- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
Autoimmune hepatitis (AIH) is a progressive liver inflammatory disease mediated by an autoimmune response, with an increasing incidence rate. In severe cases, AIH will rapidly progress to liver cirrhosis and liver failure and even lead to death. The gut microbiota is a complex ecosystem that significantly regulates physiological and pathological processes among various digestive system diseases. It is widely acknowledged that there is a critical correlation between AIH and the gut microbiota. Numerous studies have demonstrated that the composition of gut microbiota in individuals with AIH differs markedly from that of healthy subjects. Immune cells, especially T cells, are pivotal in the development of AIH, closely interacting with the gut microbiota. In this review, we discuss the regulatory role of the gut microbiota in T cell-mediated development of AIH, as well as the effect of T cells on the composition of the gut microbiota in AIH. By modulating gut microbiota or immunity pathways, novel opportunities are provided to regulate the balance of the immune-microbial microenvironment, targeting the dual factor for autoimmune hepatitis therapies.
Introduction
In recent years, there has been an increase in the incidence of autoimmune diseases such as autoimmune hepatitis (AIH) and inflammatory bowel disease (IBD), type 1 diabetes (T1D), rheumatoid arthritis (RA), and multiple sclerosis (MS), attributed to changes in lifestyle and environmental factors (1, 2). In exploring the occurrence of autoimmune diseases, there is an amounting recognition of the metagenome of all colonized microorganisms in the body, known as the microbiome (3). Although both the microbial and host genomes are regulated by diet and environmental factors, the microbial genome exhibits a rapid adaptability to environmental shifts due to its shorter duplication cycles. Therefore, pieces of evidence support that gut microbiota plays an essential role in autoimmune diseases, which regulates the taxonomic composition of microbial communities and their metagenomic functions (4).
The co-evolution of the immune system and microbiota has been identified to participate in complex immune decision-making processes. The immune system communicates with the microbiota through several recognition patterns, among which T cells possess the most extensive microbial recognition capability, ranging from detecting the entire microbial community to recognizing specific antigens. These microorganisms are not only an important source of antigen variation but also provide necessary signals for the normal development, maturation, and epigenetic modulation of T cells, while mature T cells regulate microbial responses (5). Furthermore, gut microbiota dysbiosis can also regulate liver pathophysiology and induce AIH (6).
AIH is a serious and progressive autoimmune disease characterized by high levels of gamma globulin and hepatic inflammatory infiltration. If not timely diagnosed and treated, AIH increases the risk of liver cirrhosis and end-stage liver failure (7). Although the exact pathogenesis of AIH remains unclear, studies indicate that genetic factors, environmental factors, and aberrant immunological regulatory mechanisms are involved (8). The mechanisms by which the gut microbiota influences AIH involve multiple levels, including gut microecological imbalance, which leads to the production of autoantibodies and hepatocyte damage (9). The degree of dysbiosis correlates with the severity of liver inflammation, and changes in microbial metabolites affect the hepatic immune microenvironment through the gut-liver axis (10). Furthermore, individuals with AIH frequently exhibit impaired intestinal barrier integrity, facilitating the transfer of gut bacteria or their structural components and metabolites to the liver, which activates innate immune responses, thereby exacerbating liver inflammation and autoimmune reactions (11). Alterations in the gut microbiota regulate the composition and circulation of short-chain fatty acids and bile acids, directly impacting the liver’s metabolic and inflammatory states (12). However, the specific mechanisms by which the gut microbiota and their metabolites are involved in the development of AIH have not been fully elucidated. More clinical and basic studies are being dedicated to improving AIH therapy through the gut microbiota (13).
This review discusses the roles of gut microbiota and their metabolites in the regulation of T cells during the development of AIH, as well as how T cells, in turn, regulate the composition of the gut microbiota within the AIH microenvironment. By modulating the gut microbiota, it is possible to modify the characteristics and functions of immune T cells, providing novel potential targets for the treatment of AIH.
The gut microbiota: an indispensable player in AIH
The gut microbiota, when continuously challenged by external dietary and environmental antigens, maintain normal physiological functions and dynamic equilibrium within the body (14). In normal circumstances, these microorganisms play a critical role in maintaining the structure of the gut, enhancing barrier integrity, and regulating mucosal immune responses by preserving cellular junctions and promoting epithelial repair (15). Changes in gut permeability and bacterial translocation in AIH patients are associated with disease progression (16). A recent study indicates that the gut microbiota and their metabolites, as a reservoir of exogenous antigens, are crucial for maintaining liver immune homeostasis (17). For instance, Akkermansia muciniphila enhances intestinal barrier integrity by degrading mucins within the intestinal mucus layer, thereby stimulating mucin renewal (18). This barrier-protective mechanism effectively reduces the translocation of pathogenic organisms and bacterial metabolites. Consequently, it mitigates hepatic immune activation triggered by gut-derived inflammatory signals, while preserving hepatic immune tolerance (19). HBXN2020, isolated from healthy black pigs, is a microorganism with potent stress resistance and broad-spectrum antibacterial activity. HBXN2020 significantly modulates cytokine levels and maintains the expression of tight junction proteins and mucin proteins, thereby enhancing the stability of gut microbiota and increasing the population of beneficial bacteria (20). Nicotinamide adenine dinucleotide (NAD) activates the ARTC2/P2RX7 pathway in vivo and in vitro, depleting P2RX7-sensitive unconventional T cells (21). Succinic acid improves ConA-induced liver injury by modulating immune balance, inhibiting pro-inflammatory factors, and promoting anti-apoptotic proteins in the liver (22). To gain an intensive understanding of the composition and relative abundance of the gut microbiota, 16S rRNA gene sequencing has been widely used to characterize these microbiomes and infer the pathogenesis of AIH. Studies have revealed significant changes in the gut microbiota abundance of AIH patients and animal models, including decreased bacterial diversity and increased relative abundance of aerobic or facultative anaerobic microorganisms (23, 24).
Dysbiosis of the gut microbiota is characterized by an imbalance in microbial composition or function, such as reduced diversity, abnormal abundance of specific microbial taxa, or altered metabolic activity. When this imbalance exceeds the host’s capacity for self-regulation, it may lead to inflammation, metabolic disorders, or immune dysfunctions. In patients with AIH, both alpha diversity and beta diversity of the gut microbiota are significantly reduced. The gut microbiota richness, as measured by the Chao1 index, and evenness, as measured by the Shannon index, in AIH patients is much lower than that of healthy controls. The decline in diversity is especially evident during active disease phases or in patients with abdominal symptoms (25). The composition of the gut microbiota in AIH patients shows significant differences compared to healthy individuals, indicating a disruption in the overall microbial structure (26). There is a notable decrease in beneficial bacteria within the gut microbiota of AIH patients, such as a significant reduction in the abundance of Bifidobacterium and Faecalibacterium, which are usually associated with anti-inflammatory effects and intestinal barrier functions (27). Conversely, there is an increase in potential pathogenic bacteria in AIH patients, with genera such as Veillonella, Streptococcus, and Lactobacillus overgrowing, potentially exacerbating liver damage through immune activation or production of pro-inflammatory metabolites (27, 28). The decreased diversity of the gut microbiota in AIH patients may also be accompanied by reductions in functional genes involved in the metabolism of short-chain fatty acids, amino acid synthesis, and vitamin metabolism, thereby affecting the host’s immune and metabolic functions (26). The etiology of gut microbiota dysbiosis in AIH patients arises from intricate interactions among immunological dysregulation, metabolic disturbances, and environmental factors (Table 1).
The involvement of gut microbiota and its metabolites in the development of AIH
The pathophysiology of AIH is far from being fully clarified. However, current studies have indicated that the gut microbiota and its metabolites play a significant role in the development of AIH (Figure 1). These microorganisms modulate immune responses in both the gut and liver through specific signaling pathways. For example, disruptions in the gut microbiota interfere with the WNT/β-catenin signaling pathway, resulting in damage to the gut vascular barrier (GVB) and facilitating bacterial translocation to the liver (38). Impairment of intestinal barrier function and bacterial translocation activates the NLRP3 inflammasome in the liver, leading to inflammatory responses (39). HIF-1α plays a crucial role in the transcriptional regulation of intestinal barrier integrity and inflammation and is essential for maintaining gut microbiota homeostasis and the integrity of the intestinal barrier (40). Dysbiosis of the gut microbiota disrupts the intestinal barrier, leading to the translocation of microbes to the liver (41). Acetylated bacterial lipoproteins are integral components of bacterial cell walls, increasing when the gut microbiota balance is disrupted, thus exacerbating the immune response of the liver (42). Double-stranded DNA represents the genetic material of certain bacteria, recognized by antigen-presenting cells (APCs) in the liver. The recognition of double-stranded DNA triggers the activation of the NF-κB and Mitogen-Activated Protein Kinase (MAPK) pathways, thereby promoting the release of pro-inflammatory cytokines (42). In the liver, lipopolysaccharides (LPS) can activate hepatocytes and hepatic stellate cells (HSCs) through the TLR4-mediated signaling pathway. The activation of hepatocytes and HSCs results in the production of inflammatory and fibrogenic factors, thereby promoting the progression of liver injury and fibrosis (43). An excess of gut microbial toxins abnormally activates the innate immune system and triggers signaling pathways associated with hepatic inflammatory response, thereby disrupting liver homeostasis (23). Due to damage to the intestinal barrier and immune homeostasis imbalance, the gut microbiota continuously act as antigens, initiating and maintaining the autoimmune response in AIH (44).

Figure 1. Mechanisms of gut microbiota involved in the development of AIH. Translocation of gut microbiota, alterations in microbial metabolism, and the destabilization of immune homeostasis play crucial roles in the development of AIH. Dysbiosis in the gut microbiome leads to changes in metabolic products and breaks tight junctions of the intestinal barrier. Gut microbiota migrates to the liver and exacerbates hepatic inflammation. Alterations in microbial metabolites stimulate ERK and mTOR, activating the NF-κB signaling pathway, which triggers the expression of inflammatory cytokines and enhances the response of immune cells. Changes in the gut microbiome and its metabolites disrupt the balance among immune cells such as Treg, Th17, Th1, Th2, TFH, and TFR. AIH, autoimmune hepatitis; ERK, extracellular regulated protein kinases; mTOR, mammalian target of rapamycin; NF-kB, nuclear factor-kappa B; Treg, regulatory T; Th17, T helper 17; Th1, T helper 1; Th2, T helper 2; TFH, follicular helper T; TFR, follicular regulatory T; LPS, lipopolysaccharide; SCFAs, short-chain fatty acids; AHR, arylhydrocarbon receptor; BCAA, branched-chain amino acids; TLR4, toll-like receptor 4.
The imbalance of gut microbiota regulates the production of metabolites within the gut, including short-chain fatty acids (SCFAs), amino acids, and bile acids. These changes alter the permeability and integrity of the gut barrier and the immune homeostasis of the body. The increase of SCFAs lowers the pH in the gut, thereby inhibiting the growth of pathogens (45), which helps restore the imbalanced gut microbiota in AIH. Studies have demonstrated that SCFAs reduce T helper 1 (Th1) cells and increase the number of Treg cells, thus alleviating inflammatory responses in multiple autoimmune diseases. This suggests that SCFAs relieve inflammatory damage in AIH (46). Additionally, the main components of SCFAs include acetate, propionate, and butyrate, among which butyrate plays a key role in enhancing mucin expression in intestinal epithelial cells (IECs), improving the integrity of tight junctions and altering bacterial adhesion (47). At the same time, butyrate lowers levels of pro-inflammatory factors induced by LPS, such as tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6, while promoting the secretion of anti-inflammatory factors like IL-10 (48). Therefore, the decrease in SCFAs appears to be related to the disruption of gut barrier function and immune homeostasis, potentially accelerating the progression of AIH. Interestingly, while SCFAs suppress T cell-mediated adaptive immunity, they enhance inflammation induced by innate immune cells. For instance, SCFAs ameliorate the disease severity in collagen-induced arthritis (CIA), yet exacerbate disease severity in antigen-induced arthritis (AIA). The CIA model requires both adaptive and innate immune responses to induce the disease, where the balance among pathogenic Th1 and T helper 17 (Th17) cells, as well as Treg cells, plays a role in inhibiting disease progression. In contrast, AIA does not require adaptive immune responses; the inflammation induced by pathological antibodies in the joints is independent of T cells and relies on innate immune components such as mast cells, neutrophils, and macrophages (46).
Dysbiosis in the gut microbiota triggers changes in arginine metabolism, leading to reduced serum polyamine levels, which hinder the differentiation and maturation of gut immune cells and subsequently regulate the immune response in AIH patients (23). Furthermore, increased levels of branched-chain amino acids (BCAAs) such as leucine, valine, and isoleucine in AIH patients help enhance both innate and adaptive immune responses and regulate gut barrier function through multiple critical signaling pathways (26). The mammalian Target of Rapamycin (mTOR) signaling pathway serves as a crucial nexus for nutrient sensing and metabolic signaling. The modulation of cellular growth and metabolism by BCAAs, predominantly leucine, occurs through the activation of mTOR complex 1 (mTORC1) (49). Under conditions of amino acid limitation, the brain-specific eIF2α kinase general control nonderepressible-2 (GCN2) is activated, leading to inhibited food intake, indicating that BCAAs regulate food consumption and metabolism via the GCN2 pathway. BCAAs also respond to cellular stress by regulating the phosphorylation of eIF2α, which in turn influences the autophagy process (50). BCAAs contribute to regulating gut barrier function by impacting immune cell activation and function. For instance, BCAAs interact with immunomodulatory molecules derived from the gut microbiota, regulating intestinal immune maturation and regulation. BCAAs modulate immune responses by influencing the regulation of natural killer T (NKT) cells (51). From an inflammatory response standpoint, BCAAs modulate inflammation by influencing macrophage polarization; leucine facilitates the transition from M1 (pro-inflammatory) to M2 (anti-inflammatory) macrophages through the mTORC1/LXR signaling pathway, thereby reducing inflammation (52). BCAAs regulate T cell activation and immune responses through specific amino acid transporters such as alanine-serine-cysteine transporter 2 (ASCT2) (53).
Secondary bile acids are ligands for the G-protein coupled bile acid receptor 1 (GPBAR1), which is typically expressed in NKT cells (54). In AIH, the reduced abundance of Clostridium leads to the lack of secondary bile acids (55), resulting in GPBAR1 inactivation, the inhibition of polarization from NKT10 cells to NKT cells, and a decrease in the secretion of the anti-inflammatory cytokine IL-10 (56). GPBAR1, also known as TGR5, is expressed in a variety of cell types, including metabolically active tissues, cholangiocytes, intestinal epithelial cells, brown adipose tissue, muscle, enteric and endocrine cells, primary sensory neurons, biliary cells, and the hypothalamus (57). These expression sites highlight the indispensable role of TGR5 in metabolic regulation, bile acid signaling, and energy expenditure (58). Notably, TGR5 expression in intestinal epithelial cells allows bile acids, as metabolites in the gut, to signal nutrient availability through TGR5 activation (54). TGR5 is highly expressed in cholangiocytes, where it enhances liver protection against bile acid overload by regulating the permeability of the biliary epithelium (59).
Gut microbiota metabolites modulate microbiota composition through innate immunity activation in AIH
The intestinal microbiota and its metabolites play an indispensable role in the development of AIH through immune-microbial feedback loops (60, 61). Excessive gut-derived metabolites abnormally activate the innate immune system, subsequently leading to alterations in microbiota abundance associated with AIH (36). Firstly, the gut microbiota activate the immune system by influencing the differentiation and function of T cells. RORγt+ Th17 cells are crucial for mucosal defense; they accumulate in the gut in response to the microbiota and produce IL-17 cytokines. Specific symbiotic bacteria, such as segmented filamentous bacteria (SFB), induce the production of Th17 cells and exacerbate autoimmune responses in mice (62). This suggests that the gut microbiota directly activates specific T cell subsets, thereby regulating immune responses. Conversely, the role of the immune system in regulating the composition of the gut microbiota is also indispensable. Soluble lymphotoxin alpha (sLTα3), produced by RORγt+ innate lymphoid cells (ILCs), controls the T cell-dependent induction of IgA in the lamina propria by regulating T cells, thereby influencing the composition of the gut microbiota (63). Notably, bile acids are critical metabolites of the intestinal microbiota and influence the composition of the intestinal microbiota directly or indirectly by activating the innate immune system (64). Gut bacteria that regulate bile acid metabolism include Bacteroides, Clostridium, Lactobacillus, Bifidobacterium, and Eubacterium, which are commonly enriched in AIH patients (26).
Gut microbiota influence T cell differentiation and function in AIH
Disruption of the gut microbiota activates autoreactive T cells, particularly CD4+ T cells and CD8+ T cells, thus exacerbating autoimmune reactions in target organs (65, 66). In AIH, APCs misrecognize self-antigens from the liver as foreign antigens, promote T cells, and induce their overactivation and tissue lesions (67). Hepatocytes, acting as unconventional APCs, enhance the autoimmune response in AIH through major histocompatibility complex (MHC) class II molecules on their surfaces (68). In vitro studies have shown that low concentrations of butyrate inhibit the proliferation of CD4+ T cells and CD8+ T cells (69). There exists a complex and dynamic interplay between the gut microbiota and CD4+ T cells, regulating adaptive and innate immunity during both homeostasis and inflammation (70). This interaction is crucial for maintaining health and disease states in the local gut and extraintestinal tissues. Activated CD4+ T cells are generally found in tissues with persistent microbial colonization, with the gastrointestinal tract being the most well-studied area. Interaction between the microbiota and self-recognition is common for CD4+ T cell populations and, if properly regulated, generally does not induce disease. Clearly, the microbiota plays an active role in this regulation, promoting the activation, polarization, and function of CD4+ T cells (71). Signals from microbes guide CD4+ T cells to polarize into four functionally distinct cell subtypes: T-bet+ Th1, GATA3+ Th2, RORγt+ Th17, and FOXP3+ Tregs. Numerous signals are conveyed through epithelial or dendritic cells (DCs) (1). Aberrant composition and metabolic imbalance of the gut microbiota contribute to the pathogenesis of AIH by disturbing immune homeostasis, such as the imbalance between Tregs/Th17, T follicular regulatory (TFR)/T follicular helper (TFH), and Th1/Th2 immune cells (Figure 2). Additionally, they also regulate the effector of NKT cells and γδT cells.

Figure 2. Signals from the gut microbiota lead to the differentiation of T cells into various subtypes. Many of these signals are mediated through epithelial cells or DCs. The gut microbiota promotes Treg generation through the presentation of antigens by goblet cells and CD103+ DCs, along with the secretion of high levels of TGF-β and IL-2. CD11c+ DCs detect gut microbiota and secrete TGF-β and IL-6, which drive Th17 polarization, leading to the secretion of TNF-α and IL-22. The gut microbiota also initiates TFR cell differentiation through mTOR-mediated activation of the TCF-1-Bcl-6 axis. Furthermore, the microbiota regulates the expression of microRNAs, which in turn regulates TFR cell differentiation and function. In AIH, elevated levels of LPS inhibit TFR cells and activate TFH cells via the TLR4/MyD88 signaling pathway, promoting the production of high-affinity antibodies by secreting IL-4 and IL-21. Gut microbiota is phagocytosed by DCs or stimulates them to release pro-inflammatory cytokines like IL-6 and TNF, leading to Th1 polarization. DCs produce IL-4 and TGF-β to facilitate Th2 cell differentiation. Th2 cells release cytokines such as IL-4, IL-5, and IL-13, which are associated with AIH. DC, dendritic cell; Treg, regulatory T cell; TGF-β, transforming growth factor-β; IL, interleukin; Th17, T helper 17; Th1, T helper 1; Th2, T helper 2; TFH, follicular helper T; TFR, follicular regulatory T; mTOR, mammalian target of rapamycin; TCF-1, T-cell-specific transcription factor 1; Bcl-6, B-cell lymphoma 6; LPS, lipopolysaccharide; CTLA-4, cytotoxic T-lymphocyte-associated protein 4; TLR4, toll-like receptor 4; MyD88, myeloid differentiation primary response gene 88.
Treg/Th17 cells
Tregs and Th17 cells are pivotal subsets of T cells within the immune response framework. Tregs function mainly as immunosuppressants, aiding in the preservation of immune tolerance and preventing autoimmune maladies. Conversely, Th17 cells are strongly associated with inflammation and the progression of autoimmune diseases (72). A hallmark of autoimmune disorders is the dysregulated equilibrium between Tregs and Th17 cells (73). The gut microbiota modulates the differentiation and function of Tregs and Th17 cells by producing metabolic byproducts such as SCFAs. Notably, butyrate, a type of SCFAs, has been shown to promote the differentiation and functionality of Tregs while simultaneously restraining the development of Th17 cells (72, 74). The gut microbiota interacts with immune cells through pattern recognition receptors (PRRs), impacting the differentiation of Tregs and Th17 cells. For instance, certain components of gut microbiota activate TLRs on DCs, subsequently influencing the differentiation and functionality of Tregs and Th17 cells. The gut microbiota influences the balance of Tregs and Th17 cells by modifying the functionality of DCs. In cases of Helicobacter pylori infection, DC-induced Treg skews aid in the immune evasion of Helicobacter pylori while concurrently diminishing the immune response of Th17 cells (75). By influencing T cell metabolic pathways, the gut microbiota plays a role in the differentiation process of Tregs and Th17 cells. For example, the expression and inhibition of PDHK1 in Th17 cells regulate the Th17/Tregs ratio, thereby regulating immune responses and the progression of autoimmune disorders (76). The gut microbiota also sway the production of cytokines such as IL-6, which are instrumental in the differentiation of Tregs and Th17 cells. IL-6 is a key factor for Th17 cell differentiation, whereas Tregs differentiation relies on transforming growth factor-β (TGF-β) and IL-2 (77). Additionally, the gut microbiota regulates Tregs and Th17 cell differentiation by influencing TGF-β activation, a shared factor crucial for maintaining the balance between these cell types (78). In summary, the gut microbiota modulates the balance between Tregs and Th17 cells through various mechanisms, which include the production of metabolic byproducts, direct interaction with immune cells, regulation of cytokine production, and influence on key transcription factors involved in T cell differentiation. These mechanisms collectively regulate the development and progression of autoimmune diseases.
In patients with AIH, dysbiosis of the gut microbiota alters the metabolism of gut contents, increasing the proportion of Th17 cells while reducing the proportion of Tregs (46, 79). In rodent models of inflammation, administration of SCFAs suppresses inflammatory diseases by increasing the number of Tregs (80, 81). Butyrate promotes Tregs differentiation by inhibiting histone deacetylase and promoting acetylation of histone H3 of Foxp3 (82). Studies have shown that the aberrant Th17 cell response in AIH is associated with altered aryl hydrocarbon receptor (AHR) signaling, leading to reduced responsiveness of Th17 cells to AHR activation. Furthermore, AHR activation induces the upregulation of ectonucleoside triphosphate diphosphohydrolase 1 (CD39). CD39 is an extracellular enzyme that hydrolyzes ATP to produce the immunosuppressive adenosine, and the imbalance between Tregs and Th17 is closely related to low levels of CD39 (83). The increase in IL-17 drives newly generated Tregs to polarize into pro-inflammatory Tregs with immune activation functions, impairing the function of Tregs. This promotes immune cell infiltration and liver damage, further leading to liver inflammation and fibrosis (67). Th17 cells exacerbate liver inflammatory damage and immune attacks by promoting the secretion of pro-inflammatory cytokines such as TNF-α and IL-22 (84). Conversely, Tregs maintain immune homeostasis by inhibiting the activation of immune effector cells through the release of TGF-β and IL-10, or by interacting with DCs to inhibit their function (85, 86). Therefore, the increase in Th17 cells and decrease in Tregs induced by gut microbiota dysbiosis in AIH patients disrupts immune homeostasis, exacerbates inflammatory damage, and promotes disease progression.
Th1/Th2 cells
Th1 and Th2 cells are two primary subsets of CD4+ T cells, essential in modulating adaptive immune responses and potentially inducing certain types of autoimmune diseases. Th1 cells predominantly secrete interferon-γ (IFN-γ) and TNF-α and are involved in cell-mediated immunity and defense against intracellular pathogens. In contrast, Th2 cells release cytokines such as IL-4, IL-5, and IL-13, which are associated with allergic reactions and anti-parasitic immunity (87). During the differentiation of Th1/Th2 cells, SCFAs produced by the gut microbiota, such as butyrate, promote the generation of Treg cells, thereby inhibiting the differentiation of Th1 and Th2 cells (88). The gut microbiota directly interacts with immune cells in the gut-associated lymphoid tissue (GALT), influencing the Th1/Th2 balance. For instance, certain gut microbes activate DCs to secrete cytokines favoring a Th2 response, like IL-4, thereby promoting Th2 cell differentiation (88). The gut microbiota is involved in various metabolic pathways, including those of carbohydrates and fats, which regulate Th2 cell differentiation. Several studies suggest that metabolic pathways associated with Th2 cells are more abundant in individuals with obesity and asthma (89). The gut microbiota enhances the development and function of Treg cells, which secrete cytokines like IL-10 and TGF-β to suppress excessive Th1 and Th2 responses, hence maintaining immune tolerance (88). The gut microbiota influence host signaling pathways, such as the JAK-STAT pathway, which plays a crucial role in Th1/Th2 differentiation (90). In summary, the gut microbiota regulates the Th1/Th2 balance through multiple mechanisms, impacting the onset and progression of autoimmune diseases. These mechanisms include regulating T cell differentiation, interacting with immune cells, participating in metabolic pathways, enhancing Treg cell function, and influencing signaling pathways.
TFR/TFH cells
TFH cells are crucial for B cell activation, antibody production, class switch recombination, and affinity maturation (91). TFH cells drive differentiation by expressing Bcl-6 and assisting B cell regions (92). TFR cells, a subset of regulatory T cells, control antibody production by inhibiting TFH-mediated help. TFR cells fine-tune antibody responses and prevent excessive immune reactions through their suppressive functions (93). The gut microbiota regulate TFR cell differentiation by regulating signaling pathways. For instance, mTORC1 (mammalian target of rapamycin complex 1) initiates TFR cell differentiation during immune responses or infections by activating the TCF-1-Bcl-6 axis (94). The gut microbiota indirectly influences TFR cells by regulating TFH cell functions. TFH cells are key mediators of germinal center (GC) formation, whereas TFR cells suppress TFH-mediated GC responses (91). The gut microbiota regulates TFH and TFR cells by influencing the expression of immune regulatory molecules. The microbiota has the potential to modulate the expression of pro-inflammatory microRNAs (miRNAs), like miR-155, and anti-inflammatory miRNAs, like miR-146a, by activating host cell signaling pathways through metabolites or pathogen-associated molecular patterns (PAMPs). This regulation is crucial for the generation of TFH cells and influences the differentiation and function of TFR cells (91). For example, dysbiosis in the gut microbiota leads to the upregulation of pro-inflammatory miRNAs, like miR-21, which suppress TFR cell function and enhance TFH cell activity. This imbalance potentially exacerbates the production of autoantibodies during GC reactions (95). In summary, the gut microbiota regulates the balance between TFR and TFH cells through various mechanisms, including influencing TFR cell differentiation, TFH cell function, and the expression of immune regulatory molecules.
In the pathogenesis of AIH, the abnormal selection of high-affinity autoreactive plasma cells in the GC plays a central role. TFH cells provide signals for B cells survival and differentiation through the expression of inducible T-cell co-stimulator (ICOS) and CD40 ligand (CD40L). Additionally, TFH cells promote the production of high-affinity antibodies by secreting soluble factors such as IL-4 and IL-21. Thus, uncontrolled proliferation of TFH cells leads to excessive production of autoreactive B cells and autoantibodies, triggering an autoimmune response (96). Studies have shown that TFR cells express proteins typical of both TFH cells (ICOS and PD-1) and Tregs (CD25 and CTLA-4). Consistent with this, the study indicates that TFR cells are thymic-derived Tregs that migrate to the follicles in a TFH cell-dependent manner. Similar to TFH cells, levels of PD-1+ TFR and ICOS+ TFR cells are significantly elevated in AIH patients, suggesting that activated TFR cells suppress aberrant B cell activation and differentiation by providing negative signals (97). Furthermore, AIH patients show reduced expression of CTLA-4 in TFR cells; as CTLA-4 is crucial for maintaining immune tolerance and homeostasis, its reduction enhances cell-mediated immune responses and antibody production (98, 99). In summary, the increase of PD-1 and ICOS and the decrease of CTLA-4 on TFR cells involve regulating B cell responses in the pathogenesis of AIH. Elevated LPS in AIH disease models inhibits TFR cells and activates TFH cells through the TLR4/MyD88 signaling pathway (100, 101). The overactivation of TFH cells is closely associated with hypergammaglobulinemia, which accelerates the immunopathological process of AIH (102). TFR cells indirectly inhibit the activation of TFH cells by recognizing CTLA-4, thereby reducing autoantibody production. Therefore, the imbalance between TFR and TFH cells leads to disrupted immune homeostasis and excessive autoantibody secretion, participating in the immunopathological process of AIH (103).
NKT cells
Studies have shown that the microbiota regulates susceptibility to liver injury through the Fas/FasL pathway, and liver injury mediated by NKT cells largely involves this pathway. This suggests that the impact of the microbiota on liver injury is closely related to the function of NKT cells (104). In the mechanism by which pathogenic bacteria exacerbate Con-A-induced liver injury, NKT cells play a key role, and their activation partially depends on IL-12 produced primarily by DCs (105). In the Con-A-induced fulminant hepatitis model, similar to the situation in AIH patients, NKT cells in the liver can be activated by gut pathogens through two pathways. One possible pathway is that intestinal pathogens trigger the activation of gut DCs, which then migrate to the liver via Peyer’s patches (PPs), promoting the activation of NKT cells. Another possible pathway involves a large number of translocated intestinal antigens that first enter the liver and activate hepatic DCs, thereby activating NKT cells (105). The activated NKT cells further stimulate Kupffer cells and recruit macrophages, which secrete large amounts of inflammatory cytokines, subsequently initiating repair responses through activated hepatic stellate cells (HSCs), including hepatocyte regeneration and fibrosis (106). These responses collectively exacerbate the progression of liver inflammatory damage and fibrosis in AIH patients.
γδT cells
γδT cells are abundant in the human liver but represent a small proportion of immune cells in peripheral blood (107). As unconventional T lymphocytes, γδ T cells function by expressing the γδ T cell receptor (TCR) and are independent of antigen presentation by MHC. They are capable of recognizing MHC class I chain-related antigens A and B (MICA and MICB) as well as non-peptide metabolites produced by the isoprenoid biosynthesis pathway. Even in the absence of TCR stimulation, γδ T cells rapidly act upon activation by cytokines, allowing them to respond earlier than αβ T cells. In the mucosal barrier, microbial communities provide chronic stimulation to γδ T cells, thereby constraining their effector functions. This regulatory role is crucial for maintaining immune homeostasis at the mucosal barrier and for preventing excessive immune responses (108). The gut microbiota also influences the development and maturation of γδ T cells. Studies have shown that the presence of the gut microbial community is essential for the maturation and functional expression of γδ T cells, particularly during the early stages of life (109). Additionally, the gut microbiota regulates the tissue residency characteristics of γδ T cells. Similar to tissue-resident memory T cells, the gut microbiota modulate the transcriptional programs associated with the tissue residency of γδ T cells (110). The regulatory influence of the gut microbiota on γδ T cells also manifests in the heterogeneity of γδ T cells. Different subsets of γδ T cells respond differently to the gut microbiota, resulting in functional and effector diversity among γδ T cells (110). In summary, the gut microbiota regulates γδ T cells through multiple mechanisms, including directly regulating their effector functions, influencing their development and maturation, modulating their tissue-residency characteristics, and contributing to their heterogeneity.
The microbiota is crucial for maintaining the homeostasis of hepatic γδT17 cells, potentially involving lipid antigens provided by the microbiota. Liver cells present these antigens through CD1d, activating γδ T cells and inducing the production of IL-17A. Study shows that activated γδT17 cells exhibit pro-inflammatory and anti-infective abilities (111). A recent study shows that certain bacterial groups significantly stimulate the proliferation of colonic lamina propria γδT17 cells. For example, Bifidobacterium and Bacillus enhance barrier function by promoting the expression of TLR2 on γδ T cells. However, in vitro experiments show that only Bacillus promotes the expression of TLR2 and IL-17, while Bifidobacterium does not have this effect. Additionally, the study found a positive correlation between Bifidobacteriaceae and γδT17 cells, while Prevotellaceae, Rhodospirillaceae, and Flavobacteriaceae showed a negative correlation with γδT17 cells in the gut (112).
T cells induce gut microbiota abundance and followed AIH progression
Although the gut microbiota has an indispensable impact on the function of the mucosal immune system, particularly the functions of T lymphocytes and B lymphocytes (113), whether lymphocytes can, in turn, regulate the microbiota remains insufficiently studied. Recently, scholars have begun to focus on the relationship between T cell subsets and variations in the relative abundance of intestinal bacteria. Kierasinska M et al. explored whether dysregulation of the gut microbiota is a precursor or a consequence of the development of autoimmune diseases. However, it is well established that dysbiosis is associated with an increase in pro-inflammatory lymphocytes, particularly within the Th17 cell population. This suggests that T cells play a role in modulating changes in the gut microbiota, especially in the context of autoimmune disorders such as AIH (114).
The impact of T cells on gut microbiota composition
T cells can respond to changes in the gut microbiota and regulate immune responses by secreting cytokines like interferon-gamma and IL-17, which regulate the composition of the gut microbiota (Figure 3) (115). Specific gut microbes induce specific T-cell responses, potentially regulating the abundance and composition of the gut microbiota. For example, T cell response patterns are attributed to changes in the abundance of certain strains within the gut microbial community (116).

Figure 3. T cells modulate the composition and abundance of gut microbiota through the secretion of cytokines in AIH. The hyperactivation of Th17 cells leads to an increased secretion of pro-inflammatory cytokines such as IL-17 and IL-22, which stimulates epithelial cells to release antimicrobial peptides and triggers local inflammation. Concurrently, a dysfunction in Treg cells results in decreased secretion of anti-inflammatory cytokines like IL-10 and TGF-β, disrupting intestinal immune tolerance. This imbalance exacerbates gut dysbiosis, promoting the proliferation of pathogenic bacteria such as Enterobacteriaceae, while inhibiting the growth of beneficial bacteria like Lactobacillus. MAIT cells recognize riboflavin metabolites and monitor disturbances within the intestinal microbiome. AIH, autoimmune hepatitis; Th17, T helper 17; IL, interleukin; Treg, regulatory T cell; TGF-β, transforming growth factor-β; MAIT, mucosal-associated invariant T.
Tregs secrete anti-inflammatory cytokines, such as IL-10 and TGF-β, playing a crucial role in modulating immune responses and promoting immune tolerance, consequently reducing immune attacks against intestinal microbial antigens (117). There is a positive correlation between the expression of Foxp3 in Tregs and specific gut microbiota like the order Clostridiales and the phylum Verrucomicrobia (118). It was revealed that the gut microbiota of DEREG mice showed multidimensional separation after Tregs depletion compared to pre-depletion and wild-type mice samples. Remarkably, following Tregs depletion, there was increased relative abundance of the phylum Firmicutes associated with intestinal inflammation (119).
Th17 cells produce cytokines such as IL-17 and IL-22, which play a crucial role in stimulating epithelial cells to secrete antimicrobial peptides. These peptides are vital for the control of harmful bacteria proliferation and balance of the gut microbiota (120). Beyond their direct impact on the gut flora, Th17 cells also influence the intestinal microbial environment through interactions with other immune cells. For instance, modulating the Th17/Tregs balance augmented the abundance of beneficial bacteria while reducing the prevalence of harmful counterparts (121).
It was revealed that modulating Th1 cell-related signaling pathways, such as the TLR4-myD88/TRIF pathway, altered the distribution of gut microbiota, thus exerting a protective effect against autoimmune diseases like T1D (122). Adjusting Th1 cells ameliorated the imbalance of gut microbiota, alleviating the symptoms of IBD (123). In colitis models, the modulation of Th1/Th2 and Tregs/Th17 cell balance, alongside the inhibition of the NLRP3 inflammasome, remodeled the gut microbiota, providing new therapeutic strategies for patients with colitis (124). Th1 and Th2 cells regulated immune responses by influencing the composition of gut microbiota, thereby participating in the progression of AIH (125, 126).
Additionally, inflammation induces the composition and metabolism of the microbiome, but how the host monitors and responds to alterations remains unclear. Studies have described a protective mechanism where mucosal-associated invariant T (MAIT) cells detect microbiome-derived metabolites resulting from inflammation and promote tissue repair (127). MAIT cells recognize microbiome-derived metabolites, particularly those associated with riboflavin (vitamin B) metabolism. By detecting these metabolites, MAIT cells play a crucial role in monitoring imbalances within the gut microbiome (128).
Alterations in the thymic T cell compartment influence gut microbiota distribution
Abnormalities in the thymic T cell compartment not only promote the development of AIH but also regulate the distribution of the gut microbiota. The thymic T cell compartment regulates autoimmune susceptibility by inducing complex changes in the gut microbiota and is a potent genetic determinant. Improper selection of T cells in the thymus leads to changes in the gut microbiota, exacerbating organ-specific autoimmunity and the worsening of AIH. The study by Monica Centa et al. provides evidence that endogenous T cell compartment characteristics regulate the formation of the gut microbiota, resulting in organ-specific autoimmunity. The study reveals a connection between abnormal T cells and altered gut microbiota distribution in Traf6ΔTEC mice, due to the lack of mTECs, emphasizing the interaction between T cells and the gut microbiota. In Traf6ΔTEC mice, changes in antigen presentation lead to specific changes in gene mutations within the T cell repertoire, which in turn shapes a unique gut microbiota (129). Studies have demonstrated that the absence of TOX profoundly impacts the thymic microenvironment, particularly during the maturation of mTECs. Furthermore, the loss of TOX significantly promotes the differentiation and formation of IL-17A-secreting γδ T cells, known as γδT17 cells. The abnormal increase of these cells is considered a critical factor contributing to the development of fatal AIH (130). However, whether the increasing γδT17 cells directly damage liver tissues or indirectly promote AIH by altering the composition and distribution of the gut microbiota remains to be investigated.
Promising therapy of AIH based on the mechanisms of immune-microbial microenvironment
Currently, there is no effective treatment strategy for AIH. Glucocorticoids alone or in combination with azathioprine are the main treatments for AIH. These therapies effectively alleviate symptoms in part of AIH patients and extend their survival time (131). However, other patients remain unable to achieve symptom relief with standard therapies or develop medicine resistance. In addition, these drugs probably cause side effects such as osteoporosis, bone marrow suppression, central obesity, and liver function damage (132). Given the significant role of the gut microbiota in the occurrence and development of AIH, therapies targeting the gut microbiota are considered a new direction for AIH treatment. These methods include probiotics (133), fecal microbiota transplantation (FMT) (134), and certain drugs targeting gut microbiota-related signaling pathways (135, 136). Studies have suggested that these three strategies have demonstrated the potential to reduce autoimmune hepatitis symptoms in AIH models, suggesting that microbiota-targeted therapies offer new hope for AIH patients.
In recent years, Chimeric Antigen Receptor (CAR) -T cell therapy has increasingly drawn attention. Its basic principle involves the ex vivo genetic modification of a patient’s T cells to express specific chimeric antigen receptors. These modified T cells are then reintroduced into the patient’s body, enabling them to recognize and destroy tumor cells expressing the corresponding antigen. Although CAR-T cell therapy has made significant progress in cancer treatment, its application in autoimmune diseases is still in the early study stage. CAR-T cell therapy significantly reduces the number and functionality of B cells by profoundly depleting them within tissues, thereby decreasing the production of autoantibodies. Additionally, it inhibits the antigen-presenting function of B cells, which aids in disrupting pathogenic immune cyclic responses. Furthermore, it reduces cytokine production by B cells, diminishing inflammatory responses. In autoimmune diseases such as multiple sclerosis, B cells trigger inflammatory T cell responses by presenting self-peptides. By depleting B cells, CAR-T cell therapy indirectly reduces T cell activation (137). Potential applications of CAR-T cell therapy in autoimmune diseases include: 1) Modulating autoreactive T cells: through genetic modification, T cells express receptors that inhibit autoreactive T cells to alleviate autoimmune responses. 2) Inducing immune tolerance: modified T cells express receptors that promote immune tolerance, preventing the immune system from attacking self-tissues. 3) Targeting specific immune cells: by specifically recognizing and modulating receptors on certain immune cells such as Th17 and Treg cells, the progression of autoimmune diseases be influenced (138). Recent studies have shown that CAR-T cell therapy targeting CD19 is safe and effective for treating certain autoimmune diseases, such as systemic lupus erythematosus, systemic sclerosis, and idiopathic inflammatory myopathies (139). However, numerous challenges remain in the treatment of autoimmune diseases with CAR-T cell therapy, including precise targeting of pathological tissues, avoiding damage to normal tissues, and controlling the activity of modified T cells (137). Additionally, further studies will be needed to verify their long-term safety and efficacy. It is noteworthy that studies and applications of CAR-T cell therapy are in progress, and it is expected to provide innovative strategies and methods for the treatment of autoimmune diseases in the future. Considering the application of T cells modified by intestinal microbiota and their metabolites is inspired by natural reactions, there will possibly be more advantages than CAR-T therapies alone in terms of safety, after considerable effectiveness evaluation.
Conclusion
The gut microbiota and the host maintain a mutually beneficial symbiotic dynamic balance, playing a crucial role in numerous physiological and pathological processes, such as maintaining mucosal immune homeostasis and regulating the development of autoimmune and inflammatory diseases. The gut-liver axis profoundly influences the normal physiological functions of the liver through the dynamic changes in the gut microbiota and their metabolic products. In recent years, increasing studies have focused on the potential interaction between the gut microbiota and AIH. When the gut microbiota shifts from an anaerobic to an aerobic environment, it provokes immune responses and alters metabolites, including imbalances between Tregs/Th17, TFR/TFH, and Th1/Th2 cells, activation of NKT cells, and reductions in SCFAs and secondary bile acids, ultimately leading to the disruption of the gut barrier and immune homeostasis. Notably, the critical role of γδT cells in the pathogenesis of AIH is gradually gaining recognition. However, studies into the interaction between the gut microbiota and γδT cells remain insufficient.
During the progression of AIH, alterations in the gut microbiota modulate the differentiation and function of T cells, leading to an imbalance among T cells. This imbalance further exacerbates the inflammatory response. Conversely, the imbalance in T cells regulates the abundance of the gut microbiota, creating a vicious cycle that further exacerbates AIH. Although it remains unclear whether dysbiosis of the gut microbiota or T-cell abnormalities represent the initial trigger in the development of AIH, it is undeniable that the interaction between the gut microbiota and T cells plays an indispensable role.
Currently, targeting the gut microbiota as a treatment for AIH has become a study highlight. The importance of CAR-T cell therapy in the treatment of autoimmune diseases has been widely recognized. Although this therapy’s effectiveness in AIH treatment has not been fully verified, it could potentially open new avenues for AIH treatment. Clarification of the mutual regulatory effects between the gut microbiota and T cells is crucial for revealing the pathogenesis underlying AIH and identifying another effective therapeutic target.
Author contributions
QW: Investigation, Writing – original draft. ZG: Investigation, Writing – original draft. CL: Conceptualization, Writing – review & editing. QH: Conceptualization, Project administration, Funding acquisition, Writing – review & editing.
Funding
The author(s) declare that financial support was received for the research and/or publication of this article. This work was supported by grants from the National Natural Science Foundation (82303892 to QH). This work was also supported by grants from the China Postdoctoral Science Foundation (2023M731743), the Natural Science Foundation of Jiangsu Province (BK20230156) and the Basic Science Research Fund of Jiangsu Higher Education Institutions (23KJB320007).
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Generative AI statement
The author(s) declare that no Generative AI was used in the creation of this manuscript.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
References
1. Brown EM, Kenny DJ, and Xavier RJ. Gut microbiota regulation of T cells during inflammation and autoimmunity. Annu Rev Immunol. (2019) 37:599–624. doi: 10.1146/annurev-immunol-042718-041841
2. Sharma R, Verna EC, Soderling J, Roelstraete B, Hagstrom H, and Ludvigsson JF. Increased mortality risk in autoimmune hepatitis: A nationwide population-based cohort study with histopathology. Clin Gastroenterol Hepatol. (2021) 19:2636–47 e13. doi: 10.1016/j.cgh.2020.10.006
3. Scher JU, Nayak R, and Clemente JC. Microbiome research in autoimmune and immune-mediated inflammatory diseases: lessons, advances and unmet needs. Ann Rheum Dis. (2025) 84:9–13. doi: 10.1136/ard-2024-225735
4. Bhutta NK, Xu X, Jian C, Wang Y, Liu Y, Sun J, et al. Gut microbiota mediated T cells regulation and autoimmune diseases. Front Microbiol. (2024) 15:1477187. doi: 10.3389/fmicb.2024.1477187
5. Xia L, Li C, Zhao J, Sun Q, and Mao X. Rebalancing immune homeostasis in combating disease: The impact of medicine food homology plants and gut microbiome. Phytomedicine. (2025) 136:156150. doi: 10.1016/j.phymed.2024.156150
6. Zhang H, Liu M, Zhong W, Zheng Y, Li Y, Guo L, et al. Leaky gut driven by dysbiosis augments activation and accumulation of liver macrophages via RIP3 signaling pathway in autoimmune hepatitis. Front Immunol. (2021) 12:624360. doi: 10.3389/fimmu.2021.624360
7. Muratori L, Lohse AW, and Lenzi M. Diagnosis and management of autoimmune hepatitis. BMJ. (2023) 380:e070201. doi: 10.1136/bmj-2022-070201
8. Floreani A, Restrepo-Jimenez P, Secchi MF, De Martin S, Leung PSC, Krawitt E, et al. Etiopathogenesis of autoimmune hepatitis. J Autoimmun. (2018) 95:133–43. doi: 10.1016/j.jaut.2018.10.020
9. Ma L, Zhang L, Zhuang Y, Ding Y, and Chen J. Lactobacillus improves the effects of prednisone on autoimmune hepatitis via gut microbiota-mediated follicular helper T cells. Cell Commun Signal. (2022) 20:83. doi: 10.1186/s12964-021-00819-7
10. Schneider KM, Kummen M, Trivedi PJ, and Hov JR. Role of microbiome in autoimmune liver diseases. Hepatology. (2024) 80:965–87. doi: 10.1097/HEP.0000000000000506
11. Lin H, Lin J, Pan T, Li T, Jiang H, Fang Y, et al. Polymeric immunoglobulin receptor deficiency exacerbates autoimmune hepatitis by inducing intestinal dysbiosis and barrier dysfunction. Cell Death Dis. (2023) 14:68. doi: 10.1038/s41419-023-05589-3
12. Schnabl B and Brenner DA. Interactions between the intestinal microbiome and liver diseases. Gastroenterology. (2014) 146:1513–24. doi: 10.1053/j.gastro.2014.01.020
13. Li L and Kang Y. The gut microbiome and autoimmune hepatitis: implications for early diagnostic biomarkers and novel therapies. Mol Nutr Food Res. (2023) 67:e2300043. doi: 10.1002/mnfr.202300043
14. Airola C, Severino A, Porcari S, Fusco W, Mullish BH, Gasbarrini A, et al. Future modulation of gut microbiota: from eubiotics to FMT, engineered bacteria, and phage therapy. Antibiot (Basel). (2023) 12(5):868. doi: 10.3390/antibiotics12050868
15. Gupta U and Dey P. Rise of the guardians: Gut microbial maneuvers in bacterial infections. Life Sci. (2023) 330:121993. doi: 10.1016/j.lfs.2023.121993
16. Plaza-Diaz J, Solis-Urra P, Rodriguez-Rodriguez F, Olivares-Arancibia J, Navarro-Oliveros M, Abadia-Molina F, et al. The gut barrier, intestinal microbiota, and liver disease: molecular mechanisms and strategies to manage. Int J Mol Sci. (2020) 21(21):8351. doi: 10.3390/ijms21218351
17. Cai W, Ran Y, Li Y, Wang B, and Zhou L. Intestinal microbiome and permeability in patients with autoimmune hepatitis. Best Pract Res Clin Gastroenterol. (2017) 31:669–73. doi: 10.1016/j.bpg.2017.09.013
18. Guan H, Zhang X, Kuang M, and Yu J. The gut-liver axis in immune remodeling of hepatic cirrhosis. Front Immunol. (2022) 13:946628. doi: 10.3389/fimmu.2022.946628
19. Bruneau A, Shevchenko Y, Tacke F, and Hammerich L. A comprehensive 26-color immunophenotyping panel to study the role of the gut-liver axis in chronic liver diseases. Cytomet B Clin Cytom. (2025) 108:15–22. doi: 10.1002/cyto.b.v108.1
20. Wang L, Wang H, Li X, Zhu M, Gao D, Hu D, et al. Bacillus velezensis HBXN2020 alleviates Salmonella Typhimurium infection in mice by improving intestinal barrier integrity and reducing inflammation. Elife. (2024) 13:RP93423. doi: 10.7554/eLife.93423
21. Xu C, Obers A, Qin M, Brandli A, Wong J, Huang X, et al. Selective regulation of IFN-gamma and IL-4 co-producing unconventional T cells by purinergic signaling. J Exp Med. (2024) 221(12):e20240354. doi: 10.1084/jem.20240354
22. Cai Y, Chen Z, Chen E, Zhang D, Wei T, Sun M, et al. Succinic acid ameliorates concanavalin A-induced hepatitis by altering the inflammatory microenvironment and expression of BCL-2 family proteins. Inflammation. (2024) 47(6):2000–12. doi: 10.1007/s10753-024-02021-6
23. Wei Y, Li Y, Yan L, Sun C, Miao Q, Wang Q, et al. Alterations of gut microbiome in autoimmune hepatitis. Gut. (2020) 69:569–77. doi: 10.1136/gutjnl-2018-317836
24. Yuksel M, Wang Y, Tai N, Peng J, Guo J, Beland K, et al. A novel “humanized mouse” model for autoimmune hepatitis and the association of gut microbiota with liver inflammation. Hepatology. (2015) 62:1536–50. doi: 10.1002/hep.27998
25. Zheng Y, Ran Y, Zhang H, Wang B, and Zhou L. The microbiome in autoimmune liver diseases: metagenomic and metabolomic changes. Front Physiol. (2021) 12:715852. doi: 10.3389/fphys.2021.715852
26. Elsherbiny NM, Rammadan M, Hassan EA, Ali ME, El-Rehim ASA, Abbas WA, et al. Autoimmune hepatitis: shifts in gut microbiota and metabolic pathways among Egyptian patients. Microorganisms. (2020) 8(7):1011. doi: 10.3390/microorganisms8071011
27. Liwinski T, Casar C, Ruehlemann MC, Bang C, Sebode M, Hohenester S, et al. A disease-specific decline of the relative abundance of Bifidobacterium in patients with autoimmune hepatitis. Aliment Pharmacol Ther. (2020) 51:1417–28. doi: 10.1111/apt.v51.12
28. Kim SS, Eun JW, Cho HJ, Song DS, Kim CW, Kim YS, et al. Microbiome as a potential diagnostic and predictive biomarker in severe alcoholic hepatitis. Aliment Pharmacol Ther. (2021) 53:540–51. doi: 10.1111/apt.16200
29. Fang H, Anhe FF, and Schertzer JD. Dietary sugar lowers immunity and microbiota that protect against metabolic disease. Cell Metab. (2022) 34:1422–4. doi: 10.1016/j.cmet.2022.09.006
30. Wang J, Mei L, Hao Y, Xu Y, Yang Q, Dai Z, et al. Contemporary perspectives on the role of vitamin D in enhancing gut health and its implications for preventing and managing intestinal diseases. Nutrients. (2024) 16(14):2352. doi: 10.3390/nu16142352
31. Kunasol C, Chattipakorn N, and Chattipakorn SC. Impact of calcineurin inhibitors on gut microbiota: Focus on tacrolimus with evidence from in vivo and clinical studies. Eur J Pharmacol. (2025) 987:177176. doi: 10.1016/j.ejphar.2024.177176
32. Liu Q, He W, Tang R, and Ma X. Intestinal homeostasis in autoimmune liver diseases. Chin Med J (Engl). (2022) 135:1642–52. doi: 10.1097/CM9.0000000000002291
33. Cui H, Wang N, Li H, Bian Y, Wen W, Kong X, et al. The dynamic shifts of IL-10-producing Th17 and IL-17-producing Treg in health and disease: a crosstalk between ancient “Yin-Yang” theory and modern immunology. Cell Commun Signal. (2024) 22:99. doi: 10.1186/s12964-024-01505-0
34. Berryman MA, Ilonen J, Triplett EW, and Ludvigsson J. Important denominator between autoimmune comorbidities: a review of class II HLA, autoimmune disease, and the gut. Front Immunol. (2023) 14:1270488. doi: 10.3389/fimmu.2023.1270488
35. Li Y, Zhou L, Huang Z, Yang Y, Zhang J, Yang L, et al. Fine mapping identifies independent HLA associations in autoimmune hepatitis type 1. JHEP Rep. (2024) 6:100926. doi: 10.1016/j.jhepr.2023.100926
36. Sun C, Zhu D, Zhu Q, He Z, Lou Y, and Chen D. The significance of gut microbiota in the etiology of autoimmune hepatitis: a narrative review. Front Cell Infect Microbiol. (2024) 14:1337223. doi: 10.3389/fcimb.2024.1337223
37. Contreras GV, Marugan MT, and Cuervas-Mons V. Autoimmune extrahepatic disorders in patients with autoimmune liver disease. Transplant Proc. (2021) 53:2695–7. doi: 10.1016/j.transproceed.2021.06.031
38. Mouries J, Brescia P, Silvestri A, Spadoni I, Sorribas M, Wiest R, et al. Microbiota-driven gut vascular barrier disruption is a prerequisite for non-alcoholic steatohepatitis development. J Hepatol. (2019) 71:1216–28. doi: 10.1016/j.jhep.2019.08.005
39. Liao L, Schneider KM, Galvez EJC, Frissen M, Marschall HU, Su H, et al. Intestinal dysbiosis augments liver disease progression via NLRP3 in a murine model of primary sclerosing cholangitis. Gut. (2019) 68:1477–92. doi: 10.1136/gutjnl-2018-316670
40. Shao T, Zhao C, Li F, Gu Z, Liu L, Zhang L, et al. Intestinal HIF-1alpha deletion exacerbates alcoholic liver disease by inducing intestinal dysbiosis and barrier dysfunction. J Hepatol. (2018) 69:886–95. doi: 10.1016/j.jhep.2018.05.021
41. Hsu CL and Schnabl B. The gut-liver axis and gut microbiota in health and liver disease. Nat Rev Microbiol. (2023) 21:719–33. doi: 10.1038/s41579-023-00904-3
42. Chen D, Le TH, Shahidipour H, Read SA, and Ahlenstiel G. The role of gut-derived microbial antigens on liver fibrosis initiation and progression. Cells. (2019) 8(11):1324. doi: 10.3390/cells8111324
43. Sharma A, Wang J, and Gandhi CR. CD14 is not required for carbon tetrachloride-induced hepatic inflammation and fibrosis with or without lipopolysaccharide challenge. J Cell Physiol. (2023) 238:1530–41. doi: 10.1002/jcp.v238.7
44. Ignacio A, Morales CI, Camara NO, and Almeida RR. Innate sensing of the gut microbiota: modulation of inflammatory and autoimmune diseases. Front Immunol. (2016) 7:54. doi: 10.3389/fimmu.2016.00054
45. Sorbara MT, Dubin K, Littmann ER, Moody TU, Fontana E, Seok R, et al. Inhibiting antibiotic-resistant Enterobacteriaceae by microbiota-mediated intracellular acidification. J Exp Med. (2019) 216:84–98. doi: 10.1084/jem.20181639
46. Mizuno M, Noto D, Kaga N, Chiba A, and Miyake S. The dual role of short fatty acid chains in the pathogenesis of autoimmune disease models. PloS One. (2017) 12:e0173032. doi: 10.1371/journal.pone.0173032
47. Jung TH, Park JH, Jeon WM, and Han KS. Butyrate modulates bacterial adherence on LS174T human colorectal cells by stimulating mucin secretion and MAPK signaling pathway. Nutr Res Pract. (2015) 9:343–9. doi: 10.4162/nrp.2015.9.4.343
48. Wang F, Liu J, Weng T, Shen K, Chen Z, Yu Y, et al. The Inflammation Induced by Lipopolysaccharide can be Mitigated by Short-chain Fatty Acid, Butyrate, through Upregulation of IL-10 in Septic Shock. Scand J Immunol. (2017) 85:258–63. doi: 10.1111/sji.2017.85.issue-4
49. Cangelosi AL, Puszynska AM, Roberts JM, Armani A, Nguyen TP, Spinelli JB, et al. Zonated leucine sensing by Sestrin-mTORC1 in the liver controls the response to dietary leucine. Science. (2022) 377:47–56. doi: 10.1126/science.abi9547
50. Maurin AC, Benani A, Lorsignol A, Brenachot X, Parry L, Carraro V, et al. Hypothalamic eIF2alpha signaling regulates food intake. Cell Rep. (2014) 6:438–44. doi: 10.1016/j.celrep.2014.01.006
51. Oh SF, Praveena T, Song H, Yoo JS, Jung DJ, Erturk-Hasdemir D, et al. Host immunomodulatory lipids created by symbionts from dietary amino acids. Nature. (2021) 600:302–7. doi: 10.1038/s41586-021-04083-0
52. Yan H, Liu Y, Li X, Yu B, He J, Mao X, et al. Leucine alleviates cytokine storm syndrome by regulating macrophage polarization via the mTORC1/LXRalpha signaling pathway. Elife. (2024) 12:RP89750. doi: 10.7554/eLife.89750
53. Nakaya M, Xiao Y, Zhou X, Chang JH, Chang M, Cheng X, et al. Inflammatory T cell responses rely on amino acid transporter ASCT2 facilitation of glutamine uptake and mTORC1 kinase activation. Immunity. (2014) 40:692–705. doi: 10.1016/j.immuni.2014.04.007
54. Sorrentino G, Perino A, Yildiz E, El Alam G, Bou Sleiman M, Gioiello A, et al. Bile acids signal via TGR5 to activate intestinal stem cells and epithelial regeneration. Gastroenterology. (2020) 159:956–68 e8. doi: 10.1053/j.gastro.2020.05.067
55. Kayama H, Okumura R, and Takeda K. Interaction between the microbiota, epithelia, and immune cells in the intestine. Annu Rev Immunol. (2020) 38:23–48. doi: 10.1146/annurev-immunol-070119-115104
56. Biagioli M, Carino A, Fiorucci C, Marchiano S, Di Giorgio C, Roselli R, et al. GPBAR1 functions as gatekeeper for liver NKT cells and provides counterregulatory signals in mouse models of immune-mediated hepatitis. Cell Mol Gastroenterol Hepatol. (2019) 8:447–73. doi: 10.1016/j.jcmgh.2019.06.003
57. van Nierop FS, Scheltema MJ, Eggink HM, Pols TW, Sonne DP, Knop FK, et al. Clinical relevance of the bile acid receptor TGR5 in metabolism. Lancet Diabetes Endocrinol. (2017) 5:224–33. doi: 10.1016/S2213-8587(16)30155-3
58. Jansen PL. A new life for bile acids. J Hepatol. (2010) 52:937–8. doi: 10.1016/j.jhep.2010.02.003
59. Merlen G, Kahale N, Ursic-Bedoya J, Bidault-Jourdainne V, Simerabet H, Doignon I, et al. TGR5-dependent hepatoprotection through the regulation of biliary epithelium barrier function. Gut. (2020) 69:146–57. doi: 10.1136/gutjnl-2018-316975
60. Song X, Sun X, Oh SF, Wu M, Zhang Y, Zheng W, et al. Microbial bile acid metabolites modulate gut RORgamma(+) regulatory T cell homeostasis. Nature. (2020) 577:410–5. doi: 10.1038/s41586-019-1865-0
61. Longhi MS. Lactobacillus reuteri joins the liver autoimmune arena. Cell Host Microbe. (2022) 30:901–3. doi: 10.1016/j.chom.2022.06.004
62. Sano T, Huang W, Hall JA, Yang Y, Chen A, Gavzy SJ, et al. An IL-23R/IL-22 circuit regulates epithelial serum amyloid A to promote local effector th17 responses. Cell. (2015) 163:381–93. doi: 10.1016/j.cell.2015.08.061
63. Kruglov AA, Grivennikov SI, Kuprash DV, Winsauer C, Prepens S, Seleznik GM, et al. Nonredundant function of soluble LTalpha3 produced by innate lymphoid cells in intestinal homeostasis. Science. (2013) 342:1243–6. doi: 10.1126/science.1243364
64. Guo X, Okpara ES, Hu W, Yan C, Wang Y, Liang Q, et al. Interactive relationships between intestinal flora and bile acids. Int J Mol Sci. (2022) 23(15):8343. doi: 10.3390/ijms23158343
65. Horai R, Zarate-Blades CR, Dillenburg-Pilla P, Chen J, Kielczewski JL, Silver PB, et al. Microbiota-dependent activation of an autoreactive T cell receptor provokes autoimmunity in an immunologically privileged site. Immunity. (2015) 43:343–53. doi: 10.1016/j.immuni.2015.07.014
66. Ruff WE, Dehner C, Kim WJ, Pagovich O, Aguiar CL, Yu AT, et al. Pathogenic autoreactive T and B cells cross-react with mimotopes expressed by a common human gut commensal to trigger autoimmunity. Cell Host Microbe. (2019) 26:100–13 e8. doi: 10.1016/j.chom.2019.05.003
67. Longhi MS, Liberal R, Holder B, Robson SC, Ma Y, Mieli-Vergani G, et al. Inhibition of interleukin-17 promotes differentiation of CD25(-) cells into stable T regulatory cells in patients with autoimmune hepatitis. Gastroenterology. (2012) 142:1526–35 e6. doi: 10.1053/j.gastro.2012.02.041
68. Gong J, Tu W, Liu J, and Tian D. Hepatocytes: A key role in liver inflammation. Front Immunol. (2022) 13:1083780. doi: 10.3389/fimmu.2022.1083780
69. Correa-Oliveira R, Fachi JL, Vieira A, Sato FT, and Vinolo MA. Regulation of immune cell function by short-chain fatty acids. Clin Transl Immunol. (2016) 5:e73. doi: 10.1038/cti.2016.17
70. Lee N and Kim WU. Microbiota in T-cell homeostasis and inflammatory diseases. Exp Mol Med. (2017) 49:e340. doi: 10.1038/emm.2017.36
71. Honda K and Littman DR. The microbiome in infectious disease and inflammation. Annu Rev Immunol. (2012) 30:759–95. doi: 10.1146/annurev-immunol-020711-074937
72. Antignano F, Burrows K, Hughes MR, Han JM, Kron KJ, Penrod NM, et al. Methyltransferase G9A regulates T cell differentiation during murine intestinal inflammation. J Clin Invest. (2014) 124:1945–55. doi: 10.1172/JCI69592
73. Fujiwara M, Raheja R, Garo LP, Ajay AK, Kadowaki-Saga R, Karandikar SH, et al. microRNA-92a promotes CNS autoimmunity by modulating the regulatory and inflammatory T cell balance. J Clin Invest. (2022) 132(10):e155693. doi: 10.1172/JCI155693
74. Aki D, Hayakawa T, Srirat T, Shichino S, Ito M, Saitoh SI, et al. The Nr4a family regulates intrahepatic Treg proliferation and liver fibrosis in MASLD models. J Clin Invest. (2024) 134(23):e175305. doi: 10.1172/JCI175305
75. Kao JY, Zhang M, Miller MJ, Mills JC, Wang B, Liu M, et al. Helicobacter pylori immune escape is mediated by dendritic cell-induced Treg skewing and Th17 suppression in mice. Gastroenterology. (2010) 138:1046–54. doi: 10.1053/j.gastro.2009.11.043
76. Gerriets VA, Kishton RJ, Nichols AG, Macintyre AN, Inoue M, Ilkayeva O, et al. Metabolic programming and PDHK1 control CD4+ T cell subsets and inflammation. J Clin Invest. (2015) 125:194–207. doi: 10.1172/JCI76012
77. Korn T, Bettelli E, Oukka M, and Kuchroo VK. IL-17 and th17 cells. Annu Rev Immunol. (2009) 27:485–517. doi: 10.1146/annurev.immunol.021908.132710
78. Melton AC, Bailey-Bucktrout SL, Travis MA, Fife BT, Bluestone JA, and Sheppard D. Expression of alphavbeta8 integrin on dendritic cells regulates Th17 cell development and experimental autoimmune encephalomyelitis in mice. J Clin Invest. (2010) 120:4436–44. doi: 10.1172/JCI43786
79. Lou J, Jiang Y, Rao B, Li A, Ding S, Yan H, et al. Fecal microbiomes distinguish patients with autoimmune hepatitis from healthy individuals. Front Cell Infect Microbiol. (2020) 10:342. doi: 10.3389/fcimb.2020.00342
80. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. (2013) 341:569–73. doi: 10.1126/science.1241165
81. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. (2013) 504:451–5. doi: 10.1038/nature12726
82. Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. (2013) 504:446–50. doi: 10.1038/nature12721
83. Vuerich M, Harshe R, Frank LA, Mukherjee S, Gromova B, Csizmadia E, et al. Altered aryl-hydrocarbon-receptor signalling affects regulatory and effector cell immunity in autoimmune hepatitis. J Hepatol. (2021) 74:48–57. doi: 10.1016/j.jhep.2020.06.044
84. de Oliveira GLV, Leite AZ, Higuchi BS, Gonzaga MI, and Mariano VS. Intestinal dysbiosis and probiotic applications in autoimmune diseases. Immunology. (2017) 152:1–12. doi: 10.1111/imm.2017.152.issue-1
85. Chen J, Liu W, and Zhu W. Foxp3(+) treg cells are associated with pathological process of autoimmune hepatitis by activating methylation modification in autoimmune hepatitis patients. Med Sci Monit. (2019) 25:6204–12. doi: 10.12659/MSM.915408
86. Wang H, Feng X, Yan W, and Tian D. Regulatory T cells in autoimmune hepatitis: unveiling their roles in mouse models and patients. Front Immunol. (2020) 11:575572. doi: 10.3389/fimmu.2020.575572
87. Yamane H and Paul WE. Early signaling events that underlie fate decisions of naive CD4(+) T cells toward distinct T-helper cell subsets. Immunol Rev. (2013) 252:12–23. doi: 10.1111/imr.2013.252.issue-1
88. Yin X, Chen S, and Eisenbarth SC. Dendritic cell regulation of T helper cells. Annu Rev Immunol. (2021) 39:759–90. doi: 10.1146/annurev-immunol-101819-025146
89. Tibbitt CA, Stark JM, Martens L, Ma J, Mold JE, Deswarte K, et al. Single-cell RNA sequencing of the T helper cell response to house dust mites defines a distinct gene expression signature in airway th2 cells. Immunity. (2019) 51:169–84 e5. doi: 10.1016/j.immuni.2019.05.014
90. Ma CS, Wong N, Rao G, Nguyen A, Avery DT, Payne K, et al. Unique and shared signaling pathways cooperate to regulate the differentiation of human CD4+ T cells into distinct effector subsets. J Exp Med. (2016) 213:1589–608. doi: 10.1084/jem.20151467
91. Maul J, Alterauge D, and Baumjohann D. MicroRNA-mediated regulation of T follicular helper and T follicular regulatory cell identity. Immunol Rev. (2019) 288:97–111. doi: 10.1111/imr.2019.288.issue-1
92. Linterman MA, Liston A, and Vinuesa CG. T-follicular helper cell differentiation and the co-option of this pathway by non-helper cells. Immunol Rev. (2012) 247:143–59. doi: 10.1111/j.1600-065X.2012.01121.x
93. Hou S, Clement RL, Diallo A, Blazar BR, Rudensky AY, Sharpe AH, et al. FoxP3 and Ezh2 regulate Tfr cell suppressive function and transcriptional program. J Exp Med. (2019) 216:605–20. doi: 10.1084/jem.20181134
94. Xu L, Huang Q, Wang H, Hao Y, Bai Q, Hu J, et al. The kinase mTORC1 promotes the generation and suppressive function of follicular regulatory T cells. Immunity. (2017) 47:538–51 e5. doi: 10.1016/j.immuni.2017.08.011
95. Ding T, Niu H, Zhao X, Gao C, Li X, and Wang C. T-follicular regulatory cells: potential therapeutic targets in rheumatoid arthritis. Front Immunol. (2019) 10:2709. doi: 10.3389/fimmu.2019.02709
96. Park HJ, Kim DH, Lim SH, Kim WJ, Youn J, Choi YS, et al. Insights into the role of follicular helper T cells in autoimmunity. Immune Netw. (2014) 14:21–9. doi: 10.4110/in.2014.14.1.21
97. Sage PT and Sharpe AH. T follicular regulatory cells in the regulation of B cell responses. Trends Immunol. (2015) 36:410–8. doi: 10.1016/j.it.2015.05.005
98. Paterson AM, Lovitch SB, Sage PT, Juneja VR, Lee Y, Trombley JD, et al. Deletion of CTLA-4 on regulatory T cells during adulthood leads to resistance to autoimmunity. J Exp Med. (2015) 212:1603–21. doi: 10.1084/jem.20141030
99. Sage PT, Paterson AM, Lovitch SB, and Sharpe AH. The coinhibitory receptor CTLA-4 controls B cell responses by modulating T follicular helper, T follicular regulatory, and T regulatory cells. Immunity. (2014) 41:1026–39. doi: 10.1016/j.immuni.2014.12.005
100. Liu J, Kang R, and Tang D. Lipopolysaccharide delivery systems in innate immunity. Trends Immunol. (2024) 45:274–87. doi: 10.1016/j.it.2024.02.003
101. Burke TP, Engstrom P, Chavez RA, Fonbuena JA, Vance RE, and Welch MD. Inflammasome-mediated antagonism of type I interferon enhances Rickettsia pathogenesis. Nat Microbiol. (2020) 5:688–96. doi: 10.1038/s41564-020-0673-5
102. Liang M, Liwen Z, Juan D, Yun Z, Yanbo D, and Jianping C. Dysregulated TFR and TFH cells correlate with B-cell differentiation and antibody production in autoimmune hepatitis. J Cell Mol Med. (2020) 24:3948–57. doi: 10.1111/jcmm.14997
103. Liang M, Liwen Z, Jianguo S, Juan D, Fei D, Yin Z, et al. Fecal microbiota transplantation controls progression of experimental autoimmune hepatitis in mice by modulating the TFR/TFH immune imbalance and intestinal microbiota composition. Front Immunol. (2021) 12:728723. doi: 10.3389/fimmu.2021.728723
104. Celaj S, Gleeson MW, Deng J, O’Toole GA, Hampton TH, Toft MF, et al. The microbiota regulates susceptibility to Fas-mediated acute hepatic injury. Lab Invest. (2014) 94:938–49. doi: 10.1038/labinvest.2014.93
105. Chen J, Wei Y, He J, Cui G, Zhu Y, Lu C, et al. Natural killer T cells play a necessary role in modulating of immune-mediated liver injury by gut microbiota. Sci Rep. (2014) 4:7259. doi: 10.1038/srep07259
106. Liu C, Wang YL, Yang YY, Zhang NP, Niu C, Shen XZ, et al. Novel approaches to intervene gut microbiota in the treatment of chronic liver diseases. FASEB J. (2021) 35:e21871. doi: 10.1096/fj.202100939R
107. Doherty DG. Immunity, tolerance and autoimmunity in the liver: A comprehensive review. J Autoimmun. (2016) 66:60–75. doi: 10.1016/j.jaut.2015.08.020
108. Huang HI, Xue Y, Jewell ML, Tan CY, Theriot B, Aggarwal N, et al. A binary module for microbiota-mediated regulation of gammadelta17 cells, hallmarked by microbiota-driven expression of programmed cell death protein 1. Cell Rep. (2023) 42:112951. doi: 10.1016/j.celrep.2023.112951
109. Roy Chowdhury R, Valainis JR, Dubey M, von Boehmer L, Sola E, Wilhelmy J, et al. NK-like CD8(+) gammadelta T cells are expanded in persistent Mycobacterium tuberculosis infection. Sci Immunol. (2023) 8:eade3525. doi: 10.1126/sciimmunol.ade3525
110. du Halgouet A, Bruder K, Peltokangas N, Darbois A, Obwegs D, Salou M, et al. Multimodal profiling reveals site-specific adaptation and tissue residency hallmarks of gammadelta T cells across organs in mice. Nat Immunol. (2024) 25:343–56. doi: 10.1038/s41590-023-01710-y
111. Khairallah C, Chu TH, and Sheridan BS. Tissue adaptations of memory and tissue-resident gamma delta T cells. Front Immunol. (2018) 9:2636. doi: 10.3389/fimmu.2018.02636
112. Li M, Wang B, Sun X, Tang Y, Wei X, Ge B, et al. Upregulation of intestinal barrier function in mice with DSS-induced colitis by a defined bacterial consortium is associated with expansion of IL-17A producing gamma delta T cells. Front Immunol. (2017) 8:824. doi: 10.3389/fimmu.2017.00824
113. Sorini C, Cardoso RF, Gagliani N, and Villablanca EJ. Commensal bacteria-specific CD4(+) T cell responses in health and disease. Front Immunol. (2018) 9:2667. doi: 10.3389/fimmu.2018.02667
114. Kierasinska M and Donskow-Lysoniewska K. Both the microbiome and the macrobiome can influence immune responsiveness in psoriasis. Cent Eur J Immunol. (2021) 46:502–8. doi: 10.5114/ceji.2021.110314
115. Zhao Q and Elson CO. Adaptive immune education by gut microbiota antigens. Immunology. (2018) 154:28–37. doi: 10.1111/imm.2018.154.issue-1
116. Nagashima K, Zhao A, Atabakhsh K, Bae M, Blum JE, Weakley A, et al. Mapping the T cell repertoire to a complex gut bacterial community. Nature. (2023) 621:162–70. doi: 10.1038/s41586-023-06431-8
117. Sanchez-Alcoholado L, Castellano-Castillo D, Jordan-Martinez L, Moreno-Indias I, Cardila-Cruz P, Elena D, et al. Role of gut microbiota on cardio-metabolic parameters and immunity in coronary artery disease patients with and without type-2 diabetes mellitus. Front Microbiol. (2017) 8:1936. doi: 10.3389/fmicb.2017.01936
118. Lindenberg F, Krych L, Fielden J, Kot W, Frokiaer H, van Galen G, et al. Expression of immune regulatory genes correlate with the abundance of specific Clostridiales and Verrucomicrobia species in the equine ileum and cecum. Sci Rep. (2019) 9:12674. doi: 10.1038/s41598-019-49081-5
119. Kehrmann J, Effenberg L, Wilk C, Schoemer D, Ngo Thi Phuong N, Adamczyk A, et al. Depletion of Foxp3(+) regulatory T cells is accompanied by an increase in the relative abundance of Firmicutes in the murine gut microbiome. Immunology. (2020) 159:344–53. doi: 10.1111/imm.v159.3
120. Lin M, Piao L, Zhao Z, Liao L, Wang D, Zhang H, et al. Therapeutic potential of cajanus cajan (L.) millsp. Leaf extract in modulating gut microbiota and immune response for the treatment of inflammatory bowel disease. Pharmaceut (Basel). (2025) 18(1):67. doi: 10.3390/ph18010067
121. Yan Y, Li K, Jiang J, Jiang L, Ma X, Ai F, et al. Perinatal tissue-derived exosomes ameliorate colitis in mice by regulating the Foxp3 + Treg cells and gut microbiota. Stem Cell Res Ther. (2023) 14:43. doi: 10.1186/s13287-023-03263-1
122. Luo C, Yang D, Hou C, Tan T, and Chao C. Paeoniflorin protects NOD mice from T1D through regulating gut microbiota and TLR4 mediated myD88/TRIF pathway. Exp Cell Res. (2023) 422:113429. doi: 10.1016/j.yexcr.2022.113429
123. Yang F, Zhang M, Xu R, Yu Y, Feng H, Li D, et al. SDH, a novel diarylheptane compound, alleviates dextran sulfate sodium (DSS)-induced colitis by reducing Th1/Th2/Th17 induction and regulating the gut microbiota in mice. Int Immunopharmacol. (2024) 134:112234. doi: 10.1016/j.intimp.2024.112234
124. Hu Y, Tang J, Xie Y, Xu W, Zhu W, Xia L, et al. Gegen Qinlian decoction ameliorates TNBS-induced ulcerative colitis by regulating Th2/Th1 and Tregs/Th17 cells balance, inhibiting NLRP3 inflammasome activation and reshaping gut microbiota. J Ethnopharmacol. (2024) 328:117956. doi: 10.1016/j.jep.2024.117956
125. Liang M, Liwen Z, Yun Z, Yanbo D, and Jianping C. Serum levels of IL-33 and correlation with IL-4, IL-17A, and hypergammaglobulinemia in patients with autoimmune hepatitis. Mediators Inflamm. (2018) 2018:7964654. doi: 10.1155/2018/7964654
126. Liang M, Liwen Z, Yun Z, Yanbo D, and Jianping C. The imbalance between foxp3(+)Tregs and th1/th17/th22 cells in patients with newly diagnosed autoimmune hepatitis. J Immunol Res. (2018) 2018:3753081. doi: 10.1155/2018/3753081
127. El Morr Y, Furstenheim M, Mestdagh M, Franciszkiewicz K, Salou M, Morvan C, et al. MAIT cells monitor intestinal dysbiosis and contribute to host protection during colitis. Sci Immunol. (2024) 9:eadi8954. doi: 10.1126/sciimmunol.adi8954
128. Germain L, Veloso P, Lantz O, and Legoux F. MAIT cells: Conserved watchers on the wall. J Exp Med. (2025) 222(1):e20232298. doi: 10.1084/jem.20232298
129. Centa M, Weinstein EG, Clemente JC, Faith JJ, Fiel MI, Lyallpuri R, et al. Impaired central tolerance induces changes in the gut microbiota that exacerbate autoimmune hepatitis. J Autoimmun. (2022) 128:102808. doi: 10.1016/j.jaut.2022.102808
130. He Q, Lu Y, Tian W, Jiang R, Yu W, Liu Y, et al. TOX deficiency facilitates the differentiation of IL-17A-producing gammadelta T cells to drive autoimmune hepatitis. Cell Mol Immunol. (2022) 19:1102–16. doi: 10.1038/s41423-022-00912-y
131. Mack CL, Adams D, Assis DN, Kerkar N, Manns MP, Mayo MJ, et al. Diagnosis and management of autoimmune hepatitis in adults and children: 2019 practice guidance and guidelines from the american association for the study of liver diseases. Hepatology. (2020) 72:671–722. doi: 10.1002/hep.31065
132. Lee YJ, Choi WY, Park KS, Kim YJ, Cho KB, Kim ES, et al. Differences in the adverse effects of azathioprine between inflammatory bowel disease and autoimmune hepatitis in Korean patients. Korean J Gastroenterol. (2014) 64:348–55. doi: 10.4166/kjg.2014.64.6.348
133. Zhang H, Liu M, Liu X, Zhong W, Li Y, Ran Y, et al. Bifidobacterium animalis ssp. Lactis 420 Mitigates Autoimmune Hepatitis Through Regulating Intestinal Barrier and Liver Immune Cells. Front Immunol. (2020) 11:569104. doi: 10.3389/fimmu.2020.569104
134. Wang H, Banerjee N, Liang Y, Wang G, Hoffman KL, and Khan MF. Gut microbiome-host interactions in driving environmental pollutant trichloroethene-mediated autoimmunity. Toxicol Appl Pharmacol. (2021) 424:115597. doi: 10.1016/j.taap.2021.115597
135. Telesford KM, Yan W, Ochoa-Reparaz J, Pant A, Kircher C, Christy MA, et al. A commensal symbiotic factor derived from Bacteroides fragilis promotes human CD39(+)Foxp3(+) T cells and Treg function. Gut Microbes. (2015) 6:234–42. doi: 10.1080/19490976.2015.1056973
136. Hsu MC, Liu SH, Wang CW, Hu NY, Wu ESC, Shih YC, et al. JKB-122 is effective, alone or in combination with prednisolone in Con A-induced hepatitis. Eur J Pharmacol. (2017) 812:113–20. doi: 10.1016/j.ejphar.2017.07.012
137. Schett G, Muller F, Taubmann J, Mackensen A, Wang W, Furie RA, et al. Advancements and challenges in CAR T cell therapy in autoimmune diseases. Nat Rev Rheumatol. (2024) 20:531–44. doi: 10.1038/s41584-024-01139-z
138. Schett G, Mackensen A, and Mougiakakos D. CAR T-cell therapy in autoimmune diseases. Lancet. (2023) 402:2034–44. doi: 10.1016/S0140-6736(23)01126-1
Keywords: gut microbiota, autoimmune hepatitis, T cells, interaction, immune microenvironment
Citation: Wu Q, Ge Z, Lv C and He Q (2025) Interacting roles of gut microbiota and T cells in the development of autoimmune hepatitis. Front. Immunol. 16:1584001. doi: 10.3389/fimmu.2025.1584001
Received: 26 February 2025; Accepted: 29 April 2025;
Published: 26 May 2025.
Edited by:
Taylor Sitarik Cohen, AstraZeneca, United StatesReviewed by:
Dinakaran Vasudevan, SKAN Research Trust, IndiaJorge Villacian, AstraZeneca, United Kingdom
Copyright © 2025 Wu, Ge, Lv and He. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Qifeng He, bmptdWhlcWZAMTYzLmNvbQ==; Chengyu Lv, bGN5XzEyMzRAYWxpeXVuLmNvbQ==
†These authors have contributed equally to this work