Abstract
Mucosa-associated invariant T (MAIT) cells are a large population of unconventional T cells widely distributed in the human gastrointestinal tract. Their homing to the gut is central to maintaining mucosal homeostasis and immunity. This review discusses the potential mechanisms that guide MAIT cells to the intestinal mucosa during homeostasis and inflammation, emphasizing the roles of chemokines, chemokine receptors, and tissue adhesion molecules. The potential influence of the gut microbiota on MAIT cell homing to different regions of the human gut is also discussed. Last, we introduce how organoid technology offers a potentially valuable approach to advance our understanding of MAIT cell tissue homing by providing a more physiologically relevant model that mimics the human gut tissue. These models may enable a detailed investigation of the gut-specific homing mechanisms of MAIT cells. By understanding the regulation of MAIT cell homing to the human gut, potential avenues for therapeutic interventions targeting gut inflammatory conditions such as inflammatory bowel diseases (IBD) may emerge.
MAIT cell homing to the gut in health and disease provides insights to guide therapeutic development for intestinal inflammation.
INTRODUCTION TO MAIT CELLS
Mucosal-associated invariant T (MAIT) cells constitute a large population of innate-like T cells in humans. Unlike classical T cells, MAIT cells express a semi-invariant T cell receptor (TCR) composed of an invariant TCR Vα and Jα segment (TRAV1-2-TRAJ33/12/20), paired with a limited set of TCRβ chains, predominantly TRBV20 or TRBV6 in humans (1, 2). This unique TCR arrangement allows MAIT cells to recognize various pyrimidine-related antigenic metabolites presented by the nonpolymorphic major histocompatibility complex class I–related protein (MR1) (3, 4), including those derived from microbial metabolites derived from riboflavin biosynthesis (4, 5). The MR1 gene is highly evolutionarily conserved among mammals, and of all the MHC class I–like proteins, MR1 shows the highest conservation among eutherian mammals, including in various ungulates and chiropterans (6–8). Among the antigenic metabolites of riboflavin biosynthesis that MAIT cells recognize, the most potent stimulatory MR1-binding ligands found are 5-OP-RU [5-(2-oxopropylideneamino)-5-d-ribitylaminouracil] and 5-OE-RU [5-(2-oxoethylideneamino)-5-d-ribitylaminouracil] (5).
In the human blood, the majority of MAIT cells are CD4−CD8+ subsets; ~10 to 15% CD4−CD8− subsets and less than ~5 to 10% CD4+CD8− and CD4+CD8+ subsets [reviewed in (9)]. MAIT cells exhibit innate-like immune responses, which are in part driven by the transcription factor promyelocytic leukemia zinc finger to respond quickly to infections (10). MAIT cells expressing T-box transcription factor TBX21 (T-bet), Eomesodermin (Eomes), and B lymphocyte–induced maturation protein 1 (BLIMP-1) have type 1 immunity through secretion of interferon-γ (IFN-γ), tumor necrosis factor (TNF), and cytolytic proteins including granzyme B, perforin, and granulysin when detecting bacterial infection. Activated MAIT cells can also produce type 17 immune responses, primarily identified by their interleukin-17 (IL-17) secretion that is driven by retinoic acid receptor-related orphan receptor gamma (RORγt) and signal transducer and activator of transcription 3 (STAT3), which are highly relevant in the mucosal sites (11, 12). Interestingly, IL-17–producing human MAIT cells have the capacity to switch to secreting IFN-γ following TCR stimulation in the presence of polarizing cytokines (13). Prolonged TCR stimulation can induce MAIT cells to produce type 2 cytokines, including IL-4, IL-10, and IL-13 (14–16). Follicular helper–like MAIT cells have also been identified in human tonsils, which help in B cell differentiation and the generation of high-quality antibody responses (17). In addition, there exist regulatory-type MAIT cells characterized by the expression of forkhead box (Fox) P3, Helios, and CD39 (18–21). In summary, there is substantial heterogeneity and plasticity within the human MAIT cell population, with their diverse functional roles in health and various diseases still requiring further exploration.
CELLULAR DISTRIBUTION OF MAIT CELLS IN THE HUMAN GUT MUCOSA
Because of the challenges of obtaining gut tissue samples from healthy humans, our understanding of MAIT cell distribution within the gut mucosa has been somewhat limited. However, recent research has revealed that MAIT cells are indeed present in the human gut, with varying levels of abundance across different regions and tissue compartments (21–28). This growing body of evidence provides valuable insights into the specific localization and frequency of MAIT cells within the gut, contributing to a better understanding of their role in gut immunity.
The initial discovery revealed that human TRAV1-2-TRAJ33 MAIT cells were located specifically in the lamina propria (LP) of the gut, based on analysis of gut biopsies that included tissue from the epithelium, Peyer’s patches, and LP (3). In addition, MAIT cells have been identified in the LP of the human stomach, where they exhibit a memory phenotype comparable to those found in peripheral blood (22). In healthy human jejunal tissue, only small numbers of intraepithelial (IE) MAIT cells were detected using in situ TRAV1-2+ CD161+ staining, with confirmation via MR1 tetramer staining (1, 25). Although only a subset of TRAV1-2+ CD161hi T cells in intestinal tissues reacted to the MR1-5-OP-RU tetramer, a definitive marker for MAIT cells, these cells were observed in both the LP and IE layers of the gastrointestinal mucosa, with their abundance varying across these compartments (25). Furthermore, MAIT cells were found in colectomy samples of both colon adenocarcinomas and unaffected colon tissue, with similar frequencies observed in the IE compartment and LP (23). In patients with colorectal cancer or cecal appendix cancer, a comparison of MAIT cell frequencies among intraepithelial lymphocytes (IELs) and lamina propria lymphocytes showed that the cecum harbored substantially higher frequencies of MAIT cells in the IEL compartment compared to the colon (24). Immunohistochemistry also revealed that TRAV1-2+ cells were predominantly found in the LP of the colon but were also present in IE regions of the cecum (24). Collectively, it is evident that MAIT cells are primarily located within the LP of the gut mucosa while also present at lower numbers in the IE compartment, reflecting their widespread distribution within the gut mucosa.
MAIT CELL FUNCTION WITHIN THE HUMAN GUT
MAIT cells mediate effector functions through two key mechanisms: the TCR-dependent and TCR-independent pathways. In the TCR-dependent pathway, MAIT cells recognize riboflavin metabolites presented by MR1 molecules, which triggers the release of proinflammatory cytokines such as IFN-γ and TNF, as well as cytotoxic molecules, including granulysin, granzymes, and perforin (29–32). This pathway is essential for responding to microbial infections such as riboflavin-producing bacteria and fungi (11, 30, 33). Alternatively, the TCR-independent pathway driven by cytokines such as IL-12 and IL-18 enables MAIT cells to respond even when riboflavin metabolites are absent, broadening their role to include infection caused by viruses and nonriboflavin-producing microbes (10, 27, 34–36), cancer, and noninfectious inflammation in the intestine (37). Other cytokines, including IL-7, IL-15, and type I IFNs, further enhance MAIT cell activity, allowing them to produce cytotoxic molecules and inflammatory cytokines (12, 29, 38). These two activation pathways often work together, resulting in stronger effector responses, with MAIT cells producing higher levels of cytokines and chemokines (36, 38). In addition, glycolytic, lipid, and oxidative phosphorylative metabolic pathways regulate the development and function of human MAIT cells in circulation and the lung (39–41). Whether such metabolic programming shapes gut resident MAIT cell effector responses is yet to be established.
Although studies indicate that MAIT cells are proinflammatory and may promote tissue damage, including within the human gut (22, 26, 42–47), there is an increasing appreciation of the tissue repair and healing roles of MAIT cells [reviewed in (27)]. Such studies showed the expression of tissue repair gene signatures following TCR stimulations (34, 36, 38) and production of tissue repair factors, including the cytokines IL-17 and IL-22 (21, 26, 48, 49), FURIN, CCL3 (C-C motif chemokine ligand 3), VEGFB (vascular endothelial growth factor B), CSF2 (granulocyte-macrophage colony-stimulating factor), and AREG (amphiregulin), which initiate tissue repair and reinforce barrier integrity (34, 36, 38, 50, 51). In a mouse model of colitis, MAIT cell ligands generated in the colon were found to rapidly cross the intestinal barrier, activating MAIT cells, which in turn expressed tissue repair genes and produced barrier-promoting mediators, including HIF1A (hypoxia-inducible factor 1 subunit alpha) and FURIN (52). Mice lacking MAIT cells were shown to be more susceptible to chronic colitis and colitis-induced colorectal cancer (CRC) (52). Interestingly, topical administration of the prototypical MR1 ligand 5-OP-RU was sufficient to stimulate wound healing by recruiting cutaneous MAIT cells and production of IL-17 (50). Similarly, in a human-like mouse model of full-thickness skin injury, MAIT cells express a tissue repair transcriptomic program and promote skin wound healing through their recruitment into the wound site in a TCR-independent but CXCR6-dependent manner (51). AREG-deficient mice exhibited appreciably delayed wound healing, indicating the role of Areg in the tissue healing function of MAIT cells (51). These findings underscore the importance of MAIT cells in tissue repair and maintaining barrier integrity during both infection and inflammatory conditions. However, while MAIT cells in the appendix produce IL-17 and IL-22 in response to bacterial stimulation (26), and intestinal MAIT cells have demonstrated regulatory functions that may contribute to barrier protection (21), further research is needed to determine whether MAIT cells are involved in other tissue repair processes within the human gut.
MAIT CELL SEEDING INTO THE GUT MUCOSAL TISSUES DURING DEVELOPMENT
During thymic development in mice, MAIT cells undergo positive selection via MR1 molecules expressed by CD4+ CD8+ double-positive (DP) thymocytes (53, 54). Human MAIT cells also develop within the thymus, and their intrathymic development follows three distinct stages, as outlined in a seminal study by Koay and colleagues (55). In the first stage, MAIT cells express both the CD4 and CD8 coreceptors (CD4+CD8+ DP cells) but notably lack expression of CD27 and key MAIT cell markers, CD161 and IL-18 receptor α-subunit (IL-18Rα). These receptors are progressively up-regulated during the second and third stages of MAIT cell development (55). MAIT cells exit the thymus primarily as either CD8+ single-positive cells or as double-negative (DN; CD4− CD8−) cells in a naive state (55, 56).
In the periphery, the maturation and expansion of naive MAIT cells are primarily driven by microbial exposure, particularly through riboflavin metabolites (55, 57). The gut microbiota plays a crucial role in this process, as commensal bacteria producing the riboflavin metabolites are essential for MAIT cell maturation (50, 57, 58) and germ-free mice have substantially fewer MAIT cells (3, 55). Interestingly, MAIT cells are already present in low numbers within the gut mucosae of second-trimester human fetuses and exhibit innate-like antimicrobial responses before the establishment of commensal microflora in the gut (59). This suggests that the seeding and functionality of MAIT cells in the human gut may occur independently of microbial colonization (59). However, since MR1 ligands can enter the circulation from barrier tissues and contribute to the development and accumulation of MAIT cells at distal sites (50, 57), it is tempting to speculate that these ligands may cross the placenta during pregnancy, entering the fetal circulation. This process could facilitate the seeding and functional maturation of MAIT cells in the fetal gut. Further insights from mouse models revealed that cytotoxicity-associated genes were only expressed by MAIT1 cells in the thymus (60). However, MAIT cells in the mouse intestine not only expressed cytotoxic genes but also up-regulated a tissue repair gene signature, indicating that MAIT cells undergo additional functional maturation after leaving the thymus (60). This highlights their ability to adapt and acquire tissue-specific roles.
Within human circulation, the frequency of MAIT cells increased from birth until about 25 years of age, after which they declined (61–65). Interestingly, single-nucleotide polymorphisms of IL7RA—the gene encoding IL-7 receptor α-subunit—are associated with increased MAIT cell levels within the circulation, further highlighting the potent influence of IL-7 on MAIT cell biology (66). Following adulthood, the CD8+ MAIT cell population decreases, while the DN MAIT cell population appears to accumulate with age (63, 67, 68). Subsequent investigations reveal that this may be contributed by the differentiation of the dominant CD8+ MAIT cell population into DN MAIT cells (67). Furthermore, progressively mature DN MAIT cells accumulate in human fetal tissues, while the CD8+ subset contracts in response (67). This suggests that the differentiation of peripheral CD8+ MAIT cells into the DN subpopulation might also occur without a fully established microbiota in the mucosal tissues. The DN MAIT cells exhibit distinct functional characteristics, including enhanced IL-17 production and increased susceptibility to apoptosis (67). In contrast, CD8+ MAIT cells show increased IFN-γ and TNF production (67, 69). This enhanced function in CD8+ MAIT cells is due to the interaction of CD8 with MR1, which acts as a functional coreceptor for these cells (69). Collectively, this sequence of studies supports the importance of both microbial interactions and the local environment in the maturation and functional diversification of MAIT cells, albeit that fully established microbiota may not be essential in certain circumstances.
THE POTENTIAL ROLE OF THE GUT MICROBIOTA AND MAIT CELL DISTRIBUTION IN THE HUMAN GUT
The gut harbors complex and dynamic microbial communities, influenced by multiple factors, including environment, diet, and age (Fig. 1) (70). These microbiota communities play a crucial role in shaping the distribution and function of the gut immune cell populations. Whether varied MAIT cell population in the intestine [reviewed in (27)] is associated with various microbial compositions and related riboflavin biosynthesis (71) as the microbiota composition shifts with age, diet, and environmental factors remains little understood (Fig. 2).
Fig. 1. The changes of commensal bacteria in intestinal tract along with age.
Lactobacillales and Bacillales may be detected at low levels in meconium, although the microbiome is predominantly established only after birth. The gut is rapidly colonized by Bifidobacteriales, Bacteroidales, and Clostridiales after birth and then replaced by a diverse mixture of Clostridiales and Oscillospirales between 6 and 24 months. By around 2 to 3 years of age, the composition of bacterial community resembles adult-like biomass with the dominance of Bacteroidales, Oscillospirales, and Lactobacillales. The microbial community shifts with an increased abundance of Bacteroidales and Clostridiales in the older individuals. Created with BioRender.com.
Fig. 2. The potential association of MAIT cell distribution and commensal bacteria in the intestinal tract.
Values indicate the mean or range of MAIT cell frequency in the relevant tissues as percentages of total T cells. (+) indicates riboflavin-producing bacteria. (−) indicates nonriboflavin-producing bacteria. (+/−) indicates that parts of the bacterial Order produce riboflavin (71). The abundance of Bifidobacteriales and Lactobacillales appears to be higher in faecal/luminal samples than in the mucosa, whereas Clostridiales and Lactobacillales are detected in the mucus layer as well as crypts of intestinal epithelium (84, 85). Created with BioRender.com.
The gut microbiota composition undergoes substantial changes across the life span, beginning with rapid colonization after birth, followed by the establishment of a more diverse and stable microbiota in early childhood, and eventual shifts in older age (Fig. 1) (70, 72, 73). These age-related variations in microbial communities could influence the early seeding of MAIT cells to the gut, given that MAIT cells recognize microbial metabolites produced through the riboflavin biosynthesis pathway, which is common among gut bacteria, including Bacteroidales, Bacillales, Vellionellales, and Clostridiales (74). Notably, sulfated secondary bile acids, which represent a distinct class of MR1 ligands, are produced through the metabolic activity of the microbiome and lead to MAIT cell survival and the expression of a homeostatic gene signature (75). In neonates, the predominance of early colonizers such as Lactobacillales and Bacillales (76–78) may limit the availability of riboflavin-dependent bacteria and may influence MAIT cell activity during this period. As the infant’s gut transitions to a more diverse bacterial community with the introduction of solid foods, the presence of Clostridiales and Oscillospirales increases (79–81), potentially promoting greater MAIT cell localization and functionality to areas of high microbial density, such as the ileum and colon. In adults, the microbial community shifts toward increased Bacteroidales and Clostridiales abundance (73). These taxa are known to express the riboflavin biosynthesis pathway (71). The age-associated changes in bacterial composition may thus potentially influence MAIT cell localization and functionality in the human gut and their role in maintaining gut immune homeostasis.
The varying composition of bacteria across different sections of the gut may further influence the distribution and function of MAIT cells in different parts of the gut. In the small intestine, which has a relatively low bacterial biomass, MAIT cell localization may be limited due to the lower abundance of riboflavin-producing bacteria such as the Vellionellales, Bacillales, Clostridiales, Bacteroidales, and Enterobacterales (71). The colon contains abundant bacterial loads and is dominated by obligate anaerobes such as Bacteroidales, Clostridiales, and Bifidobacteriales (82). The variation in bacterial composition between the proximal and distal parts of the colon, with Bacteroidales being prevalent in the rectum (83), might result in localized differences in MAIT cell distribution and activity. These anaerobic bacterial taxa are known to produce riboflavin (71), potentially explaining the higher concentrations of MAIT cells in the large intestines (Fig. 2) (27). Such abundance may also lead to enhanced MAIT cell function, particularly in immune surveillance and response to microbial pathogens in these parts of the gut. In addition, the variation in bacterial distribution between the mucus layer and fecal content may also affect MAIT cell positioning and function (81, 84, 85), as different microbial niches within the gut could offer distinct signals for MAIT cell recruitment and retention (Fig. 2).
In summary, the composition of the gut microbiota, particularly the riboflavin-producing taxa, may influence the distribution and functional responses of MAIT cells within various parts of the human gut across different age groups. The link between microbial composition and MAIT cell distribution and function remains an important area for further research, with potential implications for understanding MAIT cell regulation in gut health and disease.
MECHANISMS OF MAIT CELL HOMING TO THE HUMAN GUT IN HEALTH AND PATHOLOGICAL CONDITIONS
The homing of MAIT cells to the gut and their retention involve several key factors, including chemokines, chemokine receptors, and tissue adhesion molecules (Fig. 3). These molecular signals coordinate MAIT cell migration and residence in the human gut, allowing them to perform immune effector and tissue repair roles. Dysregulation of these homing signals is often observed in pathological gut conditions, including during infection, inflammation, and malignancies. These perturbations may enhance MAIT cell recruitment and retention at these sites and promote increased effector responses that may exacerbate inflammation and tissue damage. Indeed, in multiple gut pathologies, MAIT cells frequently increase within the gut mucosal tissues, whereas their circulatory counterparts concomitantly decrease (27). Together, these observations indicate that circulating MAIT cells are commonly recruited during these gut pathologies. Here, we discuss various potential mechanisms that may guide MAIT cells to home to the gut in health and disease.
Fig. 3. MAIT cell homing to the gut in health and inflammation.
In homeostasis (left), MAIT cell homing to the stomach may rely on the chemokine receptor G protein–coupled receptor 25 (GPR25) and its ligand chemokine CXCL17, whereas in the small intestine, MAIT cells may depend on CCR9. GPR15 and its ligand GPR15LG contribute to MAIT cell homing to the large intestine. Subsequently, MAIT cells transmigrate from the gut microvasculature via the interaction of α4β7 integrin with mucosal addressin cell adhesion molecule–1 (MAdCAM-1) expressed on endothelial cells. α4β7 and α4β1 integrins may also interact with low levels of vascular cell adhesion molecule–1 (VCAM-1) expressed at these sites. MAIT cells may up-regulate αEβ7 integrin to interact with E-cadherin within the mucosal LP and IE layer. During inflammation (right), such as acute appendicitis, IBDs, and CRC, inflammatory chemokines increase at barrier sites and lead to the recruitment of MAIT cells through various chemokine receptors such as CCR1, CCR2, CCR4, CCR6, CCR9, CXCR3, and CXCR6. Increased levels of inflammatory cytokines at these sites partly contribute to the up-regulation of these chemokine receptors. Concomitantly, there is an increase in the expression of tissue adhesion molecules including MAdCAM-1, VCAM-1, and intercellular adhesion molecule–1 (ICAM-1), further enhancing MAIT cell entry into the gut mucosae. The balance between homeostasis and inflammation may lead to different recruitment mechanisms of MAIT cells and possibly different effector roles (protective versus pathogenic). Created with BioRender.com.
Chemokines and chemokine receptors
Chemokines are small signaling proteins and are among the first mobilizers of host responses in homeostasis and disease processes (86–88). The main function of chemokines is to guide the migration of immune cells during homeostasis, infection, and inflammation. By binding to G protein–coupled chemokine receptors expressed on immune cells, chemokines orchestrate the directed movement of immune cells toward sites of infection or tissue damage (89). Circulating MAIT cells express high levels of tissue-homing chemokine receptors (Table 1), including C-C motif chemokine receptor 6 (CCR6), CCR9, and C-X-C motif chemokine receptor 6 (CXCR6), as well as CCR2, CCR5, and CXCR3, allowing them to migrate (26, 90, 91). Chemokine and chemokine receptors, including CCR9 and its ligand CCL25, are important for the homing of MAIT cells to the gut, particularly to the small intestine, while other chemokine receptors including CCR6 and CXCR6 may also play roles (23, 92). The G protein–coupled receptor GPR25 and its ligand CXCL17 (C-X-C motif chemokine ligand 7) specifically recruit T cells into the upper gastrointestinal tract, such as the stomach, whereas GPR15 and its ligand GPR15LG to the colon (93–95). Human tonsillar MAIT cells migrated to both CXCL17 and GPR15LG in transwell experiments, suggesting the role of these chemokine receptor–ligand axes in MAIT cell recruitment to the stomach and colon, respectively (93). In other tissues, MAIT cells appear to migrate via different chemokine-chemokine receptor axes. For instance, circulating MAIT cells migrate to the intervillous region of the placenta via macrophage migration inhibitory factor (MIF), CCL20, and CCL25 (96). In another study, MAIT cells may egress from the tissues and recirculate within the lymphatic system, possibly via CCR7 (97).
Table 1. Expression of chemokine receptors on circulating MAIT cells at steady state and their potential chemokine ligands.
The expression of chemokine receptors by peripheral blood MAIT cells in a steady state may change during inflammatory conditions. The chemokine receptors and their ligands that are up-regulated during gut inflammation are depicted in Fig. 3.
| Chemokine receptors | Potential chemokine ligands |
|---|---|
| CCR2 (26, 91, 132) | CCL2, CCL7, CCL8, CCL13, and CCL16 |
| CCR5 (26, 91, 132) | CCL3, CCL4, CCL5, CCL14, CCL16, and CCL3L1 |
| CCR6 (26, 42, 91, 132) | CCL20 |
| CCR9 (42, 91) | CCL25 |
| CXCR3 (42, 91, 132) | CXCL9, CXCL10, and CXCL11 |
| CXCR4 (91, 132) | CXCL12 |
| CXCR6 (26, 42) | CXCL16 |
CD26/dipeptidylpeptidase IV is widely expressed in blood and tissues [reviewed in (98)]. A large number of human chemokines, including CXCL2, CXCL6, CXCL9, CXCL10, CXCL11, CXCL12, CCL3L1, CCL4, CCL5, CCL11, CCL14, and CCL22, are cleaved by CD26 (98). NH2-terminal cleavage of chemokines by CD26 has prominent effects on their receptor binding and signaling (99–102). Through modulation of chemokine activity, CD26 regulates leukocyte migration and progenitor cell release from the bone marrow (98, 103–105). Interestingly, MAIT cells uniformly express high levels of CD26 (61, 106, 107). However, it is now unclear whether CD26 expressed by MAIT cells may play roles on cleaving certain chemokines and influence MAIT cell migration to the gut.
During acute infection and inflammation of the gut, MAIT cells are also recruited to the pathological sites through different sets of chemokines and chemokine receptors. Recently, we reported the recruitment of circulating MAIT cells into the appendix tissues during acute appendicitis in children (26). Acute appendicitis is a severe inflammation as an outcome of bacterial infection in the appendix tissues, usually caused by gram-negative enteric bacteria. MAIT cell recruitment in acute appendicitis was mediated by CCR1 and CCR2 via their chemokine ligands CCL2, CCL3, and CCL8, as well as by CCR4 via their chemokine ligands CCL17 and CCL22, all of which were elevated in both plasma and inflamed appendix tissues (26). During Vibrio cholerae infection, there appears to be MAIT cell clonotype redistribution from the circulation into the duodenum following infection (45). In cases of necrotizing enterocolitis (NEC), another severe intestinal inflammation as an outcome of bacterial infection in the newly born, a decrease in circulating MAIT cells was observed, while there was an enrichment of these cells in the ileal tissues of patients with NEC (46). In a human challenge trial involving controlled infection with Salmonella enterica serovar Paratyphi A, MAIT cells were activated in the blood during infection, with an early decrease in their frequency occurring before the clinical diagnosis of enteric fever (108). Together, these bodies of studies strongly support the hypothesis that MAIT cells are actively recruited to the sites of acute infection and the accompanying inflammation.
Inflammatory Bowel Disease (IBD) is a chronic inflammatory condition affecting the intestines, characterized by repetitive episodes of inflammation of the gastrointestinal tract caused by abnormal immune responses (109). IBD primarily consists of Crohn’s disease (CD) and ulcerative colitis (UC); while CD affects the entire gastrointestinal tract, UC predominantly affects the large intestine (110). In this chronic inflammation, MAIT cells were activated, leading to enhanced recruitment to inflamed tissues, a shift in their phenotype, and alterations in their cytokine secretion patterns (42–44, 47). While MAIT cell frequency in the blood of patients with IBD was notably lower than in healthy donors, it was notably higher in inflamed colons compared to healthy colons (47). In addition, the expression of chemokines such as CCL20, CXCL10, CXCL16, and CCL25 was elevated in inflamed intestinal tissues compared to non-inflamed tissues (42). In patients with UC, the frequency of MAIT cells in peripheral blood was lower compared to healthy controls but elevated in inflamed mucosa, correlating with clinical and endoscopic disease activity (44). In another study, in both UC and CD patients, MAIT cell frequency was lower not only in peripheral blood but also in inflamed intestinal mucosa compared to non-IBD controls. This reduction may be due to the proapoptotic characteristics exhibited by MAIT cells in patients with IBD (111). In mouse models of spontaneous (112) and chemically induced colitis (113), MAIT cells appear to play a pathogenic role as MR1 deficiency or suppressing MAIT cell activation in these models lessened the severity of colitis without compromising gut integrity. In a contrasting study, MR1 ligands generated in the colon led to the secretion of tissue repair factors by MAIT cells and better colitis outcomes (52).
Patients with mucosal-associated malignancies, including gastric, colon, and lung cancers, had markedly lower circulating MAIT cell levels (114–116). MAIT cell levels in such cancer tissues were higher than in peripheral blood suggesting their recruitment (114, 115), with infiltration into the cancer tissues in some studies (114, 115). Circulating MAIT cells had high levels of CCR6 and CXCR6, and their chemokines, such as CCL20 and CXCL16, were highly expressed in colon cancer tissues (115). Interestingly, chemokine responsiveness is similar in both unaffected mucosa and tumor-associated MAIT cells (23).
Although viruses do not have riboflavin synthesis pathway and thus do not activate MAIT cells directly through the TCR-dependent pathways, they provoke various innate and proinflammatory cytokines and stimulate MAIT cells via this cytokine-driven pathway [reviewed in (117)]. In chronically infected HIV-1 patients, there is a distinct decrease of MAIT cells in the circulation (35, 118). Interestingly, MAIT cells were maintained or even increased in the colorectal tissues (35, 118, 119), suggesting that MAIT cells are recruited to the gut during chronic HIV-1 infection. Going forward, it will be important to determine the mechanisms underlying MAIT cell trafficking in chronic viral infections affecting the gut mucosa such as HIV-1 infection.
Tissue adhesion molecules
Integrins are the primary adhesion molecules that immune cells use to home into tissues. Their roles in guiding MAIT cell homing into tissues are less well understood than the more extensively studied chemokine-chemokine receptor axis that governs their migration. Given the limited studies on this subject, in this Review, we infer from the well-characterized roles of integrins in the homing mechanisms of conventional gut-resident T cells. In these T cells, integrins such as α4β1 and α4β7 play a crucial role in mediating homing to the intestinal mucosa (110), where they use these integrins to interact with the ligands on vascular endothelial cells (110). Certain chemokines activate integrins such as α4β1 and α4β7, which then bind to vascular cell adhesion molecule–1 (VCAM-1) and mucosal addressin cell adhesion molecule–1 (MAdCAM-1), respectively (110, 120). These molecules are expressed in the Peyer’s patches and mesenteric lymph nodes, facilitating the transmigration of T cells into gut tissues (121). Once in the gut, T cells adhere to the intestinal epithelium by interacting with E-cadherin through αEβ7 integrin, contributing to the immune effector processes (110, 120, 122). Other integrins, such as integrin αLβ2 (LFA-1) may additionally contribute to the accumulation of T cells into the gut tissues during inflammation through its interaction with intercellular adhesion molecule–1 (ICAM-1) [reviewed in (123)].
Transcriptomic analysis reveals that MAIT cells express multiple integrins, including ITGA1, ITGA4, ITGA5, ITGB1, and ITGB3 in the murine thymus (124), suggesting that these integrins may guide them to specific tissues. However, their exact functions are not fully understood. Indeed, human MAIT cells are known to express several integrins under normal physiology and during inflammation, including α4β7 and αEβ7 (26, 49). The presence of tissue-resident-like MAIT cells expressing CD69 and αE integrin (CD103) has been shown in the gastrointestinal tract, including in the oral mucosae (49), the appendix (26), and the intestine (21).
In diseases characterized by an overactive immune response, such as IBD, extensive infiltration of T cells into the intestinal tissue is observed. Integrins such as α4β1 and α4β7 regulate this tissue invasion, resulting in the accumulation of T cells during inflammation (120, 122). It is currently unclear what are the roles of integrins for MAIT cells during gut inflammation. However, perturbations of the α4β7 and αEβ7 expression have been observed in acute appendicitis (26). In addition, MAIT cells expressing the αLβ2 integrin can secrete IFN-γ through binding to ICAM-1, potentially targeting ICAM-rich endothelial vessels during inflammatory responses (125). During mycobacterial pulmonary infection, α4β1-expressing MAIT cells are recruited to the lungs through interaction with VCAM-1, potentially providing early immune protection in this mouse model of tuberculosis infection (126). Thus, integrin signaling in MAIT cells during inflammatory processes may not only increase the recruitment of MAIT cells but also enhance their effector functions.
The influence of gut dysbiosis on MAIT cell homing to the gut
Gut dysbiosis has been observed in various diseases, including infectious colitis, IBD, cancer, and obesity (127). Dysbiosis typically features a bloom of pathobionts and loss of commensal number or diversity, potentially leading to changes in riboflavin metabolite production in the gut (28, 128). Furthermore, dysbiosis may affect the levels and patterns of chemokines, chemokine receptors, and tissue adhesion molecules (129). Therefore, a dysbiotic microbiota may influence MAIT cell distribution and function in pathological conditions through the shift in the riboflavin metabolite antigen availability and the perturbation in the molecular signaling that governs MAIT cell homing to the gut and their retention within tissues.
Following exposure to bacteria-derived metabolic products, gut epithelial cells and immune cells residing within the intestinal epithelium express chemokines and adhesion molecules (129). The transplantation of mixed human fecal bacteria strains has been shown to up-regulate the expression of chemokines CXCL9 and CXCL10 in mouse epithelial cells (130). These chemokines are ligands for CXCR3, which is expressed by MAIT cells (91). In addition, gut dysbiosis induces the production of CCL5 from mouse colonic epithelial cells (131), potentially recruiting circulating MAIT cells to the intestine through a CCL5-CCR5 interaction. Interestingly, a high-fat diet, known to be a risk factor for gut microbiota dysbiosis, results in the activation of the CCL2-CCR2 axis in the small intestine, which may attract MAIT cells to the gut (26, 90, 132). Moreover, after antibiotic treatment, the recolonization of guts in mice by Enterocloster species down-regulates the expression of ileal MAdCAM-1 (133), the ligand for integrin α4β7 expressed by MAIT cells. In parallel, a positive correlation has been found between fecal Ruminococcus and VCAM-1 levels in the serum of obese children (134), which may enhance MAIT cell retention in the gut through interaction with integrin α4β1. Interestingly, the presence of aerotolerant bacteria in the microbiome is related to the protective function of MAIT cells in the mouse colitis model (52).
Our group recently showed that inflamed appendix tissues from most acute pediatric appendicitis patients were infected by riboflavin metabolite–producing Escherichia coli (26, 67, 135). Certain pathogenic E. coli strains, such as the adherentinvasive E. coli (136) promote the secretion of IL-1β, IL-23, and the chemokine CCL5 (137, 138). Indeed, in our study, we observed substantially increased levels of IL-1β, IL-18, and IL-23 (26). Such elevated proinflammatory cytokines activate circulating MAIT cells, leading to notable changes in the chemokine receptor expression pattern, including CCR1, CCR2, CCR4, CCR6, CXCR1, and CXCR6 (26). These changes are associated with the recruitment of MAIT cells to the appendix due to increased levels of the chemokines CCL2, CCL3, CCL8, CCL17, CCL20, CCL22, and CXCL8 within the inflamed tissues (26).
In summary, MAIT cell homing to the gut is driven by complex interactions of chemokines, chemokine receptors, and tissue adhesion molecules. These processes are crucial for immune surveillance in health but are dysregulated in many gut pathologies. Inflammation and dysbiosis may perturb the gut-homing activity of MAIT cells through changes to the riboflavin-related metabolite availability and the chemokine and tissue adhesion molecule profiles. Such conditions may promote excessive MAIT cell recruitment to affected tissues, where they often adopt inflammatory phenotypes. The redistribution of MAIT cells from circulation to inflamed sites consistently shown in multiple studies strongly supports their role in mucosal immunity and their contribution to pathogenesis. On the basis of these insights, we further discuss explorations going forward to understand better the mechanisms behind MAIT cell recruitment and function in disease contexts.
DECIPHERING POTENTIAL MECHANISMS OF MAIT CELL HOMING TO THE GUT THROUGH ORGANOID CULTURE SYSTEM
One of the primary challenges in fully understanding the mechanisms of MAIT cell homing to the human gut is the lack of effective human model systems. While MAIT cell migration and tissue homing are typically studied in vivo using animal models, there are limitations to this approach. MAIT cell biology in these animal models differs appreciably from humans, and the low number of MAIT cells in these models often necessitates genetic or biological manipulation to increase their levels, potentially altering their natural biology. To address this, a fully human system for studying MAIT cell biology is essential. However, conventional in vitro two-dimensional (2D) cell cultures and ex vivo methods are insufficient for this purpose. A potential alternative approach is the use of organoid models, which offer a more physiologically relevant environment. Unlike traditional 2D cultures, organoids can mimic in vivo tissue structures and functions (139), providing a more accurate platform for studying MAIT cell behaviors.
In a recent study, we successfully established a patient-derived intestinal organoid model where we examined the mechanism of MAIT cell recruitment during appendicitis and their interactions with the intestinal epithelial cells in 3D settings (26). Through this model system, we identified a previously unknown mechanism of MAIT cell recruitment toward inflamed human intestinal tissues, including the CCR4-CCL17/CCL22–mediated migration of MAIT cells (26). We further demonstrate the potentially pathological roles of MAIT cells in the setting of acute appendicitis through their activation and secretion of proinflammatory cytokines and cytotoxicity toward bacterially infected intestinal cells (26). Interestingly, in a separate study of MAIT cell roles in biliary atresia, biliary organoid models were used to demonstrate the tissue repair capacity of MAIT cells via the secretion of Areg (140). In addition, a murine intestinal organoid model has been used to explore the role of closely related invariant natural killer T cells in regulating intestinal epithelial cell homeostasis in mice (141).
While the use of organoid models to understand MAIT cell biology and their potential application in precision medicine and personalized treatment is still in its infancy, these models may hold considerable promise. Organoids could offer a more realistic, complex tissue-like environment that mimics human gut mucosal tissues, potentially enabling a deeper understanding of MAIT cell behavior in conditions including IBD, infections, and cancer. Patient-derived organoids could also be used to test targeted therapies that may lead to the development of more personalized treatments in diseases driven by MAIT cell dysregulation [reviewed in (27)]. As these models are further developed, refined, and integrated with tools such as drug screening, gene editing, and single-cell sequencing, they may further enhance our understanding of MAIT cells and their therapeutic potential in the gut.
FUTURE PERSPECTIVES AND CONCLUDING REMARKS
MAIT cells play dual roles in intestinal mucosal immunity, contributing to barrier integrity and tissue repair during homeostasis while driving inflammation in pathological conditions. It remains unclear whether these functions represent distinct roles of MAIT cells at different stages of tissue repairs following the precipitating insults. Tissue healing typically progresses through three phases: inflammation, regenerative tissue formation, and remodeling (142). MAIT cells may adapt their functions to support these distinct stages, or there could be distinct populations involved in different phases of tissue healing. Notably, tissue-resident MAIT cells may focus on maintaining homeostasis and repair, whereas circulating MAIT cells recruited to the pathological sites may mediate inflammation. Understanding these differences could reveal alternative therapeutic strategies to enhance repair without exacerbating inflammation. This knowledge could also inform the optimization of MAIT cell expansion protocols in vitro (132), potentially improving their therapeutic application.
Targeting specific chemokine pathways and integrin interactions may further promote MAIT cell homing to the gut, improving mucosal immunity and tissue repair. Conversely, blocking these pathways could help control inflammation and tissue damage. Various drugs have been developed to inhibit T cell infiltration into inflamed tissue, with antibody or receptor antagonist therapies targeting integrins and chemokine receptors being a key focus (110, 120, 143). For instance, the monoclonal antibody against CCR4 (mogamulizumab) approved to treat mycosis fungoides or Sézary syndrome (144, 145) and a dual antagonist targeting CCR2 and CCR5 (cenicriviroc) under evaluation for the treatment of HIV-1 infection and liver fibrosis (144, 146) may be repurposed to block MAIT cell migration in cases of acute intestinal inflammation (26). In addition, monoclonal antibodies against integrins now approved or under evaluation for the treatment of IBD, including those targeting α4β7 (vedolizumab) (147, 148), α4β7/αEβ7 (etrolizumab) (149, 150), or α4β1/α4β7 (natalizumab) (151), could also potentially block MAIT cell adhesion during intestinal inflammation (26). Combining chemokine receptor and integrin antagonists offers a potential strategy for regulating MAIT cell recruitment in inflammatory gut diseases by preventing excessive MAIT cell migration, adhesion, and retention while promoting egress from tissues. This potentially synergistic approach may help to reduce inflammation and tissue damage further.
In conclusion, targeting MAIT cell homing represents a promising therapeutic strategy for various gut-related diseases by modulating the processes that govern MAIT cell migration and activity in the gut. Future research will be crucial in translating these concepts into effective therapies.
Acknowledgments
Funding: This work was supported by Tsinghua Shenzhen International Graduate School grant QD2022018C (to E.L.); Tsinghua Shenzhen International Graduate School grant JC2022007 (to E.L.); Science, Technology and Innovation Commission of Shenzhen Municipality grant WDZC20220819153248002 (to E.L.); and Shenzhen Municipality Pengcheng Peacock Program (to E.L.).
Author contributions: Conceptualization: E.L. Investigation: Z.W., X.C., F.H., and E.L. Visualization: Z.W. and F.H. Supervision: E.L. Writing—original draft: Z.W., X.C., F.H., and E.L. Writing—review and editing: F.H. and E.L. All authors reviewed and approved the final manuscript.
Competing interests: E.L. and F.H. are named inventors on a patent application (no. 202211393019.8, China National Intellectual Property Administration, granted 12 November 2024) owned by Tsinghua Shenzhen International Graduate School. All other authors declare that they have no competing interests.
Data and materials availability: All data needed to evaluate the conclusions in the paper are present in the paper.
REFERENCES AND NOTES
- 1.Reantragoon R., Corbett A. J., Sakala I. G., Gherardin N. A., Furness J. B., Chen Z., Eckle S. B., Uldrich A. P., Birkinshaw R. W., Patel O., Kostenko L., Meehan B., Kedzierska K., Liu L., Fairlie D. P., Hansen T. H., Godfrey D. I., Rossjohn J., McCluskey J., Kjer-Nielsen L., Antigen-loaded MR1 tetramers define T cell receptor heterogeneity in mucosal-associated invariant T cells. J. Exp. Med. 210, 2305–2320 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Tilloy F., Treiner E., Park S. H., Garcia C., Lemonnier F., de la Salle H., Bendelac A., Bonneville M., Lantz O., An invariant T cell receptor alpha chain defines a novel TAP-independent major histocompatibility complex class Ib-restricted α/β T cell subpopulation in mammals. J. Exp. Med. 189, 1907–1921 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Treiner E., Duban L., Bahram S., Radosavljevic M., Wanner V., Tilloy F., Affaticati P., Gilfillan S., Lantz O., Selection of evolutionarily conserved mucosal-associated invariant T cells by MR1. Nature 422, 164–169 (2003). [DOI] [PubMed] [Google Scholar]
- 4.Kjer-Nielsen L., Patel O., Corbett A. J., Le Nours J., Meehan B., Liu L., Bhati M., Chen Z., Kostenko L., Reantragoon R., Williamson N. A., Purcell A. W., Dudek N. L., McConville M. J., O’Hair R. A., Khairallah G. N., Godfrey D. I., Fairlie D. P., Rossjohn J., McCluskey J., MR1 presents microbial vitamin B metabolites to MAIT cells. Nature 491, 717–723 (2012). [DOI] [PubMed] [Google Scholar]
- 5.Corbett A. J., Eckle S. B., Birkinshaw R. W., Liu L., Patel O., Mahony J., Chen Z., Reantragoon R., Meehan B., Cao H., Williamson N. A., Strugnell R. A., Van Sinderen D., Mak J. Y., Fairlie D. P., Kjer-Nielsen L., Rossjohn J., McCluskey J., T-cell activation by transitory neo-antigens derived from distinct microbial pathways. Nature 509, 361–365 (2014). [DOI] [PubMed] [Google Scholar]
- 6.Tsukamoto K., Deakin J. E., Graves J. A., Hashimoto K., Exceptionally high conservation of the MHC class I-related gene, MR1, among mammals. Immunogenetics 65, 115–124 (2013). [DOI] [PubMed] [Google Scholar]
- 7.Leeansyah E., Hey Y. Y., Sia W. R., Ng J. H. J., Gulam M. Y., Boulouis C., Zhu F., Ahn M., Mak J. Y. W., Fairlie D. P., Kwa A. L. H., Sandberg J. K., Wang L. F., MR1-restricted T cells with MAIT-like characteristics are functionally conserved in the pteropid bat Pteropus alecto. iScience 23, 101876 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Boudinot P., Mondot S., Jouneau L., Teyton L., Lefranc M. P., Lantz O., Restricting nonclassical MHC genes coevolve with TRAV genes used by innate-like T cells in mammals. Proc. Natl. Acad. Sci. U.S.A. 113, E2983–E2992 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Godfrey D. I., Koay H. F., McCluskey J., Gherardin N. A., The biology and functional importance of MAIT cells. Nat. Immunol. 20, 1110–1128 (2019). [DOI] [PubMed] [Google Scholar]
- 10.Ussher J. E., Bilton M., Attwod E., Shadwell J., Richardson R., de Lara C., Mettke E., Kurioka A., Hansen T. H., Klenerman P., Willberg C. B., CD161++ CD8+ T cells, including the MAIT cell subset, are specifically activated by IL-12+IL-18 in a TCR-independent manner. Eur. J. Immunol. 44, 195–203 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Provine N. M., Klenerman P., MAIT cells in health and disease. Annu. Rev. Immunol. 38, 203–228 (2020). [DOI] [PubMed] [Google Scholar]
- 12.Nel I., Bertrand L., Toubal A., Lehuen A., MAIT cells, guardians of skin and mucosa? Mucosal. Immunol. 14, 803–814 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wang H., Souter M. N. T., de Lima Moreira M., Li S., Zhou Y., Nelson A. G., Yu J., Meehan L. J., Meehan B. S., Eckle S. B. G., Lee H. J., Schröder J., Haque A., Mak J. Y. W., Fairlie D. P., McCluskey J., Wang Z., Chen Z., Corbett A. J., MAIT cell plasticity enables functional adaptation that drives antibacterial immune protection. Sci. Immunol. 9, eadp9841 (2024). [DOI] [PubMed] [Google Scholar]
- 14.Kelly J., Minoda Y., Meredith T., Cameron G., Philipp M. S., Pellicci D. G., Corbett A. J., Kurts C., Gray D. H., Godfrey D. I., Kannourakis G., Berzins S. P., Chronically stimulated human MAIT cells are unexpectedly potent IL-13 producers. Immunol. Cell. Biol. 97, 689–699 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Nel I., Beaudoin L., Gouda Z., Rousseau C., Soulard P., Rouland M., Bertrand L., Boitard C., Larger E., Lehuen A., MAIT cell alterations in adults with recent-onset and long-term type 1 diabetes. Diabetologia 64, 2306–2321 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Carolan E., Tobin L. M., Mangan B. A., Corrigan M., Gaoatswe G., Byrne G., Geoghegan J., Cody D., O’Connell J., Winter D. C., Doherty D. G., Lynch L., O’Shea D., Hogan A. E., Altered distribution and increased IL-17 production by mucosal-associated invariant T cells in adult and childhood obesity. J. Immunol. 194, 5775–5780 (2015). [DOI] [PubMed] [Google Scholar]
- 17.Jensen O., Trivedi S., Meier J. D., Fairfax K. C., Hale J. S., Leung D. T., A subset of follicular helper-like MAIT cells can provide B cell help and support antibody production in the mucosa. Sci. Immunol. 7, eabe8931 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Vorkas C. K., Krishna C., Li K., Aubé J., Fitzgerald D. W., Mazutis L., Leslie C. S., Glickman M. S., Single-cell transcriptional profiling reveals signatures of helper, effector, and regulatory MAIT cells during homeostasis and activation. J. Immunol. 208, 1042–1056 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Li S., Simoni Y., Becht E., Loh C. Y., Li N., Lachance D., Koo S. L., Lim T. P., Tan E. K. W., Mathew R., Nguyen A., Golovato J., Berkson J. D., Prlic M., Lee B., Minot S. S., Nagarajan N., Dey N., Tan D. S. W., Tan I. B., Newell E. W., Human tumor-infiltrating MAIT cells display hallmarks of bacterial antigen recognition in colorectal cancer. Cell. Rep. Med. 1, 100039 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Fu S., Liu M., Zhu C., Zhang H., Zhao C., Xie Y., Chen G., Sheng D., Pan J., He Z., Dai Y., Gao Y., Li X., Chen L., Qian Y., Jin T., Sun C., Tian Z., Wang H., Bai L., Regulatory mucosa-associated invariant T cells controlled by β1 adrenergic receptor signaling contribute to hepatocellular carcinoma progression. Hepatology 78, 72–87 (2023). [DOI] [PubMed] [Google Scholar]
- 21.Kammann T., Cai C., Sekine T., Mouchtaridi E., Boulouis C., Nilsén V., Ballesteros O. R., Müller T. R., Gao Y., Raineri E. J. M., Mily A., Adamo S., Constantz C., Niessl J., Weigel W., Kokkinou E., Stamper C., Marchalot A., Bassett J., Ferreira S., Rødahl I., Wild N., Brownlie D., Tibbitt C., Mak J. Y. W., Fairlie D. P., Leeansyah E., Michaelsson J., Marquardt N., Mjösberg J., Jorns C., Buggert M., Sandberg J. K., MAIT cell heterogeneity across paired human tissues reveals specialization of distinct regulatory and enhanced effector profiles. Sci. Immunol. 9, eadn2362 (2024). [DOI] [PubMed] [Google Scholar]
- 22.Booth J. S., Salerno-Goncalves R., Blanchard T. G., Patil S. A., Kader H. A., Safta A. M., Morningstar L. M., Czinn S. J., Greenwald B. D., Sztein M. B., Mucosal-associated invariant T cells in the human gastric mucosa and blood: Role in helicobacter pylori infection. Front. Immunol. 6, 466 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Sundström P., Ahlmanner F., Akéus P., Sundquist M., Alsén S., Yrlid U., Börjesson L., Sjöling Å., Gustavsson B., Wong S. B., Quiding-Järbrink M., Human mucosa-associated invariant T cells accumulate in colon adenocarcinomas but produce reduced amounts of IFN-γ. J. Immunol. 195, 3472–3481 (2015). [DOI] [PubMed] [Google Scholar]
- 24.Hama I., Tominaga K., Yamagiwa S., Setsu T., Kimura N., Kamimura H., Wakai T., Terai S., Different distribution of mucosal-associated invariant T cells within the human cecum and colon. Cent. Eur. J. Immunol. 44, 75–83 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Boonpattanaporn N., Kongkaew T., Sengprasert P., Souter M. N. T., Lakananurak N., Rerknimitr R., Corbett A. J., Reantragoon R., Human mucosal Vα7.2+ CD161hi T cell distribution at physiologic state and in Helicobacter pylori infection. J. Leukoc. Biol. 112, 717–732 (2022). [DOI] [PubMed] [Google Scholar]
- 26.Zheng Y., Han F., Wu Z., Wang B., Chen X., Boulouis C., Jiang Y., Ho A., He D., Sia W. R., Mak J. Y. W., Fairlie D. P., Wang L. F., Sandberg J. K., Lobie P. E., Ma S., Leeansyah E., MAIT cell activation and recruitment in inflammation and tissue damage in acute appendicitis. Sci. Adv. 10, eadn6331 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zheng Y., Han F., Ho A., Xue Y., Wu Z., Chen X., Sandberg J. K., Ma S., Leeansyah E., Role of MAIT cells in gastrointestinal tract bacterial infections in humans: More than a gut feeling. Mucosal Immunol. 16, 740–752 (2023). [DOI] [PubMed] [Google Scholar]
- 28.Schmaler M., Colone A., Spagnuolo J., Zimmermann M., Lepore M., Kalinichenko A., Bhatia S., Cottier F., Rutishauser T., Pavelka N., Egli A., Azzali E., Pieroni M., Costantino G., Hruz P., Sauer U., Mori L., De Libero G., Modulation of bacterial metabolism by the microenvironment controls MAIT cell stimulation. Mucosal Immunol. 11, 1060–1070 (2018). [DOI] [PubMed] [Google Scholar]
- 29.Leeansyah E., Svard J., Dias J., Buggert M., Nystrom J., Quigley M. F., Moll M., Sonnerborg A., Nowak P., Sandberg J. K., Arming of MAIT cell cytolytic antimicrobial activity is induced by IL-7 and defective in HIV-1 infection. PLOS Pathog. 11, e1005072 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Boulouis C., Sia W. R., Gulam M. Y., Teo J. Q. M., Png Y. T., Phan T. K., Mak J. Y. W., Fairlie D. P., Poon I. K. H., Koh T. H., Bergman P., Lim C. M., Wang L. F., Kwa A. L. H., Sandberg J. K., Leeansyah E., Human MAIT cell cytolytic effector proteins synergize to overcome carbapenem resistance in Escherichia coli. PLOS Biol. 18, e3000644 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kurioka A., Ussher J. E., Cosgrove C., Clough C., Fergusson J. R., Smith K., Kang Y. H., Walker L. J., Hansen T. H., Willberg C. B., Klenerman P., MAIT cells are licensed through granzyme exchange to kill bacterially sensitized targets. Mucosal Immunol. 8, 429–440 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Le Bourhis L., Dusseaux M., Bohineust A., Bessoles S., Martin E., Premel V., Coré M., Sleurs D., Serriari N. E., Treiner E., Hivroz C., Sansonetti P., Gougeon M. L., Soudais C., Lantz O., MAIT cells detect and efficiently lyse bacterially-infected epithelial cells. PLOS Pathog. 9, e1003681 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.van Wilgenburg B., Scherwitzl I., Hutchinson E. C., Leng T., Kurioka A., Kulicke C., de Lara C., Cole S., Vasanawathana S., Limpitikul W., Malasit P., Young D., Denney L., Moore M. D., Fabris P., Giordani M. T., Oo Y. H., Laidlaw S. M., Dustin L. B., Ho L. P., Thompson F. M., Ramamurthy N., Mongkolsapaya J., Willberg C. B., Screaton G. R., Klenerman P., MAIT cells are activated during human viral infections. Nat. Commun. 7, 11653 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Hinks T. S. C., Marchi E., Jabeen M., Olshansky M., Kurioka A., Pediongco T. J., Meehan B. S., Kostenko L., Turner S. J., Corbett A. J., Chen Z., Klenerman P., McCluskey J., Activation and in vivo evolution of the MAIT cell transcriptome in mice and humans reveals tissue repair functionality. Cell. Rep. 28, 3249–3262.e5 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Leeansyah E., Ganesh A., Quigley M. F., Sonnerborg A., Andersson J., Hunt P. W., Somsouk M., Deeks S. G., Martin J. N., Moll M., Shacklett B. L., Sandberg J. K., Activation, exhaustion, and persistent decline of the antimicrobial MR1-restricted MAIT-cell population in chronic HIV-1 infection. Blood 121, 1124–1135 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Lamichhane R., Schneider M., de la Harpe S. M., Harrop T. W. R., Hannaway R. F., Dearden P. K., Kirman J. R., Tyndall J. D. A., Vernall A. J., Ussher J. E., TCR- or cytokine-activated CD8+ mucosal-associated invariant T cells are rapid polyfunctional effectors that can coordinate immune responses. Cell Rep. 28, 3061–3076.e5 (2019). [DOI] [PubMed] [Google Scholar]
- 37.Germain L., Veloso P., Lantz O., Legoux F., MAIT cells: Conserved watchers on the wall. J. Exp. Med. 222, e20232298 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Leng T., Akther H. D., Hackstein C. P., Powell K., King T., Friedrich M., Christoforidou Z., McCuaig S., Neyazi M., Arancibia-Carcamo C. V., Hagel J., Powrie F., Oxford I. B. D. I., Peres R. S., Millar V., Ebner D., Lamichhane R., Ussher J., Hinks T. S. C., Marchi E., Willberg C., Klenerman P., TCR and inflammatory signals tune human MAIT cells to exert specific tissue repair and effector functions. Cell Rep. 28, 3077–3091.e5 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Riffelmacher T., Paynich Murray M., Wientjens C., Chandra S., Cedillo-Castelán V., Chou T. F., McArdle S., Dillingham C., Devereaux J., Nilsen A., Brunel S., Lewinsohn D. M., Hasty J., Seumois G., Benedict C. A., Vijayanand P., Kronenberg M., Divergent metabolic programmes control two populations of MAIT cells that protect the lung. Nat. Cell. Biol. 25, 877–891 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Chandra S., Ascui G., Riffelmacher T., Chawla A., Ramírez-Suástegui C., Castelan V. C., Seumois G., Simon H., Murray M. P., Seo G. Y., Premlal A. L. R., Schmiedel B., Verstichel G., Li Y., Lin C. H., Greenbaum J., Lamberti J., Murthy R., Nigro J., Cheroutre H., Ottensmeier C. H., Hedrick S. M., Lu L. F., Vijayanand P., Kronenberg M., Transcriptomes and metabolism define mouse and human MAIT cell populations. Sci. Immunol. 8, eabn8531 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kedia-Mehta N., Pisarska M. M., Rollings C., O’Neill C., De Barra C., Foley C., Wood N. A. W., Wrigley-Kelly N., Veerapen N., Besra G., Bergin R., Jones N., O’Shea D., Sinclair L. V., Hogan A. E., The proliferation of human mucosal-associated invariant T cells requires a MYC-SLC7A5-glycolysis metabolic axis. Sci. Signal. 16, eabo2709 (2023). [DOI] [PubMed] [Google Scholar]
- 42.Ju J. K., Cho Y. N., Park K. J., Kwak H. D., Jin H. M., Park S. Y., Kim H. S., Kee S. J., Park Y. W., Activation, deficiency, and reduced IFN-γ production of mucosal-associated invariant T cells in patients with inflammatory bowel disease. J. Innate. Immun. 12, 422–434 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Serriari N. E., Eoche M., Lamotte L., Lion J., Fumery M., Marcelo P., Chatelain D., Barre A., Nguyen-Khac E., Lantz O., Dupas J. L., Treiner E., Innate mucosal-associated invariant T (MAIT) cells are activated in inflammatory bowel diseases. Clin. Exp. Immunol. 176, 266–274 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Haga K., Chiba A., Shibuya T., Osada T., Ishikawa D., Kodani T., Nomura O., Watanabe S., Miyake S., MAIT cells are activated and accumulated in the inflamed mucosa of ulcerative colitis. J. Gastroenterol. Hepatol. 31, 965–972 (2016). [DOI] [PubMed] [Google Scholar]
- 45.Bhuiyan T. R., Rahman M. A., Trivedi S., Afroz T., Al Banna H., Hoq M. R., Pop I., Jensen O., Rashu R., Uddin M. I., Hossain M., Khan A. I., Chowdhury F., Harris J. B., Calderwood S. B., Ryan E. T., Qadri F., Leung D. T., Mucosal-associated invariant T (MAIT) cells are highly activated in duodenal tissue of humans with Vibrio cholerae O1 infection: A preliminary report. PLOS Negl. Trop. Dis. 16, e0010411 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Tian J., Yan C., Jiang Y., Zhou H., Li L., Shen J., Wang J., Sun H., Yang G., Sun W., Peripheral and intestinal mucosal-associated invariant T cells in premature infants with necrotizing enterocolitis. Front. Pharmacol. 13, 1008080 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Tominaga K., Yamagiwa S., Setsu T., Kimura N., Honda H., Kamimura H., Honda Y., Takamura M., Yokoyama J., Suzuki K., Wakai T., Terai S., Possible involvement of mucosal-associated invariant T cells in the progression of inflammatory bowel diseases. Biomed. Res. 38, 111–121 (2017). [DOI] [PubMed] [Google Scholar]
- 48.Gibbs A., Leeansyah E., Introini A., Paquin-Proulx D., Hasselrot K., Andersson E., Broliden K., Sandberg J. K., Tjernlund A., MAIT cells reside in the female genital mucosa and are biased towards IL-17 and IL-22 production in response to bacterial stimulation. Mucosal Immunol. 10, 35–45 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Sobkowiak M. J., Davanian H., Heymann R., Gibbs A., Emgård J., Dias J., Aleman S., Krüger-Weiner C., Moll M., Tjernlund A., Leeansyah E., Sällberg Chen M., Sandberg J. K., Tissue-resident MAIT cell populations in human oral mucosa exhibit an activated profile and produce IL-17. Eur. J. Immunol. 49, 133–143 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Constantinides M. G., Link V. M., Tamoutounour S., Wong A. C., Perez-Chaparro P. J., Han S. J., Chen Y. E., Li K., Farhat S., Weckel A., Krishnamurthy S. R., Vujkovic-Cvijin I., Linehan J. L., Bouladoux N., Merrill E. D., Roy S., Cua D. J., Adams E. J., Bhandoola A., Scharschmidt T. C., Aubé J., Fischbach M. A., Belkaid Y., MAIT cells are imprinted by the microbiota in early life and promote tissue repair. Science 366, eaax6624 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.du Halgouet A., Darbois A., Alkobtawi M., Mestdagh M., Alphonse A., Premel V., Yvorra T., Colombeau L., Rodriguez R., Zaiss D., El Morr Y., Bugaut H., Legoux F., Perrin L., Aractingi S., Golub R., Lantz O., Salou M., Role of MR1-driven signals and amphiregulin on the recruitment and repair function of MAIT cells during skin wound healing. Immunity 56, 78–92.e6 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.El Morr Y., Fürstenheim M., Mestdagh M., Franciszkiewicz K., Salou M., Morvan C., Dupré T., Vorobev A., Jneid B., Premel V., Darbois A., Perrin L., Mondot S., Colombeau L., Bugaut H., du Halgouet A., Richon S., Procopio E., Maurin M., Philippe C., Rodriguez R., Lantz O., Legoux F., MAIT cells monitor intestinal dysbiosis and contribute to host protection during colitis. Sci. Immunol. 9, eadi8954 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Seach N., Guerri L., Le Bourhis L., Mburu Y., Cui Y., Bessoles S., Soudais C., Lantz O., Double-positive thymocytes select mucosal-associated invariant T cells. J. Immunol. 191, 6002–6009 (2013). [DOI] [PubMed] [Google Scholar]
- 54.Martin E., Treiner E., Duban L., Guerri L., Laude H., Toly C., Premel V., Devys A., Moura I. C., Tilloy F., Cherif S., Vera G., Latour S., Soudais C., Lantz O., Stepwise development of MAIT cells in mouse and human. PLOS Biol. 7, e54 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Koay H. F., Gherardin N. A., Enders A., Loh L., Mackay L. K., Almeida C. F., Russ B. E., Nold-Petry C. A., Nold M. F., Bedoui S., Chen Z., Corbett A. J., Eckle S. B., Meehan B., d’Udekem Y., Konstantinov I. E., Lappas M., Liu L., Goodnow C. C., Fairlie D. P., Rossjohn J., Chong M. M., Kedzierska K., Berzins S. P., Belz G. T., McCluskey J., Uldrich A. P., Godfrey D. I., Pellicci D. G., A three-stage intrathymic development pathway for the mucosal-associated invariant T cell lineage. Nat. Immunol. 17, 1300–1311 (2016). [DOI] [PubMed] [Google Scholar]
- 56.Pellicci D. G., Koay H. F., Berzins S. P., Thymic development of unconventional T cells: How NKT cells, MAIT cells and γδ T cells emerge. Nat. Rev. Immunol. 20, 756–770 (2020). [DOI] [PubMed] [Google Scholar]
- 57.Legoux F., Bellet D., Daviaud C., El Morr Y., Darbois A., Niort K., Procopio E., Salou M., Gilet J., Ryffel B., Balvay A., Foussier A., Sarkis M., El Marjou A., Schmidt F., Rabot S., Lantz O., Microbial metabolites control the thymic development of mucosal-associated invariant T cells. Science 366, 494–499 (2019). [DOI] [PubMed] [Google Scholar]
- 58.Jabeen M. F., Hinks T. S. C., MAIT cells and the microbiome. Front. Immunol. 14, 1127588 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Leeansyah E., Loh L., Nixon D. F., Sandberg J. K., Acquisition of innate-like microbial reactivity in mucosal tissues during human fetal MAIT-cell development. Nat. Commun. 5, 3143 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Bugaut H., El Morr Y., Mestdagh M., Darbois A., Paiva R. A., Salou M., Perrin L., Fürstenheim M., du Halgouet A., Bilonda-Mutala L., Le Gac A.-L., Arnaud M., El Marjou A., Guerin C., Chaiyasitdhi A., Piquet J., Smadja D. M., Cieslak A., Ryffel B., Maciulyte V., Turner J. M. A., Bernardeau K., Montagutelli X., Lantz O., Legoux F., A conserved transcriptional program for MAIT cells across mammalian evolution. J. Exp. Med. 221, e20231487 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Gherardin N. A., Souter M. N., Koay H. F., Mangas K. M., Seemann T., Stinear T. P., Eckle S. B., Berzins S. P., d’Udekem Y., Konstantinov I. E., Fairlie D. P., Ritchie D. S., Neeson P. J., Pellicci D. G., Uldrich A. P., McCluskey J., Godfrey D. I., Human blood MAIT cell subsets defined using MR1 tetramers. Immunol. Cell. Biol. 96, 507–525 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ben Youssef G., Tourret M., Salou M., Ghazarian L., Houdouin V., Mondot S., Mburu Y., Lambert M., Azarnoush S., Diana J.-S., Virlouvet A.-L., Peuchmaur M., Schmitz T., Dalle J.-H., Lantz O., Biran V., Caillat-Zucman S., Ontogeny of human mucosal-associated invariant T cells and related T cell subsets. J. Exp. Med. 215, 459–479 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Lee O. J., Cho Y. N., Kee S. J., Kim M. J., Jin H. M., Lee S. J., Park K. J., Kim T. J., Lee S. S., Kwon Y. S., Kim N., Shin M. G., Shin J. H., Suh S. P., Ryang D. W., Park Y. W., Circulating mucosal-associated invariant T cell levels and their cytokine levels in healthy adults. Exp. Gerontol. 49, 47–54 (2014). [DOI] [PubMed] [Google Scholar]
- 64.Chen P., Deng W., Li D., Zeng T., Huang L., Wang Q., Wang J., Zhang W., Yu X., Duan D., Wang J., Xia H., Chen H., Huang W., Li J., Zhang D., Zhong X. P., Gao J., Circulating mucosal-associated invariant T cells in a large cohort of healthy chinese individuals from newborn to elderly. Front. Immunol. 10, 260 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Loh L., Gherardin N. A., Sant S., Grzelak L., Crawford J. C., Bird N. L., Koay H. F., van de Sandt C. E., Moreira M. L., Lappas M., Allen E. K., Crowe J., Loudovaris T., Flanagan K. L., Quinn K. M., Rossjohn J., Thomas P. G., Eckle S. B. G., McCluskey J., Godfrey D. I., Kedzierska K., Human mucosal-associated invariant T cells in older individuals display expanded TCRαβ clonotypes with potent antimicrobial responses. J. Immunol. 204, 1119–1133 (2020). [DOI] [PubMed] [Google Scholar]
- 66.Han F., Gulam M. Y., Zheng Y., Zulhaimi N. S., Sia W. R., He D., Ho A., Hadadi L., Liu Z., Qin P., Lobie P. E., Kamarulzaman A., Wang L. F., Sandberg J. K., Lewin S. R., Rajasuriar R., Leeansyah E., IL7RA single nucleotide polymorphisms are associated with the size and function of the MAIT cell population in treated HIV-1 infection. Front. Immunol. 13, 985385 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Dias J., Boulouis C., Gorin J. B., van den Biggelaar R., Lal K. G., Gibbs A., Loh L., Gulam M. Y., Sia W. R., Bari S., Hwang W. Y. K., Nixon D. F., Nguyen S., Betts M. R., Buggert M., Eller M. A., Broliden K., Tjernlund A., Sandberg J. K., Leeansyah E., The CD4-CD8- MAIT cell subpopulation is a functionally distinct subset developmentally related to the main CD8+ MAIT cell pool. Proc. Natl. Acad. Sci. U.S.A. 115, E11513–E11522 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Novak J., Dobrovolny J., Novakova L., Kozak T., The decrease in number and change in phenotype of mucosal-associated invariant T cells in the elderly and differences in men and women of reproductive age. Scand. J. Immunol. 80, 271–275 (2014). [DOI] [PubMed] [Google Scholar]
- 69.Souter M. N. T., Awad W., Li S., Pediongco T. J., Meehan B. S., Meehan L. J., Tian Z., Zhao Z., Wang H., Nelson A., Le Nours J., Khandokar Y., Praveena T., Wubben J., Lin J., Sullivan L. C., Lovrecz G. O., Mak J. Y. W., Liu L., Kostenko L., Kedzierska K., Corbett A. J., Fairlie D. P., Brooks A. G., Gherardin N. A., Uldrich A. P., Chen Z., Rossjohn J., Godfrey D. I., McCluskey J., Pellicci D. G., Eckle S. B. G., CD8 coreceptor engagement of MR1 enhances antigen responsiveness by human MAIT and other MR1-reactive T cells. J. Exp. Med. 219, e20210828 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Thursby E., Juge N., Introduction to the human gut microbiota. Biochem. J. 474, 1823–1836 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Rodionov D. A., Arzamasov A. A., Khoroshkin M. S., Iablokov S. N., Leyn S. A., Peterson S. N., Novichkov P. S., Osterman A. L., Micronutrient requirements and sharing capabilities of the human gut microbiome. Front. Microbiol. 10, 1316 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Rodríguez J. M., Murphy K., Stanton C., Ross R. P., Kober O. I., Juge N., Avershina E., Rudi K., Narbad A., Jenmalm M. C., Marchesi J. R., Collado M. C., The composition of the gut microbiota throughout life, with an emphasis on early life. Microb. Ecol. Health Dis. 26, 26050 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Claesson M. J., Cusack S., O’Sullivan O., Greene-Diniz R., de Weerd H., Flannery E., Marchesi J. R., Falush D., Dinan T., Fitzgerald G., Stanton C., van Sinderen D., O’Connor M., Harnedy N., O’Connor K., Henry C., O’Mahony D., Fitzgerald A. P., Shanahan F., Twomey C., Hill C., Ross R. P., O’Toole P. W., Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc. Natl. Acad. Sci. U.S.A. 108, 4586–4591 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Salazar J., Durán P., Díaz M. P., Chacín M., Santeliz R., Mengual E., Gutiérrez E., León X., Díaz A., Bernal M., Escalona D., Hernández L. A. P., Bermúdez V., Exploring the relationship between the gut microbiota and ageing: A possible age modulator. Int. J. Environ. Res. Public Health 20, 5845 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ito E., Inuki S., Izumi Y., Takahashi M., Dambayashi Y., Ciacchi L., Awad W., Takeyama A., Shibata K., Mori S., Mak J. Y. W., Fairlie D. P., Bamba T., Ishikawa E., Nagae M., Rossjohn J., Yamasaki S., Sulfated bile acid is a host-derived ligand for MAIT cells. Sci. Immunol. 9, eade6924 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Moles L., Gómez M., Heilig H., Bustos G., Fuentes S., de Vos W., Fernández L., Rodríguez J. M., Jiménez E., Bacterial diversity in meconium of preterm neonates and evolution of their fecal microbiota during the first month of life. PLOS ONE 8, e66986 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Jiménez E., Marín M. L., Martín R., Odriozola J. M., Olivares M., Xaus J., Fernández L., Rodríguez J. M., Is meconium from healthy newborns actually sterile? Res. Microbiol. 159, 187–193 (2008). [DOI] [PubMed] [Google Scholar]
- 78.Walker R. W., Clemente J. C., Peter I., Loos R. J. F., The prenatal gut microbiome: Are we colonized with bacteria in utero? Pediatr. Obes. 12, 3–17 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Bokulich N. A., Chung J., Battaglia T., Henderson N., Jay M., Li H., Lieber A. D., Wu F., Perez-Perez G. I., Chen Y., Schweizer W., Zheng X., Contreras M., Dominguez-Bello M. G., Blaser M. J., Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci. Transl. Med. 8, 343ra382 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Yatsunenko T., Rey F. E., Manary M. J., Trehan I., Dominguez-Bello M. G., Contreras M., Magris M., Hidalgo G., Baldassano R. N., Anokhin A. P., Heath A. C., Warner B., Reeder J., Kuczynski J., Caporaso J. G., Lozupone C. A., Lauber C., Clemente J. C., Knights D., Knight R., Gordon J. I., Human gut microbiome viewed across age and geography. Nature 486, 222–227 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Eckburg P. B., Bik E. M., Bernstein C. N., Purdom E., Dethlefsen L., Sargent M., Gill S. R., Nelson K. E., Relman D. A., Diversity of the human intestinal microbial flora. Science 308, 1635–1638 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Arumugam M., Raes J., Pelletier E., Le Paslier D., Yamada T., Mende D. R., Fernandes G. R., Tap J., Bruls T., Batto J. M., Bertalan M., Borruel N., Casellas F., Fernandez L., Gautier L., Hansen T., Hattori M., Hayashi T., Kleerebezem M., Kurokawa K., Leclerc M., Levenez F., Manichanh C., Nielsen H. B., Nielsen T., Pons N., Poulain J., Qin J., Sicheritz-Ponten T., Tims S., Torrents D., Ugarte E., Zoetendal E. G., Wang J., Guarner F., Pedersen O., de Vos W. M., Brunak S., Doré J., Antolín M., Artiguenave F., Blottiere H. M., Almeida M., Brechot C., Cara C., Chervaux C., Cultrone A., Delorme C., Denariaz G., Dervyn R., Foerstner K. U., Friss C., van de Guchte M., Guedon E., Haimet F., Huber W., van Hylckama-Vlieg J., Jamet A., Juste C., Kaci G., Knol J., Lakhdari O., Layec S., Le Roux K., Maguin E., Mérieux A., Melo Minardi R., M’Rini C., Muller J., Oozeer R., Parkhill J., Renault P., Rescigno M., Sanchez N., Sunagawa S., Torrejon A., Turner K., Vandemeulebrouck G., Varela E., Winogradsky Y., Zeller G., Weissenbach J., Ehrlich S. D., Bork P., Enterotypes of the human gut microbiome. Nature 473, 174–180 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Li G., Yang M., Zhou K., Zhang L., Tian L., Lv S., Jin Y., Qian W., Xiong H., Lin R., Fu Y., Hou X., Diversity of duodenal and rectal microbiota in biopsy tissues and luminal contents in healthy volunteers. J. Microbiol. Biotechnol. 25, 1136–1145 (2015). [DOI] [PubMed] [Google Scholar]
- 84.Swidsinski A., Weber J., Loening-Baucke V., Hale L. P., Lochs H., Spatial organization and composition of the mucosal flora in patients with inflammatory bowel disease. J. Clin. Microbiol. 43, 3380–3389 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Van den Abbeele P., Belzer C., Goossens M., Kleerebezem M., De Vos W. M., Thas O., De Weirdt R., Kerckhof F. M., Van de Wiele T., Butyrate-producing Clostridium cluster XIVa species specifically colonize mucins in an in vitro gut model. ISME J 7, 949–961 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Proudfoot A. E., Chemokine receptors: Multifaceted therapeutic targets. Nat. Rev. Immunol. 2, 106–115 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Griffith J. W., Sokol C. L., Luster A. D., Chemokines and chemokine receptors: Positioning cells for host defense and immunity. Annu. Rev. Immunol. 32, 659–702 (2014). [DOI] [PubMed] [Google Scholar]
- 88.Mackay C. R., Chemokines: Immunology’s high impact factors. Nat. Immunol. 2, 95–101 (2001). [DOI] [PubMed] [Google Scholar]
- 89.Ziarek J. J., Kleist A. B., London N., Raveh B., Montpas N., Bonneterre J., St-Onge G., DiCosmo-Ponticello C. J., Koplinski C. A., Roy I., Stephens B., Thelen S., Veldkamp C. T., Coffman F. D., Cohen M. C., Dwinell M. B., Thelen M., Peterson F. C., Heveker N., Volkman B. F., Structural basis for chemokine recognition by a G protein-coupled receptor and implications for receptor activation. Sci. Signal. 10, eaah5756 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Lee C. H., Zhang H. H., Singh S. P., Koo L., Kabat J., Tsang H., Singh T. P., Farber J. M., C/EBPδ drives interactions between human MAIT cells and endothelial cells that are important for extravasation. eLife 7, 32532 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Kurioka A., Jahun A. S., Hannaway R. F., Walker L. J., Fergusson J. R., Sverremark-Ekström E., Corbett A. J., Ussher J. E., Willberg C. B., Klenerman P., Shared and distinct phenotypes and functions of human CD161++ Vα7.2+ T cell subsets. Front. Immunol. 8, 1031 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Dusseaux M., Martin E., Serriari N., Péguillet I., Premel V., Louis D., Milder M., Le Bourhis L., Soudais C., Treiner E., Lantz O., Human MAIT cells are xenobiotic-resistant, tissue-targeted, CD161hi IL-17-secreting T cells. Blood 117, 1250–1259 (2011). [DOI] [PubMed] [Google Scholar]
- 93.Ocón B., Xiang M., Bi Y., Tan S., Brulois K., Ayesha A., Kunte M., Zhou C., LaJevic M., Lazarus N., Mengoni F., Sharma T., Montgomery S., Hooper J. E., Huang M., Handel T., Dawson J. R. D., Kufareva I., Zabel B. A., Pan J., Butcher E. C., A lymphocyte chemoaffinity axis for lung, non-intestinal mucosae and CNS. Nature 635, 736–745 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kim S. V., Xiang W. V., Kwak C., Yang Y., Lin X. W., Ota M., Sarpel U., Rifkin D. B., Xu R., Littman D. R., GPR15-mediated homing controls immune homeostasis in the large intestine mucosa. Science 340, 1456–1459 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Nguyen L. P., Pan J., Dinh T. T., Hadeiba H., O’Hara E. III, Ebtikar A., Hertweck A., Gökmen M. R., Lord G. M., Jenner R. G., Butcher E. C., Habtezion A., Role and species-specific expression of colon T cell homing receptor GPR15 in colitis. Nat. Immunol. 16, 207–213 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Solders M., Gorchs L., Tiblad E., Gidlöf S., Leeansyah E., Dias J., Sandberg J. K., Magalhaes I., Lundell A. C., Kaipe H., Recruitment of MAIT cells to the intervillous space of the placenta by placenta-derived chemokines. Front. Immunol. 10, 1300 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Voillet V., Buggert M., Slichter C. K., Berkson J. D., Mair F., Addison M. M., Dori Y., Nadolski G., Itkin M. G., Gottardo R., Betts M. R., Prlic M., Human MAIT cells exit peripheral tissues and recirculate via lymph in steady state conditions. JCI Insight 3, e98487 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Mortier A., Gouwy M., Van Damme J., Proost P., Struyf S., CD26/dipeptidylpeptidase IV-chemokine interactions: Double-edged regulation of inflammation and tumor biology. J. Leukoc. Biol. 99, 955–969 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Iwata S., Yamaguchi N., Munakata Y., Ikushima H., Lee J. F., Hosono O., Schlossman S. F., Morimoto C., CD26/dipeptidyl peptidase IV differentially regulates the chemotaxis of T cells and monocytes toward RANTES: Possible mechanism for the switch from innate to acquired immune response. Int. Immunol. 11, 417–426 (1999). [DOI] [PubMed] [Google Scholar]
- 100.Van Damme J., Rampart M., Conings R., Decock B., Van Osselaer N., Willems J., Billiau A., The neutrophil-activating proteins interleukin 8 and β-thromboglobulin: In vitro and in vivo comparison of NH2-terminally processed forms. Eur. J. Immunol. 20, 2113–2118 (1990). [DOI] [PubMed] [Google Scholar]
- 101.Hopsu-Havu V. K., Glenner G. G., A new dipeptide naphthylamidase hydrolyzing glycyl-prolyl-β-naphthylamide. Histochemie 7, 197–201 (1966). [DOI] [PubMed] [Google Scholar]
- 102.Proost P., Struyf S., Couvreur M., Lenaerts J. P., Conings R., Menten P., Verhaert P., Wuyts A., Van Damme J., Posttranslational modifications affect the activity of the human monocyte chemotactic proteins MCP-1 and MCP-2: Identification of MCP-2(6-76) as a natural chemokine inhibitor. J. Immunol. 160, 4034–4041 (1998). [PubMed] [Google Scholar]
- 103.Christopherson K. W. II, Hangoc G., Broxmeyer H. E., Cell surface peptidase CD26/dipeptidylpeptidase IV regulates CXCL12/stromal cell-derived factor-1 α-mediated chemotaxis of human cord blood CD34+ progenitor cells. J. Immunol. 169, 7000–7008 (2002). [DOI] [PubMed] [Google Scholar]
- 104.Christopherson K. W. II, Uralil S. E., Porecha N. K., Zabriskie R. C., Kidd S. M., Ramin S. M., G-CSF- and GM-CSF-induced upregulation of CD26 peptidase downregulates the functional chemotactic response of CD34+CD38- human cord blood hematopoietic cells. Exp. Hematol. 34, 1060–1068 (2006). [DOI] [PubMed] [Google Scholar]
- 105.Khurana S., Margamuljana L., Joseph C., Schouteden S., Buckley S. M., Verfaillie C. M., Glypican-3-mediated inhibition of CD26 by TFPI: A novel mechanism in hematopoietic stem cell homing and maintenance. Blood 121, 2587–2595 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Suliman S., Murphy M., Musvosvi M., Gela A., Meermeier E. W., Geldenhuys H., Hopley C., Toefy A., Bilek N., Veldsman A., Hanekom W. A., Johnson J. L., Boom W. H., Obermoser G., Huang H., Hatherill M., Lewinsohn D. M., Nemes E., Scriba T. J., MR1-independent activation of human mucosal-associated invariant T cells by mycobacteria. J. Immunol. 203, 2917–2927 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Bulitta B., Zuschratter W., Bernal I., Bruder D., Klawonn F., von Bergen M., Garritsen H. S. P., Jänsch L., Proteomic definition of human mucosal-associated invariant T cells determines their unique molecular effector phenotype. Eur. J. Immunol. 48, 1336–1349 (2018). [DOI] [PubMed] [Google Scholar]
- 108.Howson L. J., Napolitani G., Shepherd D., Ghadbane H., Kurupati P., Preciado-Llanes L., Rei M., Dobinson H. C., Gibani M. M., Teng K. W. W., Newell E. W., Veerapen N., Besra G. S., Pollard A. J., Cerundolo V., MAIT cell clonal expansion and TCR repertoire shaping in human volunteers challenged with Salmonella Paratyphi A. Nat. Commun. 9, 253 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Neurath M. F., Strategies for targeting cytokines in inflammatory bowel disease. Nat. Rev. Immunol. 24, 559–576 (2024). [DOI] [PubMed] [Google Scholar]
- 110.Zundler S., Günther C., Kremer A. E., Zaiss M. M., Rothhammer V., Neurath M. F., Gut immune cell trafficking: Inter-organ communication and immune-mediated inflammation. Nat. Rev. Gastroenterol. Hepatol. 20, 50–64 (2023). [DOI] [PubMed] [Google Scholar]
- 111.Hiejima E., Kawai T., Nakase H., Tsuruyama T., Morimoto T., Yasumi T., Taga T., Kanegane H., Hori M., Ohmori K., Higuchi T., Matsuura M., Yoshino T., Ikeuchi H., Kawada K., Sakai Y., Kitazume M. T., Hisamatsu T., Chiba T., Nishikomori R., Heike T., Reduced numbers and proapoptotic features of mucosal-associated invariant T cells as a characteristic finding in patients with inflammatory bowel disease. Inflamm. Bowel Dis. 21, 1529–1540 (2015). [DOI] [PubMed] [Google Scholar]
- 112.L. Loh, D. Orlicky, A. Spengler, C. Levens, S. Celli, J. Domenico, J. Klarquist, J. Onyiah, J. Matsuda, K. Kuhn, L. Gapin, MAIT cells drive chronic inflammation in a genetically diverse murine model of spontaneous colitis. bioRxiv 569225 [Preprint] (2023). 10.1101/2023.11.29.569225. [DOI]
- 113.Yasutomi Y., Chiba A., Haga K., Murayama G., Makiyama A., Kuga T., Watanabe M., Okamoto R., Nagahara A., Nagaishi T., Miyake S., Activated mucosal-associated invariant T cells have a pathogenic role in a murine model of inflammatory bowel disease. Cell Mol. Gastroenterol. Hepatol. 13, 81–93 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Ling L., Lin Y., Zheng W., Hong S., Tang X., Zhao P., Li M., Ni J., Li C., Wang L., Jiang Y., Circulating and tumor-infiltrating mucosal associated invariant T (MAIT) cells in colorectal cancer patients. Sci. Rep. 6, 20358 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Won E. J., Ju J. K., Cho Y. N., Jin H. M., Park K. J., Kim T. J., Kwon Y. S., Kee H. J., Kim J. C., Kee S. J., Park Y. W., Clinical relevance of circulating mucosal-associated invariant T cell levels and their anti-cancer activity in patients with mucosal-associated cancer. Oncotarget 7, 76274–76290 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Shao C., Zhu C., Zhu Y., Hao J., Li Y., Hu H., Si L., Zhong F., Wang X., Wang H., Decrease of peripheral blood mucosal-associated invariant T cells and impaired serum Granzyme-B production in patients with gastric cancer. Cell Biosci. 11, 12 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Han F., Zheng Y., Ho A., Ma S., Sandberg J. K., Leeansyah E., MAIT cell loss and reconstitution in HIV-1 disease. Crit. Rev. Immunol. 41, 69–82 (2021). [DOI] [PubMed] [Google Scholar]
- 118.Cosgrove C., Ussher J. E., Rauch A., Gärtner K., Kurioka A., Hühn M. H., Adelmann K., Kang Y. H., Fergusson J. R., Simmonds P., Goulder P., Hansen T. H., Fox J., Günthard H. F., Khanna N., Powrie F., Steel A., Gazzard B., Phillips R. E., Frater J., Uhlig H., Klenerman P., Early and nonreversible decrease of CD161++ /MAIT cells in HIV infection. Blood 121, 951–961 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Xia P., Xing X.-D., Yang C.-X., Liao X.-J., Liu F.-H., Huang H.-H., Zhang C., Song J.-W., Jiao Y.-M., Shi M., Jiang T.-J., Zhou C.-B., Wang X.-C., He Q., Zeng Q.-L., Wang F.-S., Zhang J.-Y., Activation-induced pyroptosis contributes to the loss of MAIT cells in chronic HIV-1 infected patients. Military Med. Res. 9, 24 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Pang X., He X., Qiu Z., Zhang H., Xie R., Liu Z., Gu Y., Zhao N., Xiang Q., Cui Y., Targeting integrin pathways: Mechanisms and advances in therapy. Sig. Transduct. Target Ther. 8, 1 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Mora J. R., Bono M. R., Manjunath N., Weninger W., Cavanagh L. L., Rosemblatt M., Von Andrian U. H., Selective imprinting of gut-homing T cells by Peyer’s patch dendritic cells. Nature 424, 88–93 (2003). [DOI] [PubMed] [Google Scholar]
- 122.Zundler S., Becker E., Schulze L. L., Neurath M. F., Immune cell trafficking and retention in inflammatory bowel disease: Mechanistic insights and therapeutic advances. Gut 68, 1688–1700 (2019). [DOI] [PubMed] [Google Scholar]
- 123.Walling B. L., Kim M., LFA-1 in T cell migration and differentiation. Front. Immunol. 9, 952 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Salou M., Legoux F., Gilet J., Darbois A., du Halgouet A., Alonso R., Richer W., Goubet A. G., Daviaud C., Menger L., Procopio E., Premel V., Lantz O., A common transcriptomic program acquired in the thymus defines tissue residency of MAIT and NKT subsets. J. Exp. Med. 216, 133–151 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Sharma A., Lawry S. M., Klein B. S., Wang X., Sherer N. M., Zumwalde N. A., Gumperz J. E., LFA-1 ligation by high-density ICAM-1 is sufficient to activate IFN-γ release by innate T lymphocytes. J. Immunol. 201, 2452–2461 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Sakala I. G., Kjer-Nielsen L., Eickhoff C. S., Wang X., Blazevic A., Liu L., Fairlie D. P., Rossjohn J., McCluskey J., Fremont D. H., Hansen T. H., Hoft D. F., Functional heterogeneity and antimycobacterial effects of mouse mucosal-associated invariant T cells specific for riboflavin metabolites. J. Immunol. 195, 587–601 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Winter S. E., Bäumler A. J., Gut dysbiosis: Ecological causes and causative effects on human disease. Proc. Natl. Acad. Sci. U.S.A. 120, e2316579120 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Tastan C., Karhan E., Zhou W., Fleming E., Voigt A. Y., Yao X., Wang L., Horne M., Placek L., Kozhaya L., Oh J., Unutmaz D., Tuning of human MAIT cell activation by commensal bacteria species and MR1-dependent T-cell presentation. Mucosal Immunol. 11, 1591–1605 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Ansaldo E., Farley T. K., Belkaid Y., Control of immunity by the microbiota. Annu. Rev. Immunol. 39, 449–479 (2021). [DOI] [PubMed] [Google Scholar]
- 130.Tanoue T., Morita S., Plichta D. R., Skelly A. N., Suda W., Sugiura Y., Narushima S., Vlamakis H., Motoo I., Sugita K., Shiota A., Takeshita K., Yasuma-Mitobe K., Riethmacher D., Kaisho T., Norman J. M., Mucida D., Suematsu M., Yaguchi T., Bucci V., Inoue T., Kawakami Y., Olle B., Roberts B., Hattori M., Xavier R. J., Atarashi K., Honda K., A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature 565, 600–605 (2019). [DOI] [PubMed] [Google Scholar]
- 131.Elinav E., Strowig T., Kau A. L., Henao-Mejia J., Thaiss C. A., Booth C. J., Peaper D. R., Bertin J., Eisenbarth S. C., Gordon J. I., Flavell R. A., NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell 145, 745–757 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Parrot T., Healy K., Boulouis C., Sobkowiak M. J., Leeansyah E., Aleman S., Bertoletti A., Sällberg Chen M., Sandberg J. K., Expansion of donor-unrestricted MAIT cells with enhanced cytolytic function suitable for TCR redirection. JCI Insight 6, e140074 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Fidelle M., Rauber C., Costa Silva C. A., Tian A.-L., Lahmar I., de La Varende A.-L. M., Zhao L., Thelemaque C., Lebhar I., Messaoudene M., Pizzato E., Birebent R., Mbogning Fonkou M. D., Zoppi S., Reni A., Dalban C., Leduc M., Ferrere G., Durand S., Ly P., Silvin A., Mulder K., Dutertre C.-A., Ginhoux F., Yonekura S., Roberti M. P., Tidjani-Alou M., Terrisse S., Chen J., Kepp O., Schippers A., Wagner N., Suárez-Gosálvez J., Kobold S., Fahrner J.-E., Richard C., Bosq J., Lordello L., Vitali G., Galleron N., Quinquis B., Le Chatelier E., Blanchard L., Girard J.-P., Jarry A., Gervois N., Godefroy E., Labarrière N., Koschny R., Daillère R., Besse B., Truntzer C., Ghiringhelli F., Coatnoan N., Mhanna V., Klatzmann D., Drubay D., Albiges L., Thomas A. M., Segata N., Danlos F.-X., Marabelle A., Routy B., Derosa L., Kroemer G., Zitvogel L., A microbiota-modulated checkpoint directs immunosuppressive intestinal T cells into cancers. Science 380, eabo2296 (2023). [DOI] [PubMed] [Google Scholar]
- 134.Nirmalkar K., Murugesan S., Pizano-Zárate M. L., Villalobos-Flores L. E., García-González C., Morales-Hernández R. M., Nuñez-Hernández J. A., Hernández-Quiroz F., Romero-Figueroa M. D. S., Hernández-Guerrero C., Hoyo-Vadillo C., García-Mena J., Gut microbiota and endothelial dysfunction markers in obese mexican children and adolescents. Nutrients 10, 2009 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Dias J., Sobkowiak M. J., Sandberg J. K., Leeansyah E., Human MAIT-cell responses to Escherichia coli: Activation, cytokine production, proliferation, and cytotoxicity. J. Leukoc. Biol. 100, 233–240 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Kamali Dolatabadi R., Feizi A., Halaji M., Fazeli H., Adibi P., The prevalence of adherent-invasive Escherichia coli and its association with inflammatory bowel diseases: A systematic review and meta-analysis. Front. Med. 8, 730243 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Viladomiu M., Metz M. L., Lima S. F., Jin W. B., Chou L., Guo C. J., Diehl G. E., Simpson K. W., Scherl E. J., Longman R. S., Adherent-invasive E. coli metabolism of propanediol in Crohn’s disease regulates phagocytes to drive intestinal inflammation. Cell Host Microbe 29, 607–619.e8 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Paroni M., Leccese G., Ranzani V., Moschetti G., Chiara M., Perillo F., Ferri S., Clemente F., Noviello D., Conforti F. S., Ferrero S., Karnani B., Bosotti R., Vasco C., Curti S., Crosti M. C., Gruarin P., Rossetti G., Conte M. P., Vecchi M., Pagani M., Landini P., Facciotti F., Abrignani S., Caprioli F., Geginat J., An intestinal Th17 subset is associated with inflammation in crohn’s disease and activated by adherent-invasive Escherichia coli. J. Crohns Colitis 17, 1988–2001 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Zhao Z., Chen X., Dowbaj A. M., Sljukic A., Bratlie K., Lin L., Fong E. L. S., Balachander G. M., Chen Z., Soragni A., Huch M., Zeng Y. A., Wang Q., Yu H., Organoids. Nat. Rev. Methods Primers 2, 94 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Xiao M. H., Wu S., Liang P., Ma D., Zhang J., Chen H., Zhong Z., Liu J., Jiang H., Feng X., Luo Z., Mucosal-associated invariant T cells promote ductular reaction through amphiregulin in biliary atresia. EBioMedicine 103, 105138 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Lebrusant-Fernandez M., Ap Rees T., Jimeno R., Angelis N., Ng J. C., Fraternali F., Li V. S. W., Barral P., IFN-γ-dependent regulation of intestinal epithelial homeostasis by NKT cells. Cell Rep. 43, 114948 (2024). [DOI] [PubMed] [Google Scholar]
- 142.Gurtner G. C., Werner S., Barrandon Y., Longaker M. T., Wound repair and regeneration. Nature 453, 314–321 (2008). [DOI] [PubMed] [Google Scholar]
- 143.Neurath M. F., Targeting immune cell circuits and trafficking in inflammatory bowel disease. Nat. Immunol. 20, 970–979 (2019). [DOI] [PubMed] [Google Scholar]
- 144.Miao M., De Clercq E., Li G., Clinical significance of chemokine receptor antagonists. Expert Opin. Drug Metab. Toxicol. 16, 11–30 (2020). [DOI] [PubMed] [Google Scholar]
- 145.Ollila T. A., Sahin I., Olszewski A. J., Mogamulizumab: A new tool for management of cutaneous T-cell lymphoma. Onco Targets Ther. 12, 1085–1094 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Tacke F., Cenicriviroc for the treatment of non-alcoholic steatohepatitis and liver fibrosis. Expert. Opin. Investig. Drugs 27, 301–311 (2018). [DOI] [PubMed] [Google Scholar]
- 147.Sandborn W. J., Feagan B. G., Rutgeerts P., Hanauer S., Colombel J. F., Sands B. E., Lukas M., Fedorak R. N., Lee S., Bressler B., Fox I., Rosario M., Sankoh S., Xu J., Stephens K., Milch C., Parikh A., Vedolizumab as induction and maintenance therapy for Crohn’s disease. N. Engl. J. Med. 369, 711–721 (2013). [DOI] [PubMed] [Google Scholar]
- 148.Feagan B. G., Rutgeerts P., Sands B. E., Hanauer S., Colombel J. F., Sandborn W. J., Van Assche G., Axler J., Kim H. J., Danese S., Fox I., Milch C., Sankoh S., Wyant T., Xu J., Parikh A., Vedolizumab as induction and maintenance therapy for ulcerative colitis. N. Engl. J. Med. 369, 699–710 (2013). [DOI] [PubMed] [Google Scholar]
- 149.Dai B., Hackney J. A., Ichikawa R., Nguyen A., Elstrott J., Orozco L. D., Sun K. H., Modrusan Z., Gogineni A., Scherl A., Gubatan J., Habtezion A., Deswal M., Somsouk M., Faubion W. A., Chai A., Sharafali Z., Hassanali A., Oh Y. S., Tole S., McBride J., Keir M. E., Yi T., Dual targeting of lymphocyte homing and retention through α4β7 and αEβ7 inhibition in inflammatory bowel disease. Cell Rep. Med. 2, 100381 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Sandborn W. J., Vermeire S., Tyrrell H., Hassanali A., Lacey S., Tole S., Tatro A. R., Etrolizumab for the treatment of ulcerative colitis and crohn’s disease: An overview of the phase 3 clinical program. Adv. Ther. 37, 3417–3431 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Ghosh S., Goldin E., Gordon F. H., Malchow H. A., Rask-Madsen J., Rutgeerts P., Vyhnálek P., Zádorová Z., Palmer T., Donoghue S., Natalizumab for active Crohn’s disease. N. Engl. J. Med. 348, 24–32 (2003). [DOI] [PubMed] [Google Scholar]



