Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Nov 1.
Published in final edited form as: Semin Immunol. 2023 Mar 30;67:101757. doi: 10.1016/j.smim.2023.101757

Immunoglobulins at the interface of the gut mycobiota and anti-fungal immunity

Itai Doron 1,2, Takato Kusakabe 1,2, Iliyan D Iliev 1,2,3,4,*
PMCID: PMC10192079  NIHMSID: NIHMS1890572  PMID: 37003056

Abstract

The dynamic and complex community of microbes that colonizes the intestines is composed of bacteria, fungi, and viruses. At the mucosal surfaces, immunoglobulins play a key role in protection against bacterial and fungal pathogens, and their toxins. Secretory immunoglobulin A (sIgA) is the most abundantly produced antibody at the mucosal surfaces, while Immunoglobulin G (IgG) isotypes play a critical role in systemic protection. IgA and IgG antibodies with reactivity to commensal fungi play an important role in shaping the mycobiota and host antifungal immunity. In this article, we review the latest evidence that establishes a connection between commensal fungi and B cell-mediated antifungal immunity as an additional layer of protection against fungal infections and inflammation.

Keywords: Antifungal antibodies, Mycobiome, secretory IgA, IgG, humoral immunity, B-cells, Germinal Centers, Candida, hyphae, Saccharomyces

1. Introduction

Known collectively as the “gut mycobiota”, the fungal component of the intestinal microbiota ranges between 1% and 2% of biomass [1], [2] or ~0.01% of DNA in human fecal material as assessed through metagenomic sequencing [3]. Recent evidence points to several fungal species associating with the gastrointestinal tract as either commensals or transients arriving through food sources or the outside environment [3], [4], [5]. Among these species and strains belonging to Candida, Saccharomyces, Pichia, Malassezia, Debaryomyces and several other genera have been more vigorously studied in the recent years to highlight the relationship of gut fungi to the gut immune system and various diseases including inflammatory bowel disease (IBD), diabetes, cancer, brain disorders, chronic kidney diseases, respiratory diseases, and liver diseases [6], [7], [8], [9], [10], [11], [12], [13], [14], [15], [16], [17], [18].

Antibodies play a pivotal role in the interface between intestinal microbes and the host immune system due to their capacity to generate potent mucosal immune responses and maintain gut homeostasis. Recent investigation revealed that C. albicans and its hyphal morphotype are particularly potent inducers of antifungal sIgA, which promotes fungal commensalism in the gut. In this review, we will examine the body of work elucidating the mechanisms that regulate antifungal antibody (aFA) responses and a therapeutic potential of antibodies to fungal commensals and pathogens.

2. Systemic and mucosal antibody responses to bacteria and fungi

The diverse community of bacteria, fungi and viruses composing the gut microbiota necessitates a constant, broad, and finely tuned management of host-microbe interactions by the mucosal immune system throughout life. In this context, an expanding body of research has identified antibodies against commensal bacteria as a critical arm of the adaptive immune response, providing protection against toxins and invasive pathogens while also promoting commensal interactions. In the gut, vast quantities of secretory immunoglobulins (sIg) are secreted daily into the gut lumen to bind [19], [20], [21], neutralize [21], [22], [23], and chaperone microbial residents to lymphoid tissues for sampling by immune cells [24], [25], [26], [27], [28]. Although IgA – the most abundant mammalian sIg isotype produced in the gut mucosa – is considered to play an important role in maintaining homeostasis at mucosal barriers, more recent work revealed that IgG also recognizes a broad range of microbial antigens that are in some cases overlapping with IgA [29]. sIg plays a particularly critical protective role in early life, as maternally transferred sIg protects the immature neonatal intestinal environment by dampening the response to colonizing microbes (with observable long-term implications) [30], [31], [32], [33], [34], as well as playing a pivotal role in protection against enteric pathogenic bacteria such as Shigella flexneri and Salmonella typhimurium, [35], [36]. Recent studies utilizing novel techniques and improved resolution uncover more nuanced homeostatic functions of sIgA, acting as a buffer against offensive colonization [37], [38], [39], and triggering transcriptional modulation of sIgA -bound microbiota species [38], [40], [41], [42].

2.1. Role of Immunoglobulins to bacteria at the mucosal surfaces

Mounting evidence has demonstrated that intestinal microbiota participates in shaping B cells and humoral immunity in health and disease settings. Germ-free (GF) mice exhibit impaired lymphoid structures, dramatically reduced serum IgG and IgA titers, and antibody-producing plasma cells- features that can be reversed by microbial colonization [43], [44]. Another study also revealed that deletion of activation-induced cytidine deaminase (AID)-dependent antibodies, which fail to undergo class switch recombination (CSR) and somatic hypermutation (SHM) necessary for affinity maturation, allowed for expansion of anaerobic bacteria such as segmented filamentous bacteria (SFB), which could penetrate the mucus layer to reach the intestinal epithelium and induce isolated lymphoid follicle (ILF) hyperplasia [45], [46]. Interestingly, the Bacteroidetes, the major phylum of gut microbiota including Bacteroides faecis, Bacteroides caccae, and Bacteroides acidifaciens, was positively associated with sIgA production in Peyer’s patches and AID activity [47]. In addition, it was recently reported that the level of sIgA production in mice monocolonized with human gut isolates of Bacteroides ovatus depends on yet to be determined strain-specific features [48].

sIgA possess broad reactivity to a variety of microorganisms in the gut to prevent systemic dissemination of gut microbes. Generally, it is estimated that approximately 50% of luminal commensal microbiota in the small intestine is bound to sIgA. A fraction of microbes bound by highly-specific sIgA antibodies contain pathobionts that aggravate colon inflammation [49]. Analysis of fecal samples from elderly people revealed that age-related changes in the composition of gut microbiota correlates with a reduction of sIgA antibodies against several pathobionts [50]. Another study reported that pediatric patients with allergy have a low proportion of sIgA bound to fecal bacteria compared with healthy individuals [51]. While sIgA coating of pathobionts can limit their pathogenesis, it can also trigger inflammation in a context-dependent manner:

For example, opsonized bacteria engulfed by neutrophils, can recruit inflammatory cells into the colonic mucosa of ulcerative colitis patients, and promote inflammation via neutrophil extracellular trap (NET) formation [52], [53], [54]. Thus, antibody-coated pathobionts can cause tissue damage and inflammation by neutrophil activation in the context of ongoing intestinal disease.

2.2. Role of secretory IgA antibodies to fungi at the mucosal surfaces

Diverse fungal communities (the mycobiota) are associated with nearly all human barrier surfaces. Recent characterization of the mycobiota biogeography along the gastrointestinal tract revealed the presence of a subset of fungi associated with the intestinal mucosa of mice and humans [55], with several Candida and Saccharomyces species making the majority of these mucosa-associated fungi. Using a multistep flow cytometry-based technique for multi-kingdom antibody profiling (multiKAP) a recent study explored the antibody pool against gut mycobiota and determined a population of intestinal fungi that was bound by both luminal (sIgA) and systemic (IgG) antibodies [56]. The human gut commensal C. albicans was determined as the primary target (Fig. 1). In contrast to bacteria, C. albicans cells in the gut exist in both yeast and hyphal morphotypes that differ in cell size and function. However, how fungal species with the potential to cause disease can safely inhabit the intestinal mucosa without causing harm has been a mystery [57]. Using mouse models and several genetic approaches affecting the process of hyphal formation in C. albicans, two studies determined that sIgA antibodies bind preferentially to the larger hyphal morphotypes [58], [59].

Fig. 1. Function of secretory IgA and systemic IgG antibodies to fungi.

Fig. 1.

Several effects of antibody binding have been described in the context of antifungal immunity. From the oral cavity to the intestinal lumen, sIgA immobilizes and thus prevents fungi from encroaching on the epithelial layer, a process known as immunologic exclusion (A, top left). sIgA also promotes commensalism in C. albicans by targeting hyphae-associated (Als1 and Als3) and secreted (Candidalysin, SAP6) virulence factors to incur a fitness penalty on the hyphal morphotype (A, top) and protect host cells (A, top right). A significant fraction of the systemic antibody repertoire in circulation also recognizes fungi, from highly reactive IgG to natural IgM (A-B, bottom). These sIg have been demonstrated to protect against fungi that disseminate from the intestinal or the lung mucosa and cause systemic infection (A-B). Figures Created with BioRender.com.

Given the importance of hyphae for host cell invasion and the presence of multiple virulence factors associated with this morphotype, recent studies focused on hyphae-associated virulence factors as a potential target of these antibodies (Fig. 1). Adhesin proteins such as members of the Als family (particularly Als3 and Als1), and Hwp1 and Hyr1 [14], [60], [61] allow hyphae to bind to intestinal epithelial cells. Among these, Als3 is also critical for regulating iron homeostasis between the iron-rich intestinal lumen and to the iron-poor bloodstream upon invasion [61]. Als3, along with Ssa1, is involved in the hyphae-associated invasion strategy of inducing endocytosis through binding to N- or E-cadherin [14], [60], [61], [62]. Recent studies demonstrated that salivary sIgA prevent C. albicans adhesion and interferes with its iron acquisition through binding of Als3, further interfering with hyphae-associated activities [63], [64]. Thus, Als proteins play several roles that foster C. albicans invasion properties in different environments. Furthermore, sIgA in the oral cavity prevented fungal epithelial adhesion and invasion leading to decrease of the proinflammatory response [65]. What are the factors targeted by sIgA in this scenario remains to be determined.

Corresponding with the preferential sIgA coating of these hyphal-associated virulence factors, numerous studies point to associations between Candida commensalism and its intestinal fitness (Fig. 1). In the gut, transcription factor WOR1, induced upon gastrointestinal passage of C. albicans, results in unique “GUT” morphotypes that excel at intestinal colonization [66]. Competitive gut colonization of mice with a library of homozygous gene disruption C. albicans mutants resulted in successful gut colonization upon disruption of transcription factor Ume6, a master regulator of filamentation. Ume6, similar to the well-known regulator of morphogenesis Efg1 [13], promotes the expression of hyphae-associated virulence factors including the secreted aspartyl proteinase protease (Sap6) and adhesin Hyr1 [14]. Sap6 mutants exhibit enhanced colonization fitness, whereas Sap6-overexpression strains exhibit reduced competitiveness in the gut. Using a screening approach of homozygous deletion mutant libraries of C. albicans, a recent study determined that transcription factor Ahr1 is necessary for sIgA binding to C. albicans. Ahr1 is involved in adhesion and hyphal formation and regulates Als3, which the study determined to be the target of sIgA [59]. Expression of Als3 or two other hyphae-associated adhesins Als1 and Hwp1 on S. cerevisiae (yeast that induces little sIgA) was sufficient for sIgA binding. Similar sIgA binding was observed to C. glabrata that also expresses these adhesins. Repeated colonization of C. albicans into wild type (WT), but not into the adaptive immunity deficient Rag1−/− mice, resulted in decreased expression of ALS1 and ALS3, demonstrating the fitness penalty conferred by the adaptive immunity [59]. It remains to be determined whether these effects on C. albicans are direct (B- and/or T-cells) or indirect (mediated by different microbiota in mice lacking adaptive immunity).

While adhesins are surface molecules with natural ability to bind to substrates allowing the attachment and penetration of pathogens into host cells, these proteins are not directly responsible for the damage caused by the pathogen itself. Indeed, it was demonstrated that during pathologies [12], C. albicans damage intestinal epithelial cells (IECs) and macrophages by secretion of pore-forming toxin candidalysin [67], which induces apoptotic cell death [68] and fuels intestinal inflammation by inducing macrophage cell damage and release of IL-1 [12]. Notably, strains of C. albicans, associated with different individuals harbor different potential to cause damage [12]. These findings underline a strain-specific nature of host–fungal interactions in the gut [12]. Candidalysin is produced by several Candida species that are present in the human gut [69], and it was identified along with Sap6 as a target of C. albicans colonization-induced sIgA that was notably decreased in Crohn’s Disease patients’ mucosal lavages [58]. Candida hyphal morphotypes and several hyphae-associated factors have a direct effect on host adaptive immunity by triggering B cell expansion and class switch recombination in Peyer’s Patches (PP) in response to intestinal C. albicans colonization [58], [59]. These findings reveal the nuanced effect of sIgA binding to C. albicans and demonstrate a role for sIgA in influencing the processes of yeast-to-hyphal transition in the intestines.

2.3. Systemic antifungal antibodies as a layer of protection against systemic fungemia

The systemic immunoglobulin response is dominated by IgG, making up ~70–75% of the total Ig in serum and tissues [70]. A continuously expanding body of research demonstrates that, in addition to the secretory anti-commensal sIgA response, a surprisingly large fraction of the systemic IgG repertoire circulating in the blood recognizes gut commensal epitopes. Multiple studies have documented the capacity of the systemic IgG to bind gut microbes under steady-state conditions and confer protection against bacteria escaping systemically from the gut [29], [30], [31], [32], [71], [72], [73]. Furthermore, extraintestinal B cells have been shown to recognize a significant fraction of the gut microbiota [74], [75].

Traditionally, the systemic antifungal antibody responses have been studied in the context of systemic Candida [76], [77] and Cryptococcus [78], [79] infections. Potent antifungal antibody responses, targeting fungal cell wall-associated epitopes, such as β-glucan and mannan [80], also arise during lung infections caused by fungal pathogens such as Pneumocystis and Aspergillus [81]. More recently, specificity or polyreactivity of antifungal antibodies present in the serum has been analyzed by flow cytometry [82] or a serological proteome analysis (SERPA) [83]. A high-throughput multi-kingdom antibody profiling (MultiKAP) was currently applied to explore the human antibody repertoires against gut commensal fungi (mycobiota), leading to several advances in the understanding of antibody mediated immunity to fungi [56].

The antifungal humoral response is necessary and sufficient for protection against systemic fungemia [56], [84], [85]. While B cell-deficient (μMT−/−) mice, similar to their WT counterparts, are resistant to primary systemic infection with C.albicans, systemic antifungal IgG appear particularly important during secondary challenges with the same pathogen, which rely on the generation of B cell memory and is particularly lethal in the absence of B-cells [32], [56], [86]. Deficiencies in C. albicans phagocytosis, IL-10 production, and Treg priming, which could be remedied by a passive transfer of antifungal serum IgG, are thought to further contibute to susceptibility of μMT−/− mice to fugnal infections [56], [85].

A recent study revealed the critical role of B-cell mediated immunity in protection against gut disseminated fungal disease [56]. The gut commensal C. albicans was the major inducer of antifungal IgG. C. albicans intestinal colonization of mice led to the induction of germinal center (GC)-dependent B cell expansion in extraintestinal lymphoid tissues and generation of systemic antibodies. Notably these circulating anti-fungal IgG antibodies conferred protection against disseminated C. albicans infection and cross-protection against C. auris. These findings revealed an important role of gut commensal fungi in shaping the human antibody repertoire and a role of antibody-mediated immunity in antifungal immunity and production.

In the lungs, Aspergillus fumigatus (Af) is one of the most frequent causes of invasive fungal disease in immunosuppressed individuals. Immunosuppressive drugs are well-recognized risk factors for developing invasive aspergillosis [87], lead to neutrophil impairment [88] and dramatic decrease in antifungal IgG antibodies in the serum [56]. Emergency of COVID-19–associated pulmonary aspergillosis (CAPA) was also reported, adding pulmonary viral infections to the risk factors for the development of fungal coinfections [89], [90]. A recent study reported that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)–driven pneumonia depletes innate B lymphocytes and led to decrease of anti-Af IgG antibodies in the serum of severe COVID19 patients [91]. Neutrophils, provide essential defense by preventing conidial germination and invasive disease caused by Af [92] while systemic IgG antibodies further limit fungal disease . SARS-CoV-2 spike protein and corticosteroid therapy both induced B1a cell apoptosis [91]. Further experiments in a mouse model revealed that neutrophil-mediated host defense against Af was dependent on antifungal IgG. Altogether, these studies highlight the role of circulating antibodies and B cells in antifungal immunity and defense.

Reflecting the unique morphological flexibility of some fungal pathogens in the mammalian hosts, a wide range of effector functions have been connected to fungal antigen binding to systemic immunoglobulins during systemic infection. Clonally gut-related IgA+ B cells, which travel to the dural sinuses of the brain in response to commensal intestinal colonization, produce protective IgA to restrict systemic candidiasis to dural sinuses [93]. Supporting these findings, neutrophils and monocytes isolated from an invasive candidiasis patient with a missense mutation in CARD9 were deficient in C. albicans yeast killing, which could be restored by serum incubation to allow serum IgG opsonization [94]. In addition to the role of CARD9 in innate immunity against fungi, it was found that [56] antifungal IgG production also depends on the CARD9, revealing an important crosstalk between gut commensal fungi, innate and adaptive immunity for the generation of host-protective antifungal IgG.

3. Mechanistic factors governing anti-fungal antibody induction

The mosaic of diverse targets and reactivities of the secretory and systemic anti-commensal antibody repertoires is facilitated by multiple induction pathways (Fig. 2). In particular, we will discuss few components including B cell subsets, phagocyte subsets, and antibody isotypes.

Fig. 2. Multiple pathways for antibody repertoire development contribute to a variety of antifungal antibody isotypes and reactivities.

Fig. 2.

In the absence of presentation with gut microbial antigens or stimulation associated with gut microbial colonization, a subset of B cells will spontaneously mature and secrete natural antibodies (top). Gut fungal colonization-induced f antifungal antibodies occur through either T cell-independent (middle) or -dependent (bottom) mechanisms, although the exact pathways are yet to be elucidated. In the former, a combination of BCR engagement, PRR co-stimulation, and TNF-family ligands BAFF and APRIL induce CSR responses in the absence of T cell help. In the latter, BCR-engaged B cells undergo CSR and harmonize with T helper cells to form germinal centers that promote cycles of expansion/SHM and selection of high-affinity BCR clones, a process known as affinity maturation.

3.1. A diversity of B cell subsets that contribute to antifungal antibody reactivities

B cell diversity arises from two processes mediated by the activation-induced cytidine deaminase (AID): 1) CSR, the process of replacing the constant μ region of the heavy chain and with other constant regions to express a different antibody isotype without altering the variable region; and 2) SHM, the process of converting cytosines in the antigen-binding variable regions to uracil to induce DNA breaks that can be randomly filled in to generate diversity of Ig binding. The degree to which B cells will undergo either or both of these processes is dependent upon the secondary lymphoid tissue, the antigen sensed by pattern recognition receptors (PRRs) on innate immune cells, and co-stimulation by other immune cells.

A fraction of naïve mature B cells will produce antibodies spontaneously with minimal to no CSR or SHM. These “natural” antibodies, which can be observed in GF mice [95] in the absence of live gut microbiota or dietary antigen exposure [73], [96], exhibit light and heavy chain regions that are or are nearly identical to the “germline sequences” of B cells leaving the bone marrow. While natural IgA [74], [97], [98] and IgG [99] isotypes have been described, these “natural” antibodies are mostly associated with IgM isotypes. Typical epitopes of natural antibodies include polysaccharides common to the cell walls of a wide range of prokaryotes and eukaryotes [81], [98], making these antibodies highly polyreactive with low binding affinity. Notably, fungal cell wall polysaccharides such as beta-glucan and chitin are a common target of anti-fungal antibodies, and some of these antibodies are identified in GF mice in the absence of microbial colonization, suggesting the existence of a robust and protective “natural antifungal” antibody response [56], [78], [81], [95].

Polyreactivity and low binding affinity is a shared quality with antibodies produced by B cells that matured without T cell co-stimulation, thus collectively identified as the T cell-independent (TI) response. In the gut, this process is characteristically initiated by co-stimulation with both microbial antigens and myeloid cell-secreted TNF family ligands BAFF and APRIL [100], [101], [102]. Assessments of the breadth of TI anti-commensal sIgA coating and comparison of sIgA commensal coating frequencies in T cell-deficient mice, show similar results relative to immunocompetent mice, suggesting that TI anti-commensal antibodies can serve as redundant and compensatory protection upon T cell dysfunction, albeit with potentially different epitopes [103].

T cell-dependent (TD) antibody induction is the source of most immunoglobulins with high specificity to microbial antigens [74], [104]. The defining feature of TD antibody responses occur in germinal centers (GC), where SHM-derived B cell clones are selectively activated for clonal expansion and optimization depending on their ability to bind to presented antigens, a process known as affinity maturation. These dynamics have recently been reviewed [105]. In brief, GCs are polirized into a light zone (LZ) and a dark zone (DZ), and are the sites where B-cell clonal expansion, hypermutation, and selection occurs. GC-B cells are repeatedly cycling in between these zones. In the DZ, densely packed GC-B cells undergo extensive divisions and SHM, while upon migrating to the LZ GC-B cells test their B cell receptor (BCR) affinity against specially presented antigens [106]. The latter action is critically dependent upon proliferation signals from T follicular helper cells (Tfh), which promote B cell proliferation and expansion through their unique profile of co-stimulation and cytokine expression [107], [108], [109]. Additionally, multiple potential models have been suggested for the selection process upon LZ BCR engagement, including the culling of low-affinity GC-B cells through apoptosis as well as GC-B cell proliferation that correlates with the degree of BCR affinity [105]. In either case, the resulting B cells exhibit high specificity and affinity for their target antigen.

Evidence for C. albicans GC engagement could be found in the presence of its delivery to the GC-laden PP by certain subsets of intestinal dendritic cells [110]. Stimulation with fungal β-glucan induces the expression of activation markers such as CD40 and CD86, and is dependent upon spleen tyrosine kinase (Syk) signaling [111]. Such activation is critical for engaging naïve T cells and inducing their transition to Tfh cells. Investigations of the intestinal PP, show that C. albicans intestinal colonization promotes expansion of IgA+ GC-B cells , while the absence of GC formation in T cell-deficient Tcrb−/− mice leads to a significant decrease in anti-C. albicans sIgA binding [58], [59] (Fig. 2). GC populations are also critical for the induction of antifungal systemic IgG, with a dramatic loss of antifungal systemic IgG observed in the serum of Tcrb−/− mice [56].

3.2. Role of innate phagocytes in the generation of antifungal antibody responses

The persistent colonization of microbes at the intestinal barriers provide an abundant source of pathogen-associated molecular patterns (PAMP) and antigens that are sensed and processed by several phagocyte subsets in the intestines. Throughout the intestine, these cell types play varying roles in direct and indirect regulation of sIg production and response to opsonized microbial antigens. Two main populations of intestinal phagocytes, characterized by the expression of C-type lectin receptors (CLR), mediate the generation of antifungal antibodies in the murine gut: CX3CR1+ macrophages (Mfs) and type 2 conventional dendritic cells (cDC2) [56], [58], [112], [113]. A recent study has further described the presence of CLRs on a population of AIRE-expressing cells with features of both innate lymphoid (ILCs) and dendritic cells (DCs), and with unknown function in these processes [114].

CX3CR1+ intestinal Mfs are characterized by high expression of the integrin CD11b and chemokine receptor CX3CR1 [115], and are in a close proximity to intestinal epithelial cells (IEC) in the colon and the small intestine [116]. CX3CR1+ Mfs are also characterized by their relatively poor ability to migrate to other tissues such as the mesenteric lymph nodes (mLNs), in stark contrast to their cDC counterparts in the gut [115], [117]. Despite their relatively static nature, CX3CR1+ Mfs are critical contributors to both cellular and humoral protective immune responses, and have the ability to directly sample antigens coming from the lumen [118], [119]. Underlining this sensitivity, the perturbation of a steady-state gut microbiota by antibiotics or invasive pathogens can shift macrophages tolerogenic program towards a pro-inflammatory program [120], [121]. Importantly, these cells are critical mediators of antifungal Th17 responses to commensal fungi in the gut [113], [122].

Several conventional Zbtb46-dependent cDC subsets, are found at higher frequencies in the small intestine [123]. These cells lack canonical macrophage-specific markers and express CD11b and/or CD103 [124]. The latter two markers are used to further distinguish between CD103+CD11b cDC1s and CD103+CD11b+ cDC2s, whose differentiation is dependent on Batf3 and Irf4 respectively [125]. cDC2s sample luminal contents in the gut to induce gut-homing tolerogenic adaptive immune responses, primarily against gut microbial and dietary antigens in a TLR-dependent manner [126]. Lacking the ability to sample antigens across the epithelium, these cells instead access antigens through gap junctions-mediated interaction with macrophages [119], through transcytosis of antigens from goblet cells [127] or through the uptake of sIg-bound antigens [24], [128]. Unique to cDCs among intestinal MNPs is the ability to use the chemokine receptor CCR7 to enter lymphatic vessels [129] and migrate to draining lymph nodes. These cell types are critical for the priming of effector Th cells during homeostasis [130], [131], and for the induction of gut-homing receptors in T cells [132]. Failure to activate effective priming often results in a significant decrease of small intestine-homing effector T cells [130], [132], [133], [134], [135].

An increasing number of studies point to the critical involvement of the CX3CR1+ subset in the induction of antibody responses to commensal microbes. In vitro experiments using ovalbumin as a model antigen have demonstrated the ability of this MNP subset to induce IgA production when cocultured with B cells. Induction of low-affinity TI IgA antibodies by CX3CR1+ macrophages required BAFF and APRIL, but was independent of retinoic acid signaling [101]. Meanwhile, cDC2s that encounter antigens in the PPs also play a role in the generation of sIgA responses. The expression of CCL20 by follicle-associated epithelium (FAE) initially draws CCR6-expressing cDCs, along with CCR6-expressing B cells, to the PP where penetrant commensals and antigens can be sampled [136], [137].

Currently, the mechanisms of MNP-mediated modulation of antibody responses to fungi remain unclear. Experiments in mice demonstrate that both the CX3CR1+ macrophages and cDC2s contribute to the overall antifungal sIgA response in the intestine, albeit in compartmentalized fashion: while depleting either population leads to significant decreases in free luminal anti-C. albicans sIgA titers, cDC2 depletion abrogates PP IgA-CSR while CX3CR1+ macrophage depletion does the same for plasmablasts [58]. These findings are supported by previous studies demonstrating that human cDC2 subset found in gut associated lymphoid tissues (GALT) were capable of C. albicans phagocytosis in the PPs [110].

3.3. Innate immune receptors and their involvement in regulation of antifungal antibody responses in the gut

While a number of PRRs sense fungal cellular components, the primary and most studied contributors are several C-type lectin receptors (CLRs). The most extensively studies CLR is Dectin-1, which recognizes cell wall b-glucan and initiates signaling through Syk and caspase recruitment domain-containing protein 9 (Card9) [111], [138]. Signaling initiated through Dectin-1 has been tied to protective antifungal responses such as the production of reactive oxygen species, phagocytosis, the production of critical pro-inflammatory cytokines and chemokines, and priming Th1 and Th17 populations [2], [139], [140], [141]. Other CLRs notable for expression on macrophages and DCs include Dectin-2 and Mincle. The former binds mannan, induces IL-1β and IL-23 to promote protective Th17 responses [142], and exhibits a proclivity for binding hyphal filaments [142], [143]. The latter, which binds mannose and appears to play a critical role during systemic candidiasis involving the induction of TNF-α [144]. Notably, neither of these receptors have an intracellular ITAM-containing tail to activate Syk, instead forming complexes with Fc receptor (FcR) adaptor molecules that are necessary for mediating antifungal signaling through Syk [145], [146]. Despite lack of observed fungi-specific clinical phenotypes [147], TLR signaling – TLR2, TLR4, and TLR9 in particular – contribute to the induction of innate antifungal immunity in response to fungal cell components [148], [149], [150], [151], [152].

PRR signaling specifically, stimulation of Dectin-1, Dectin-2 CLRs, but not mincle, on macrophages results in secretory sIgA induction by co-cultured B cells [153]. Interestingly, Dectin-1 is expressed on FAE cells and contributes to transcytosis of recognized sIgA-bound complexes, suggesting that their contribution to the secretory antibody response may not be restricted to antigen presentation to B cells or Tfhs [26]. In contrast to sIgA, depletion studies suggest the systemic IgG responses to commensal mycobiota appear to be solely dependent on CX3CR1+ macrophages and solely induced in the spleen [56], raising important questions regarding site of fungal sensing and the site of induction.

3.4. Immunoglobulin isotypes

While the mechanisms guiding B cell activation and the subsequent affinity maturation in systemic lymphoid tissues match those of GALT, the antibodies produced by the resulting plasma cells exhibit drastically different effector functions upon binding. This is due mostly to the variety among the four IgG subclasses (IgG1–4) and their variable affinity for IgG Fc receptors [70]. While mice have a different set of IgG subclasses (IgG1, IgG2a/c, IgG2b, and IgG3), their subclasses IgG2b and IgG3 closely match the human IgG1 and IgG2, respectively. Profiles of IgG subclass binding reveal IgG1 and IgG3 reactivity against proteinaceous antigens [154], [155], while IgG2 exhibits near-exclusive reactivity against surface polysaccharides (although polysaccharide reactivity is not restricted to the IgG2 subclass) [156], [157]. While IgG4 also exhibits preferential reactivity to microbial proteins, this typically occurs after extended exposure ? in non-infectious conditions, and it is more commonly associated with reactivity to allergens such as pollen [158], [159].

While all but the IgG4 subclass can recruit complement, most other IgG effector functions require Fc-gamma receptor (FcgR) binding, systemic IgG effector functions are guided by their interaction with their cognate receptors, the FcgR superfamily. Reflecting the fact that only one FcgR family member (FcgRIIB) mediates anti-inflammatory signaling, IgG effector functions are overwhelmingly pro-inflammatory. Nevertheless, FcgRIIB plays a critical role in the calibration of pro-inflammatory IgG responses: a variety of FcgRs expressed on almost every immune cell, with a higher activating-to-inhibitory ratio (A:I) effectively reducing the IgG binding threshold for raising the appropriate downstream immune responses [160]. Notably, FcgR-mediated phagocytosis in macrophages increased the A:I ratio, demonstrating a positive feedback loop of IgG-mediated pro-inflammatory responses [161]. Among the IgG subclasses, IgG1 and particularly IgG3 are the most effective inducers of effector functions due to their longer and more flexible hinge region that facilitates more efficient FcgR binding and recruitment of complement [162]. However, a single AA substitution in IgG3 results in its intracellular out-competition for transcytosis by IgG1, significantly reducing the half-life of IgG3 antibodies [163]. Among these IgG subclasses, IgG2b and IgG3 dominate the IgG response to commensal fungi with IgG3 binding the largest fraction of the intestinal fungi [56].

4. Secretory and systemic antibody responses during fungal infections and IBD

Perturbation of gut microbiota composition or deficiencies in gut immunity result in a state of intestinal dysbiosis associated with IBD disorders, such as Cohn’s Disease (CD) and Ulcerative Colitis (UC), which affect over three million Americans [164]. Increasing focus is being devoted to understanding the role of antibody responses induced against gut bacteria, which depending on the context, can have a dual role by potentiating autoimmunity or dampening immune responses to prevent inflammation [32], [165], [166]. Interestingly, dysbiosis resulting from mucosal infections and inflammation during IBD provide examples of IgA and IgG antibodies reacting to the gut microbiota with opposite effects on intestinal health. The disruption of the intestinal barrier by invasive enteric infection can lead to changes in lymphoid tissue architecture that significantly alter the secretory IgA response. In the colon, this is characterized by the formation of additional tertiary lymphoid tissues (TLS), a category of ectopic lymphoid tissue that are induced into formation on-site by immunogenic intestinal microbes [167] or the development of inflammatory lesions in IBD patients. Mouse models of human salmonellosis reveal these TLSs, mediated by colon-resident CX3CR1hi Mfs, are critical sites of Salmonella-specific IgA-CSR and generation of protective salmonella-specific sIgA (along with CD4+ T cell-mediated cellular immune responses), without which mice succumb to systemic infection within weeks [167]. Turning to chronic intestinal inflammation, sIgA binding to the gut microbiota is significantly increased in IBD patients relative to healthy individuals [49], [168]. Increases in sIgA gut microbiota coating can be transferred to healthy WT mice simply by co-housing them with littermates whose gut microbiota composition induces spontaneous colitis, demonstrating that colitogenic microbes induce robust protective sIgA responses [49]. IBD-associated sIgA also exhibits disrupted functionality, particularly with regards to the capacity in mediating Dectin-1 reverse transcytosis after binding gut microbes, the capacity of which is significantly decreased in UC patients but expanded in CD patients [169].

Meanwhile, an examination of human intestinal mucosal tissue samples revealed that the onset of inflammation in IBD patients was associated with the accumulation of IgG+ plasmablasts in the mucosa at significantly higher frequencies than healthy individuals [170]. Single-cell analyses revealed that these IBD-associated B cells exhibit immature phenotypes such as reduced diversity and lower SHM frequency [170], [171]. Notably, multiple studies have identified FcγR SNPs that significantly decrease the activating:inhibiting (A:I) ratio of MNPs resulting in greater protection during UC. These studies suggest a critical role for IgG binding to gut microbes and subsequent induction of pro-inflammatory signaling in the pathogenesis of IBD. Matched analyses of samples from UC patients and their healthy relatives sharing the same households support these observations [161]. Commensal-bound IgG complexes induced pro-inflammatory responses through the FcγR, including IL-1β production leading to neutrophil and Th17 infiltration.

Interestingly, the most prominent association between the gut mycobiota and systemic antibody responses are anti-Saccharomyces cerevisiae-antibodies (ASCA). These antibodies, which are reactive with S. cerevisiae cell wall mannan, are a robust clinical prognostic marker of Crohn’s Disease [172], [173] when titers are elevated in the blood [173], [174] and feces [175]. Although the mechanisms of ASCA induction are not precisely understood, both C. albicans and S. cerevisiae express ASCA epitopes, emphasizing the possibility that these clinically relevant antibodies could be cross-reactive with a number of fungal commensals in the intestine [176], [177]. Further complicating the ASCA induction picture, C. albicans ASCA epitope expression is dependent upon unique conditions, such as live systemic exposure (heat-killed C. albicans did not induce ASCA) [175], [176], once again highlighting the impact environmental conditions have on fungal immunogenicity. Despite these findings, the function of ASCA is currently unknown.

5. Prospects for an Antifungal Vaccination

Fungal pathogens present unique problems. Due in part to their ability to proliferate outside of their host and global travel, this fungus has spread to tens of countries since its first isolation in 1996 in South Korea. Additionally, its transmissibility appears to be positively associated by global warming [178], [179], suggesting that this threat will continue to grow. Like other Candida spp., C. auris persistently menaces hospitals worldwide by lying dormant on commonly used equipment such as catheter tubing [179], [180], [181]. However, unlike other Candida spp., it is multi-drug resistant, in some cases against all available anti-fungal drugs. Finally, within the host, the conditions inducing yeast-to-hyphae transition towards virulence are associated with immunocompromised state and/or dysbiosis, leaving little available recourse for treatment outside of passive immunization or vaccination before sickness.

While no fungal vaccines have progressed beyond clinical evaluation, a number of fungal antigens and strategies have been applied and have been extensively covered by several reviews in the past few years [7], [182]. Several rather broad targets include surface glycans, for which monoclonal antibodies have been generated and whose binding has been associated with promoting phagocytosis and killing while inhibiting filamentous growth [183], [184]. Several specific targets include hyphae-associated virulence factors that aid epithelial adhesion such as Als3 (NDV-3A) [185], Sap proteins, Hsp90, and Hyr1 include other promising targets [76], [186], [187]. The wide variety of targets reflects the primary challenge of developing these vaccines: fungi are present as a variety of morphologies as commensals [188] and during infection [189]. Additionally, systemic candidiasis can originate from any of several members of the Candida spp. including C. albicans, C. galabrata, C. auris, C. parapsilosis, each occurring in immunodeficient patients or those with a weakened immune system. A recent success of a phase 2 randomized, double-blind, placebo-controlled trial of a vaccine against Als3 (NDV-3A) for treatment of recurrent vulvovaginal candidiasis (RVVC) shows promise. NDV-3A administered to women with RVVC was safe, highly immunogenic and reduced the frequency of symptomatic episodes of vulvovaginal candidiasis for up to 12 months [185]. Thus, the most promising antifungal immunoglobulin therapy design would likely be directed against multiple fungal antigens, provide help to innate antifungal defense by activating neutrophils (although this will be challenging in neutropenic patients), or may simply rely on passive transfer of antibodies.

6. Conclusions

In this review, we discussed the existing body of literature related to the known functions of antifungal antibodies locally and at systemic levels. We also reviewed less-explored antibody induction sites, immune cell subsets, and signaling pathways involved in B cell-mediated antifungal immunity. Many challenges and roadblocks remain. For example, any extent to which the fungal and the bacterial microbiota affect each other’s metabolic activity, morphology, and fitness would have profound consequences on how antibody repertoires against specific mucosa-associated fungi will be generated. We also lack knowledge of the extent to which antifungal antibody repertoires are shaped by commensal bacteria (and vice versa) or other factors such as the diet.

While relatively unspecific and broadly cross-reactive, deeper understanding of TI antibody responses to fungi might shade light to mechanisms that limit fungal pathogenesis. Future studies will have to contend with the overlapping contributions of B cells and T cells to adaptive antifungal immunity. The evidence coming from current studies on adaptive immunity to fungi suggest that B cells , while not as critical for antifungal protection as T cell , are sufficient to boost antifungal response during systemic infection. Knowledge gaps remain regarding site-dependent mechanisms through which antifungal antibody responses are generated and regarding the specificities of such antibodies. In regard to mechanisms by which systemic antibody responses to commensal fungi are induced, it remains to be elucidated whether specific cell type(s) traffic fungal antigens from the gut or if those antigens and PAMPs passively disseminate to induce systemic IgG. Such investigations will further our knowledge and might lead to therapeutic implications.

Acknowledgments

Research in the Iliev laboratory is supported by US National Institutes of Health (R01DK113136, R01DK121977, and R01AI163007), the Leona M. and Harry B. Helmsley Charitable Trust, the Irma T. Hirschl Career Scientist Award, the Research Corporation for Science Advancement Award, the Burroughs Wellcome Fund Investigator in the Pathogenesis of Infectious Disease (PATH) Award, and the Cancer Research Institute Lloyd J. Old STAR Award. I.D.I is a fellow of the CIFAR program Fungal Kingdom: Threats and Opportunities.

References:

  • [1].Huffnagle GB, Noverr MC, The emerging world of the fungal microbiome, Trends in Microbiology. 21 (2013) 334–341. 10.1016/j.tim.2013.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Iliev ID, Funari VA, Taylor KD, Nguyen Q, Reyes CN, Strom SP, Brown J, Becker CA, Fleshner PR, Dubinsky M, Rotter JI, Wang HL, McGovern DPB, Brown GD, Underhill DM, Interactions Between Commensal Fungi and the C-Type Lectin Receptor Dectin-1 Influence Colitis, Science. 336 (2012) 1314–1317. https://doi.org/doi: 10.1126/science.1221789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Nash AK, Auchtung TA, Wong MC, Smith DP, Gesell JR, Ross MC, Stewart CJ, Metcalf GA, Muzny DM, Gibbs RA, Ajami NJ, Petrosino JF, The gut mycobiome of the Human Microbiome Project healthy cohort, Microbiome. 5 (2017) 153. 10.1186/s40168-017-0373-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Fiers WD, Gao IH, Iliev ID, Gut mycobiota under scrutiny: fungal symbionts or environmental transients?, Current Opinion in Microbiology. 50 (2019) 79–86. 10.1016/j.mib.2019.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Suhr MJ, Banjara N, Hallen-Adams HE, Sequence-based methods for detecting and evaluating the human gut mycobiome, Letters in Applied Microbiology. 62 (2016) 209–215. 10.1111/lam.12539. [DOI] [PubMed] [Google Scholar]
  • [6].Fan D, Coughlin LA, Neubauer MM, Kim J, Kim MS, Zhan X, Simms-Waldrip TR, Xie Y, Hooper LV, Koh AY, Activation of HIF-1α and LL-37 by commensal bacteria inhibits Candida albicans colonization, Nat Med. 21 (2015) 808–14. 10.1038/nm.3871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Tso GHW, Reales-Calderon JA, Tan ASM, Sem X, Le GTT, Tan TG, Lai GC, Srinivasan KG, Yurieva M, Liao W, Poidinger M, Zolezzi F, Rancati G, Pavelka N, Experimental evolution of a fungal pathogen into a gut symbiont, Science. 362 (2018) 589. 10.1126/science.aat0537. [DOI] [PubMed] [Google Scholar]
  • [8].Shao T-Y, Ang WXG, Jiang TT, Huang FS, Andersen H, Kinder JM, Pham G, Burg AR, Ruff B, Gonzalez T, Khurana Hershey GK, Haslam DB, Way SS, Commensal Candida albicans Positively Calibrates Systemic Th17 Immunological Responses, Cell Host & Microbe. 25 (2019) 404–417.e6. 10.1016/j.chom.2019.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Jain U, Ver Heul AM, Xiong S, Gregory MH, Demers EG, Kern JT, Lai CW, Muegge BD, Barisas DAG, Leal-Ekman JS, Deepak P, Ciorba MA, Liu TC, Hogan DA, Debbas P, Braun J, McGovern DPB, Underhill DM, Stappenbeck TS, Debaryomyces is enriched in Crohn’s disease intestinal tissue and impairs healing in mice, Science. 371 (2021) 1154–1159. 10.1126/science.abd0919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Limon JJ, Tang J, Li D, Wolf AJ, Michelsen KS, Funari V, Gargus M, Nguyen C, Sharma P, Maymi VI, Iliev ID, Skalski JH, Brown J, Landers C, Borneman J, Braun J, Targan SR, McGovern DPB, Underhill DM, Malassezia Is Associated with Crohn’s Disease and Exacerbates Colitis in Mouse Models, Cell Host Microbe. 25 (2019) 377–388.e6. 10.1016/j.chom.2019.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].van Tilburg Bernardes E, Pettersen VK, Gutierrez MW, Laforest-Lapointe I, Jendzjowsky NG, Cavin J-B, Vicentini FA, Keenan CM, Ramay HR, Samara J, MacNaughton WK, Wilson RJA, Kelly MM, McCoy KD, Sharkey KA, Arrieta M-C, Intestinal fungi are causally implicated in microbiome assembly and immune development in mice, Nature Communications. 11 (2020) 2577. 10.1038/s41467-020-16431-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Li XV, Leonardi I, Putzel GG, Semon A, Fiers WD, Kusakabe T, Lin WY, Gao IH, Doron I, Gutierrez-Guerrero A, DeCelie MB, Carriche GM, Mesko M, Yang C, Naglik JR, Hube B, Scherl EJ, Iliev ID, Immune regulation by fungal strain diversity in inflammatory bowel disease, Nature. 603 (2022) 672–678. 10.1038/s41586-022-04502-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Liang S-H, Anderson MZ, Hirakawa MP, Wang JM, Frazer C, Alaalm LM, Thomson GJ, Ene IV, Bennett RJ, Hemizygosity Enables a Mutational Transition Governing Fungal Virulence and Commensalism, Cell Host & Microbe. 25 (2019) 418–431.e6. 10.1016/j.chom.2019.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Witchley JN, Penumetcha P, Abon NV, Woolford CA, Mitchell AP, Noble SM, Candida albicans Morphogenesis Programs Control the Balance between Gut Commensalism and Invasive Infection, Cell Host Microbe. 25 (2019) 432–443.e6. 10.1016/j.chom.2019.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Petersen KM, Westall S, Jespersen L, Microbial succession of Debaryomyces hansenii strains during the production of Danish surfaced-ripened cheeses, J Dairy Sci. 85 (2002) 478–86. 10.3168/jds.S0022-0302(02)74098-8. [DOI] [PubMed] [Google Scholar]
  • [16].Pattaroni C, Macowan M, Chatzis R, Daunt C, Custovic A, Shields MD, Power UF, Grigg J, Roberts G, Ghazal P, Schwarze J, Gore M, Turner S, Bush A, Saglani S, Lloyd CM, Marsland BJ, Early life inter-kingdom interactions shape the immunological environment of the airways, Microbiome. 10 (2022) 34. 10.1186/s40168-021-01201-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Boutin RCT, Petersen C, Woodward SE, Serapio-Palacios A, Bozorgmehr T, Loo R, Chalanuchpong A, Cirstea M, Lo B, Huus KE, Barcik W, Azad MB, Becker AB, Mandhane PJ, Moraes TJ, Sears MR, Subbarao P, McNagny KM, Turvey SE, Finlay BB, Bacterial–fungal interactions in the neonatal gut influence asthma outcomes later in life, ELife. 10 (2021) e67740. 10.7554/eLife.67740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Kumamoto CA, Gresnigt MS, Hube B, The gut, the bad and the harmless: Candida albicans as a commensal and opportunistic pathogen in the intestine, Curr Opin Microbiol. 56 (2020) 7–15. 10.1016/j.mib.2020.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Blanchard TG, Czinn SJ, Maurer R, Thomas WD, Soman G, Nedrud JG, Urease-specific monoclonal antibodies prevent Helicobacter felis infection in mice, Infection and Immunity. 63 (1995) 1394–1399. https://doi.org/doi: 10.1128/iai.63.4.1394-1399.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Michetti P, Mahan MJ, Slauch JM, Mekalanos JJ, Neutra MR, Monoclonal secretory immunoglobulin A protects mice against oral challenge with the invasive pathogen Salmonella typhimurium, Infection and Immunity. 60 (1992) 1786–1792. https://doi.org/doi: 10.1128/iai.60.5.1786-1792.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Phalipon A, Kaufmann M, Michetti P, Cavaillon JM, Huerre M, Sansonetti P, Kraehenbuhl JP, Monoclonal immunoglobulin A antibody directed against serotype-specific epitope of Shigella flexneri lipopolysaccharide protects against murine experimental shigellosis, Journal of Experimental Medicine. 182 (1995) 769–778. 10.1084/jem.182.3.769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Apter FM, Michetti P, Winner LS, Mack JA, Mekalanos JJ, Neutra MR, Analysis of the roles of antilipopolysaccharide and anti-cholera toxin immunoglobulin A (IgA) antibodies in protection against Vibrio cholerae and cholera toxin by use of monoclonal IgA antibodies in vivo, Infection and Immunity. 61 (1993) 5279–5285. https://doi.org/doi: 10.1128/iai.61.12.5279-5285.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Lycke N, Eriksen L, Holmgren J, Protection against cholera toxin after oral immunization is thymus-dependent and associated with intestinal production of neutralizing IgA antitoxin, Scand J Immunol. 25 (1987) 413–9. 10.1111/j.1365-3083.1987.tb02208.x. [DOI] [PubMed] [Google Scholar]
  • [24].Kadaoui KA, Corthésy B, Secretory IgA Mediates Bacterial Translocation to Dendritic Cells in Mouse Peyer’s Patches with Restriction to Mucosal Compartment, The Journal of Immunology. 179 (2007) 7751. 10.4049/jimmunol.179.11.7751. [DOI] [PubMed] [Google Scholar]
  • [25].Lycke N, Erlandsson L, Ekman L, Schön K, Leanderson T, Lack of J chain inhibits the transport of gut IgA and abrogates the development of intestinal antitoxic protection, J Immunol. 163 (1999) 913–9. [PubMed] [Google Scholar]
  • [26].Rochereau N, Drocourt D, Perouzel E, Pavot V, Redelinghuys P, Brown GD, Tiraby G, Roblin X, Verrier B, Genin C, Corthésy B, Paul S, Dectin-1 Is Essential for Reverse Transcytosis of Glycosylated SIgA-Antigen Complexes by Intestinal M Cells, PLOS Biology. 11 (2013) e1001658. 10.1371/journal.pbio.1001658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Schwartz-Cornil I, Benureau Y, Greenberg H, Hendrickson BA, Cohen J, Heterologous protection induced by the inner capsid proteins of rotavirus requires transcytosis of mucosal immunoglobulins, Journal of Virology. 76 (2002) 8110–8117. 10.1128/jvi.76.16.8110-8117.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Sun K, Johansen FE, Eckmann L, Metzger DW, An important role for polymeric Ig receptor-mediated transport of IgA in protection against Streptococcus pneumoniae nasopharyngeal carriage, J Immunol. 173 (2004) 4576–81. 10.4049/jimmunol.173.7.4576. [DOI] [PubMed] [Google Scholar]
  • [29].Fadlallah J, Sterlin D, Fieschi C, Parizot C, Dorgham K, El Kafsi H, Autaa G, Ghillani-Dalbin P, Juste C, Lepage P, Malphettes M, Galicier L, Boutboul D, Clément K, André S, Marquet F, Tresallet C, Mathian A, Miyara M, Oksenhendler E, Amoura Z, Yssel H, Larsen M, Gorochov G, Synergistic convergence of microbiota-specific systemic IgG and secretory IgA, Journal of Allergy and Clinical Immunology. 143 (2019) 1575–1585.e4. 10.1016/j.jaci.2018.09.036. [DOI] [PubMed] [Google Scholar]
  • [30].de Agüero MG, Ganal-Vonarburg SC, Fuhrer T, Rupp S, Uchimura Y, Li H, Steinert A, Heikenwalder M, Hapfelmeier S, Sauer U, McCoy KD, Macpherson AJ, The maternal microbiota drives early postnatal innate immune development, Science. 351 (2016) 1296–1302. https://doi.org/doi: 10.1126/science.aad2571. [DOI] [PubMed] [Google Scholar]
  • [31].Gopalakrishna KP, Macadangdang BR, Rogers MB, Tometich JT, Firek BA, Baker R, Ji J, Burr AHP, Ma C, Good M, Morowitz MJ, Hand TW, Maternal IgA protects against the development of necrotizing enterocolitis in preterm infants, Nature Medicine. 25 (2019) 1110–1115. 10.1038/s41591-019-0480-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Koch MA, Reiner GL, Lugo KA, Kreuk LSM, Stanbery AG, Ansaldo E, Seher TD, Ludington WB, Barton GM, Maternal IgG and IgA Antibodies Dampen Mucosal T Helper Cell Responses in Early Life, Cell. 165 (2016) 827–841. 10.1016/j.cell.2016.04.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Zheng W, Zhao W, Wu M, Song X, Caro F, Sun X, Gazzaniga F, Stefanetti G, Oh S, Mekalanos JJ, Kasper DL, Microbiota-targeted maternal antibodies protect neonates from enteric infection, Nature. 577 (2020) 543–548. 10.1038/s41586-019-1898-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Usami K, Niimi K, Matsuo A, Suyama Y, Sakai Y, Sato S, Fujihashi K, Kiyono H, Uchino S, Furukawa M, Islam J, Ito K, Moriya T, Kusumoto Y, Tomura M, Hovey RC, Sugawara J, Yoneyama H, Kitazawa H, Watanabe K, Aso H, Nochi T, The gut microbiota induces Peyer’s-patch-dependent secretion of maternal IgA into milk, Cell Reports. 36 (2021). 10.1016/j.celrep.2021.109655. [DOI] [PubMed] [Google Scholar]
  • [35].Robin G, Cohen D, Orr N, Markus I, Slepon R, Ashkenazi S, Keisari Y, Characterization and Quantitative Analysis of Serum IgG Class and Subclass Response to Shigella sonnei and Shigella flexneri 2a Lipopolysaccharide following Natural Shigella Infection, The Journal of Infectious Diseases. 175 (1997) 1128–1133. 10.1086/516452. [DOI] [PubMed] [Google Scholar]
  • [36].Lee S-J, Liang L, Juarez S, Nanton MR, Gondwe EN, Msefula CL, Kayala MA, Necchi F, Heath JN, Hart P, Tsolis RM, Heyderman RS, MacLennan CA, Felgner PL, Davies DH, McSorley SJ, Identification of a common immune signature in murine and human systemic Salmonellosis, Proceedings of the National Academy of Sciences. 109 (2012) 4998–5003. 10.1073/pnas.1111413109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Moor K, Diard M, Sellin ME, Felmy B, Wotzka SY, Toska A, Bakkeren E, Arnoldini M, Bansept F, Co AD, Völler T, Minola A, Fernandez-Rodriguez B, Agatic G, Barbieri S, Piccoli L, Casiraghi C, Corti D, Lanzavecchia A, Regoes RR, Loverdo C, Stocker R, Brumley DR, Hardt W-D, Slack E, High-avidity IgA protects the intestine by enchaining growing bacteria, Nature. 544 (2017) 498–502. 10.1038/nature22058. [DOI] [PubMed] [Google Scholar]
  • [38].Nakajima A, Vogelzang A, Maruya M, Miyajima M, Murata M, Son A, Kuwahara T, Tsuruyama T, Yamada S, Matsuura M, Nakase H, Peterson DA, Fagarasan S, Suzuki K, IgA regulates the composition and metabolic function of gut microbiota by promoting symbiosis between bacteria, Journal of Experimental Medicine. 215 (2018) 2019–2034. 10.1084/jem.20180427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Donaldson GP, Ladinsky MS, Yu KB, Sanders JG, Yoo BB, Chou W-C, Conner ME, Earl AM, Knight R, Bjorkman PJ, Mazmanian SK, Gut microbiota utilize immunoglobulin A for mucosal colonization, Science. 360 (2018) 795–800. https://doi.org/doi: 10.1126/science.aaq0926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Porter NT, Canales P, Peterson DA, Martens EC, A Subset of Polysaccharide Capsules in the Human Symbiont Bacteroides thetaiotaomicron Promote Increased Competitive Fitness in the Mouse Gut, Cell Host & Microbe. 22 (2017) 494–506.e8. 10.1016/j.chom.2017.08.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Peterson DA, McNulty NP, Guruge JL, Gordon JI, IgA Response to Symbiotic Bacteria as a Mediator of Gut Homeostasis, Cell Host & Microbe. 2 (2007) 328–339. 10.1016/j.chom.2007.09.013. [DOI] [PubMed] [Google Scholar]
  • [42].Rollenske T, Burkhalter S, Muerner L, von Gunten S, Lukasiewicz J, Wardemann H, Macpherson AJ, Parallelism of intestinal secretory IgA shapes functional microbial fitness, Nature. 598 (2021) 657–661. 10.1038/s41586-021-03973-7. [DOI] [PubMed] [Google Scholar]
  • [43].Wilmore JR, Gaudette BT, Gomez Atria D, Hashemi T, Jones DD, Gardner CA, Cole SD, Misic AM, Beiting DP, Allman D, Commensal Microbes Induce Serum IgA Responses that Protect against Polymicrobial Sepsis, Cell Host Microbe. 23 (2018) 302–311.e3. 10.1016/j.chom.2018.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Kozakova H, Schwarzer M, Tuckova L, Srutkova D, Czarnowska E, Rosiak I, Hudcovic T, Schabussova I, Hermanova P, Zakostelska Z, Aleksandrzak-Piekarczyk T, Koryszewska-Baginska A, Tlaskalova-Hogenova H, Cukrowska B, Colonization of germ-free mice with a mixture of three lactobacillus strains enhances the integrity of gut mucosa and ameliorates allergic sensitization, Cell Mol Immunol. 13 (2016) 251–262. 10.1038/cmi.2015.09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Fagarasan S, Muramatsu M, Suzuki K, Nagaoka H, Hiai H, Honjo T, Critical roles of activation-induced cytidine deaminase in the homeostasis of gut flora, Science. 298 (2002) 1424–1427. 10.1126/science.1077336. [DOI] [PubMed] [Google Scholar]
  • [46].Wei M, Shinkura R, Doi Y, Maruya M, Fagarasan S, Honjo T, Mice carrying a knock-in mutation of Aicda resulting in a defect in somatic hypermutation have impaired gut homeostasis and compromised mucosal defense, Nat Immunol. 12 (2011) 264–270. 10.1038/ni.1991. [DOI] [PubMed] [Google Scholar]
  • [47].Nakajima A, Sasaki T, Itoh K, Kitahara T, Takema Y, Hiramatsu K, Ishikawa D, Shibuya T, Kobayashi O, Osada T, Watanabe S, Nagahara A, A Soluble Fiber Diet Increases Bacteroides fragilis Group Abundance and Immunoglobulin A Production in the Gut, Applied and Environmental Microbiology. 86 (2020) e00405–20. 10.1128/AEM.00405-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Yang C, Mogno I, Contijoch EJ, Borgerding JN, Aggarwala V, Li Z, Siu S, Grasset EK, Helmus DS, Dubinsky MC, Mehandru S, Cerutti A, Faith JJ, Fecal IgA Levels Are Determined by Strain-Level Differences in Bacteroides ovatus and Are Modifiable by Gut Microbiota Manipulation, Cell Host & Microbe. 27 (2020) 467–475.e6. 10.1016/j.chom.2020.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Palm NW, de Zoete MR, Cullen TW, Barry NA, Stefanowski J, Hao L, Degnan PH, Hu J, Peter I, Zhang W, Ruggiero E, Cho JH, Goodman AL, Flavell RA, Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease, Cell. 158 (2014) 1000–1010. 10.1016/j.cell.2014.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Sugahara H, Okai S, Odamaki T, Wong CB, Kato K, Mitsuyama E, Xiao J-Z, Shinkura R, Decreased Taxon-Specific IgA Response in Relation to the Changes of Gut Microbiota Composition in the Elderly, Frontiers in Microbiology. 8 (2017). https://www.frontiersin.org/articles/10.3389/fmicb.2017.01757 (accessed January 27, 2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Dzidic M, Abrahamsson TR, Artacho A, Björkstén B, Collado MC, Mira A, Jenmalm MC, Aberrant IgA responses to the gut microbiota during infancy precede asthma and allergy development, J Allergy Clin Immunol. 139 (2017) 1017–1025.e14. 10.1016/j.jaci.2016.06.047. [DOI] [PubMed] [Google Scholar]
  • [52].Fournier BM, Parkos CA, The role of neutrophils during intestinal inflammation, Mucosal Immunol. 5 (2012) 354–366. 10.1038/mi.2012.24. [DOI] [PubMed] [Google Scholar]
  • [53].van der Steen L, Tuk CW, Bakema JE, Kooij G, Reijerkerk A, Vidarsson G, Bouma G, Kraal G, de Vries HE, Beelen RHJ, van Egmond M, Immunoglobulin A: FcαRI Interactions Induce Neutrophil Migration Through Release of Leukotriene B4, Gastroenterology. 137 (2009) 2018–2029.e3. 10.1053/j.gastro.2009.06.047. [DOI] [PubMed] [Google Scholar]
  • [54].Muthas D, Reznichenko A, Balendran CA, Böttcher G, Clausen IG, Kärrman Mårdh C, Ottosson T, Uddin M, MacDonald TT, Danese S, Berner Hansen M, Neutrophils in ulcerative colitis: a review of selected biomarkers and their potential therapeutic implications, Scand J Gastroenterol. 52 (2017) 125–135. 10.1080/00365521.2016.1235224. [DOI] [PubMed] [Google Scholar]
  • [55].Leonardi I, Gao IH, Lin W-Y, Allen M, Li XV, Fiers WD, De Celie MB, Putzel GG, Yantiss RK, Johncilla M, Colak D, Iliev ID, Mucosal fungi promote gut barrier function and social behavior via Type 17 immunity, Cell. 185 (2022) 831–846.e14. 10.1016/j.cell.2022.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Doron I, Leonardi I, Li XV, Fiers WD, Semon A, Bialt-DeCelie M, Migaud M, Gao IH, Lin W-Y, Kusakabe T, Puel A, Iliev ID, Human gut mycobiota tune immunity via CARD9-dependent induction of anti-fungal IgG antibodies, Cell. 184 (2021) 1017–1031.e14. 10.1016/j.cell.2021.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Dambuza IM, Brown GD, Managing the mycobiota with IgA, Nature Microbiology. 6 (2021) 1471–1472. 10.1038/s41564-021-01006-7. [DOI] [PubMed] [Google Scholar]
  • [58].Doron I, Mesko M, Li XV, Kusakabe T, Leonardi I, Shaw DG, Fiers WD, Lin W-Y, Bialt-DeCelie M, Román E, Longman RS, Pla J, Wilson PC, Iliev ID, Mycobiota-induced IgA antibodies regulate fungal commensalism in the gut and are dysregulated in Crohn’s disease, Nature Microbiology. (2021). 10.1038/s41564-021-00983-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Ost KS, O’Meara TR, Stephens WZ, Chiaro T, Zhou H, Penman J, Bell R, Catanzaro JR, Song D, Singh S, Call DH, Hwang-Wong E, Hanson KE, Valentine JF, Christensen KA, O’Connell RM, Cormack B, Ibrahim AS, Palm NW, Noble SM, Round JL, Adaptive immunity induces mutualism between commensal eukaryotes, Nature. (2021). 10.1038/s41586-021-03722-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Liu Y, Filler SG, Candida albicans Als3, a multifunctional adhesin and invasin, Eukaryotic Cell. 10 (2011) 168–173. 10.1128/EC.00279-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Phan QT, Myers CL, Fu Y, Sheppard DC, Yeaman MR, Welch WH, Ibrahim AS Jr. Edwards John E., S.G. Filler, Als3 Is a Candida albicans Invasin That Binds to Cadherins and Induces Endocytosis by Host Cells, PLOS Biology. 5 (2007) e64. 10.1371/journal.pbio.0050064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Sun JN, Solis NV, Phan QT, Bajwa JS, Kashleva H, Thompson A, Liu Y, Dongari-Bagtzoglou A, Edgerton M, Filler SG, Host Cell Invasion and Virulence Mediated by Candida albicans Ssa1, PLOS Pathogens. 6 (2010) e1001181. 10.1371/journal.ppat.1001181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Brena S, Cabezas-Olcoz J, Moragues María D, Fernández de Larrinoa I, Domínguez A, Quindós G, Pontón J, Fungicidal Monoclonal Antibody C7 Interferes with Iron Acquisition in Candida albicans, Antimicrobial Agents and Chemotherapy. 55 (2011) 3156–3163. 10.1128/AAC.00892-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].San Millán R, Elguezabal N, Regúlez P, Moragues MD, Quindós G, Pontón J, Effect of salivary secretory IgA on the adhesion of Candida albicans to polystyrene, Microbiology. 146 (2000) 2105–2112. 10.1099/00221287-146-9-2105. [DOI] [PubMed] [Google Scholar]
  • [65].Millet N, Solis NV, Swidergall M, Mucosal IgA Prevents Commensal Candida albicans Dysbiosis in the Oral Cavity, Frontiers in Immunology. 11 (2020). 10.3389/fimmu.2020.555363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Pande K, Chen C, Noble SM, Passage through the mammalian gut triggers a phenotypic switch that promotes Candida albicans commensalism, Nature Genetics. 45 (2013) 1088–1091. 10.1038/ng.2710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Moyes DL, Wilson D, Richardson JP, Mogavero S, Tang SX, Wernecke J, Höfs S, Gratacap RL, Robbins J, Runglall M, Murciano C, Blagojevic M, Thavaraj S, Förster TM, Hebecker B, Kasper L, Vizcay G, Iancu SI, Kichik N, Häder A, Kurzai O, Luo T, Krüger T, Kniemeyer O, Cota E, Bader O, Wheeler RT, Gutsmann T, Hube B, Naglik JR, Candidalysin is a fungal peptide toxin critical for mucosal infection, Nature. 532 (2016) 64–68. 10.1038/nature17625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Allert S, Förster TM, Svensson C-M, Richardson JP, Pawlik T, Hebecker B, Rudolphi S, Juraschitz M, Schaller M, Blagojevic M, Morschhäuser J, Figge MT, Jacobsen ID, Naglik JR, Kasper L, Mogavero S, Hube B, Kronstad JW, Bignell E, Lorenz M, Candida albicans-Induced Epithelial Damage Mediates Translocation through Intestinal Barriers, MBio. 9 (2018) e00915–18. https://doi.org/doi: 10.1128/mBio.00915-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Richardson Jonathan P, Brown R, Kichik N, Lee S, Priest E, Mogavero S, Maufrais C, Wickramasinghe Don N, Tsavou A, Kotowicz Natalia K, Hepworth Olivia W, Gallego-Cortés A, Ponde Nicole O, Ho J, Moyes David L, Wilson D, D’Enfert C, Hube B, Naglik Julian R, Candidalysins Are a New Family of Cytolytic Fungal Peptide Toxins, MBio. 13 (2022) e03510–21. 10.1128/mbio.03510-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Vidarsson G, Dekkers G, Rispens T, IgG Subclasses and Allotypes: From Structure to Effector Functions, Frontiers in Immunology. 5 (2014). 10.3389/fimmu.2014.00520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Ansaldo E, Slayden LC, Ching KL, Koch MA, Wolf NK, Plichta DR, Brown EM, Graham DB, Xavier RJ, Moon JJ, Barton GM, Akkermansia muciniphila induces intestinal adaptive immune responses during homeostasis, Science. 364 (2019) 1179. 10.1126/science.aaw7479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Zeng MY, Cisalpino D, Varadarajan S, Hellman J, Warren HS, Cascalho M, Inohara N, Núñez G, Gut Microbiota-Induced Immunoglobulin G Controls Systemic Infection by Symbiotic Bacteria and Pathogens, Immunity. 44 (2016) 647–658. 10.1016/j.immuni.2016.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Christmann BS, Abrahamsson TR, Bernstein CN, Duck LW, Mannon PJ, Berg G, Björkstén B, Jenmalm MC, Elson CO, Human seroreactivity to gut microbiota antigens, Journal of Allergy and Clinical Immunology. 136 (2015) 1378–1386.e5. 10.1016/j.jaci.2015.03.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Bunker JJ, Erickson SA, Flynn TM, Henry C, Koval JC, Meisel M, Jabri B, Antonopoulos DA, Wilson PC, Bendelac A, Natural polyreactive IgA antibodies coat the intestinal microbiota, Science. 358 (2017) eaan6619. 10.1126/science.aan6619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Li H, Limenitakis JP, Greiff V, Yilmaz B, Schären O, Urbaniak C, Zünd M, Lawson MAE, Young ID, Rupp S, Heikenwälder M, McCoy KD, Hapfelmeier S, Ganal-Vonarburg SC, Macpherson AJ, Mucosal or systemic microbiota exposures shape the B cell repertoire, Nature. 584 (2020) 274–278. 10.1038/s41586-020-2564-6. [DOI] [PubMed] [Google Scholar]
  • [76].Matthews RC, Burnie JP, Howat D, Rowland T, Walton F, Autoantibody to heat-shock protein 90 can mediate protection against systemic candidosis, Immunology. 74 (1991) 20–24. [PMC free article] [PubMed] [Google Scholar]
  • [77].Viudes A, Lazzell A, Perea S, Kirkpatrick WR, Peman J, Patterson TF, Martinez JP, López-Ribot JL, The C-terminal antibody binding domain of Candida albicans mp58 represents a protective epitope during candidiasis, FEMS Microbiology Letters. 232 (2004) 133–138. 10.1016/S0378-1097(04)00042-4. [DOI] [PubMed] [Google Scholar]
  • [78].McClelland EE, Nicola AM, Prados-Rosales R, Casadevall A, Ab binding alters gene expression in Cryptococcus neoformans and directly modulates fungal metabolism, The Journal of Clinical Investigation. 120 (2010) 1355–1361. 10.1172/JCI38322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Trevijano-Contador N, Pianalto KM, Nichols CB, Zaragoza O, Alspaugh JA, Pirofski L, Bäumler AJ, Human IgM Inhibits the Formation of Titan-Like Cells in Cryptococcus neoformans, Infection and Immunity. 88 (2020) e00046–20. https://doi.org/doi: 10.1128/IAI.00046-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Chiani P, Bromuro C, Cassone A, Torosantucci A, Anti-β-glucan antibodies in healthy human subjects, Vaccine. 27 (2009) 513–519. 10.1016/j.vaccine.2008.11.030. [DOI] [PubMed] [Google Scholar]
  • [81].Cywes-Bentley C, Skurnik D, Zaidi T, Roux D, DeOliveira RB, Garrett WS, Lu X, O’Malley J, Kinzel K, Zaidi T, Rey A, Perrin C, Fichorova RN, Kayatani AKK, Maira-Litràn T, Gening ML, Tsvetkov YE, Nifantiev NE, Bakaletz LO, Pelton SI, Golenbock DT, Pier GB, Antibody to a conserved antigenic target is protective against diverse prokaryotic and eukaryotic pathogens, Proceedings of the National Academy of Sciences. 110 (2013) E2209. 10.1073/pnas.1303573110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Moreno-Sabater A, Autaa G, Sterlin D, Jerbi A, Villette R, Holm JB, Parizot C, Selim S, Senghor Y, Ghillani-Dalbin P, Bachmeyer C, Hennequin C, Gorochov G, Larsen M, Systemic anti-commensal response to fungi analyzed by flow cytometry is related to gut mycobiome ecology, Microbiome. 8 (2020) 159. 10.1186/s40168-020-00924-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Huertas B, Prieto D, Pitarch A, Gil C, Pla J, Díez-Orejas R, Serum Antibody Profile during Colonization of the Mouse Gut by Candida albicans: Relevance for Protection during Systemic Infection, Journal of Proteome Research. 16 (2017) 335–345. 10.1021/acs.jproteome.6b00383. [DOI] [PubMed] [Google Scholar]
  • [84].Jensen J, Warner T, Johnson C, Balish E, Oral Immunization of Mice against Candidiasis, The Journal of Infectious Diseases. 174 (1996) 133–140. 10.1093/infdis/174.1.133. [DOI] [PubMed] [Google Scholar]
  • [85].Montagnoli C, Bozza S, Bacci A, Gaziano R, Mosci P, Morschhäuser J, Pitzurra L, Kopf M, Cutler J, Romani L, A role for antibodies in the generation of memory antifungal immunity, European Journal of Immunology. 33 (2003) 1193–1204. 10.1002/eji.200323790. [DOI] [PubMed] [Google Scholar]
  • [86].Wang L, Ray A, Jiang X, y Wang J, Basu S, Liu X, Qian T, He R, Dittel BN, Chu Y, T regulatory cells and B cells cooperate to form a regulatory loop that maintains gut homeostasis and suppresses dextran sulfate sodium-induced colitis, Mucosal Immunology. 8 (2015) 1297–1312. 10.1038/mi.2015.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Herbrecht R, Bories P, Moulin J-C, Ledoux M-P, Letscher-Bru V, Risk stratification for invasive aspergillosis in immunocompromised patients, Annals of the New York Academy of Sciences. 1272 (2012) 23–30. 10.1111/j.1749-6632.2012.06829.x. [DOI] [PubMed] [Google Scholar]
  • [88].Lionakis MS, Kontoyiannis DP, Glucocorticoids and invasive fungal infections, Lancet. 362 (2003) 1828–1838. 10.1016/S0140-6736(03)14904-5. [DOI] [PubMed] [Google Scholar]
  • [89].Schauwvlieghe AFAD, Rijnders BJA, Philips N, Verwijs R, Vanderbeke L, Tienen CV, Lagrou K, Verweij PE, de Veerdonk FLV, Gommers D, Spronk P, Bergmans DCJJ, Hoedemaekers A, Andrinopoulou E-R, van den Berg CHSB, Juffermans NP, Hodiamont CJ, Vonk AG, Depuydt P, Boelens J, Wauters J, Invasive aspergillosis in patients admitted to the intensive care unit with severe influenza: a retrospective cohort study, The Lancet Respiratory Medicine. 6 (2018) 782–792. 10.1016/S2213-2600(18)30274-1. [DOI] [PubMed] [Google Scholar]
  • [90].Janssen NAF, Nyga R, Vanderbeke L, Jacobs C, Ergün M, Buil JB, van Dijk K, Altenburg J, Bouman CSC, van der Spoel HI, Rijnders BJA, Dunbar A, Schouten JA, Lagrou K, Bourgeois M, Reynders M, van Regenmortel N, Rutsaert L, Lormans P, Feys S, Debavaye Y, Tamion F, Costa D, Maizel J, Dupont H, Chouaki T, Nseir S, Sendid B, Brüggemann RJM, van de Veerdonk FL, Wauters J, Verweij PE, Multinational Observational Cohort Study of COVID-19-Associated Pulmonary Aspergillosis1, Emerg Infect Dis. 27 (2021) 2892–2898. 10.3201/eid2711.211174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Sarden N, Sinha S, Potts KG, Pernet E, Hiroki CH, Hassanabad MF, Nguyen AP, Lou Y, Farias R, Winston BW, Bromley A, Snarr BD, Zucoloto AZ, Andonegui G, Muruve DA, McDonald B, Sheppard DC, Mahoney DJ, Divangahi M, Rosin N, Biernaskie J, Yipp BG, A B1a–natural IgG–neutrophil axis is impaired in viral- and steroid-associated aspergillosis, Science Translational Medicine. 14 (2022) eabq6682. 10.1126/scitranslmed.abq6682. [DOI] [PubMed] [Google Scholar]
  • [92].Mircescu MM, Lipuma L, van Rooijen N, Pamer EG, Hohl TM, Essential Role for Neutrophils but not Alveolar Macrophages at Early Time Points following Aspergillus fumigatus Infection, The Journal of Infectious Diseases. 200 (2009) 647–656. 10.1086/600380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Fitzpatrick Z, Frazer G, Ferro A, Clare S, Bouladoux N, Ferdinand J, Tuong ZK, Negro-Demontel ML, Kumar N, Suchanek O, Tajsic T, Harcourt K, Scott K, Bashford-Rogers R, Helmy A, Reich DS, Belkaid Y, Lawley TD, McGavern DB, Clatworthy MR, Gut-educated IgA plasma cells defend the meningeal venous sinuses, Nature. 587 (2020) 472–476. 10.1038/s41586-020-2886-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Drummond RA, Collar AL, Swamydas M, Rodriguez CA, Lim JK, Mendez LM, Fink DL, Hsu AP, Zhai B, Karauzum H, Mikelis CM, Rose SR, Ferre EMN, Yockey L, Lemberg K, Kuehn HS, Rosenzweig SD, Lin X, Chittiboina P, Datta SK, Belhorn TH, Weimer ET, Hernandez ML, Hohl TM, Kuhns DB, Lionakis MS, CARD9-Dependent Neutrophil Recruitment Protects against Fungal Invasion of the Central Nervous System, PLOS Pathogens. 11 (2015) e1005293. 10.1371/journal.ppat.1005293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Rapaka RR, Ricks DM, Alcorn JF, Chen K, Khader SA, Zheng M, Plevy S, Bengtén E, Kolls JK, Conserved natural IgM antibodies mediate innate and adaptive immunity against the opportunistic fungus Pneumocystis murina, Journal of Experimental Medicine. 207 (2010) 2907–2919. 10.1084/jem.20100034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Bos NA, Kimura H, Meeuwsen CG, Visser HD, Hazenberg MP, Wostmann BS, Pleasants JR, Benner R, Marcus DM, Serum immunoglobulin levels and naturally occurring antibodies against carbohydrate antigens in germ-free BALB/c mice fed chemically defined ultrafiltered diet, European Journal of Immunology. 19 (1989) 2335–2339. 10.1002/eji.1830191223. [DOI] [PubMed] [Google Scholar]
  • [97].Bunker JJ, Bendelac A, IgA Responses to Microbiota, Immunity. 49 (2018) 211–224. 10.1016/j.immuni.2018.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].New JS, Dizon BLP, Fucile CF, Rosenberg AF, Kearney JF, King RG, Neonatal Exposure to Commensal-Bacteria-Derived Antigens Directs Polysaccharide-Specific B-1 B Cell Repertoire Development, Immunity. 53 (2020) 172–186.e6. 10.1016/j.immuni.2020.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Panda S, Zhang J, Tan NS, Ho B, Ding JL, Natural IgG antibodies provide innate protection against ficolin-opsonized bacteria, Embo j. 32 (2013) 2905–19. 10.1038/emboj.2013.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].He B, Xu W, Santini PA, Polydorides AD, Chiu A, Estrella J, Shan M, Chadburn A, Villanacci V, Plebani A, Knowles DM, Rescigno M, Cerutti A, Intestinal Bacteria Trigger T Cell-Independent Immunoglobulin A2 Class Switching by Inducing Epithelial-Cell Secretion of the Cytokine APRIL, Immunity. 26 (2007) 812–826. 10.1016/j.immuni.2007.04.014. [DOI] [PubMed] [Google Scholar]
  • [101].Kim Y-I, Song J-H, Ko H-J, Kweon M-N, Kang C-Y, Reinecker H-C, Chang S-Y, CX3CR1+ Macrophages and CD8+ T Cells Control Intestinal IgA Production, The Journal of Immunology. 201 (2018) 1287. 10.4049/jimmunol.1701459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Grasset EK, Chorny A, Casas-Recasens S, Gutzeit C, Bongers G, Thomsen I, Chen L, He Z, Matthews DB, Oropallo MA, Veeramreddy P, Uzzan M, Mortha A, Carrillo J, Reis BS, Ramanujam M, Sintes J, Magri G, Maglione PJ, Cunningham-Rundles C, Bram RJ, Faith J, Mehandru S, Pabst O, Cerutti A, Gut T cell–independent IgA responses to commensal bacteria require engagement of the TACI receptor on B cells, Science Immunology. 5 (2020) eaat7117. 10.1126/sciimmunol.aat7117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Bunker JJ, Flynn TM, Koval JC, Shaw DG, Meisel M, McDonald BD, Ishizuka IE, Dent AL, Wilson PC, Jabri B, Antonopoulos DA, Bendelac A, Innate and Adaptive Humoral Responses Coat Distinct Commensal Bacteria with Immunoglobulin A, Immunity. 43 (2015) 541–53. 10.1016/j.immuni.2015.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Kabbert J, Benckert J, Rollenske T, Hitch TCA, Clavel T, Cerovic V, Wardemann H, Pabst O, High microbiota reactivity of adult human intestinal IgA requires somatic mutations, Journal of Experimental Medicine. 217 (2020). 10.1084/jem.20200275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Victora GD, Nussenzweig MC, Germinal Centers, Annual Review of Immunology. 40 (2022) null. 10.1146/annurev-immunol-120419-022408. [DOI] [PubMed] [Google Scholar]
  • [106].Camacho SA, Kosco-Vilbois MH, Berek C, The dynamic structure of the germinal center, Immunol Today. 19 (1998) 511–4. 10.1016/s0167-5699(98)01327-9. [DOI] [PubMed] [Google Scholar]
  • [107].Ise W, Fujii K, Shiroguchi K, Ito A, Kometani K, Takeda K, Kawakami E, Yamashita K, Suzuki K, Okada T, Kurosaki T, T Follicular Helper Cell-Germinal Center B Cell Interaction Strength Regulates Entry into Plasma Cell or Recycling Germinal Center Cell Fate, Immunity. 48 (2018) 702–715.e4. 10.1016/j.immuni.2018.03.027. [DOI] [PubMed] [Google Scholar]
  • [108].Liu D, Xu H, Shih C, Wan Z, Ma X, Ma W, Luo D, Qi H, T–B-cell entanglement and ICOSL-driven feed-forward regulation of germinal centre reaction, Nature. 517 (2015) 214–218. 10.1038/nature13803. [DOI] [PubMed] [Google Scholar]
  • [109].Shulman Z, Gitlin Alexander D, Weinstein Jason S, Lainez B, Esplugues E, Flavell Richard A, Craft Joseph E, Nussenzweig Michel C, Dynamic signaling by T follicular helper cells during germinal center B cell selection, Science. 345 (2014) 1058–1062. 10.1126/science.1257861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].De Jesus M, Rodriguez AE, Yagita H, Ostroff GR, Mantis NJ, Sampling of Candida albicans and Candida tropicalis by Langerin-positive dendritic cells in mouse Peyer’s patches, Immunology Letters. 168 (2015) 64–72. 10.1016/j.imlet.2015.09.008. [DOI] [PubMed] [Google Scholar]
  • [111].LeibundGut-Landmann S, Groß O, Robinson MJ, Osorio F, Slack EC, Tsoni SV, Schweighoffer E, Tybulewicz V, Brown GD, Ruland J, Reis e Sousa C, Syk- and CARD9-dependent coupling of innate immunity to the induction of T helper cells that produce interleukin 17, Nature Immunology. 8 (2007) 630–638. 10.1038/ni1460. [DOI] [PubMed] [Google Scholar]
  • [112].Leonardi I, Li X, Iliev ID, Macrophage interactions with fungi and bacteria in inflammatory bowel disease, Current Opinion in Gastroenterology. 34 (2018). https://journals.lww.com/co-gastroenterology/Fulltext/2018/11000/Macrophage_interactions_with_fungi_and_bacteria_in.5.aspx. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Leonardi I, Li X, Semon A, Li D, Doron I, Putzel G, Bar A, Prieto D, Rescigno M, McGovern DPB, Pla J, Iliev ID, CX3CR1(+) mononuclear phagocytes control immunity to intestinal fungi, Science. 359 (2018) 232–236. 10.1126/science.aao1503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Dobeš J, Ben-Nun O, Binyamin A, Stoler-Barak L, Oftedal BE, Goldfarb Y, Kadouri N, Gruper Y, Givony T, Zalayat I, Kováčová K, Böhmová H, Valter E, Shulman Z, Filipp D, Husebye ES, Abramson J, Extrathymic expression of Aire controls the induction of effective TH17 cell-mediated immune response to Candida albicans, Nature Immunology. 23 (2022) 1098–1108. 10.1038/s41590-022-01247-6. [DOI] [PubMed] [Google Scholar]
  • [115].Schulz O, Jaensson E, Persson EK, Liu X, Worbs T, Agace WW, Pabst O, Intestinal CD103+, but not CX3CR1+, antigen sampling cells migrate in lymph and serve classical dendritic cell functions, Journal of Experimental Medicine. 206 (2009) 3101–3114. 10.1084/jem.20091925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Mowat AM, Agace WW, Regional specialization within the intestinal immune system, Nature Reviews Immunology. 14 (2014) 667–685. 10.1038/nri3738. [DOI] [PubMed] [Google Scholar]
  • [117].Diehl GE, Longman RS, Zhang J-X, Breart B, Galan C, Cuesta A, Schwab SR, Littman DR, Microbiota restricts trafficking of bacteria to mesenteric lymph nodes by CX3CR1hi cells, Nature. 494 (2013) 116–120. 10.1038/nature11809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Fransen F, Zagato E, Mazzini E, Fosso B, Manzari C, El Aidy S, Chiavelli A, D’Erchia AM, Sethi MK, Pabst O, Marzano M, Moretti S, Romani L, Penna G, Pesole G, Rescigno M, BALB/c and C57BL/6 Mice Differ in Polyreactive IgA Abundance, which Impacts the Generation of Antigen-Specific IgA and Microbiota Diversity, Immunity. 43 (2015) 527–40. 10.1016/j.immuni.2015.08.011. [DOI] [PubMed] [Google Scholar]
  • [119].Mazzini E, Massimiliano L, Penna G, Rescigno M, Oral Tolerance Can Be Established via Gap Junction Transfer of Fed Antigens from CX3CR1+ Macrophages to CD103+ Dendritic Cells, Immunity. 40 (2014) 248–261. 10.1016/j.immuni.2013.12.012. [DOI] [PubMed] [Google Scholar]
  • [120].Kim M, Galan C, Hill AA, Wu W-J, Fehlner-Peach H, Song HW, Schady D, Bettini ML, Simpson KW, Longman RS, Littman DR, Diehl GE, Critical Role for the Microbiota in CX3CR1+ Intestinal Mononuclear Phagocyte Regulation of Intestinal T Cell Responses, Immunity. 49 (2018) 151–163.e5. 10.1016/j.immuni.2018.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Panea C, Farkas AM, Goto Y, Abdollahi-Roodsaz S, Lee C, Koscsó B, Gowda K, Hohl TM, Bogunovic M, Ivanov II, Intestinal Monocyte-Derived Macrophages Control Commensal-Specific Th17 Responses, Cell Reports. 12 (2015) 1314–1324. 10.1016/j.celrep.2015.07.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Doron I, Leonardi I, Iliev ID, Profound mycobiome differences between segregated mouse colonies do not influence Th17 responses to a newly introduced gut fungal commensal, Fungal Genet Biol. 127 (2019) 45–49. 10.1016/j.fgb.2019.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Satpathy AT, KC W, Albring JC, Edelson BT, Kretzer NM, Bhattacharya D, Murphy TL, Murphy KM, Zbtb46 expression distinguishes classical dendritic cells and their committed progenitors from other immune lineages, Journal of Experimental Medicine. 209 (2012) 1135–1152. 10.1084/jem.20120030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Bogunovic M, Ginhoux F, Helft J, Shang L, Hashimoto D, Greter M, Liu K, Jakubzick C, Ingersoll MA, Leboeuf M, Stanley ER, Nussenzweig M, Lira SA, Randolph GJ, Merad M, Origin of the Lamina Propria Dendritic Cell Network, Immunity. 31 (2009) 513–525. 10.1016/j.immuni.2009.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Gross M, Salame T-M, Jung S, Guardians of the Gut – Murine Intestinal Macrophages and Dendritic Cells, Frontiers in Immunology. 6 (2015). 10.3389/fimmu.2015.00254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Farache J, Koren I, Milo I, Gurevich I, Kim K-W, Zigmond E, Furtado GC, Lira SA, Shakhar G, Luminal Bacteria Recruit CD103+ Dendritic Cells into the Intestinal Epithelium to Sample Bacterial Antigens for Presentation, Immunity. 38 (2013) 581–595. 10.1016/j.immuni.2013.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].McDole JR, Wheeler LW, McDonald KG, Wang B, Konjufca V, Knoop KA, Newberry RD, Miller MJ, Goblet cells deliver luminal antigen to CD103+ dendritic cells in the small intestine, Nature. 483 (2012) 345–349. 10.1038/nature10863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Yoshida M, Claypool SM, Wagner JS, Mizoguchi E, Mizoguchi A, Roopenian DC, Lencer WI, Blumberg RS, Human Neonatal Fc Receptor Mediates Transport of IgG into Luminal Secretions for Delivery of Antigens to Mucosal Dendritic Cells, Immunity. 20 (2004) 769–783. 10.1016/j.immuni.2004.05.007. [DOI] [PubMed] [Google Scholar]
  • [129].Jang MH, Sougawa N, Tanaka T, Hirata T, Hiroi T, Tohya K, Guo Z, Umemoto E, Ebisuno Y, Yang B-G, Seoh J-Y, Lipp M, Kiyono H, Miyasaka M, CCR7 Is Critically Important for Migration of Dendritic Cells in Intestinal Lamina Propria to Mesenteric Lymph Nodes, The Journal of Immunology. 176 (2006) 803. 10.4049/jimmunol.176.2.803. [DOI] [PubMed] [Google Scholar]
  • [130].Schlitzer A, McGovern N, Teo P, Zelante T, Atarashi K, Low D, Ho AW, See P, Shin A, Wasan PS, Hoeffel G, Malleret B, Heiseke A, Chew S, Jardine L, Purvis HA, Hilkens CM, Tam J, Poidinger M, Stanley ER, Krug AB, Renia L, Sivasankar B, Ng LG, Collin M, Ricciardi-Castagnoli P, Honda K, Haniffa M, Ginhoux F, IRF4 transcription factor-dependent CD11b+ dendritic cells in human and mouse control mucosal IL-17 cytokine responses, Immunity. 38 (2013) 970–83. 10.1016/j.immuni.2013.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Worbs T, Bode U, Yan S, Hoffmann MW, Hintzen G, Bernhardt G, Förster R, Pabst O, Oral tolerance originates in the intestinal immune system and relies on antigen carriage by dendritic cells, The Journal of Experimental Medicine. 203 (2006) 519–527. 10.1084/jem.20052016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Welty NE, Staley C, Ghilardi N, Sadowsky MJ, Igyártó BZ, Kaplan DH, Intestinal lamina propria dendritic cells maintain T cell homeostasis but do not affect commensalism, Journal of Experimental Medicine. 210 (2013) 2011–2024. 10.1084/jem.20130728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Flores-Langarica A, Cook C, Müller Luda K, Persson EK, Marshall JL, Beristain-Covarrubias N, Yam-Puc JC, Dahlgren M, Persson JJ, Uematsu S, Akira S, Henderson IR, Lindbom BJ, Agace W, Cunningham AF, Intestinal CD103+CD11b+ cDC2 Conventional Dendritic Cells Are Required for Primary CD4+ T and B Cell Responses to Soluble Flagellin, Frontiers in Immunology. 9 (2018). 10.3389/fimmu.2018.02409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Johansson-Lindbom B, Svensson M, Pabst O, Palmqvist C, Marquez G, Förster R, Agace WW, Functional specialization of gut CD103+ dendritic cells in the regulation of tissue-selective T cell homing, The Journal of Experimental Medicine. 202 (2005) 1063–1073. 10.1084/jem.20051100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Persson EK, Uronen-Hansson H, Semmrich M, Rivollier A, Hägerbrand K, Marsal J, Gudjonsson S, Håkansson U, Reizis B, Kotarsky K, Agace WW, IRF4 Transcription-Factor-Dependent CD103+CD11b+ Dendritic Cells Drive Mucosal T Helper 17 Cell Differentiation, Immunity. 38 (2013) 958–969. 10.1016/j.immuni.2013.03.009. [DOI] [PubMed] [Google Scholar]
  • [136].Salazar-Gonzalez RM, Niess JH, Zammit DJ, Ravindran R, Srinivasan A, Maxwell JR, Stoklasek T, Yadav R, Williams IR, Gu X, McCormick BA, Pazos MA, Vella AT, Lefrancois L, Reinecker H-C, McSorley SJ, CCR6-Mediated Dendritic Cell Activation of Pathogen-Specific T Cells in Peyer’s Patches, Immunity. 24 (2006) 623–632. 10.1016/j.immuni.2006.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Sierro F, Dubois B, Coste A, Kaiserlian D, Kraehenbuhl J-P, Sirard J-C, Flagellin stimulation of intestinal epithelial cells triggers CCL20-mediated migration of dendritic cells, Proceedings of the National Academy of Sciences. 98 (2001) 13722. 10.1073/pnas.241308598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Gross O, Gewies A, Finger K, Schäfer M, Sparwasser T, Peschel C, Förster I, Ruland J, Card9 controls a non-TLR signalling pathway for innate anti-fungal immunity, Nature. 442 (2006) 651–656. 10.1038/nature04926. [DOI] [PubMed] [Google Scholar]
  • [139].Agrawal S, Gupta S, Agrawal A, Human Dendritic Cells Activated via Dectin-1 Are Efficient at Priming Th17, Cytotoxic CD8 T and B Cell Responses, PLOS ONE. 5 (2010) e13418. 10.1371/journal.pone.0013418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Goodridge HS, Underhill DM, Touret N, Mechanisms of Fc Receptor and Dectin-1 Activation for Phagocytosis, Traffic. 13 (2012) 1062–1071. 10.1111/j.1600-0854.2012.01382.x. [DOI] [PubMed] [Google Scholar]
  • [141].Taylor PR, Tsoni SV, Willment JA, Dennehy KM, Rosas M, Findon H, Haynes K, Steele C, Botto M, Gordon S, Brown GD, Dectin-1 is required for β-glucan recognition and control of fungal infection, Nature Immunology. 8 (2007) 31–38. 10.1038/ni1408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Saijo S, Ikeda S, Yamabe K, Kakuta S, Ishigame H, Akitsu A, Fujikado N, Kusaka T, Kubo S, Chung S, Komatsu R, Miura N, Adachi Y, Ohno N, Shibuya K, Yamamoto N, Kawakami K, Yamasaki S, Saito T, Akira S, Iwakura Y, Dectin-2 Recognition of α-Mannans and Induction of Th17 Cell Differentiation Is Essential for Host Defense against Candida albicans, Immunity. 32 (2010) 681–691. 10.1016/j.immuni.2010.05.001. [DOI] [PubMed] [Google Scholar]
  • [143].Bi L, Gojestani S, Wu W, Hsu Y-MS, Zhu J, Ariizumi K, Lin X, CARD9 mediates dectin-2-induced IkappaBalpha kinase ubiquitination leading to activation of NF-kappaB in response to stimulation by the hyphal form of Candida albicans, The Journal of Biological Chemistry. 285 (2010) 25969–25977. 10.1074/jbc.M110.131300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [144].Wells CA, Salvage-Jones JA, Li X, Hitchens K, Butcher S, Murray RZ, Beckhouse AG, Lo Y-L-S, Manzanero S, Cobbold C, Schroder K, Ma B, Orr S, Stewart L, Lebus D, Sobieszczuk P, Hume DA, Stow J, Blanchard H, Ashman RB, The Macrophage-Inducible C-Type Lectin, Mincle, Is an Essential Component of the Innate Immune Response to Candida albicans, The Journal of Immunology. 180 (2008) 7404. 10.4049/jimmunol.180.11.7404. [DOI] [PubMed] [Google Scholar]
  • [145].Sato K, Yang XL, Yudate T, Chung JS, Wu J, Luby-Phelps K, Kimberly RP, Underhill D, Cruz PD Jr., Ariizumi K, Dectin-2 is a pattern recognition receptor for fungi that couples with the Fc receptor gamma chain to induce innate immune responses, J Biol Chem. 281 (2006) 38854–66. 10.1074/jbc.M606542200. [DOI] [PubMed] [Google Scholar]
  • [146].Lobato-Pascual A, Saether PC, Fossum S, Dissen E, Daws MR, Mincle, the receptor for mycobacterial cord factor, forms a functional receptor complex with MCL and FcεRI-γ, European Journal of Immunology. 43 (2013) 3167–3174. 10.1002/eji.201343752. [DOI] [PubMed] [Google Scholar]
  • [147].von Bernuth H, Picard C, Jin Z, Pankla R, Xiao H, Ku C-L, Chrabieh M, Mustapha IB, Ghandil P, Camcioglu Y, Vasconcelos J, Sirvent N, Guedes M, Vitor AB, Herrero-Mata MJ, Aróstegui JI, Rodrigo C, Alsina L, Ruiz-Ortiz E, Juan M, Fortuny C, Yagüe J, Antón J, Pascal M, Chang H-H, Janniere L, Rose Y, Garty B-Z, Chapel H, Issekutz A, Maródi L, Rodriguez-Gallego C, Banchereau J, Abel L, Li X, Chaussabel D, Puel A, Casanova J-L, Pyogenic Bacterial Infections in Humans with MyD88 Deficiency, Science. 321 (2008) 691–696. https://doi.org/doi: 10.1126/science.1158298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [148].Ferreira-Gomes M, Wich M, Böde S, Hube B, Jacobsen ID, Jungnickel B, B Cell Recognition of Candida albicans Hyphae via TLR 2 Promotes IgG1 and IL-6 Secretion for TH17 Differentiation, Frontiers in Immunology. 12 (2021). 10.3389/fimmu.2021.698849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Dillon S, Agrawal S, Banerjee K, Letterio J, Denning TL, Oswald-Richter K, Kasprowicz DJ, Kellar K, Pare J, van Dyke T, Ziegler S, Unutmaz D, Pulendran B, Yeast zymosan, a stimulus for TLR2 and dectin-1, induces regulatory antigen-presenting cells and immunological tolerance, The Journal of Clinical Investigation. 116 (2006) 916–928. 10.1172/JCI27203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Sutmuller RPM, den Brok MHMGM, Kramer M, Bennink EJ, Toonen LWJ, Kullberg B-J, Joosten LA, Akira S, Netea MG, Adema GJ, Toll-like receptor 2 controls expansion and function of regulatory T cells, The Journal of Clinical Investigation. 116 (2006) 485–494. 10.1172/JCI25439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Tada H, Nemoto E, Shimauchi H, Watanabe T, Mikami T, Matsumoto T, Ohno N, Tamura H, Shibata K, Akashi S, Miyake K, Sugawara S, Takada H, Saccharomyces cerevisiae- and Candida albicans-Derived Mannan Induced Production of Tumor Necrosis Factor Alpha by Human Monocytes in a CD14- and Toll-Like Receptor 4-Dependent Manner, Microbiology and Immunology. 46 (2002) 503–512. 10.1111/j.1348-0421.2002.tb02727.x. [DOI] [PubMed] [Google Scholar]
  • [152].van de Veerdonk FL, Netea MG, Jansen TJ, Jacobs L, Verschueren I, van der Meer JWM, Kullberg BJ, Redundant role of TLR9 for anti-Candida host defense, Immunobiology. 213 (2008) 613–620. 10.1016/j.imbio.2008.05.002. [DOI] [PubMed] [Google Scholar]
  • [153].Dzharullaeva AS, Tukhvatulin AI, Erokhova AS, Bandelyuk AS, Polyakov NB, Solovyev AI, Nikitenko NA, Shcheblyakov DV, Naroditsky BS, Logunov DY, Gintsburg AL, Stimulation of Dectin-1 and Dectin-2 during Parenteral Immunization, but Not Mincle, Induces Secretory IgA in Intestinal Mucosa, Journal of Immunology Research. 2018 (2018) 3835720. 10.1155/2018/3835720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Ferrante A, Beard LJ, Feldman RG, IgG subclass distribution of antibodies to bacterial and viral antigens, The Pediatric Infectious Disease Journal. 9 (1990). https://journals.lww.com/pidj/Fulltext/1990/08001/IgG_subclass_distribution_of_antibodies_to.4.aspx. [PubMed] [Google Scholar]
  • [155].Hjelholt A, Christiansen G, Sørensen US, Birkelund S, IgG subclass profiles in normal human sera of antibodies specific to five kinds of microbial antigens, Pathog Dis. 67 (2013) 206–13. 10.1111/2049-632x.12034. [DOI] [PubMed] [Google Scholar]
  • [156].Kuijpers TW, Weening RS, Out TA, IgG subclass deficiencies and recurrent pyogenic infections, unresponsiveness against bacterial polysaccharide antigens, Allergol Immunopathol (Madr). 20 (1992) 28–34. [PubMed] [Google Scholar]
  • [157].von Gunten S, Smith DF, Cummings RD, Riedel S, Miescher S, Schaub A, Hamilton RG, Bochner BS, Intravenous immunoglobulin contains a broad repertoire of anticarbohydrate antibodies that is not restricted to the IgG2 subclass, Journal of Allergy and Clinical Immunology. 123 (2009) 1268–1276.e15. 10.1016/j.jaci.2009.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [158].Nouri-Aria KT, Wachholz PA, Francis JN, Jacobson MR, Walker SM, Wilcock LK, Staple SQ, Aalberse RC, Till SJ, Durham SR, Grass pollen immunotherapy induces mucosal and peripheral IL-10 responses and blocking IgG activity, J Immunol. 172 (2004) 3252–9. 10.4049/jimmunol.172.5.3252. [DOI] [PubMed] [Google Scholar]
  • [159].Aalberse RC, Stapel SO, Schuurman J, Rispens T, Immunoglobulin G4: an odd antibody, Clin Exp Allergy. 39 (2009) 469–77. 10.1111/j.1365-2222.2009.03207.x. [DOI] [PubMed] [Google Scholar]
  • [160].Nimmerjahn F, Ravetch JV, Fcγ receptors as regulators of immune responses, Nature Reviews Immunology. 8 (2008) 34–47. 10.1038/nri2206. [DOI] [PubMed] [Google Scholar]
  • [161].Castro-Dopico T, Dennison TW, Ferdinand JR, Mathews RJ, Fleming A, Clift D, Stewart BJ, Jing C, Strongili K, Labzin LI, Monk EJM, Saeb-Parsy K, Bryant CE, Clare S, Parkes M, Clatworthy MR, Anti-commensal IgG Drives Intestinal Inflammation and Type 17 Immunity in Ulcerative Colitis, Immunity. 50 (2019) 1099–1114.e10. 10.1016/j.immuni.2019.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [162].Fleming A, Castro-Dopico T, Clatworthy MR, B cell class switching in intestinal immunity in health and disease, Scandinavian Journal of Immunology. 95 (2022) e13139. 10.1111/sji.13139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].Stapleton NM, Andersen JT, Stemerding AM, Bjarnarson SP, Verheul RC, Gerritsen J, Zhao Y, Kleijer M, Sandlie I, de Haas M, Jonsdottir I, van der Schoot CE, Vidarsson G, Competition for FcRn-mediated transport gives rise to short half-life of human IgG3 and offers therapeutic potential, Nature Communications. 2 (2011) 599. 10.1038/ncomms1608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].Glocker E-O, Hennigs A, Nabavi M, Schäffer AA, Woellner C, Salzer U, Pfeifer D, Veelken H, Warnatz K, Tahami F, Jamal S, Manguiat A, Rezaei N, Amirzargar AA, Plebani A, Hannesschläger N, Gross O, Ruland J, Grimbacher B, A Homozygous CARD9 Mutation in a Family with Susceptibility to Fungal Infections, New England Journal of Medicine. 361 (2009) 1727–1735. 10.1056/NEJMoa0810719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Rogiers O, Frising Ulrika C, Kucharíková S, Jabra-Rizk Mary A, van Loo G, Van Dijck P, Wullaert A, Lorenz M, Candidalysin Crucially Contributes to Nlrp3 Inflammasome Activation by Candida albicans Hyphae, MBio. 10 (n.d.) e02221–18. 10.1128/mBio.02221-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [166].Teng F, Klinger CN, Felix KM, Bradley CP, Wu E, Tran NL, Umesaki Y, Wu H-JJ, Gut Microbiota Drive Autoimmune Arthritis by Promoting Differentiation and Migration of Peyer’s Patch T Follicular Helper Cells, Immunity. 44 (2016) 875–888. 10.1016/j.immuni.2016.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Koscsó B, Kurapati S, Rodrigues RR, Nedjic J, Gowda K, Shin C, Soni C, Ashraf AZ, Purushothaman I, Palisoc M, Xu S, Sun H, Chodisetti SB, Lin E, Mack M, Kawasawa YI, He P, Rahman ZSM, Aifantis I, Shulzhenko N, Morgun A, Bogunovic M, Gut-resident CX3CR1hi macrophages induce tertiary lymphoid structures and IgA response in situ, Science Immunology. 5 (2020) eaax0062. 10.1126/sciimmunol.aax0062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [168].Viladomiu M, Kivolowitz C, Abdulhamid A, Dogan B, Victorio D, Castellanos JG, Woo V, Teng F, Tran NL, Sczesnak A, Chai C, Kim M, Diehl GE, Ajami NJ, Petrosino JF, Zhou XK, Schwartzman S, Mandl LA, Abramowitz M, Jacob V, Bosworth B, Steinlauf A, Scherl EJ, Wu H-JJ, Simpson KW, Longman RS, IgA-coated E coli enriched in Crohn’s disease spondyloarthritis promote TH17-dependent inflammation, Science Translational Medicine. 9 (2017) eaaf9655. 10.1126/scitranslmed.aaf9655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Michaud E, Waeckel L, Gayet R, Goguyer-Deschaumes R, Chanut B, Jospin F, Bathany K, Monnoye M, Genet C, Prier A, Tokarski C, Gérard P, Roblin X, Rochereau N, Paul S, Alteration of microbiota antibody-mediated immune selection contributes to dysbiosis in inflammatory bowel diseases, EMBO Molecular Medicine. n/a (2022) e15386. 10.15252/emmm.202115386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [170].Uo M, Hisamatsu T, Miyoshi J, Kaito D, Yoneno K, Kitazume MT, Mori M, Sugita A, Koganei K, Matsuoka K, Kanai T, Hibi T, Mucosal CXCR4+ IgG plasma cells contribute to the pathogenesis of human ulcerative colitis through FcγR-mediated CD14 macrophage activation, Gut. 62 (2013) 1734–44. 10.1136/gutjnl-2012-303063. [DOI] [PubMed] [Google Scholar]
  • [171].Uzzan M, Martin JC, Mesin L, Livanos AE, Castro-Dopico T, Huang R, Petralia F, Magri G, Kumar S, Zhao Q, Rosenstein AK, Tokuyama M, Sharma K, Ungaro R, Kosoy R, Jha D, Fischer J, Singh H, Keir ME, Ramamoorthi N, Gorman WEO, Cohen BL, Rahman A, Cossarini F, Seki A, Leyre L, Vaquero ST, Gurunathan S, Grasset EK, Losic B, Dubinsky M, Greenstein AJ, Gottlieb Z, Legnani P, George J, Irizar H, Stojmirovic A, Brodmerkel C, Kasarkis A, Sands BE, Furtado G, Lira SA, Tuong ZK, Ko HM, Cerutti A, Elson CO, Clatworthy MR, Merad M, Suárez-Fariñas M, Argmann C, Hackney JA, Victora GD, Randolph GJ, Kenigsberg E, Colombel JF, Mehandru S, Ulcerative colitis is characterized by a plasmablast-skewed humoral response associated with disease activity, Nature Medicine. 28 (2022) 766–779. 10.1038/s41591-022-01680-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [172].Hoarau G, Mukherjee PK, Gower-Rousseau C, Hager C, Chandra J, Retuerto MA, Neut C, Vermeire S, Clemente J, Colombel JF, Fujioka H, Poulain D, Sendid B, Ghannoum MA, Bacteriome and Mycobiome Interactions Underscore Microbial Dysbiosis in Familial Crohn’s Disease, MBio. 7 (2016). 10.1128/mBio.01250-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [173].Israeli E, Grotto I, Gilburd B, Balicer RD, Goldin E, Wiik A, Shoenfeld Y, Anti-Saccharomyces cerevisiae and antineutrophil cytoplasmic antibodies as predictors of inflammatory bowel disease, Gut. 54 (2005) 1232–6. 10.1136/gut.2004.060228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Riente L, Chimenti D, Pratesi F, Delle Sedie A, Tommasi S, Tommasi C, Bombardieri S, Migliorini P, Antibodies to tissue transglutaminase and Saccharomyces cerevisiae in ankylosing spondylitis and psoriatic arthritis, The Journal of Rheumatology. 31 (2004) 920. [PubMed] [Google Scholar]
  • [175].Tang V, Valim C, Moman R, Richman A, Zhou J, Ramgopal V, Albert R, Boone JH, Rufo PA, Assessment of Fecal ASCA Measurement as a Biomarker of Crohn Disease in Pediatric Patients, J Pediatr Gastroenterol Nutr. 64 (2017) 248–253. 10.1097/mpg.0000000000001244. [DOI] [PubMed] [Google Scholar]
  • [176].Standaert–Vitse A, Jouault T, Vandewalle P, Mille C, Seddik M, Sendid B, Mallet J, Colombel J, Poulain D, Candida albicans Is an Immunogen for Anti–Saccharomyces cerevisiae Antibody Markers of Crohn’s Disease, Gastroenterology. 130 (2006) 1764–1775. 10.1053/j.gastro.2006.02.009. [DOI] [PubMed] [Google Scholar]
  • [177].Standaert-Vitse A, Sendid B, Joossens M, François N, Vandewalle-El Khoury P, Branche J, Van Kruiningen H, Jouault T, Rutgeerts P, Gower-Rousseau C, Libersa C, Neut C, Broly F, Chamaillard M, Vermeire S, Poulain D, Colombel JF, Candida albicans colonization and ASCA in familial Crohn’s disease, Am J Gastroenterol. 104 (2009) 1745–53. 10.1038/ajg.2009.225. [DOI] [PubMed] [Google Scholar]
  • [178].Casadevall A, Kontoyiannis DP, Robert V, Kronstad JW, On the Emergence of Candida auris: Climate Change, Azoles, Swamps, and Birds, MBio. 10 (2019) e01397–19. https://doi.org/doi: 10.1128/mBio.01397-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Fisher MC, Henk DA, Briggs CJ, Brownstein JS, Madoff LC, McCraw SL, Gurr SJ, Emerging fungal threats to animal, plant and ecosystem health, Nature. 484 (2012) 186–194. 10.1038/nature10947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [180].Eyre DW, Sheppard AE, Madder H, Moir I, Moroney R, Quan TP, Griffiths D, George S, Butcher L, Morgan M, Newnham R, Sunderland M, Clarke T, Foster D, Hoffman P, Borman AM, Johnson EM, Moore G, Brown CS, Walker AS, Peto TEA, Crook DW, Jeffery KJM, A Candida auris Outbreak and Its Control in an Intensive Care Setting, New England Journal of Medicine. 379 (2018) 1322–1331. 10.1056/NEJMoa1714373. [DOI] [PubMed] [Google Scholar]
  • [181].Proctor DM, Dangana T, Sexton DJ, Fukuda C, Yelin RD, Stanley M, Bell PB, Baskaran S, Deming C, Chen Q, Conlan S, Park M, Mullikin J, Thomas J, Young A, Bouffard G, Barnabas B, Brooks S, Han J, Ho S, Kim J, Legaspi R, Maduro Q, Marfani H, Montemayor C, Riebow N, Schandler K, Schmidt B, Sison C, Stantripop M, Black S, Dekhtyar M, Masiello C, McDowell J, Thomas P, Vemulapalli M, Welsh RM, Vallabhaneni S, Chiller T, Forsberg K, Black SR, Pacilli M, Kong HH, Lin MY, Schoeny ME, Litvintseva AP, Segre JA, Hayden MK, Program NCS, Integrated genomic, epidemiologic investigation of Candida auris skin colonization in a skilled nursing facility, Nature Medicine. 27 (2021) 1401–1409. 10.1038/s41591-021-01383-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Wang X, Sui X, Yan L, Wang Y, Cao Y, Jiang Y, Vaccines in the treatment of invasive candidiasis, Virulence. 6 (2015) 309–315. 10.4161/21505594.2014.983015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Han Y, Kanbe T, Cherniak R, Cutler JE, Biochemical characterization of Candida albicans epitopes that can elicit protective and nonprotective antibodies, Infection and Immunity. 65 (1997) 4100–4107. 10.1128/iai.65.10.4100-4107.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [184].Torosantucci A, Chiani P, Bromuro C, De Bernardis F, Palma AS, Liu Y, Mignogna G, Maras B, Colone M, Stringaro A, Zamboni S, Feizi T, Cassone A, Protection by Anti-β-Glucan Antibodies Is Associated with Restricted β-1,3 Glucan Binding Specificity and Inhibition of Fungal Growth and Adherence, PLOS ONE. 4 (2009) e5392. 10.1371/journal.pone.0005392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Edwards John E J., Schwartz MM, Schmidt CS, Sobel JD, Nyirjesy P, Schodel F, Marchus E, Lizakowski M, DeMontigny EA, Hoeg J, Holmberg T, Cooke MT, Hoover K, Edwards L, Jacobs M, Sussman S, Augenbraun M, Drusano M, Yeaman MR, Ibrahim AS, Filler SG, Hennessey John P J., A Fungal Immunotherapeutic Vaccine (NDV-3A) for Treatment of Recurrent Vulvovaginal Candidiasis—A Phase 2 Randomized, Double-Blind, Placebo-Controlled Trial, Clinical Infectious Diseases. 66 (2018) 1928–1936. 10.1093/cid/ciy185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Rudkin FM, Raziunaite I, Workman H, Essono S, Belmonte R, MacCallum DM, Johnson EM, Silva LM, Palma AS, Feizi T, Jensen A, Erwig LP, Gow NAR, Single human B cell-derived monoclonal anti-Candida antibodies enhance phagocytosis and protect against disseminated candidiasis, Nature Communications. 9 (2018) 5288. 10.1038/s41467-018-07738-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Sandini S, La Valle R, Deaglio S, Malavasi F, Cassone A, De Bernardis F, A highly immunogenic recombinant and truncated protein of the secreted aspartic proteases family (rSap2t) of Candida albicans as a mucosal anticandidal vaccine, FEMS Immunology & Medical Microbiology. 62 (2011) 215–224. 10.1111/j.1574-695X.2011.00802.x. [DOI] [PubMed] [Google Scholar]
  • [188].Noble SM, Gianetti BA, Witchley JN, Candida albicans cell-type switching and functional plasticity in the mammalian host, Nature Reviews Microbiology. 15 (2017) 96–108. 10.1038/nrmicro.2016.157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [189].Lionakis MS, Lim JK, Lee CC, Murphy PM, Organ-specific innate immune responses in a mouse model of invasive candidiasis, J Innate Immun. 3 (2011) 180–99. 10.1159/000321157. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES