023, 11(4), 1127; https://doi.org/10.3390/biomedicines11041127
Submission received: 10 March 2023 / Revised: 30 March 2023 / Accepted: 5 April 2023 / Published: 8 April 2023

Abstract

:
Monogenic autoimmune disorders represent an important tool to understand the mechanisms behind central and peripheral immune tolerance. Multiple factors, both genetic and environmental, are known to be involved in the alteration of the immune activation/immune tolerance homeostasis typical of these disorders, making it difficult to control the disease. The latest advances in genetic analysis have contributed to a better and more rapid diagnosis, although the management remains confined to the treatment of clinical manifestations, as there are limited studies on rare diseases. Recently, the correlation between microbiota composition and the onset of autoimmune disorders has been investigated, thus opening up new perspectives on the cure of monogenic autoimmune diseases. In this review, we will summarize the main genetic features of both organ-specific and systemic monogenic autoimmune diseases, reporting on the available literature data on microbiota alterations in these patients.

1. Introduction

A breach of immunological tolerance is defined as the loss of the ability of our immune system to distinguish self from non-self, and it is the basis for autoimmune disease. Autoimmune diseases are believed to be mainly triggered by infections through several mechanism, such as molecular mimicry, the release of sequestered autoantigens, or the induction of inflammation (adjuvant effect); however, multiple factors, both genetic and environmental (nutrition, microbiota, and xenobiotics), can favor this breakdown, making difficult to determine the exact pathogenesis of the disease [1].
Epidemiologic studies have demonstrated that genetic factors are crucial determinants of susceptibility to autoimmune disease. Commonly, autoimmune disorders involve multiple genes, each of them exerting a minimal predisposing effect; however, rare monogenic autoimmune diseases have been identified, allowing for a better understanding of the mechanisms behind central and peripheral immune tolerance and suggesting new potential therapeutic strategies. Most of the genes causing these monogenic autoimmune diseases can alter the key mechanisms of tolerance, such as the negative selection of autoreactive T cells in the thymus or T cell apoptosis involved in switching off the immune response, or defects of T regulatory cell development or functions [1,2,3].
A contribution can also be provided by certain drugs, such as procainamide and hydralazine, by reducing DNA methylation in T cells and increasing the expression of the genes that are involved in lymphocyte activation, such as lymphocyte function-associated antigen (LFA-1), thus favoring autoimmunity; furthermore, some treatments can also alter the development of central thymic tolerance, causing the escape of autoreactive lymphocytes in the periphery [1].
Autoimmune disorders can be either organ-specific or systemic, and they may involve both adaptive and innate immune response mechanisms; although, in some cases, these differences are not so well defined.
The altered immune activation leads to tissue destruction through several mechanisms, such as the production of autoantibodies directed against surface molecules, nucleic acid, and other cellular components, which leads to cytotoxicity through antibody-dependent cell-mediated cytotoxicity (ADCC) and complements activation mechanisms, which recruit inflammatory cells, thus causing tissue damage [1]. Alternatively, autoreactive cytotoxic and helper T cells may recognize the autoantigen-derived peptides that are expressed in the target tissue and consequently induce tissue damage through the release of cytotoxic granules or the production of inflammatory cytokines (organ-specific autoimmunity). However, the role of the different types of autoreactive cells in systemic autoimmune disease is still unclear [1].
This review will focus on some of these rare monogenic diseases, such as autoimmune polyglandular syndrome (APS1), immunedysregulation polyendocrinopathy enteropathy X-linked syndrome (IPEX), autoimmune lymphoproliferative syndrome (ALPS), and monogenic systemic lupus erythematosus (SLE). In addition, we will discuss the involvement of microbiota in the pathophysiology of these monogenic diseases and the role of vitamin D in the interplay between microbiota and the onset of autoimmunity.

2. Microbiota and the Immune System

2.1. The Close Relationship between Microbiota and Immunity

Thousands of different species of microorganisms colonize the gut, the skin, and other mucosal environments in the human body. It is now known that the human microbiome plays an important role in multiple physiological functions, including metabolism, circadian rhythmicity, and shaping the immune system, contributing to health and disease [4]. In particular, microbiota promote both humoral immunity (B cell development and proinflammatory T cell responses) and immune regulation (regulatory B and T cells) [5]. A close relationship establishes between the microbiota and the immune system that continuously evolves in order to preserve the symbiosis and allows the induction of appropriate responses to pathogens and a tolerance to safe agents [5]. The first interaction between the immune system and the microbiota starts in early life, during the passage through the birth canal. Live microbes, metabolites, cells, cytokines, and immunoglobulin (Ig) A contained in maternal milk will then shape the neonatal microbiota, promoting the expansion of specific components, such as Bifidobacterium, or restricting immune activation against commensal antigens [6]. The establishment of microbiota is favored by an immature regulatory environment of the developing immune system. On the other hand, commensals influence early-life immunity through different mechanisms. For example, the suppression of invariant natural killer T (iNKT) cells that are involved in the inflammatory response occurs because of the interaction with inhibitory sphingolipids derived from commensals [7].
The homeostatic host/commensal relationship is fundamental and is guaranteed by the large subsets of immune cells that are resident in the sites that are colonized by commensals, such as the skin and the gastrointestinal (GI) tract. Here, the contact between microorganisms and the epithelial cell surface is minimized by mucus and the cell surface glycocalyx in order to avoid tissue inflammation and microbe translocation [8,9]. The induction of commensal-specific T regulatory cells (Tregs) preferentially occurs in GI in order to maintain the tolerance to commensal antigens and other oral-intake antigens [10]. Hence, commensals induce lamina propria resident, not inflammatory macrophages, and the local expansion of Tregs, which further promote IgA class-switching in an antigen-specific manner [11].
Interestingly, commensal-derived molecules, such as the polysaccharide A (PSA) produced by Bacteroides fragilis, could also induce regulatory responses. Specifically, PSA induces IL-10-producing Tregs, through the engagement of toll-like receptor (TLR) 2 on T cells, which further inhibits T helper (Th) 17 activity [12]. Besides the TLRs, additional pattern-recognition receptors (PRRs) alter the gut microbiota, such as NOD-like receptor (NLR) 2, which avoids small intestine inflammation by inhibiting the growth of the commensal Bacteroides vulgatus [13]. Some of the NLRs are part of multiprotein complexes, termed inflammasomes, such as for NLRP6, whose impact on the microbiota depends on the configuration of the microbiome itself, and its inflammation signaling, which affects IL-18 production by epithelial cells and, in turn, the antimicrobial peptide (AMP) expression profiles, which are influenced by microbiota-derived metabolites such as taurine, histamine, and spermine [14]. Accordingly, mouse models have shown that commensal metabolites coming from the fermentation of indigestible fibers, such as short-chain fatty acids (SCFA), can directly affect the activity of immune cells, contributing to arthritis, allergy [15], and the extrathymic generation of Tregs [16]. In addition, SCFAs promote the memory potential of antigen-activated CD8+ cells [17].
Commensal microbes control the production of cytokines, such as IL-1β and IL-18, which enhance the innate immune response against infections [18].
Among the complexity of microbiota, some species have recently emerged with a dominant role in the immune response, such as segmented filamentous bacteria (SFB) and Gram-positive anaerobic bacteria, which promote the accumulation of Th1 cells and atypical non-inflammatory Th17 cells in the small intestine, thus inducing IgA secretion [19,20]. Of note, distinct Th17 populations are supported by distinct microbes, since Citrobacter induce typical Th17 cells producing high amounts of pro-inflammatory cytokines [21].
The gut microbiota also interact with extra-intestinal sites, thus modulating the immune response in other organs [22,23]. Indeed, the gut microbiota and the associated metabolites can translocate through the circulatory system to different organs (such as the liver, the brain, the lungs, and even secondary lymphoid tissues), where they are recognized by local sensors and regulate the immune responses [22,23].
Overall, these data suggest the existence of a complex network of bidirectional interactions between microbiota and both innate and adaptive immunity cells, which is fundamental to prevent autoimmune reactions and a high inflammatory status with consequent tissue injury. Since the immune system and the microbiome are tightly interconnected in a complex network, the growing interest in the possible role of the microbiome in autoimmune diseases is not surprising.

2.2. The Role of Vitamin D in the Interplay between Microbiota and the Immune System

In the context of microbiota and the immune system interconnection, vitamin D has recently emerged as one of the regulators of this complex network [24].
Interestingly, vitamin D shortage has been often reported in several autoimmune diseases, such as type I diabetes mellitus, systemic lupus erythematosus, rheumatoid arthritis, and multiple sclerosis, and it is suspected to play a role in the pathogenesis of these diseases [25,26,27]. A lack of vitamin D may in fact impair the integrity of the gut epithelial barrier and, consequently, the microbiota balance [28]. Vitamin D/vitamin D receptor (VDR) signaling normally enhances the expression of several components of tight junctions and adherent junctions, such as occludin and claudins, in the intestinal epithelium [29]. The loss of such epithelial junctional proteins due to vitamin D deficiency could contribute to “leaky gut” syndrome, which favors the absorption of microbic components, alters the microbial composition, and promotes immune cell activation. A meta-analysis on patients with inflammatory bowel disease has interestingly indicated that the correction of hypovitaminosis D contributes to better maintenance and reduces the risk of disease relapse [28,30].
VDR is a nuclear receptor that can be found in about 30 different tissues, and it is able to regulate the expression of more than 1000 genes in the genome, after binding to the active form of vitamin D, 11α,25(OH)2D3 [31]. Studies have identified four single nucleotide variants (SNVs) in the VDR gene, named FokI in exon 2, BsmI and ApaI in intron 8, and TaqI in exon 9, which affect the structure of the receptor and, consequently, the response to vitamin D, and seem to be involved in autoimmune disease [32,33].
Within the immune system, 1α,25(OH)2D3 modulates the immune response via the VDR expressed in naïve and CD4+ and CD8+ T cells, B cells, neutrophils, and antigen-presenting cells, such as monocytes, macrophages, and dendritic cells [34]. Vitamin D promotes Tregs, inhibits the differentiation of proinflammatory Th1 and Th17 cells, impairs the development and the function of B cells, modulates the regulatory B cells, reduces monocyte activation, downregulates IL-17 and IL-23 production, and promotes IL-10 production [34]. Since Th1, Th2, and Th17 cells can support mucosal inflammation and tissue injury, while Tregs are important players of immune tolerance, as they exert anti-inflammatory functions and stimulate tissue repair [35], vitamin D substantially influences the immune response by balancing inflammation versus anti-inflammation, and vitamin D deficiency disrupts this balance, favoring inflammation [36].
Vitamin D and microbiota display many similarities in the modulation of the immune system, downregulating the proinflammatory pathways and the inflammatory markers, and upregulating anti-inflammatory cytokine synthesis [35]. These similarities could be due to the synergistic effect between vitamin D and microbiota metabolites such as SCFAs. In fact, both can exert anti-inflammatory activity by promoting Treg function, increasing the levels of the anti-inflammatory cytokine IL-10, affecting the maturation of DC, and inhibiting the expression of pro inflammatory cytokines [35] (Figure 1).
In addition, human studies have reported significant associations between vitamin D and the microbiome genus composition [37]. A recent genome-wide association study has identified two VDR polymorphisms significantly influencing the abundance of the genus Parabacterioides (phylum: Bacteroidetes), which has been confirmed in VDR−/− mice showing an increased abundance of Parabacteroides compared to wild-type mice [33].
In conclusion, vitamin D appears to be a critical player in the dynamic relationship between the immune system and the microbiome, with predominantly immunosuppressive properties; therefore, vitamin D supplementation has been suggested to be of benefit in the treatment of autoimmune diseases. However, little is known about how vitamin D supplementation impacts the microbiome in autoimmune patients since few studies have addressed this question [37].
In the following section, we analyze both organ-specific and systemic monogenic autoimmune disorders, summarizing their genetic features, the involvement of microbiota, and the available data on the role of vitamin D in their pathogenesis in order to provide a comprehensive review and potential suggestions for further studies.

3. Pathophysiology of Organ-Specific Monogenic Autoimmune Disorders and Microbiota Implication

3.1. Autoimmune Polyendocrine Syndrome Type 1 (APS1)

Autoimmune polyendocrine syndrome type 1, which is also known as autoimmune polyendocrinopathy candidiasis-ectodermal dystrophy (APECED), is a rare recessive disorder that mainly manifests with mucositis (from the mouth to the gastrointestinal tract), hypoparathyroidism, adrenal insufficiency, and displays a great variability in the timing of the onset [38]. However, other various symptoms have been described, such as alopecia, vitiligo, premature ovarian failure, and type I diabetes [39].
The autoimmune regulator gene (AIRE) was identified in 1997 as the causal gene on chromosomal region 21q22.3 by positional cloning [40,41]. AIRE encodes a 545 amino acid protein that contains several domains that are characteristic of transcription factors, including a homogeneously staining region (HSR) domain (involved in dimerization), followed by a SAND domain (DNA binding), and two plant homeodomain (PHD) domains (probably E3 ubiquitin ligase activity) [42].
AIRE is mainly expressed in the thymus (medullary thymic epithelial cells (mTEC) and dendritic cells (DCs)), and, to a lesser extent, in secondary lymphoid organs. It regulates the ectopic expression of tissue-specific proteins, thus contributing to the deletion of autoreactive T cells by both central and peripheral tolerance [43,44].
More than 60 different mutations have been described, and the most frequent is the 1094-1106del13 [45]. Generally, heterozygous patients for other APS1 mutations do not develop the typical clinical features of APS1, but may manifest different uncommon phenotypes. A G228W pathogenic variant with dominant inheritance was discovered in an Italian family, displaying a nonclassical autoimmune thyroiditis [46,47]. Overall, a low correlation between the genotype and the phenotype has been reported and, even in siblings, the same mutation may cause different clinical manifestations [48].
Defects in the number and the function of Tregs characterize APS1 patients, as well as abnormal DCs [49,50]. In addition, the infiltration of lymphocytes in affected organs (the stomach, lungs, and eyes), as well as the production of autoantibodies (autoAbs) against antigens (i.e., insulin and cytochrome p450 1A2) of the affected tissues, have been described [51]. The early production of high titer of autoAbs against interferon (IFN)-ω and -α2 are also used in diagnosis [52]. More recently, the NACHT leucine-rich repeat protein NALP5 (also known as NLRP5) has been identified as a putative parathyroid autoantigen to be evaluated in patients with hypoparathyroidism [53].
Furthermore, autoAbs to interleukin (IL)-17A, IL-17F, and IL-22 have been detected in the sera of all APS1 patients with candidiasis, thus suggesting an impaired role of the Th17 cells in fungal immunity [54]. In addition, a recent study showed AIRE interaction with Dectin-1, which is involved in the inflammasome-activating pathway during the anti-fungal response, which suggests a possible impairment of inflammasome activity in APS1 patients [55].
Among the clinical manifestations, gastrointestinal symptoms frequently occur in APS1 patients [56]. Studies have demonstrated that they correlate with the lack of tolerance to self-antigens such as tryptophan hydroxylase and histidine decarboxylase, which are produced by the enteroendocrine cells (EECs) of the gut epithelium [57]. The consequent loss of EECs causes low serum levels of serotonin and typical constipation symptoms [57].
Interestingly, APS1 patients show serum reactivity against the secretory granules of Paneth cells containing enteric α-defensins and the loss of Paneth cells themselves, together with the infiltration of T cells in the gut [58]. Enteric defensins have antimicrobial activity and influence the ecology of the commensal microbiota [59]. Specifically, a study by Petersen A. et al. [60] analyzed the fecal microbiomes of APS1 patients and found a correlation between several taxa, mainly belonging to the Clostridiales, and gastrointestinal symptoms. Notably, the depletion of a commonly used probiotic L acidophilus correlated with an increased Bacteroidetes–Firmicutes ratio in APS1 patients [60]. Moreover, Hetemaki I et al. [61] reported high levels of Abs against Saccharomyces cerevisiae (S. cerevisiae) and several species of commensal gut bacteria, which did not correlate with gastrointestinal autoAbs and neutralizing anti–IL-17 or –IL-22 autoAbs, or gastrointestinal symptoms. Conversely, anti–S. cerevisiae Ab levels were inversely correlated with the Foxp3 expression levels in Tregs, which are crucial for the maintenance of mucosal tolerance and are numerically decreased in the gut biopsies of APS1 patients [61]. The Treg defect may allow the resident T and B cells to mount a response against commensals supported by the defects in the Th17 cytokines. Of note, a shift toward an immunoglobulin (Ig) G response has been described in these patients, in contrast with the local IgA response, which characterizes the normal gut with normal flora [61]. Anti–S. cerevisiae Abs have been also associated with Crohn’s disease [62], thus suggesting that AIRE plays a role in regulating gut homeostasis.
Studies on Foxp3-deficient scurfy (SF) mice have shown that autoimmunity is accompanied by decreased gut bacterial diversity, which is restored by Foxp3+ Tregs administration [63]. Moreover, the treatment with Lactobacillus reuterii (DSM 17938, LR 17938), a human-derived probiotic, which has been used to treat infantile colic and acute infectious diarrhea, repristinated the flora heterogeneity and decreased the disease severity [64]. Nevertheless, whether dysbiosis is a response to or a driver of the gastrointestinal symptoms that are characteristic of APS1 remains to be determined.
Currently, APS1 treatment is based on targeting organ-specific manifestations. Antifungals are used in the case of candidiasis as a replacement therapy for endocrinopathies, with cortisol and fludrocortisone being given for adrenal insufficiency, and calcium and vitamin D as a replacement for hypoparathyroidism [39]. The deficit of vitamin D has been described in several patients with APS1 who also showed severe hypocalcemia [65]. The authors suggest that the loss of EECs in the gut epithelium contributed to vitamin D malabsorption, making useless the oral administration of calcitriol, while suggesting a better efficacy of parental vitamin D analogs in controlling hypoparathyroidism in these patients [65,66]. In light of the role of vitamin D in autoimmunity and its effects on microbiota, it is conceivable that its deficit in APS1 patients might be also involved in the pathogenesis of the disease or correlated with the severity of the clinical manifestations. However, studies on the involvement of vitamin D in APS1 are currently missing, thus opening interesting new research fields.

3.2. Immunodysregulation Polyendocrinopathy Enteropathy X-Linked (IPEX) Syndrome

IPEX is a rare X-linked recessive disorder, first described in 1982 [67], resulting in early onset type 1 diabetes mellitus (T1DM), severe enteropathy, eczema, anemia, thrombocytopenia, hypothyroidism, and a hyperreaction to viral infections [68]. If it is not diagnosed early, IPEX has a very severe onset and is usually lethal in infancy or childhood, independently from the type of causative mutation. Patients frequently show high levels of IgA and IgE and eosinophilia [68]. Furthermore, autoAbs targeting pancreatic islets, thyroid, erythrocytes, platelets, and the small intestine are frequently detected [68].
In 2000 [69], IPEX was mapped to the Xp11.23-Xq.13.3 chromosomal region where FOXP3 (Forkhead box protein 3) was identified as the responsible gene.
FOXP3 is a highly conserved gene that is composed of 12 exons, encoding the 431 amino acid protein Scurfin, which is the master transcription factor for Treg functions, is involved in the expression of the main markers of these cells, such as CTLA-4 and CD25, and is responsible for the suppression of proinflammatory cytokine production [70]. Over 70 pathogenic variants have been reported so far, mainly at the C-terminal forkhead (FKH) DNA-binding domain. Mutations of the polyadenylation site are causative of unstable FOXP3 mRNA, resulting in severe and early onset disease [71]. However, the severity of the disease is not strictly dependent on the absence of the protein, and most of the mutations (missense) cause an expression of normal or reduced levels of mutant protein. Normal percentages of CD4+CD25+FOXP3+ T cells can be detected in the peripheral blood of IPEX 1 patients; however, these mutant FOXP3 Tregs are functionally impaired and are the main pathogenetic cause of the multi-organ autoimmunity in IPEX patients [72]. In vitro studies have described a high heterogeneity in the severity of functional impairments among Tregs from different IPEX patients [72]; moreover, a lack of correlation between the genotype and the phenotype has been reported, which reflects the complexity of FOXP3 interactions and the role of non-genetic factors in the clinical manifestations of the disease, such as environmental and epigenetic factors [73]. In addition to the loss of the suppressive functions, an inflammatory shift toward the Th17 phenotype has been reported in patients with mutant FOXP3 Tregs, which might be involved in organ damage [74]. Furthermore, a decreased production of Th1 cytokines and increased levels of Th2 cytokines have been shown by several authors [72,75,76], which suggests that FOXP3 mutations may also affect T effector cell activities.
At least 50% of IPEX patients do not carry mutations in FOXP3. In children who are diagnosed with an IPEX-like syndrome, the genetic defect is either unknown or alterations in other genes that are related to Treg functions have been detected, such as in IL2RA, STAT 5b, ITCH, STAT1, STAT3, CTLA4, LRBA, TTC7A, and TTC37 [77].
The therapeutic strategies depend on the clinical manifestations and include replacement and supportive therapy targeting the damaged organs, immunosuppressive agents, and hematopoietic stem cell transplantation (HSCT) [73]. Early HSCT can lead to the best outcome when autoimmunity has not yet damaged the organs, hence making an early diagnosis fundamental. A future cell/gene therapy approach may represent an option to replace mutant cells with wild-type Tregs, which seem to be sufficient to control the disease. A recent study has suggested a CRISPR-based gene correction that can specifically target FOXP3 in autologous hematopoietic stem and progenitor cells (HSPCs). This approach allows us to edit HPSCs while preserving their differentiation potential and represents a promising strategy for IPEX patients with different types of mutations [78].
Fecal microbiota transplantation (FMT) has been suggested as an alternative strategy to cure IPEX patients with enteropathy symptoms [79]. Wu et al. specifically described a significantly decreased diversity in the gut microbiota composition in a child with IPEX, which was repristinated after receiving FMT along with remission of diarrhea [79]. A correlation between gut dysbiosis and gut inflammation has been previously reported in the IPEX model FoxP3+ Treg cell-deficient SF mouse [80,81]. Indeed, these studies showed significant differences in the microbiota composition in 22-day-old SF mice as compared with wild-type (WT) mice by analyzing stool 16S rRNAs. Specifically, the SF mice showed a decreased relative abundance of Lactobacillus at day 8 and increased Bacteroides at day 22, thus suggesting a role of Treg deficiency in shaping the gut microbiota composition [64]. Moreover, the treatment with 107 CFU/day of Lactobacillus reuterii, which was orally fed by gavage, reset SF dysbiosis, increasing the relative abundance of the phylum Firmicutes and the genera Lactobacillus and Oscillospira, and decreasing the relative abundance of the phylum Tenericutes and the genus Bacteroides [64].
In addition to remodeling the gut microbiota, the treatment with Lactobacillus reuterii significantly increased the survival of the SF mice, which usually die after 21 days. It reduced the T lymphocyte infiltration in the lungs and the liver and decreased the IFN-γ and IL-4 plasma levels. This indicates that Lactobacillus reuterii also suppresses the autoimmunity that is caused by Treg deficiency [64]. An analysis of the plasma metabolites also demonstrated significant effects on alterations that were correlated to Treg deficiency; for example, the immunomodulatory purine metabolite inosine, but not adenosine, hypoxanthine, or xanthine, was reduced 5-fold in the SF mice and the treatment with Lactobacillus reuterii restored these levels [64]. In addition, decreased multiorgan inflammation and prolonged survival was described after inosine feeding, which inhibited the Th1/Th2 differentiation in an adenosine A2A-receptor-dependent manner [64].
IPEX dysbiosis was also restored by the administration of Foxp3 + Tregs in T-cell-deficient mice [63].
Overall, these data suggest multiple pathways that could be targeted in order to treat the disease, and respective therapeutic strategies that could be investigated.

4. Pathophysiology of Systemic Monogenic Autoimmune Disorders and Microbiota Implication

4.1. Autoimmune Lymphoproliferative Syndrome (ALPS)

ALPS is a disorder of T cell-apoptosis dysregulation that leads to chronic polyclonal, but not neoplastic lymphoproliferation, autoimmune manifestations and an increased development of lymphomas [82,83,84].
ALPS patients show multiple manifestations, such as lymphadenopathy, splenomegaly, hypergammaglobulinemia, and autoimmune features that are often represented by hemolytic anemia and thrombocytopenia, but also hepatitis, uveitis, and vasculitis [84]. Systemic autoimmunity frequently requires medical intervention, as the destruction of several blood cell lines often occurs, leading to chronic cytopenia, which can range from mild to severe, and is treated with immunosuppression [82]. Autoimmunity usually manifests in early childhood, months or years after lymphoproliferation.
ALPS was first described in the 1990s in a cohort of patients with chronic lymphoproliferation and an increased number of double negative T cell populations (DNTs; cell phenotype CD4/CD8, CD3+, TCRαβ+), which reach <1% in the peripheral blood of healthy controls (vs. >2.5% in ALPS patients) [85]. Increased DNTs in the peripheral blood and the lymphoid tissues is a hallmark of ALPS, together with defective in vitro Fas-mediated apoptosis. However, mild increases in DNTs have been also described in other autoimmune diseases, including SLE and immune thrombocytopenic purpura [86,87]. The origin of DNTs is still not clear, though a significant overlap of TCR Vβ-Jβ transcripts between DNTs and CD8 T cells has emerged from CDR3 sequencing in ALPS patients, thus suggesting at least a partial CD8 origin [88,89].
Elevated serum levels of IL-10, vitamin B12, and Fas ligand have been also reported in ALPS patients [90,91].
As for IPEX and APS1, mouse models with MRLlpr/lpr and MRLgld/gld mice with similar clinical features have contributed to a deep understanding of the diseases, also highlighting the importance of apoptosis in the maintenance of immunologic homeostasis and tolerance. These mice carry loss-of-function mutations of either the FAS (lpr mutation) or the FASLG (gld mutation) genes, causing defective FasL/Fas-induced apoptosis [92,93]. Pathogenic variants in these genes are detected in approximately 70% of ALPS patients and are usually inherited with autosomal dominant mechanisms [94]. These mutations alter the formation of the Fas/FasL functional trimolecular complex, which is required to induce apoptosis in effector lymphocytes and controls their lifespan [95]. As a consequence, lymphocytes accumulate in the secondary lymphoid tissues, which favors development of autoimmunity and, possibly, lymphomas.
The most frequent mutations that hit FAS and are often characterized by incomplete penetrance, and the inheritance pattern can be dominant with incomplete penetrance, recessive, or can be manifested when it is associated to variants in other genes, thus suggesting a possible digenic/oligogenic inheritance [96,97]. ALPS that is caused by germline mutations of FAS is classified as ALPS-FAS, but some cases are caused by somatic mutations of this gene (ALPSsFAS). Rare cases are due to mutations of FASL (ALPS-FASL) or CASP10 (ALPS-CASP10) that are involved in Fas signaling, whereas the mutated gene is undetermined in about 30% of cases (ALPS-U) [98]. Some studies report that mutations at the heterozygous state in the FAS intracellular domain affect the canonical death domain that is required for initiating the apoptotic cascade and are associated with a higher penetrance, compared with heterozygous mutations of the extracellular domain [99]. However, there is no consensus about the penetrance of the disease and its correlation with the type of mutation, thus suggesting a recommendation of genetic counselling for all of the unaffected family members who inherited the pathogenic variant.
In addition, it may be difficult to diagnose ALPS, since a biological and clinical overlap exists with other diseases, which may show lymphoproliferation, autoimmunity, and apoptosis defects, such as caspase-8 deficiency (CEDS), RAS-associated autoimmune leukoproliferative disease (RALD), Dianzani autoimmune lymphoproliferative disease (DALD), and even X-linked lymphoproliferative syndrome (XLP) [84,86,100,101,102,103]. Moreover, a defective Fas function in the absence of FAS mutations can be detected in subsets of patients with common autoimmune diseases such as multiple sclerosis, type I diabetes mellitus, and SLE [104,105,106].
Furthermore, a tissue biopsy (bone marrow and/or lymph node) is recommended at the initial presentation in order to distinguish ALPS from malignancy [107].
More recently, a role of antigen-presenting cells (APCs) has been suggested in the onset of ALPS, since defective apoptosis of APCs has been reported in ALPS patients [108]. Moreover, studies on murine models describe autoimmunity signs after conditional inactivation of Fas in APCs (DCs or B lymphocytes), with the production of high levels of antinuclear Abs (ANA), hyper-immunoglobulinemia, and splenomegaly, without DNT expansion [109]. These findings suggest that chronic antigen presentation, from either activated DCs or B cells with defective apoptosis, can also disrupt immune tolerance.
To our knowledge, no data are currently available on the involvement of microbiota in ALPS patients, leaving space for future research.
ALPS patients may not require any treatment, but rather clinical surveillance. In the presence of clinical manifestations, immunosuppressive agents are used to treat autoimmune manifestations and chemotherapy is used for malignancy [107].

4.2. Monogenic Forms of SLE

SLE is a complex multisystem autoimmune disease with heterogeneous clinical manifestations. The pathogenesis is correlated to different genetic backgrounds, immunologic alterations, and environmental factors, which makes it difficult to fully understand the SLE mechanisms and, consequently, to develop therapeutic strategies [110,111].
However, the identification of a rare monogenic form of SLE, characterized by familial segregation, as well as early-onset juvenile SLE, allowed for a better understanding of the involved pathways, i.e., complement, apoptosis, nucleic acid degradation, nucleic acid sensing, self-tolerance, and type I IFN production [112].

4.2.1. Genetic Alterations

The Complement System

The complement system plays a key role in the innate and adaptive immune response. Some components act as opsonins, which allow the clearance of immune complexes by red blood cells and phagocytes, thus avoiding tissue vascular deposition. Moreover, early components of the classical pathway (C1q, C1r, C1s, C2, and C4) decrease the release of autoantigens by promoting the clearance of apoptotic cells by phagocytes (efferocytosis) [113]. Its role in T and B cell activation has been described as well, which may alter the balance of lymphoid cell activation [114,115]. C1q also reduces type I IFN production by plasmocytoid DCs [116] and modulates CD8+ T cell mitochondrial metabolism in order to avoid self-antigen autoreactivity and promote viral control [117].
Complement C1q, C1s, and C1r deficiencies are associated with early-onset SLE, although they are very rare (<1%). Most of these patients show skin involvement and a severe SLE phenotype, together with recurrent life-threatening infections, such as meningitis; moreover, high levels of ANA and rarely anti-double-stranded DNA (dsDNA) have been described [118]. SLE patients with homozygous C2 deficiencies are characterized by severe arthritis and cutaneous vasculitis, while nephritis is frequent in those patients with C4 defects [119].
Dysregulations of apoptosis, as well as NETosis, i.e., the release of neutrophil extracellular traps (NETs), are also thought to be involved in monogenic SLE, increasing the autoantigen load and disrupting tolerance [120]. As mentioned previously, ALPS, FAS, and FASL mutations are implicated in the development of autoimmune responses, which may mimic SLE in some patients. MRLlpr/lpr mice have been often used as models of SLE [121,122].

PRKCD

A study by Belot et al. [123] identified PRKCD mutations in three siblings with juvenile-onset SLE. PRKCD encodes protein kinase C (PKCδ), which is involved in the deletion of autoreactive B cells [124]. The mutation causes a reduction in PKCδ expression and activity, thus leading to B cell expansion and defective apoptosis [123]. More recently, other studies have supported PRKCD as a candidate gene that is associated with SLE risk [125,126]. Monogenic pathogenic variants in RAG2 (recombination activating 2 gene) that are involved in V(D)J recombination and BCR to TCR variability generation also cause SLE manifestations, including lupus nephritis and erosive arthritis [127].
T cell expansion and defective apoptosis also occur in some patients with Noonan syndrome, a neurodevelopment disorder that is caused by pathogenic variants in one of the several RAS/MAPK pathway genes, including PTPN11, SOS1, RIT1, and KRAS, encoding for a small GTPase that is activated by TCR signaling [128,129]. This disease manifests with a short stature, dysmorphic features, congenital heart disease, bleeding diathesis, and an increased risk of malignancy and lupus-like symptoms [130].

TREX1

The degradation of nuclear materials is another mechanism that prevents autoantigen load. Several intracellular enzymes, such as three prime repair exonuclease 1 (TREX1) and RNase H2 complex, are involved in this function. TREX1 encodes an IFN-inducible exonuclease that is responsible for genomic DNA degradation in response to DNA damage. Therefore, it is involved in the immune response to single-stranded (ss)-DNA and dsDNA and in the maintenance of immune tolerance to cytosolic self-DNA [131]. Dysfunctional TREX1 causes an accumulation of self-DNA, which represents a damage-associated molecular pattern (DAMP), activating systemic inflammation and triggering autoimmunity [132]. Similarly, RNase H2 degrades RNA, DNA heteroduplexes and contributes to the ribonucleotide excision repair pathway [133].
Alterations of genes encoding for these enzymes have been reported in Aicardi–Goutières syndrome (AGS), an autosomal recessive disease affecting the skin, the brain, and the immune system, which phenotypically overlap with SLE [134]. Biallelic mutations of TREX1 cause a complete loss of protein function in AGS patients, who show high levels of IgG autoAbs directed against the nuclear antigens, the basement membrane components, the gliadin and brain endothelial cells, and the astrocytes [135]. Partial loss-of-function biallelic mutations in RNase H2 also occur in AGS, leading to an intracellular accumulation of aberrant nucleic acid species and the consequent activation of nucleic acid sensor and IFN cascade [136].
The loss-of-function mutations of TREX1 (also called DNAse type III) and genes of the RNase H2 complex have been found in SLE patients as well [134,137,138].
AGS patients also show high levels of type I IFN in the serum and the cerebrospinal fluid [139] because of the mutated TREX1 enzyme, which is not able to prevent intracellular DNA detection by innate immune sensors, thus inducing type I IFN activation and accumulation. The excessive production of type I IFNs is the hallmark of SLE too [140]. This signature induces immature DCs towards an effector phenotype (APCs), suppresses Tregs, increases the differentiation of Th17 cells, and stimulates B cell proliferation [141].

DNASE1L3

A study by Al-Mayouf SM et al. [142] identified another gene, DNASE1L3, which was mutated in familial cases of juvenile SLE, with anti-dsDNA reactivity. DNASE1L3 is an extracellular DNA-degrading enzyme, a homologue of DNASE1, with a positively charged C-terminal peptide, which confers the ability to remove DNA chromatin that is associated to the extracellular microparticles [142]. It has been recently shown, indeed, that, in these patients, DNA accumulates in the circulating microparticles containing high levels of long polynucleosomal cell-free DNA (cfDNA) fragments, with an increased affinity to auto-Abs and capacity to induce type I IFN production after recognition by DNA/RNA sensors such as Toll-like receptors (TLR7 and TLR9) [143]. The amount of auto-Abs against DNASE1L3-sensitive antigens on microparticles correlates with disease severity and is frequent in SLE nephritis patients [143]. Among these antigens, the authors describe the high-mobility group B1 (HMGB1) protein, belonging to structural transcription factors, that can be incorporated into chromatin or released from activated or dying cells [143]. Pisetsky D et al. [144] previously described the exposure of HMGB1 on apoptotic microparticles, while Hartl J and colleagues [143] demonstrated that DNASE1L3 controls this phenomenon through the digestion of HMGB1–DNA complexes, overall suggesting a new potential therapeutic target in the monogenic forms of SLE [143]. Mutations in DNASE1 have also been described in patients with SLE manifestations and high titer of ANA [145].

ACP5

Another disease that is usually associated with autoimmune SLE is spondyloenchondrodysplasia (SPENCD), a rare autosomal recessive skeletal dysplasia caused by mutations in the ACP5 (acid phosphatase 5) gene [146,147]. ACP5 encodes tartrate-resistant acid phosphatase (TRAP), which uses osteopontin (OPN) as a primary substrate [148]. OPN is also required for type I IFN production in response to TLR9 stimulation by plasmacytoid DCs [149]. ACP5 mutations alter the TRAP activity, causing OPN hyperphosphorylation, which in turn induces excessive type I IFN production and SLE-like pathologies [147].
Overall, the monogenic forms of SLE provide opportunities for a better understanding of the mechanisms behind the disease and allow for the design of precise therapeutic strategies. SLE patients with an IFN signature could benefit from a human anti-IFN Ab, which competes with IFN-α binding to the receptor IFNAR2, thus preventing the activation of the IFN-α mediated pathways [150]. Interestingly, a systematic literature review by Rodríguez-Carrio et al. [151] has revealed that measuring type I IFN pathway activation in these patients could potentially help in monitoring the disease activity, the prognosis, and the response to treatment, although assay harmonization and clinical validation are still needed [151].
Since nucleic acid sensors, such as the retinoic acid inducible gene (RIG)-1 and the melanoma differentiation-associated gene 5 (MDA5), activate type I IFN via TBK-1 signaling, the use of TBK inhibitors might represent an alternative strategy in SLE patients, with fewer side effects than the conventional immunosuppressive drugs [152]. The reverse transcriptase inhibitors that are currently used to treat human immunodeficiency virus (HIV) could be also used in monogenic forms of SLE with mutated TREX1 or RNase H2 complex, which alter the metabolism of the nucleic acids that are generated during reverse transcription [153].
The use of NGS (next-generation sequencing) in early-onset SLE will provide further insights in understanding the molecular mechanisms of the disease, but also of tolerance maintenance, leading to a personalized medicine with less adverse events and increased efficacy [154].

4.2.2. Microbiota Alterations

Due to the extensive involvement of multiple organs, a higher level of fatigue is observed in patients with SLE who also show a deficit in vitamin D, which worsens the condition [155,156]. Several studies have explored the association between calcidiol (the main vitamin D metabolite) serum levels and the disease activity and susceptibility, although the results are still inconclusive, because of the wide calcidiol range that has been reported in the different analyzed populations. Indeed, its levels are influenced by environmental and genetic factors, making it difficult to compare different studies [32,155,157]. Moreover, high levels of the active form, calcitriol, have been detected in SLE patients and could be indicative of active inflammation and Th2 polarization [156].
Several recent studies, on both mouse models and humans, suggest a correlation between alterations of the gut microbiota composition and SLE disease manifestations [158]. The main feature is that the ratio of Firmicutes/Bacteroides (F/B) is significantly reduced, and the decreased abundance of some families of Firmicutes may be related to remissive SLE [159]. Synergistetes negatively correlate with anti-dsDNA Abs [160]; meanwhile, anti-Ruminococcusgnavus Abs positively correlate with lupus nephritis and anti-dsDNA Abs [161]. A positive correlation was found between Streptococcus, Campylobacter, Streptococcus anginosus, and Veillonella dispar and lupus activity, while Bifidobacterium was negatively associated [162]. Low levels of Lactobacillus and increased Lachnospiraceae, with inflammatory properties, have been described. Gender differences in the gut microbiota of SLE mice have also emerged [159].
Interestingly, microbiota isolated from SLE patient stool samples induce lymphocyte activation and Th17 differentiation from naïve CD4+ lymphocytes [160].
These data suggest that the modulation of gut microbiota via diet, probiotics/prebiotics, and FMT might be a promising additional therapeutic strategy for SLE, possibly also including the monogenic forms of SLE.

4.3. The Role of Microbiota in Murine Models of Systemic Autoimmune Disorders

As mentioned previously, studies on murine models have helped to clarify the mechanisms behind the microbiota involvement in monogenic autoimmune diseases. Comparable results have been reported from different groups, using well-established mouse models that show similar features of both ALPS and SLE patients, as follows: MRL/lpr, which spontaneously develops autoimmune disease, with hypergammaglobulinemia, the production of numerous ANA, anti-dsDNA, circulating immune complexes, lymphadenopathy, and glomerulonephritis; and MRL+/+, with a similar background, but with a much slower onset of SLE. Wang et al. [163] used 6- and 18-week-old mice and highlighted several differences in terms of the microbial composition, the intestinal barrier integrity, and the inflammatory markers among the models. Specifically, a lower Firmicutes/Bacteroidetes ratio was reported in the 6-week-old MRL/lpr mice compared to the age-matched MRL+/+ mice, with decreased levels of Peptostreptococcaceae (Firmicutes phylum) and increased Rikenellaceae (Bacteroidetes phylum) [163]. No correlation was found with the advanced disease activity in the 18-week-old mice; however, a decreased number of Lactobacillaceae was reported in these mice compared with their 6-week-old counterparts. Interestingly, previous studies have shown that the oral administration of a mixture of five strains of Lactobacillus spp (L. reuteri, L. oris, L. johnsonii, L. gasseri, and L. rhamnosus) restored the microbiota in the MRL/lpr mice and attenuated the disease symptoms, thus improving the renal functions, splenomegaly, and lymphadenopathy [164]. This group further investigated the mechanisms behind Lactobacillus effect, demonstrating the restoration of the Treg-Th17 balance, with a decreased percentage of central memory T cells and increased effector memory T cells, along with reduced IL-6 and augmented IL-10 production in the gut. Of note, the strains acted synergistically in order to ameliorate the disease in a CX3CR1-dependent manner [165]. CX3CR1 is involved in the translocation of the APCs from the blood to the mesenteric lymph nodes, where they can activate T cells, thus contributing to the homeostasis between the commensal bacteria and the immune system [166]. Furthermore, the same benefits of the mixed Lactobacilli were obtained using culture supernatants, suggesting a potential use of both probiotics and postbiotics in the management of SLE as a cost-effective approach in support of the conventional treatment strategies. In this regard, Da Som Kim et al. [167] demonstrated an improved efficacy of the immunosuppressant Tacrolimus, which was administered in combination with Lactobacillus acidophilus in MRL/lpr mice. A decreased number of double negative T cells and lower serum levels of anti-dsDNA Abs and IgG2a were detected after the treatment, along with improved renal functions [167]. A higher production of indoleamine-2,3-dioxygenase (IDO), PD-1, and IL-10 was reported after the in vitro treatment with Lactobacillus, and it seems to be mediated by the specific intracellular adhesion molecule-3 grabbing the non-integrin homolog-related 3 receptor (SIGN3) signals, which is a homologue of the human DC-SIGN [167]. The positive effects of Lactobacillus treatments were confirmed in other SLE mouse models, such as the NZBWF1 model, which showed reduced lupus disease activity, blood pressure, cardiac and renal hypertrophy, and splenomegaly after Lactobacillus fermentum CECT5716 (LC40) intake. Endothelial dysfunctions, which characterize SLE disease, were also solved, and decreased plasma levels of LPS were detected, suggesting an improved gut barrier integrity [168].
A study by Hanchang He et al. [169] gave evidence of dysbiosis improvement after sodium butyrate treatment in MRL/lpr mice. Butyrate belongs to the SCFA, which are the products of intestinal bacterial fermentation. It increases the expression of the tight junction proteins, contributing to the integrity of the gut barrier, and it reduces oxidative stress and mucosal inflammation. In the MRL/lpr mice, the butyrate treatment enhanced the gut microbiota diversity, increasing the abundance of Firmicutes, Clostridia, Clostridiales, and Lachnospiraceae, while reducing Bacteroidetes, Bacteroidia, and Bacteroidales [169]. In addition, the promoting effect of butyrate on fat oxidation through brown adipose tissue activation [170] further supports its beneficial activity on SLE, according to the efficacy of caloric restriction in preventing disease progression that has been shown in NZB and (NZBxNZW) F1 mice, which spontaneously develop lupus, slowly.
High levels of oxidative stress molecules, such as 4-hydroxynonenal (HNE)-adducts and HNE specific immune complexes, were also detected in MRL/lpr mice, similarly to human SLE [171]. Protein oxidation or lipid peroxidation can affect the intestinal permeability in vitro and in vivo, thus worsening the already altered gut barrier, as shown by the decreased expression of the tight junction protein ZO-2, the increased fecal albumin, and the IgA levels that have been described in 18-week-old MRL/lpr mice [171]. Inflammatory markers such as phos-NFkB, IL-6, and IgG levels also characterized these mice. Intriguingly, the treatment with a ROS scavenger, N-acetylcysteine, not only downregulated the hepatic inflammasome activation markers, but also changed the microbiota composition in these mice by reducing Rikenellaceae, while supporting Akkeransiaceae, Erysipelotrichaceae, and Muribaculaceae colonization, finally attenuating autoimmunity [171].
Additional studies have investigated the involvement of the gut microbiota in the therapeutic response to glucocorticoids in SLE. Prednisone upregulated the concentration of Proteobacteria, Parabacteroides, and Escherichia Shigella [172]. Moreover, experiments of FMT from prednisone-treated MRL/lpr mice revealed that the altered microbiota after the glucocorticoid treatment recapitulate the therapeutic effects of the glucocorticoid itself on SLE, without showing side effects [172].
All together, these data support the involvement of the gut microbiota within SLE and autoimmune lymphoproliferative disease progression, thus suggesting a new potential therapeutic strategy, which should be further investigated.

5. Conclusions

The study of monogenic autoimmune diseases has importantly contributed to our understanding of the immune tolerance mechanisms.
The complex causes of autoimmune diseases present a challenge to the development and testing of novel prognostic markers and therapies. Mouse models have provided data on the molecular mechanisms of autoimmunity and have suggested new potential therapeutic approaches. For example, the adoptive transfer of Tregs for the treatment of type I diabetes has emerged from mouse studies, and several groups are moving toward using this strategy even in other diseases, such as graft vs. host disease [173]. Likewise, the diagnosis of more common diseases, such as interstitial lung disease or autoimmune gonadal failure, has been facilitated by the discovery of autoAbs in APS1 patients [174]. Overall, these data highlight the importance and the contribution of studying monogenic autoimmune disease in the diagnosis and treatment of common autoimmune diseases.
In addition, recent advances in deep sequencing have disclosed new defective genes in isolated rare families, thus providing new insights into the development of future personalized therapies [46]. Genetic testing, together with laboratory biomarkers and autoimmune features, is crucial to accurately diagnose monogenic diseases (see Table 1 for summarized disease features); however, it remains challenging, due to the variability in disease progression and overlapping clinical manifestations. However, massive NGS has been introduced in diagnostic laboratories, thus revolutionizing the screening of these disorders. In addition, genetic diagnosis provides important information of the recurrence risk, allows the monitoring of disease progression, and gives the opportunity of timely treatment decisions, thus minimizing complications. Finally, the use of NGS to characterize microbiota may add further knowledge to a possible key factor that may be involved in the development and the progression of these diseases and open the way to novel therapeutic approaches.
The study of the microbiome, indeed, has provided further insights into the understanding of the immune mechanisms behind these diseases, thus representing a target for new therapeutic strategies. However, future studies are needed in order to deeply understand if a direct causal implication of the microbiome exists before the onset, or during, the early phases of autoimmune monogenic diseases. Several limitations also characterize the study of microbiota, since the mouse models that are mostly used in this field do not develop microbiota that are similar to humans. On the other hand, human individuals themselves show highly variable configurations of the microbiome, making the study of potential correlations between disease and microbe composition very challenging. This complexity opens the possibility of applying artificial intelligence and machine learning to characterize individual microbe patterns and thus create personalized therapies.
In addition to FMT, which has been widely used to treat Clostridium difficilis infections, new, more precise strategies are currently under investigation. For example, the elimination of specific pathobionts using bacteriophage therapy [175]; or the use of a personalized diet that alters the nutrient availability for specific strains, along with the intake of postbiotics, which represent the microbe-derived metabolites that are highly present in the systemic circulation, able to influence the immune system, as described in this review.
However, standardized and unbiased preclinical and clinical studies are needed before translating the microbiome-based treatments into clinical practice.
Lastly, the role of vitamin D in modulating the immune response and influencing microbiota composition suggests its potential use as a supplement within conventional therapies that are currently used to treat monogenic autoimmune disorders. Indeed, several studies have reported a reduced rate of autoimmune diseases, such as SLE, psoriasis, and rheumatoid arthritis, in patients under vitamin D treatment [176], thus encouraging the same approach in monogenic autoimmune disorders. However, studies in this field are currently missing and should be further investigated.
All together, these data highlight the importance of studying these rare diseases, which still leave numerous open questions.

Author Contributions

F.C., R.R., and M.G. wrote the manuscript; R.R., M.G., and U.D. supervised the work. F.C., E.B. (Eleonora Beltrami), S.M., S.S., E.B. (Elena Boggio), C.L.G., I.S., U.D., R.R., and M.G. have provided critical feedback and helped to shape the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by IG 27154 Associazione Italiana per la Ricerca sul Cancro, Milan, Italy.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Associazione Italiana per la Ricerca sul Cancro, Milan, Italy for supporting the work.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wang, L.; Wang, F.-S.; Gershwin, M.E.; Wang, A.; Wang, L.; Me, G. Human Autoimmune Diseases: A Comprehensive Update. J. Intern. Med. 2015, 278, 369–395. [Google Scholar] [CrossRef] [PubMed]
  2. Christen, U.; Herrath, M.G.V. Initiation of Autoimmunity. Curr. Opin. Immunol. 2004, 16, 759–767. [Google Scholar] [CrossRef] [PubMed]
  3. Marson, A.; Housley, W.J.; Hafler, D.A. Genetic Basis of Autoimmunity. J. Clin. Investig. 2015, 125, 2234–2241. [Google Scholar] [CrossRef] [PubMed]
  4. Lynch, J.B.; Hsiao, E.Y. Microbiomes as Sources of Emergent Host Phenotypes. Science 2019, 365, 1405–1409. [Google Scholar] [CrossRef]
  5. Zheng, D.; Liwinski, T.; Elinav, E. Interaction between Microbiota and Immunity in Health and Disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef]
  6. Marcobal, A.; Sonnenburg, J.L. Human Milk Oligosaccharide Consumption by Intestinal Microbiota. Clin. Microbiol. Infect. 2012, 18 (Suppl. 4), 12–15. [Google Scholar] [CrossRef] [Green Version]
  7. An, D.; Oh, S.F.; Olszak, T.; Neves, J.F.; Avci, F.Y.; Erturk-Hasdemir, D.; Lu, X.; Zeissig, S.; Blumberg, R.S.; Kasper, D.L. Sphingolipids from a Symbiotic Microbe Regulate Homeostasis of Host Intestinal Natural Killer T Cells. Cell 2014, 156, 123. [Google Scholar] [CrossRef] [Green Version]
  8. McGuckin, M.A.; Lindén, S.K.; Sutton, P.; Florin, T.H. Mucin Dynamics and Enteric Pathogens. Nat. Rev. Microbiol. 2011, 9, 265–278. [Google Scholar] [CrossRef]
  9. MacPherson, A.J.; Slack, E.; Geuking, M.B.; McCoy, K.D. The Mucosal Firewalls against Commensal Intestinal Microbes. Semin. Immunopathol. 2009, 31, 145–149. [Google Scholar] [CrossRef]
  10. Sun, C.M.; Hall, J.A.; Blank, R.B.; Bouladoux, N.; Oukka, M.; Mora, J.R.; Belkaid, Y. Small Intestine Lamina Propria Dendritic Cells Promote de Novo Generation of Foxp3 T Reg Cells via Retinoic Acid. J. Exp. Med. 2007, 204, 1775. [Google Scholar] [CrossRef] [Green Version]
  11. Chieppa, M.; Rescigno, M.; Huang, A.Y.C.; Germain, R.N. Dynamic Imaging of Dendritic Cell Extension into the Small Bowel Lumen in Response to Epithelial Cell TLR Engagement. J. Exp. Med. 2006, 203, 2841. [Google Scholar] [CrossRef] [Green Version]
  12. Round, J.L.; Lee, S.M.; Li, J.; Tran, G.; Jabri, B.; Chatila, T.A.; Mazmanian, S.K. The Toll-like Receptor Pathway Establishes Commensal Gut Colonization. Science 2011, 332, 974. [Google Scholar] [CrossRef] [Green Version]
  13. Ramanan, D.; Tang, M.S.; Bowcutt, R.; Loke, P.; Cadwell, K. Bacterial Sensor Nod2 Prevents Inflammation of the Small Intestine by Restricting the Expansion of the Commensal Bacteroides Vulgatus. Immunity 2014, 41, 311–324. [Google Scholar] [CrossRef] [Green Version]
  14. Levy, M.; Thaiss, C.A.; Zeevi, D.; Dohnalová, L.; Zilberman-Schapira, G.; Mahdi, J.A.; David, E.; Savidor, A.; Korem, T.; Herzig, Y.; et al. Microbiota-Modulated Metabolites Shape the Intestinal Microenvironment by Regulating NLRP6 Inflammasome Signaling. Cell 2015, 163, 1428. [Google Scholar] [CrossRef] [Green Version]
  15. Maslowski, K.M.; Vieira, A.T.; Ng, A.; Kranich, J.; Sierro, F.; Di, Y.; Schilter, H.C.; Rolph, M.S.; MacKay, F.; Artis, D.; et al. Regulation of Inflammatory Responses by Gut Microbiota and Chemoattractant Receptor GPR43. Nature 2009, 461, 1282–1286. [Google Scholar] [CrossRef] [Green Version]
  16. 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] [Green Version]
  17. Bachem, A.; Makhlouf, C.; Binger, K.J.; de Souza, D.P.; Tull, D.; Hochheiser, K.; Whitney, P.G.; Fernandez-Ruiz, D.; Dähling, S.; Kastenmüller, W.; et al. Microbiota-Derived Short-Chain Fatty Acids Promote the Memory Potential of Antigen-Activated CD8+ T Cells. Immunity 2019, 51, 285–297.e5. [Google Scholar] [CrossRef]
  18. Franchi, L.; Kamada, N.; Nakamura, Y.; Burberry, A.; Kuffa, P.; Suzuki, S.; Shaw, M.H.; Kim, Y.G.; Núñez, G. NLRC4-Driven Production of IL-1β Discriminates between Pathogenic and Commensal Bacteria and Promotes Host Intestinal Defense. Nat. Immunol. 2012, 13, 449–456. [Google Scholar] [CrossRef]
  19. Ivanov, I.I.; de Llanos Frutos, R.; Manel, N.; Yoshinaga, K.; Rifkin, D.B.; Sartor, R.B.; Finlay, B.B.; Littman, D.R. Specific Microbiota Direct the Differentiation of IL-17-Producing T-Helper Cells in the Mucosa of the Small Intestine. Cell Host Microbe 2008, 4, 337–349. [Google Scholar] [CrossRef] [Green Version]
  20. Gaboriau-Routhiau, V.; Rakotobe, S.; Lécuyer, E.; Mulder, I.; Lan, A.; Bridonneau, C.; Rochet, V.; Pisi, A.; De Paepe, M.; Brandi, G.; et al. The Key Role of Segmented Filamentous Bacteria in the Coordinated Maturation of Gut Helper T Cell Responses. Immunity 2009, 31, 677–689. [Google Scholar] [CrossRef] [Green Version]
  21. 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.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Corbitt, N.; Kimura, S.; Isse, K.; Specht, S.; Chedwick, L.; Rosborough, B.R.; Lunz, J.G.; Murase, N.; Yokota, S.; Demetris, A.J. Gut Bacteria Drive Kupffer Cell Expansion via MAMP-Mediated ICAM-1 Induction on Sinusoidal Endothelium and Influence Preservation-Reperfusion Injury after Orthotopic Liver Transplantation. Am. J. Pathol. 2013, 182, 180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Sharon, G.; Sampson, T.R.; Geschwind, D.H.; Mazmanian, S.K. The Central Nervous System and the Gut Microbiome. Cell 2016, 167, 915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Fakhoury, H.M.A.; Kvietys, P.R.; AlKattan, W.; Al Anouti, F.; Elahi, M.A.; Karras, S.N.; Grant, W.B. Vitamin D and Intestinal Homeostasis: Barrier, Microbiota, and Immune Modulation. J. Steroid Biochem. Mol. Biol. 2020, 200, 105663. [Google Scholar] [CrossRef]
  25. Hiremath, G.S.; Cettomai, D.; Baynes, M.; Ratchford, J.N.; Newsome, S.; Harrison, D.; Kerr, D.; Greenberg, B.M.; Calabresi, P.A. Vitamin D Status and Effect of Low-Dose Cholecalciferol and High-Dose Ergocalciferol Supplementation in Multiple Sclerosis. Mult. Scler. 2009, 15, 735–740. [Google Scholar] [CrossRef]
  26. Bener, A.; Alsaied, A.; Al-Ali, M.; Al-Kubaisi, A.; Basha, B.; Abraham, A.; Guiter, G.; Mian, M. High Prevalence of Vitamin D Deficiency in Type 1 Diabetes Mellitus and Healthy Children. Acta Diabetol. 2009, 46, 183–189. [Google Scholar] [CrossRef]
  27. Amital, H.; Szekanecz, Z.; Szücs, G.; Dankó, K.; Nagy, E.; Csépány, T.; Kiss, E.; Rovensky, J.; Tuchynova, A.; Kozakova, D.; et al. Serum Concentrations of 25-OH Vitamin D in Patients with Systemic Lupus Erythematosus (SLE) Are Inversely Related to Disease Activity: Is It Time to Routinely Supplement Patients with SLE with Vitamin D? Ann. Rheum. Dis. 2010, 69, 1155–1157. [Google Scholar] [CrossRef]
  28. Singh, P.; Kumar, M.; Al Khodor, S. Vitamin D Deficiency in the Gulf Cooperation Council: Exploring the Triad of Genetic Predisposition, the Gut Microbiome and the Immune System. Front. Immunol. 2019, 10, 1042. [Google Scholar] [CrossRef]
  29. Chen, S.W.; Wang, P.Y.; Zhu, J.; Chen, G.W.; Zhang, J.L.; Chen, Z.Y.; Zuo, S.; Liu, Y.C.; Pan, Y.S. Protective Effect of 1,25-Dihydroxyvitamin D3 on Lipopolysaccharide-Induced Intestinal Epithelial Tight Junction Injury in Caco-2 Cell Monolayers. Inflammation 2015, 38, 375–383. [Google Scholar] [CrossRef]
  30. Li, J.; Chen, N.; Wang, D.; Zhang, J.; Gong, X. Efficacy of Vitamin D in Treatment of Inflammatory Bowel Disease A Meta-Analysis. Medicine 2018, 97, e12662. [Google Scholar] [CrossRef]
  31. Nurminen, V.; Neme, A.; Seuter, S.; Carlberg, C. The Impact of the Vitamin D-Modulated Epigenome on VDR Target Gene Regulation. Biochim. Biophys. Acta Gene Regul. Mech. 2018, 1861, 697–705. [Google Scholar] [CrossRef]
  32. Mahto, H.; Tripathy, R.; Das, B.K.; Panda, A.K. Association between Vitamin D Receptor Polymorphisms and Systemic Lupus Erythematosus in an Indian Cohort. Int. J. Rheum. Dis. 2018, 21, 468–476. [Google Scholar] [CrossRef]
  33. Wang, J.; Thingholm, L.B.; Skiecevičienė, J.; Rausch, P.; Kummen, M.; Hov, J.R.; Degenhardt, F.; Heinsen, F.-A.; Rühlemann, M.C.; Szymczak, S.; et al. Genome-Wide Association Analysis Identifies Variation in Vitamin D Receptor and Other Host Factors Influencing the Gut Microbiota HHS Public Access Author Manuscript. Nat. Genet. 2016, 48, 1396–1406. [Google Scholar] [CrossRef]
  34. Yamamoto, E.; Jørgensen, T.N. Immunological Effects of Vitamin D and Their Relations to Autoimmunity. J. Autoimmun. 2019, 100, 7–16. [Google Scholar] [CrossRef]
  35. Malaguarnera, L. Vitamin D and Microbiota: Two Sides of the Same Coin in the Immunomodulatory Aspects. Int. Immunopharmacol. 2020, 79, 106112. [Google Scholar] [CrossRef]
  36. L Bishop, E.; Ismailova, A.; Dimeloe, S.; Hewison, M.; White, J.H. Vitamin D and Immune Regulation: Antibacterial, Antiviral, Anti-Inflammatory. JBMR Plus 2020, 5. [Google Scholar] [CrossRef]
  37. Yamamoto, E.A.; Jørgensen, T.N. Relationships Between Vitamin D, Gut Microbiome, and Systemic Autoimmunity. Front. Immunol. 2020, 10, 3141. [Google Scholar] [CrossRef]
  38. Perheentupa, J. APS-I/APECED: The Clinical Disease and Therapy. Endocrinol. Metab. Clin. N. Am. 2002, 31, 295–320. [Google Scholar] [CrossRef]
  39. Husebye, E.S.; Perheentupa, J.; Rautemaa, R.; Kämpe, O. Clinical Manifestations and Management of Patients with Autoimmune Polyendocrine Syndrome Type I. J. Intern. Med. 2009, 265, 514–529. [Google Scholar] [CrossRef]
  40. Aaltonen, J.; Björses, P.; Perheentupa, J.; Horelli-Kuitunen, N.; Palotie, A.; Peltonen, L.; Lee Su, Y.; Francis, F.; Hennig, S.; Thiel, C.; et al. An Autoimmune Disease, APECED, Caused by Mutations in a Novel Gene Featuring Two PHD-Type Zinc-Finger Domains. Nat. Genet. 1997, 17, 399–403. [Google Scholar] [CrossRef]
  41. Nagamine, K.; Peterson, P.; Scott, H.S.; Kudoh, J.; Minoshima, S.; Heino, M.; Krohn, K.J.E.; Lalioti, M.D.; Mullis, P.E.; Antonarakis, S.E.; et al. Positional Cloning of the APECED Gene. Nat. Genet. 1997, 17, 393–398. [Google Scholar] [CrossRef] [PubMed]
  42. Halonen, M.; Kangas, H.; Rüppell, T.; Ilmarinen, T.; Ollila, J.; Kolmer, M.; Vihinen, M.; Palvimo, J.; Saarela, J.; Ulmanen, I.; et al. APECED-Causing Mutations in AIRE Reveal the Functional Domains of the Protein. Hum. Mutat. 2004, 23, 245–257. [Google Scholar] [CrossRef] [PubMed]
  43. Derbinski, J.; Schulte, A.; Kyewski, B.; Klein, L. Promiscuous Gene Expression in Medullary Thymic Epithelial Cells Mirrors the Peripheral Self. Nat. Immunol. 2001, 2, 2915–2922. [Google Scholar] [CrossRef] [PubMed]
  44. Anderson, M.S.; Venanzi, E.S.; Klein, L.; Chen, Z.; Berzins, S.P.; Turley, S.J.; Von Boehmer, H.; Bronson, R.; Dierich, A.; Benoist, C.; et al. Projection of an Immunological Self Shadow within the Thymus by the Aire Protein. Science 2002, 298, 1395–1401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Ruan, Q.G.; She, J.X. Autoimmune Polyglandular Syndrome Type 1 and the Autoimmune Regulator. Clin. Lab. Med. 2004, 24, 305–317. [Google Scholar] [CrossRef]
  46. Cetani, F.; Barbesino, G.; Borsari, S.; Pardi, E.; Cianferotti, L.; Pinchera, A.; Marcocci, C. A Novel Mutation of the Autoimmune Regulator Gene in an Italian Kindred with Autoimmune Polyendocrinopathy-Candidiasis-Ectodermal Dystrophy, Acting in a Dominant Fashion and Strongly Cosegregating with Hypothyroid Autoimmune Thyroiditis. J. Clin. Endocrinol. Metab. 2001, 86, 4747–4752. [Google Scholar] [CrossRef]
  47. Cervato, S.; Mariniello, B.; Lazzarotto, F.; Morlin, L.; Zanchetta, R.; Radetti, G.; De Luca, F.; Valenzise, M.; Giordano, R.; Rizzo, D.; et al. Evaluation of the Autoimmune Regulator (AIRE) Gene Mutations in a Cohort of Italian Patients with Autoimmune-Polyendocrinopathy-Candidiasis-Ectodermal-Dystrophy (APECED) and in Their Relatives. Clin. Endocrinol. 2009, 70, 421–428. [Google Scholar] [CrossRef]
  48. Kogawa, K.; Kudoh, J.; Nagafuchi, S.; Ohga, S.; Katsuta, H.; Ishibashi, H.; Harada, M.; Hara, T.; Shimizu, N. Distinct Clinical Phenotype and Immunoreactivity in Japanese Siblings with Autoimmune Polyglandular Syndrome Type 1 (APS-1) Associated with Compound Heterozygous Novel AIRE Gene Mutations. Clin. Immunol. 2002, 103, 277–283. [Google Scholar] [CrossRef]
  49. Laakso, S.M.; Laurinolli, T.T.; Rossi, L.H.; Lehtoviita, A.; Sairanen, H.; Perheentupa, J.; Kekäläinen, E.; Arstila, T.P. Regulatory T Cell Defect in APECED Patients Is Associated with Loss of Naive FOXP3+ Precursors and Impaired Activated Population. J. Autoimmun. 2010, 35, 351–357. [Google Scholar] [CrossRef]
  50. Pöntynen, N.; Strengell, M.; Sillanpää, N.; Saharinen, J.; Ulmanen, I.; Julkunen, I.; Peltonen, L. Critical Immunological Pathways Are Downregulated in APECED Patient Dendritic Cells. J. Mol. Med. 2008, 86, 1139–1152. [Google Scholar] [CrossRef] [Green Version]
  51. Gebre-Medhin, G.; Husebye, E.S.; Gustafsson, J.; Winqvist, O.; Goksøyr, A.; Rorsman, F.; Kämpe, O. Cytochrome P450IA2 and Aromatic L-Amino Acid Decarboxylase Are Hepatic Autoantigens in Autoimmune Polyendocrine Syndrome Type I. FEBS Lett. 1997, 412, 439–445. [Google Scholar] [CrossRef] [Green Version]
  52. Meloni, A.; Furcas, M.; Cetani, F.; Marcocci, C.; Falorni, A.; Perniola, R.; Pura, M.; Bøe Wolff, A.S.; Husebye, E.S.; Lilic, D.; et al. Autoantibodies against Type I Interferons as an Additional Diagnostic Criterion for Autoimmune Polyendocrine Syndrome Type I. J. Clin. Endocrinol. Metab. 2008, 93, 4389–4397. [Google Scholar] [CrossRef] [Green Version]
  53. Alimohammadi, M.; Björklund, P.; Hallgren, Å.; Pöntynen, N.; Szinnai, G.; Shikama, N.; Keller, M.P.; Ekwall, O.; Kinkel, S.A.; Husebye, E.S.; et al. Autoimmune Polyendocrine Syndrome Type 1 and NALP5, a Parathyroid Autoantigen. N. Engl. J. Med. 2008, 358, 1018–1028. [Google Scholar] [CrossRef] [Green Version]
  54. Puel, A.; Döffinger, R.; Natividad, A.; Chrabieh, M.; Barcenas-Morales, G.; Picard, C.; Cobat, A.; Ouachée-Chardin, M.; Toulon, A.; Bustamante, J.; et al. Autoantibodies against IL-17A, IL-17F, and IL-22 in Patients with Chronic Mucocutaneous Candidiasis and Autoimmune Polyendocrine Syndrome Type I. J. Exp. Med. 2010, 207, 291–297. [Google Scholar] [CrossRef] [Green Version]
  55. de Albuquerque, J.A.T.; Banerjee, P.P.; Castold, A.; Ma, R.; Zurro, N.B.; Ynoue, L.H.; Arslanian, C.; Barbosa-Carvalho, M.U.W.; de Menezes Correia-Deur, J.E.; Weiler, F.G.; et al. The Role of AIRE in the Immunity Against Candida Albicans in a Model of Human Macrophages. Front. Immunol. 2018, 9, 567. [Google Scholar] [CrossRef] [Green Version]
  56. Kluger, N.; Jokinen, M.; Krohn, K.; Ranki, A. Gastrointestinal Manifestations in APECED Syndrome. J. Clin. Gastroenterol. 2013, 47, 112–120. [Google Scholar] [CrossRef]
  57. Kluger, N.; Jokinen, M.; Lintulahti, A.; Krohn, K.; Ranki, A. Gastrointestinal Immunity against Tryptophan Hydroxylase-1, Aromatic L-Amino-Acid Decarboxylase, AIE-75, Villin and Paneth Cells in APECED. Clin. Immunol. 2015, 158, 212–220. [Google Scholar] [CrossRef]
  58. Dobeš, J.; Neuwirth, A.; Dobešová, M.; Vobořil, M.; Balounová, J.; Ballek, O.; Lebl, J.; Meloni, A.; Krohn, K.; Kluger, N.; et al. Gastrointestinal Autoimmunity Associated with Loss of Central Tolerance to Enteric α-Defensins. Gastroenterology 2015, 149, 139–150. [Google Scholar] [CrossRef] [Green Version]
  59. Salzman, N.H.; Hung, K.; Haribhai, D.; Chu, H.; Karlsson-Sjöberg, J.; Amir, E.; Teggatz, P.; Barman, M.; Hayward, M.; Eastwood, D.; et al. Enteric Defensins Are Essential Regulators of Intestinal Microbial Ecology. Nat. Immunol. 2009, 11, 76–82. [Google Scholar] [CrossRef] [PubMed]
  60. Petersen, A.; Jokinen, M.; Plichta, D.R.; Liebisch, G.; Gronwald, W.; Dettmer, K.; Oefner, P.J.; Vlamakis, H.; Chung, D.C.; Ranki, A.; et al. Cytokine-Specific Autoantibodies Shape the Gut Microbiome in Autoimmune Polyendocrine Syndrome Type 1. J. Allergy Clin. Immunol. 2021, 148, 876–888. [Google Scholar] [CrossRef] [PubMed]
  61. Hetemäki, I.; Jarva, H.; Kluger, N.; Baldauf, H.-M.; Laakso, S.; Bratland, E.; Husebye, E.S.; Kisand, K.; Ranki, A.; Peterson, P.; et al. Anticommensal Responses Are Associated with Regulatory T Cell Defect in Autoimmune Polyendocrinopathy-Candidiasis-Ectodermal Dystrophy Patients. J. Immunol. 2016, 196, 2955–2964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Zhang, Z.; Li, C.; Zhao, X.; Lv, C.; He, Q.; Lei, S.; Guo, Y.; Zhi, F. Anti-Saccharomyces Cerevisiae Antibodies Associate with Phenotypes and Higher Risk for Surgery in Crohn’s Disease: A Meta-Analysis. Dig. Dis. Sci. 2012, 57, 2944–2954. [Google Scholar] [CrossRef]
  63. Kawamoto, S.; Maruya, M.; Kato, L.M.; Suda, W.; Atarashi, K.; Doi, Y.; Tsutsui, Y.; Qin, H.; Honda, K.; Okada, T.; et al. Foxp3(+) T Cells Regulate Immunoglobulin a Selection and Facilitate Diversification of Bacterial Species Responsible for Immune Homeostasis. Immunity 2014, 41, 152–165. [Google Scholar] [CrossRef] [Green Version]
  64. He, B.; Hoang, T.K.; Wang, T.; Ferris, M.; Taylor, C.M.; Tian, X.; Luo, M.; Tran, D.Q.; Zhou, J.; Tatevian, N.; et al. Resetting Microbiota by Lactobacillus Reuteri Inhibits T Reg Deficiency-Induced Autoimmunity via Adenosine A2A Receptors. J. Exp. Med. 2017, 214, 107–123. [Google Scholar] [CrossRef]
  65. Winer, K.K.; Schmitt, M.M.; Ferre, E.M.N.; Fennelly, K.P.; Olivier, K.N.; Heller, T.; Lionakis, M.S. Impact of Periprocedural Subcutaneous Parathyroid Hormone on Control of Hypocalcaemia in APS-1/APECED Patients Undergoing Invasive Procedures. Clin. Endocrinol. 2021, 94, 377–383. [Google Scholar] [CrossRef]
  66. Halabi, I.; Barohom, M.N.; Peleg, S.; Trougouboff, P.; Elias-Assad, G.; Agbaria, R.; Tenenbaum-Rakover, Y. Case Report: Severe Hypocalcemic Episodes Due to Autoimmune Enteropathy. Front. Endocrinol. 2021, 12, 674. [Google Scholar] [CrossRef]
  67. Powell, B.R.; Buist, N.R.M.; Stenzel, P. An X-Linked Syndrome of Diarrhea, Polyendocrinopathy, and Fatal Infection in Infancy. J. Pediatr. 1982, 100, 731–737. [Google Scholar] [CrossRef] [PubMed]
  68. Wildin, R.S.; Smyk-Pearson, S.; Filipovich, A.H. Clinical and Molecular Features of the Immunodysregulation, Polyendocrinopathy, Enteropathy, X Linked (IPEX) Syndrome. J. Med. Genet. 2002, 39, 537–545. [Google Scholar] [CrossRef] [Green Version]
  69. Bennett, C.L.; Yoshioka, R.; Kiyosawa, H.; Barker, D.F.; Fain, P.R.; Shigeoka, A.O.; Chance, P.F. X-Linked Syndrome of Polyendocrinopathy, Immune Dysfunction, and Diarrhea Maps to Xp11.23-Xq13.3. Am. J. Hum. Genet. 2000, 66, 461–468. [Google Scholar] [CrossRef] [Green Version]
  70. Passerini, L.; Santoni De Sio, F.R.; Roncarolo, M.G.; Bacchetta, R. Forkhead Box P3: The Peacekeeper of the Immune System. Int. Rev. Immunol. 2014, 33, 129–145. [Google Scholar] [CrossRef] [Green Version]
  71. Bennett, C.L.; Christie, J.; Ramsdell, F.; Brunkow, M.E.; Ferguson, P.J.; Whitesell, L.; Kelly, T.E.; Saulsbury, F.T.; Chance, P.F.; Ochs, H.D. The Immune Dysregulation, Polyendocrinopathy, Enteropathy, X-Linked Syndrome (IPEX) Is Caused by Mutations of FOXP3. Nat. Genet. 2001, 27, 20–21. [Google Scholar] [CrossRef] [PubMed]
  72. Bacchetta, R.; Passerini, L.; Gambineri, E.; Dai, M.; Allan, S.E.; Perroni, L.; Dagna-Bricarelli, F.; Sartirana, C.; Matthes-Martin, S.; Lawitschka, A.; et al. Defective Regulatory and Effector T Cell Functions in Patients with FOXP3 Mutations. J. Clin. Investig. 2006, 116, 1713–1722. [Google Scholar] [CrossRef] [PubMed]
  73. Gambineri, E.; Perroni, L.; Passerini, L.; Bianchi, L.; Doglioni, C.; Meschi, F.; Bonfanti, R.; Sznajer, Y.; Tommasini, A.; Lawitschka, A.; et al. Clinical and Molecular Profile of a New Series of Patients with Immune Dysregulation, Polyendocrinopathy, Enteropathy, X-Linked Syndrome: Inconsistent Correlation between Forkhead Box Protein 3 Expression and Disease Severity. J. Allergy Clin. Immunol. 2008, 122. [Google Scholar] [CrossRef] [PubMed]
  74. Passerini, L.; Olek, S.; Di Nunzio, S.; Barzaghi, F.; Hambleton, S.; Abinun, M.; Tommasini, A.; Vignola, S.; Cipolli, M.; Amendola, M.; et al. Forkhead Box Protein 3 (FOXP3) Mutations Lead to Increased TH17 Cell Numbers and Regulatory T-Cell Instability. J. Allergy Clin. Immunol. 2011, 128, 1376–1379. [Google Scholar] [CrossRef]
  75. Nieves, D.S.; Phipps, R.P.; Pollock, S.J.; Ochs, H.D.; Zhu, Q.; Scott, G.A.; Ryan, C.K.; Kobayashi, I.; Rossi, T.M.; Goldsmith, L.A. Dermatologic and Immunologic Findings in the Immune Dysregulation, Polyendocrinopathy, Enteropathy, X-Linked Syndrome. Arch. Derm. 2004, 140, 466–472. [Google Scholar] [CrossRef] [Green Version]
  76. Chatila, T.A.; Blaeser, F.; Ho, N.; Lederman, H.M.; Voulgaropoulos, C.; Helms, C.; Bowcock, A.M. JM2, Encoding a Fork Head-Related Protein, Is Mutated in X-Linked Autoimmunity-Allergic Disregulation Syndrome. J. Clin. Investig. 2000, 106, R75–R81. [Google Scholar] [CrossRef] [Green Version]
  77. Ben-skowronek, I. IPEX Syndrome: Genetics and Treatment Options. Genes 2021, 12, 323. [Google Scholar] [CrossRef]
  78. Goodwin, M.; Lee, E.; Lakshmanan, U.; Shipp, S.; Froessl, L.; Barzaghi, F.; Passerini, L.; Narula, M.; Sheikali, A.; Lee, C.M.; et al. CRISPR-Based Gene Editing Enables FOXP3 Gene Repair in IPEX Patient Cells. Sci. Adv. 2020, 6, eaaz0571. [Google Scholar] [CrossRef]
  79. Wu, W.; Shen, N.; Luo, L.; Deng, Z.; Chen, J.; Tao, Y.; Mo, X.; Cao, Q. Fecal Microbiota Transplantation before Hematopoietic Stem Cell Transplantation in a Pediatric Case of Chronic Diarrhea with a FOXP3 Mutation. Pediatr. Neonatol. 2021, 62, 172–180. [Google Scholar] [CrossRef]
  80. Chinen, T.; Volchkov, P.Y.; Chervonsky, A.V.; Rudensky, A.Y. A Critical Role for Regulatory T Cell-Mediated Control of Inflammation in the Absence of Commensal Microbiota. J. Exp. Med. 2010, 207, 2323–2330. [Google Scholar] [CrossRef] [Green Version]
  81. Wildin, R.S.; Ramsdell, F.; Peake, J.; Faravelli, F.; Casanova, J.L.; Buist, N.; Levy-Lahad, E.; Mazzella, M.; Goulet, O.; Perroni, L.; et al. X-Linked Neonatal Diabetes Mellitus, Enteropathy and Endocrinopathy Syndrome Is the Human Equivalent of Mouse Scurfy. Nat. Genet. 2001, 27, 18–20. [Google Scholar] [CrossRef]
  82. Bleesing, J.J.H. Autoimmune Lymphoproliferative Syndrome: A Genetic Disorder of Abnormal Lymphocyte Apoptosis. Immunol. Allergy Clin. N. Am. 2002, 22, 339–355. [Google Scholar] [CrossRef]
  83. Straus, S.E.; Jaffe, E.S.; Puck, J.M.; Dale, J.K.; Elkon, K.B.; Rösen-Wolff, A.; Peters, A.M.J.; Sneller, M.C.; Hallahan, C.W.; Wang, J.; et al. The Development of Lymphomas in Families with Autoimmune Lymphoproliferative Syndrome with Germline Fas Mutations and Defective Lymphocyte Apoptosis. Blood 2001, 98, 194–200. [Google Scholar] [CrossRef]
  84. Oliveira, J.B.; Bleesing, J.J.; Dianzani, U.; Fleisher, T.A.; Jaffe, E.S.; Lenardo, M.J.; Rieux-Laucat, F.; Siegel, R.M.; Su, H.C.; Teachey, D.T.; et al. Revised Diagnostic Criteria and Classification for the Autoimmune Lymphoproliferative Syndrome (ALPS): Report from the 2009 NIH International Workshop. Blood 2010, 116, e35. [Google Scholar] [CrossRef] [Green Version]
  85. Sneller, M.C.; Straus, S.E.; Jaffe, E.S.; Jaffe, J.S.; Fleisher, T.A.; Stetler- Stevenson, M.; Strober, W. A Novel Lymphoproliferative/Autoimmune Syndrome Resembling Murine Lpr/Gld Disease. J. Clin. Investig. 1992, 90, 334–341. [Google Scholar] [CrossRef] [Green Version]
  86. Dianzani, U.; Bragardo, M.; DiFranco, D.; Alliaudi, C.; Scagni, P.; Buonfiglio, D.; Redoglia, V.; Bonissoni, S.; Correra, A.; Dianzani, I.; et al. Deficiency of the Fas Apoptosis Pathway Without Fas Gene Mutations in Pediatric Patients With Autoimmunity/Lymphoproliferation. Blood 1997, 89, 2871–2879. [Google Scholar] [CrossRef]
  87. Dean, G.S.; Anand, A.; Blofeld, A.; Isenberg, D.A.; Lydyard, P.M. Characterization of CD3+ CD4- CD8- (Double Negative) T Cells in Patients with Systemic Lupus Erythematosus: Production of IL-4. Lupus 2002, 11, 501–507. [Google Scholar] [CrossRef]
  88. Rensing-Ehl, A.; Völkl, S.; Speckmann, C.; Lorenz, M.R.; Ritter, J.; Janda, A.; Abinun, M.; Pircher, H.; Bengsch, B.; Thimme, R.; et al. Abnormally Differentiated CD4+ or CD8+ T Cells with Phenotypic and Genetic Features of Double Negative T Cells in Human Fas Deficiency. Blood 2014, 124, 851–860. [Google Scholar] [CrossRef] [Green Version]
  89. Bristeau-Leprince, A.; Mateo, V.; Lim, A.; Magerus-Chatinet, A.; Solary, E.; Fischer, A.; Rieux-Laucat, F.; Gougeon, M.-L. Human TCR Alpha/Beta+ CD4-CD8- Double-Negative T Cells in Patients with Autoimmune Lymphoproliferative Syndrome Express Restricted Vbeta TCR Diversity and Are Clonally Related to CD8+ T Cells. J. Immunol. 2008, 181, 440–448. [Google Scholar] [CrossRef] [Green Version]
  90. Bowen, R.A.R.; Dowdell, K.C.; Dale, J.K.; Drake, S.K.; Fleisher, T.A.; Hortin, G.L.; Remaley, A.T.; Nexo, E.; Rao, V.K. Elevated Vitamin B₁₂ Levels in Autoimmune Lymphoproliferative Syndrome Attributable to Elevated Haptocorrin in Lymphocytes. Clin. Biochem. 2012, 45, 490–492. [Google Scholar] [CrossRef] [Green Version]
  91. Magerus-Chatinet, A.; Stolzenberg, M.C.; Loffredo, M.S.; Neven, B.; Schaffner, C.; Ducrot, N.; Arkwright, P.D.; Bader-Meunier, B.; Barbot, J.; Blanche, S.; et al. FAS-L, IL-10, and Double-Negative CD4- CD8- TCR Alpha/Beta+ T Cells Are Reliable Markers of Autoimmune Lymphoproliferative Syndrome (ALPS) Associated with FAS Loss of Function. Blood 2009, 113, 3027–3030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Watanabe-Fukunaga, R.; Brannan, C.I.; Copeland, N.G.; Jenkins, N.A.; Nagata, S. Lymphoproliferation Disorder in Mice Explained by Defects in Fas Antigen That Mediates Apoptosis. Nature 1992, 356, 314–317. [Google Scholar] [CrossRef] [PubMed]
  93. Takahashi, T.; Tanaka, M.; Brannan, C.I.; Jenkins, N.A.; Copeland, N.G.; Suda, T.; Nagata, S. Generalized Lymphoproliferative Disease in Mice, Caused by a Point Mutation in the Fas Ligand. Cell 1994, 76, 969–976. [Google Scholar] [CrossRef] [PubMed]
  94. Consonni, F.; Gambineri, E.; Favre, C. ALPS, FAS, and beyond: From Inborn Errors of Immunity to Acquired Immunodeficiencies. Ann. Hematol. 2022, 101, 469–484. [Google Scholar] [CrossRef]
  95. Strasser, A.; Jost, P.J.; Nagata, S. The Many Roles of FAS Receptor Signaling in the Immune System. Immunity 2009, 30, 180–192. [Google Scholar] [CrossRef] [Green Version]
  96. Clementi, R.; Dagna, L.; Dianzani, U.; Dupré, L.; Dianzani, I.; Ponzoni, M.; Cometa, A.; Chiocchetti, A.; Sabbadini, M.G.; Rugarli, C.; et al. Inherited Perforin and Fas Mutations in a Patient with Autoimmune Lymphoproliferative Syndrome and Lymphoma. N. Engl. J. Med. 2004, 351, 1419–1424. [Google Scholar] [CrossRef]
  97. Cerutti, E.; Campagnoli, M.F.; Ferretti, M.; Garelli, E.; Crescenzio, N.; Rosolen, A.; Chiocchetti, A.; Lenardo, M.J.; Ramenghi, U.; Dianzani, U. Co-Inherited Mutations of Fas and Caspase-10 in Development of the Autoimmune Lymphoproliferative Syndrome. BMC Immunol. 2007, 8, 1–9. [Google Scholar] [CrossRef] [Green Version]
  98. Holzelova, E.; Vonarbourg, C.; Stolzenberg, M.-C.; Arkwright, P.D.; Selz, F.; Prieur, A.-M.; Blanche, S.; Bartunkova, J.; Vilmer, E.; Fischer, A.; et al. Autoimmune Lymphoproliferative Syndrome with Somatic Fas Mutations. N. Engl. J. Med. 2004, 351, 1409–1418. [Google Scholar] [CrossRef]
  99. Dowdell, K.C.; Niemela, J.E.; Price, S.; Davis, J.; Hornung, R.L.; Oliveira, J.B.; Puck, J.M.; Jaffe, E.S.; Pittaluga, S.; Cohen, J.I.; et al. Somatic FAS Mutations Are Common in Patients with Genetically Undefined Autoimmune Lymphoproliferative Syndrome. Blood 2010, 115, 5164–5169. [Google Scholar] [CrossRef] [Green Version]
  100. Campagnoli, M.F.; Garbarini, L.; Quarello, P.; Garelli, E.; Carando, A.; Baravalle, V.; Doria, A.; Biava, A.; Chiocchetti, A.; Rosolen, A.; et al. The Broad Spectrum of Autoimmune Lymphoproliferative Disease: Molecular Bases, Clinical Features and Long-Term Follow-up in 31 Patients. Haematologica 2006, 91, 538–541. [Google Scholar]
  101. Aricò, M.; Boggio, E.; Cetica, V.; Melensi, M.; Orilieri, E.; Clemente, N.; Cappellano, G.; Buttini, S.; Soluri, M.F.; Comi, C.; et al. Variations of the UNC13D Gene in Patients with Autoimmune Lymphoproliferative Syndrome. PLoS ONE 2013, 8, e68045. [Google Scholar] [CrossRef]
  102. Clementi, R.; Chiocchetti, A.; Cappellano, G.; Cerutti, E.; Ferretti, M.; Orilieri, E.; Dianzani, I.; Ferrarini, M.; Bregni, M.; Danesino, C.; et al. Variations of the Perforin Gene in Patients with Autoimmunity/Lymphoproliferation and Defective Fas Function. Blood 2006, 108, 3079–3084. [Google Scholar] [CrossRef]
  103. Booth, C.; Gilmour, K.C.; Veys, P.; Gennery, A.R.; Slatter, M.A.; Chapel, H.; Heath, P.T.; Steward, C.G.; Smith, O.; O’Meara, A.; et al. X-Linked Lymphoproliferative Disease Due to SAP/SH2D1A Deficiency: A Multicenter Study on the Manifestations, Management and Outcome of the Disease. Blood 2011, 117, 53–62. [Google Scholar] [CrossRef] [Green Version]
  104. Comi, C.; Leone, M.; Bonissoni, S.; DeFranco, S.; Bottarel, F.; Mezzatesta, C.; Chiocchetti, A.; Perla, F.; Monaco, F.; Dianzani, U. Defective T Cell Fas Function in Patients with Multiple Sclerosis. Neurology 2000, 55, 921–927. [Google Scholar] [CrossRef]
  105. DeFranco, S.; Bonissoni, S.; Cerutti, F.; Bona, G.; Bottarel, F.; Cadario, F.; Brusco, A.; Loffredo, G.; Rabbone, I.; Corrias, A.; et al. Defective Function of Fas in Patients with Type 1 Diabetes Associated with Other Autoimmune Diseases. Diabetes 2001, 50, 483–488. [Google Scholar] [CrossRef] [Green Version]
  106. Cheng, J.; Zhou, T.; Liu, C.; Shapiro, J.P.; Brauer, M.J.; Kiefer, M.C.; Barr, P.J.; Mountz, J.D. Protection from Fas-Mediated Apoptosis by a Soluble Form of the Fas Molecule. Science 1994, 263, 1759–1762. [Google Scholar] [CrossRef]
  107. Teachey, D.T.; Seif, A.E.; Grupp, S.A. Advances in the Management and Understanding of Autoimmune Lymphoproliferative Syndrome (ALPS). Br. J. Haematol. 2010, 148, 205–216. [Google Scholar] [CrossRef]
  108. Wang, J.; Zheng, L.; Lobito, A.; Chan, F.K.M.; Dale, J.; Sneller, M.; Yao, X.; Puck, J.M.; Straus, S.E.; Lenardo, M.J. Inherited Human Caspase 10 Mutations Underlie Defective Lymphocyte and Dendritic Cell Apoptosis in Autoimmune Lymphoproliferative Syndrome Type II. Cell 1999, 98, 47–58. [Google Scholar] [CrossRef] [Green Version]
  109. Stranges, P.B.; Watson, J.; Cooper, C.J.; Choisy-Rossi, C.M.; Stonebraker, A.C.C.; Beighton, R.A.; Hartig, H.; Sundberg, J.P.; Servick, S.; Kaufmann, G.; et al. Elimination of Antigen-Presenting Cells and Autoreactive T Cells by Fas Contributes to Prevention of Autoimmunity. Immunity 2007, 26, 629–641. [Google Scholar] [CrossRef] [Green Version]
  110. Yu, C.; Gershwin, M.E.; Chang, C. Diagnostic Criteria for Systemic Lupus Erythematosus: A Critical Review. J. Autoimmun. 2014, 48–49, 10–13. [Google Scholar] [CrossRef]
  111. Namjou, B.; Kilpatrick, J.; Harley, J.B. Genetics of Clinical Expression in SLE. Autoimmunity 2007, 40, 602–612. [Google Scholar] [CrossRef] [PubMed]
  112. Belot, A.; Cimaz, R. Monogenic Forms of Systemic Lupus Erythematosus: New Insights into SLE Pathogenesis. Pediatr. Rheumatol. Online J. 2012, 10, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Leffler, J.; Bengtsson, A.A.; Blom, A.M. The Complement System in Systemic Lupus Erythematosus: An Update. Ann. Rheum. Dis. 2014, 73, 1601–1606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Tuveson, D.A.; Ahearn, J.M.; Matsumoto, A.K.; Fearon, D.T. Molecular Interactions of Complement Receptors on B Lymphocytes: A CR1/CR2 Complex Distinct from the CR2/CD19 Complex. J. Exp. Med. 1991, 173, 1083–1089. [Google Scholar] [CrossRef] [PubMed]
  115. Kemper, C.; Chan, A.C.; Green, J.M.; Brett, K.A.; Murphy, K.M.; Atkinson, J.P. Activation of Human CD4+ Cells with CD3 and CD46 Induces a T-Regulatory Cell 1 Phenotype. Nature 2003, 421, 388–392. [Google Scholar] [CrossRef]
  116. Lood, C.; Gullstrand, B.; Truedsson, L.; Olin, A.I.; Alm, G.V.; Rönnblom, L.; Sturfelt, G.; Eloranta, M.L.; Bengtsson, A.A. C1q Inhibits Immune Complex-Induced Interferon-α Production in Plasmacytoid Dendritic Cells. Arthritis Rheum. 2009, 60, 3081–3090. [Google Scholar] [CrossRef]
  117. Ling, G.S.; Crawford, G.; Buang, N.; Bartok, I.; Tian, K.; Thielens, N.M.; Bally, I.; Harker, J.A.; Ashton-Rickardt, P.G.; Rutschmann, S.; et al. C1q Restrains Autoimmunity and Viral Infection by Regulating CD8+ T Cell Metabolism. Science (1979) 2018, 360, 558–563. [Google Scholar] [CrossRef] [Green Version]
  118. Stegert, M.; Bock, M.; Trendelenburg, M. Clinical Presentation of Human C1q Deficiency: How Much of a Lupus? Mol. Immunol. 2015, 67, 3–11. [Google Scholar] [CrossRef]
  119. Macedo, A.C.L.; Isaac, L. Systemic Lupus Erythematosus and Deficiencies of Early Components of the Complement Classical Pathway. Front. Immunol. 2016, 7, 55. [Google Scholar] [CrossRef] [Green Version]
  120. Elkon, K.B. Cell Death, Nucleic Acids, and Immunity: Inflammation Beyond the Grave. Arthritis Rheumatol. 2018, 70, 805–816. [Google Scholar] [CrossRef] [Green Version]
  121. Wu, J.; Wilson, J.; He, J.; Xiang, L.; Schur, P.H.; Mountz, J.D. Fas Ligand Mutation in a Patient with Systemic Lupus Erythematosus and Lymphoproliferative Disease. J. Clin. Investig. 1996, 98, 1107–1113. [Google Scholar] [CrossRef] [Green Version]
  122. Vaishnaw, A.K.; Toubi, E.; Ohsako, S.; Drappa, J.; Buys, S.; Estrada, J.; Sitarz, A.; Zemel, L.; Chu, J.-L.; Elkon, K.B. THE SPECTRUM OF APOPTOTIC DEFECTS AND CLINICAL MANIFESTATIONS, INCLUDING SYSTEMIC LUPUS ERYTHEMATOSUS, IN HUMANS WITH CD95 (Fas/APO-1) MUTATIONS. Arthritis Rheum. 1999, 42, 1833–1842. [Google Scholar] [CrossRef]
  123. Belot, A.; Kasher, P.R.; Trotter, E.W.; Foray, A.P.; Debaud, A.L.; Rice, G.I.; Szynkiewicz, M.; Zabot, M.T.; Rouvet, I.; Bhaskar, S.S.; et al. Protein Kinase Cδ Deficiency Causes Mendelian Systemic Lupus Erythematosus with B Cell-Defective Apoptosis and Hyperproliferation. Arthritis Rheum. 2013, 65, 2161–2171. [Google Scholar] [CrossRef] [Green Version]
  124. Limnander, A.; Zikherman, J.; Lau, T.; Leitges, M.; Weiss, A.; Roose, J.P. Protein Kinase Cδ Promotes Transitional B Cell-Negative Selection and Limits Proximal B Cell Receptor Signaling to Enforce Tolerance. Mol. Cell Biol. 2014, 34, 1474–1485. [Google Scholar] [CrossRef] [Green Version]
  125. Kiykim, A.; Ogulur, I.; Baris, S.; Salzer, E.; Karakoc-Aydiner, E.; Ozen, A.O.; Garncarz, W.; Hirschmugl, T.; Krolo, A.; Yucelten, A.D.; et al. Potentially Beneficial Effect of Hydroxychloroquine in a Patient with a Novel Mutation in Protein Kinase Cδ Deficiency. J. Clin. Immunol. 2015, 35, 523–526. [Google Scholar] [CrossRef]
  126. Pullabhatla, V.; Roberts, A.L.; Lewis, M.J.; Mauro, D.; Morris, D.L.; Odhams, C.A.; Tombleson, P.; Liljedahl, U.; Vyse, S.; Simpson, M.A.; et al. De Novo Mutations Implicate Novel Genes in Systemic Lupus Erythematosus. Hum. Mol. Genet. 2018, 27, 421–429. [Google Scholar] [CrossRef] [Green Version]
  127. Walter, J.E.; Lo, M.S.; Kis-Toth, K.; Tirosh, I.; Frugoni, F.; Lee, Y.N.; Csomos, K.; Chen, K.; Pillai, S.; Dunham, J.; et al. Impaired Receptor Editing and Heterozygous RAG2 Mutation in a Patient with Systemic Lupus Erythematosus and Erosive Arthritis. J. Allergy Clin. Immunol. 2015, 135, 272–273. [Google Scholar] [CrossRef] [Green Version]
  128. Ragotte, R.J.; Dhanrajani, A.; Pleydell-Pearce, J.; Del Bel, K.L.; Tarailo-Graovac, M.; van Karnebeek, C.; Terry, J.; Senger, C.; McKinnon, M.L.; Seear, M.; et al. The Importance of Considering Monogenic Causes of Autoimmunity: A Somatic Mutation in KRAS Causing Pediatric Rosai-Dorfman Syndrome and Systemic Lupus Erythematosus. Clin. Immunol. 2017, 175, 143–146. [Google Scholar] [CrossRef]
  129. Hebron, K.E.; Hernandez, E.R.; Yohe, M.E. The RASopathies: From Pathogenetics to Therapeutics. Dis. Model. Mech. 2022, 15. [Google Scholar] [CrossRef]
  130. Leventopoulos, G.; Denayer, E.; Makrythanasis, P.; Papapolychroniou, C.; Fryssira, H. Noonan Syndrome and Systemic Lupus Erythematosus in a Patient with a Novel KRAS Mutation. Clin. Exp. Rheumatol. 2010, 28, 556–557. [Google Scholar]
  131. Hiraki, L.T.; Silverman, E.D. Genomics of Systemic Lupus Erythematosus: Insights Gained by Studying Monogenic Young-Onset Systemic Lupus Erythematosus. Rheum. Dis. Clin. N. Am. 2017, 43, 415–434. [Google Scholar] [CrossRef] [PubMed]
  132. Stetson, D.B.; Ko, J.S.; Heidmann, T.; Medzhitov, R. Trex1 Prevents Cell-Intrinsic Initiation of Autoimmunity. Cell 2008, 134, 587–598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Sparks, J.L.; Chon, H.; Cerritelli, S.M.; Kunkel, T.A.; Johansson, E.; Crouch, R.J.; Burgers, P.M. RNase H2-Initiated Ribonucleotide Excision Repair. Mol. Cell 2012, 47, 980–986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Rice, G.I.; Rodero, M.P.; Crow, Y.J. Human Disease Phenotypes Associated with Mutations in TREX1. J. Clin. Immunol. 2015, 35, 235–243. [Google Scholar] [CrossRef] [PubMed]
  135. Crow, Y.J.; Chase, D.S.; Lowenstein Schmidt, J.; Szynkiewicz, M.; Forte, G.M.A.; Gornall, H.L.; Oojageer, A.; Anderson, B.; Pizzino, A.; Helman, G.; et al. Characterization of Human Disease Phenotypes Associated with Mutations in TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, ADAR, and IFIH1. Am. J. Med. Genet. A 2015, 167A, 296–312. [Google Scholar] [CrossRef] [Green Version]
  136. Mackenzie, K.J.; Carroll, P.; Lettice, L.; Tarnauskait, Z.; Reddy, K.; Dix, F.; Revuelta, A.; Abbondati, E.; Rigby, R.E.; Rabe, B.; et al. Ribonuclease H2 Mutations Induce a CGAS/STING-Dependent Innate Immune Response. EMBO J. 2016, 35, 831–844. [Google Scholar] [CrossRef]
  137. Ramantani, G.; Kohlhase, J.; Hertzberg, C.; Micheil Innes, A.; Engel, K.; Hunger, S.; Borozdin, W.; Mah, J.K.; Ungerath, K.; Walkenhorst, H.; et al. Expanding the Phenotypic Spectrum of Lupus Erythematosus in Aicardi-Goutières Syndrome. Arthritis Rheum. 2010, 62, 1469–1477. [Google Scholar] [CrossRef]
  138. Ellyard, J.I.; Jerjen, R.; Martin, J.L.; Lee, A.Y.S.; Field, M.A.; Jiang, S.H.; Cappello, J.; Naumann, S.K.; Andrews, T.D.; Scott, H.S.; et al. Brief Report: Identification of a Pathogenic Variant in Trex1 in Early-Onset Cerebral Systemic Lupus Erythematosus by Whole-Exome Sequencing. Arthritis Rheumatol.. 2014, 66, 3382–3386. [Google Scholar] [CrossRef]
  139. Lebon, P.; Badoual, J.; Ponsot, G.; Goutières, F.; Hémeury-Cukier, F.; Aicardi, J. Intrathecal Synthesis of Interferon-Alpha in Infants with Progressive Familial Encephalopathy. J. Neurol. Sci. 1988, 84, 201–208. [Google Scholar] [CrossRef]
  140. Crow, M.K. Pathogenesis of Systemic Lupus Erythematosus: Risks, Mechanisms and Therapeutic Targets. Ann. Rheum. Dis. 2023, ard-2022-223741. [Google Scholar] [CrossRef]
  141. Bengtsson, A.A.; Rönnblom, L. Role of Interferons in SLE. Best. Pract. Res. Clin. Rheumatol. 2017, 31, 415–428. [Google Scholar] [CrossRef]
  142. Al-Mayouf, S.M.; Sunker, A.; Abdwani, R.; Al Abrawi, S.; Almurshedi, F.; Alhashmi, N.; Al Sonbul, A.; Sewairi, W.; Qari, A.; Abdallah, E.; et al. Loss-of-Function Variant in DNASE1L3 Causes a Familial Form of Systemic Lupus Erythematosus. Nat. Genet. 2011, 43, 1186–1188. [Google Scholar] [CrossRef]
  143. Hartl, J.; Serpas, L.; Wang, Y.; Rashidfarrokhi, A.; Perez, O.A.; Sally, B.; Sisirak, V.; Soni, C.; Khodadadi-Jamayran, A.; Tsirigos, A.; et al. Autoantibody-Mediated Impairment of DNASE1L3 Activity in Sporadic Systemic Lupus Erythematosus. J. Exp. Med. 2021, 218, e20201138. [Google Scholar] [CrossRef]
  144. Pisetsky, D.S. The Expression of HMGB1 on Microparticles Released during Cell Activation and Cell Death in Vitro and in Vivo. Mol. Med. 2014, 20, 158–163. [Google Scholar] [CrossRef]
  145. Bodaño, A.; Amarelo, J.; González, A.; Gómez-Reino, J.J.; Conde, C. Novel DNASE I Mutations Related to Systemic Lupus Erythematosus. Arthritis Rheum. 2004, 50, 4070–4071. [Google Scholar] [CrossRef]
  146. Kara, B.; Ekinci, Z.; Sahin, S.; Gungor, M.; Gunes, A.S.; Ozturk, K.; Adrovic, A.; Cefle, A.; Inanç, M.; Gul, A.; et al. Monogenic Lupus Due to Spondyloenchondrodysplasia with Spastic Paraparesis and Intracranial Calcification: Case-Based Review. Rheumatol. Int. 2020, 40, 1903–1910. [Google Scholar] [CrossRef]
  147. Bilginer, Y.; Düzova, A.; Topaloaylu, R.; Batu, E.D.; Boduroaylu, K.; Güçer, A.; Bodur, A.; Alanay, Y. Three Cases of Spondyloenchondrodysplasia (SPENCD) with Systemic Lupus Erythematosus: A Case Series and Review of the Literature. Lupus 2016, 25, 760–765. [Google Scholar] [CrossRef]
  148. Weber, G.F.; Zawaideh, S.; Hikita, S.; Kumar, V.A.; Cantor, H.; Ashkar, S. Phosphorylation-Dependent Interaction of Osteopontin with Its Receptors Regulates Macrophage Migration and Activation. J. Leukoc. Biol. 2002, 72, 752–761. [Google Scholar] [CrossRef]
  149. Shinohara, M.L.; Lu, L.; Bu, J.; Werneck, M.B.F.; Kobayashi, K.S.; Glimcher, L.H.; Cantor, H. Osteopontin Expression Is Essential for Interferon-α Production by Plasmacytoid Dendritic Cells. Nat. Immunol. 2006, 7, 498–506. [Google Scholar] [CrossRef]
  150. Ouyang, S.; Gong, B.; Li, J.Z.; Zhao, L.X.; Wu, W.; Zhang, F.S.; Sun, L.; Wang, S.J.; Pan, M.; Li, C.; et al. Structural Insights into a Human Anti-IFN Antibody Exerting Therapeutic Potential for Systemic Lupus Erythematosus. J. Mol. Med. 2012, 90, 837–846. [Google Scholar] [CrossRef]
  151. Rodríguez-Carrio, J.; Burska, A.; Conaghan, P.G.; Dik, W.A.; Biesen, R.; Eloranta, M.-L.; Cavalli, G.; Visser, M.; Boumpas, D.T.; Bertsias, G.; et al. Association between Type I Interferon Pathway Activation and Clinical Outcomes in Rheumatic and Musculoskeletal Diseases: A Systematic Literature Review Informing EULAR Points to Consider. RMD Open 2023, 9, e002864. [Google Scholar] [CrossRef] [PubMed]
  152. Wahadat, M.J.; Bodewes, I.L.A.; Maria, N.I.; van Helden-Meeuwsen, C.G.; van Dijk-Hummelman, A.; Steenwijk, E.C.; Kamphuis, S.; Versnel, M.A. Type I IFN Signature in Childhood-Onset Systemic Lupus Erythematosus: A Conspiracy of DNA- and RNA-Sensing Receptors? Arthritis Res. 2018, 20, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Crow, Y.J.; Vanderver, A.; Orcesi, S.; Kuijpers, T.W.; Rice, G.I. Therapies in Aicardi–Goutières Syndrome. Clin. Exp. Immunol. 2013, 175, 1–8. [Google Scholar] [CrossRef] [PubMed]
  154. Dasdemir, S.; Yildiz, M.; Celebi, D.; Sahin, S.; Aliyeva, N.; Haslak, F.; Gunalp, A.; Adrovic, A.; Barut, K.; Artim Esen, B.; et al. Genetic Screening of Early-Onset Patients with Systemic Lupus Erythematosus by a Targeted next-Generation Sequencing Gene Panel. Lupus 2022, 31, 330–337. [Google Scholar] [CrossRef]
  155. Eloi, M.; Horvath, D.V.; Ortega, C.; Prado, S.; Andrade, L.E.C.; Lú Cia Szejnfeld, V.; Heldan, C.; Castro, M. 25-Hydroxivitamin D Serum Concentration, Not Free and Bioavailable Vitamin D, Is Associated with Disease Activity in Systemic Lupus Erythematosus Patients. PLoS ONE 2017, 12, e0170323. [Google Scholar] [CrossRef] [Green Version]
  156. Meza-Meza, M.R.; Francisco Muñoz-Valle, J.; Ruiz-Ballesteros, A.I.; Vizmanos-Lamotte, B.; Parra-Rojas, I.; Martínez-López, E.; Oregon-Romero, E.; Fabiola Márquez-Sandoval, Y.; Cerpa-Cruz, S.; De La Cruz-Mosso, U. Association of High Calcitriol Serum Levels and Its Hydroxylation Efficiency Ratio with Disease Risk in SLE Patients with Vitamin D Deficiency. Hindawi J. Immunol. Res. 2021, 2021, 16. [Google Scholar] [CrossRef]
  157. Swee Gaik, O.; Hui Jen, D. Vitamin D Status in a Monocentric Cohort of Systemic Lupus Erythematosus (SLE) Patients and Correlations with Clinical and Immunological Profile. Med. J. Malays. 2019, 74, 493. [Google Scholar]
  158. di Chen, B.; Jia, X.; Xu, J.; Zhao, L.; Ji, J.; Wu, B.; Ma, Y.; Li, H.; Zuo, X.; Pan, W.; et al. An Autoimmunogenic and Proinflammatory Profile Defined by the Gut Microbiota of Patients with Untreated Systemic Lupus Erythematosus. Arthritis Rheumatol. 2021, 73, 232–243. [Google Scholar] [CrossRef]
  159. Zhang, L.; Qing, P.; Yang, H.; Wu, Y.; Liu, Y.; Luo, Y. Gut Microbiome and Metabolites in Systemic Lupus Erythematosus: Link, Mechanisms and Intervention. Front. Immunol. 2021, 12, 686501. [Google Scholar] [CrossRef]
  160. López, P.; De Paz, B.; Rodríguez-Carrio, J.; Hevia, A.; Sánchez, B.; Margolles, A.; Suárez, A. Th17 Responses and Natural IgM Antibodies Are Related to Gut Microbiota Composition in Systemic Lupus Erythematosus Patients. Sci. Rep. 2016, 6, 24072. [Google Scholar] [CrossRef] [Green Version]
  161. 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] [Green Version]
  162. Li, Y.; Wang, H.F.; Li, X.; Li, H.X.; Zhang, Q.; Zhou, H.W.; He, Y.; Li, P.; Fu, C.; Zhang, X.H.; et al. Disordered Intestinal Microbes Are Associated with the Activity of Systemic Lupus Erythematosus. Clin. Sci. 2019, 133, 821–838. [Google Scholar] [CrossRef]
  163. 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, 1. [Google Scholar] [CrossRef]
  164. Mu, Q.; Zhang, H.; Liao, X.; Lin, K.; Liu, H.; Edwards, M.R.; Ahmed, S.A.; Yuan, R.; Li, L.; Cecere, T.E.; et al. Control of Lupus Nephritis by Changes of Gut Microbiota. Microbiome 2017, 5, 73. [Google Scholar] [CrossRef] [Green Version]
  165. Cabana-Puig, X.; Mu, Q.; Lu, R.; Swartwout, B.; Abdelhamid, L.; Zhu, J.; Prakash, M.; Cecere, T.E.; Wang, Z.; Callaway, S.; et al. Lactobacillus Spp. Act in Synergy to Attenuate Splenomegaly and Lymphadenopathy in Lupus-Prone MRL/Lpr Mice. Front. Immunol. 2022, 13, 4103. [Google Scholar] [CrossRef]
  166. Niess, J.H. What Are CX3CR1+ Mononuclear Cells in the Intestinal Mucosa? Gut. Microbes 2010, 1, 396–400. [Google Scholar] [CrossRef] [Green Version]
  167. Kim, D.S.; Park, Y.; Choi, J.W.; Park, S.H.; La Cho, M.; 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, 5246. [Google Scholar] [CrossRef]
  168. Toral, M.; Robles-Vera, I.; Romero, M.; de la Visitación, N.; Sánchez, M.; O’Valle, F.; Rodriguez-Nogales, A.; Gálvez, J.; Duarte, J.; Jiménez, R. Lactobacillus Fermentum CECT5716: A Novel Alternative for the Prevention of Vascular Disorders in a Mouse Model of Systemic Lupus Erythematosus. FASEB J. 2019, 33, 10005–10018. [Google Scholar] [CrossRef] [Green Version]
  169. He, H.; Xu, H.; Xu, J.; Zhao, H.; Lin, Q.; Zhou, Y.; Nie, Y. Sodium Butyrate Ameliorates Gut Microbiota Dysbiosis in Lupus-Like Mice. Front. Nutr. 2020, 7, 604283. [Google Scholar] [CrossRef]
  170. Li, Z.; Yi, C.X.; Katiraei, S.; Kooijman, S.; Zhou, E.; Chung, C.K.; Gao, Y.; Van Den Heuvel, J.K.; Meijer, O.C.; Berbée, J.F.P.; et al. Butyrate Reduces Appetite and Activates Brown Adipose Tissue via the Gut-Brain Neural Circuit. Gut 2018, 67, 1269–1279. [Google Scholar] [CrossRef] [Green Version]
  171. Frostegård, J.; Svenungsson, E.; Wu, R.; Gunnarsson, I.; Lundberg, I.E.; Klareskog, L.; Hörkkö, S.; Witztum, J.L. Lipid Peroxidation Is Enhanced in Patients with Systemic Lupus Erythematosus and Is Associated with Arterial and Renal Disease Manifestations. Arthritis Rheum. 2005, 52, 192–200. [Google Scholar] [CrossRef] [PubMed]
  172. Wang, M.; Zhu, Z.; Lin, X.; Li, H.; Wen, C.; Bao, J.; He, Z. Gut Microbiota Mediated the Therapeutic Efficacies and the Side Effects of Prednisone in the Treatment of MRL/Lpr Mice. Arthritis Res. 2021, 23, 240. [Google Scholar] [CrossRef] [PubMed]
  173. Trzonkowski, P.; Bieniaszewska, M.; Juścińska, J.; Dobyszuk, A.; Krzystyniak, A.; Marek, N.; Myśliwska, J.; Hellmann, A. First-in-Man Clinical Results of the Treatment of Patients with Graft versus Host Disease with Human Ex Vivo Expanded CD4+CD25+CD127- T Regulatory Cells. Clin. Immunol. 2009, 133, 22–26. [Google Scholar] [CrossRef] [PubMed]
  174. Shum, A.K.; DeVoss, J.; Tan, C.L.; Hou, Y.; Johannes, K.; O’Gorman, C.S.; Jones, K.D.; Sochett, E.B.; Fong, L.; Anderson, M.S. Identification of an Autoantigen Demonstrates a Link between Interstitial Lung Disease and a Defect in Central Tolerance. Sci. Transl. Med. 2009, 1, 9ra20. [Google Scholar] [CrossRef] [Green Version]
  175. Van Belleghem, J.D.; Dąbrowska, K.; Vaneechoutte, M.; Barr, J.J.; Bollyky, P.L. Interactions between Bacteriophage, Bacteria, and the Mammalian Immune System. Viruses 2019, 11, 10. [Google Scholar] [CrossRef] [Green Version]
  176. Hahn, J.; Cook, N.R.; Alexander, E.K.; Friedman, S.; Walter, J.; Bubes, V.; Kotler, G.; Lee, I.M.; Manson, J.A.E.; Costenbader, K.H. Vitamin D and Marine Omega 3 Fatty Acid Supplementation and Incident Autoimmune Disease: VITAL Randomized Controlled Trial. BMJ 2022, 376, e066452. [Google Scholar] [CrossRef]
Figure 1. Gut microbiota and vitamin D modulation of the immune system. On the right side of the figure, an anti-inflammatory milieu, with a balanced microbiota composition, in the presence of normal vitamin D levels, which ensure intestinal barrier integrity, support a tolerogenic immune system setting. On the left side, a pathologic status with high levels of inflammation, low levels of vitamin D, and a decreased heterogeneity of microbiota, which promote immune cell activation while suppressing a regulatory phenotype. (AMPs, antimicrobial peptides; PsA, polysaccharide A; SCFA, short chain fatty acid; and VDR, vitamin D receptor). ↑ increased levels; ↓ decreased levels.
Figure 1. Gut microbiota and vitamin D modulation of the immune system. On the right side of the figure, an anti-inflammatory milieu, with a balanced microbiota composition, in the presence of normal vitamin D levels, which ensure intestinal barrier integrity, support a tolerogenic immune system setting. On the left side, a pathologic status with high levels of inflammation, low levels of vitamin D, and a decreased heterogeneity of microbiota, which promote immune cell activation while suppressing a regulatory phenotype. (AMPs, antimicrobial peptides; PsA, polysaccharide A; SCFA, short chain fatty acid; and VDR, vitamin D receptor). ↑ increased levels; ↓ decreased levels.
Biomedicines 11 01127 g001
Table 1. Summary of monogenic autoimmune diseases.
Table 1. Summary of monogenic autoimmune diseases.
DiseaseDefective Gene(s)InheritanceAutoimmunityMechanism of AutoimmunityMajor SymptomsMicrobiota Changes
APS1 [42,49,50,61]AIREAutosomal recessiveOrgan specificDefective Tregs
Abnormal DCs
Lymphocyte Infiltration
AutoAbs
Hypoparathyroidism
Mucocutaneous
candidiasis
Adrenal insufficiency
↓Firmicutes
↑Proteobacteria
↑Bacteroidetes
IPEX [69,72,80]FOXP3X-linkedOrgan specificDysfunctional Tregs
AutoAbs
Diarrhea
Type I diabetes
Thyroiditis
Eczema
↓Firmicutes
↑Bacteroidetes
ALPS [92,97,98,110]FAS
FASL
CASP10
Autosomal dominant Autosomal recessive
(variable penetrance)
Systemic, organ specificDefective lymphocyte apoptosisLymphadenopathy
Splenomegaly
Autoimmune Cytopenias
Malignancy
N.A.
SLE [120,124,132,143,161]Clq, Cls, Clr, C2, C4
TREX1
RNase H2
DNASE1L3
ACP5
Autosomal recessiveSystemicComplement deficiencies
Impaired apoptosis
Nucleic acid degradation
Nucleic acid sensing self-tolerance
type I interferonopathies
Arthritis
Cutaneous vasculitis
Nephritis
Infections
↓Firmicutes *
↓Lactobacillus *
↑Bacteroidetes *
↑Lachnospiraceae *
Abbreviations: ALPS, Autoimmune lymphoproliferative syndrome; APS1, Autoimmune polyendocrine syndrome type 1; AutoAbs, Autoantibodies; DCs, Dendritic cells; IPEX, Immunodysregulation polyendocrinopathy enteropathy X-linked syndrome; N.A., Not available; SLE, Systemic lupus erythematosus; Tregs, T regulatory cells; *, In polygenic SLE; ↑ increased levels; ↓ decreased levels.
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

Costa, F.; Beltrami, E.; Mellone, S.; Sacchetti, S.; Boggio, E.; Gigliotti, C.L.; Stoppa, I.; Dianzani, U.; Rolla, R.; Giordano, M. Genes and Microbiota Interaction in Monogenic Autoimmune Disorders. Biomedicines 2023, 11, 1127. https://doi.org/10.3390/biomedicines11041127

AMA Style

Costa F, Beltrami E, Mellone S, Sacchetti S, Boggio E, Gigliotti CL, Stoppa I, Dianzani U, Rolla R, Giordano M. Genes and Microbiota Interaction in Monogenic Autoimmune Disorders. Biomedicines. 2023; 11(4):1127. https://doi.org/10.3390/biomedicines11041127

Chicago/Turabian Style

Costa, Federica, Eleonora Beltrami, Simona Mellone, Sara Sacchetti, Elena Boggio, Casimiro Luca Gigliotti, Ian Stoppa, Umberto Dianzani, Roberta Rolla, and Mara Giordano. 2023. "Genes and Microbiota Interaction in Monogenic Autoimmune Disorders" Biomedicines 11, no. 4: 1127. https://doi.org/10.3390/biomedicines11041127

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