Keywords autoimmune liver diseases - gut–liver axis - gut microbiota - intestinal dysbiosis
- microbial metabolites - microbial biomarkers
Autoimmune liver diseases (AILDs), including autoimmune hepatitis, primary biliary
cholangitis, and primary sclerosing cholangitis, are chronic conditions caused by
the immune system attacking the liver. Recent studies have revealed that the gut microbiota,
the vast community of microbes living in the intestine, plays a critical role in shaping
liver immunity and disease progression. These microbes influence liver health by affecting
intestinal barrier integrity, modulating immune responses, and producing bioactive
metabolites. Moreover, disturbances in gut microbial composition have been associated
with the onset and severity of AILDs. This review summarizes the latest findings on
how gut–liver communication contributes to autoimmunity and explores emerging microbiota-based
therapies, such as probiotics, prebiotics, and fecal microbiota transplantation. Understanding
the gut–liver axis may pave the way for novel diagnostic and therapeutic approaches
in AILDs.
Autoimmune liver diseases (AILDs) comprise a group of chronic, nonsuppurative inflammatory
disorders of the hepatobiliary system. These conditions are generally classified into
two main categories based on the primary target of autoimmune injury: autoimmune hepatitis
(AIH), which primarily affects hepatocytes, and autoimmune cholangitis, which includes
cholestatic diseases such as primary biliary cholangitis (PBC) and primary sclerosing
cholangitis (PSC). More recently, IgG4-related hepatobiliary diseases have also been
recognized as part of the AILD spectrum.
The etiology of AILDs remains unclear, and current research efforts are focused on
elucidating their pathogenesis through investigations into the influence of the gut
microbiota on hepatobiliary health. Accumulating evidence indicates that the effects
of intestinal microbes on the immune system extend beyond their local colonization
sites.[1 ]
[2 ] The liver, which receives blood from the gastrointestinal tract via the portal vein,
is the first extraintestinal organ exposed to microbial metabolites and endotoxins.
When the intestinal barrier is compromised, potentially pathogenic bacteria and microbial
products can translocate to the liver through the gut–liver axis, initiating a cascade
of pathological immune responses. This axis enables bidirectional communication via
both the portal vein and systemic circulation, thereby regulating hepatic metabolism,
immune homeostasis, and inflammatory signaling.
In the following review, we explore the complex interplay between intestinal microorganisms
and AILDs, with a particular focus on the pivotal role of the microbiota–intestine–hepatobiliary
triangle in disease pathogenesis. Finally, we discuss emerging diagnostic and therapeutic
strategies for AILDs that target this interconnected axis.
Basic Characteristics of Gut Microbiota
Basic Characteristics of Gut Microbiota
The gut microbiota represents a highly complex ecosystem composed of a diverse range
of microorganisms, including bacteria, archaea, viruses, eukaryotes, and fungi ([Table 1 ]). Genomic analyses have revealed that the collective gene pool of the gut microbiome
far exceeds that of the human genome, earning it the designation of the “second human
genome.” Notably, more than 99% of the genes within the human gut microbiota are derived
from bacteria.[3 ] Although fungal biomes represent less than 1% of the total human gut microbiota,[4 ] and the Human Microbiome Project has indicated that the diversity of fungi in the
human intestine is considerably lower than that of bacteria, with notable inter- and
intraindividual variations,[5 ] numerous early investigations have illustrated the role of fungi in disease development
and their significant influence on the host's immune system.[6 ]
[7 ] Wu et al[8 ] suggested core microbiome characteristics that can serve as indicators of health.
By examining metagenomic data from a high-fiber dietary intervention aimed at type
2 diabetes, along with 26 case–control studies across 15 diseases, they discovered
genome pairs consistently linked in coabundance networks during dietary interventions
and disease disruptions. These genomes formed a framework of “two competitive gut
communities” that effectively distinguished cases from controls for various diseases
and forecasted immunotherapy results. The key functional group (C1A) comprises solely
species from the phylum Firmicutes, which are specialized in fermenting fiber and
producing butyrate, thereby benefitting host health. Members of C1A are rich in carbohydrate-active
enzyme (CAZy) genes, enabling them to break down complex plant polysaccharides, as
well as in genes linked to butyrate synthesis, including the but gene. Supporting
this notion, Lau et al further illustrated how specific microbial alterations and
metabolites—including short-chain fatty acids (SCFAs)—affect immune responses and
shape cancer immunotherapy outcomes.[9 ]
Table 1
The primary constituents of the intestinal microbiota
Category
Species
Effection
References
Bacteria
Fungi
Firmicutes
Bacteroidetes
Actinobacteria
Proteobacteria
Fusobacteria
Verrucomicrobia
Candida
Saccharomyces
Penicillium
Aspergillus
Cryptococcus
Malassezia
Cladosporium
Galactomyces
Debaryomyces
Trichosporon
The Lactobacillus and Clostridium are included, as they have been reported to assist with the digestion of complex
carbohydrates, the fermentation of simple sugars, and the metabolism of short-chain
fatty acids. These bacteria are frequently observed in greater abundance in obese
individuals.
The Bacteroides spp. is of particular significance within the colon, occupying an important position
and was observed predominantly in adults like the Firmicutes.
The Bifidobacterium is the primary focus, especially B. longum , is capable of utilizing oligosaccharides present in breast milk as a carbon source,
which might inhibit the growth of other bacteria which enables it to flourish in the
infant gut.
A small proportion of a healthy intestine, but its diversity and ability to respond
to environmental changes make it unique in the intestinal microecology. An increase
in the abundance of it is considered to be one of the characteristics of intestinal
flora imbalance.
Fusobacteria account for a small proportion of healthy intestines, but studies have
found that their abundance increases significantly in some pathological conditions,
such as IBD and colorectal cancer.
The primary representative is Akkermansia muciniphila , a bacterium that inhabits the mucus layer of the intestine, breaks down mucin glycoproteins,
encourages the production of mucus, and supports the intestinal barrier's integrity.
Furthermore, it aids in the production of SCFAs and vitamin B12.
Ten core species of intestinal fungal pathogens have been identified. They are capable
of synthesizing vitamins B and vitamin D, which affects and shapes the host's immune
system. Furthermore, the body is capable of mounting an immune response against pathogens
and developing tolerance to beneficial bacteria by activating fungal-specific pathogen
recognition receptors (PRRs).
[115 ]
[116 ]
[117 ]
[118 ]
[119 ]
[120 ]
[121 ]
[122 ]
[123 ]
[124 ]
[125 ]
[126 ]
[127 ]
Virus
Bacteriophage
Phages play a role in regulating the structure of bacterial communities and promoting
horizontal gene transfer in bacterial populations.
[128 ]
Archaeon
Methanogenic archaea
Methanogenica smithia is closely related to adults and plays an important role in regulating the host's
energy balance.
[129 ]
[130 ]
Notes: This table summarizes the microbial species and the general functions they
perform as described in numerous peer-reviewed research and review articles on the
healthy human gut microbiota. Due to the variations in the designs of the original
studies including the use of clinical cohorts, animal models, and in vitro studies,
we do not have detailed information on sample size, subject characteristics and type
of studies. As a result, this table presents a broad overview of commonly reported
taxa and their functions, not a quantitative meta-analysis.
Mechanisms Linking Intestinal Microecology to Autoimmune Liver Diseases
Mechanisms Linking Intestinal Microecology to Autoimmune Liver Diseases
The detrimental impacts of intestinal dysbiosis on the health of the host have been
recognized for a significant duration. Evidence has shown that intestinal dysbiosis
can provoke autoimmune reactions via several mechanisms, such as helper T cell bias,
paracrine activation, antigen-determining site spreading, cross-reactivity, and the
recognition of microbial antigens by dual T cell receptors.[10 ] Recent studies have indicated that the gut microbiota, as a key environmental factor,
is crucial in the development of AILDs. Presently, it is believed that intestinal
dysbiosis may lead to the onset of AILDs by activating specific signaling pathways,
modifying gut microbiota metabolism, and affecting the regulation of the intestinal
barrier, among other factors.
Intestinal Barrier Function and Microbial Translocation
Intestinal Barrier Function
Bacterial translocation, which refers to the movement of viable bacteria or their
products from the intestinal lumen to mesenteric lymph nodes or other extra-intestinal
organs, has been shown to be commonly linked with hepatobiliary diseases. It is believed
that dysfunction of the intestinal barrier serves as the underlying pathological basis
for this occurrence. Preserving the integrity of the gut barrier is essential to stop
the microbiota from triggering an adaptive immune response in healthy individuals.
This barrier consists of four primary components that occur in succession: biological,
chemical, mechanical, and immunological ([Fig. 1 ]). Research has indicated that the liver, particularly the Kupffer cells, functions
as an “intravascular immune firewall” (via the gut–liver axis of microbial D-lactate)
to restrict the further dissemination of enteric pathogens by capturing and eliminating
bacteria during instances of increased intestinal permeability, a condition commonly
referred to as “leaky gut syndrome.”[11 ] However, this can also render hepatocytes susceptible to immune stimulation from
gut microbiota and their metabolites. The inflammasome pathway is currently recognized
as a key molecular mechanism for sustaining epithelial integrity and intestinal homeostasis.
Among these pathways, the NLRP3 inflammasome enhances inflammation through caspase-1-dependent
activation, which involves the cleavage of pro-inflammatory cytokines interleukin
(IL)-1β and IL-18, in addition to inducing pyroptotic cell death.[12 ] Prior studies have indicated that a leaky gut resulting from NLRP3 deficiency can
lead to elevated levels of toll-like receptor 4 (TLR4) and TLR9 agonists (such as
lipopolysaccharides [LPS]) within the portal vein, consequently prompting hepatocytes
to generate tumor necrosis factor (TNF) and advancing the progression of liver diseases.[13 ] Moreover, scholars propose that additional mechanisms could also contribute to the
development of a leaky gut. The currently investigated mechanisms include physical
trauma, toxins, disruption of tight junctions (TJs), alterations in epithelial stem
cell turnover, and modifications in the consistency of the mucus layer.[14 ]
[15 ]
Fig. 1 The multilayered intestinal barrier: a collaborative defense system maintaining homeostasis.
Created in BioRender. ShiHui, W. (2025)
https://BioRender.com/u73b050
.
The intestinal barrier comprises several layers of defense mechanisms—biological,
chemical, mechanical, and immune—that collaborate to sustain intestinal homeostasis.
The biological component includes gut microbiota, which is essential for shielding
the host from pathogens. Meanwhile, the chemical aspect features mucus, antimicrobial
peptides, and digestive secretions like bile and gastric acid, which deter the attachment
and invasion of pathogens. The mechanical barrier is established by tightly linked
intestinal epithelial cells, bolstered by proteins such as occludin and claudin, which
uphold the integrity of the intestinal mucosa. The immune layer is characterized by
gut-associated lymphoid tissue, which includes Peyer's patches, where Treg cells and
Th17 cells play a role in managing immune responses. Treg cells release TGF-β and
IL-10 to foster immune tolerance and curb excessive inflammation, whereas Th17 cells
generate IL-17 and IL-22 to boost mucosal immunity. Additionally, plasma cells within
the intestinal immune barrier produce sIgA, aiding in the establishment of the chemical
barrier. Immune cells, including macrophages, further modulate the immune response
in the gastrointestinal tract ([Fig. 1 ]).
Microbial Translocation: Gateways to Liver Inflammation through the Gut Barrier
Damage to the intestinal barrier enhances the dynamic translocation and modification
of gut microbiota, thereby affecting the development and progression of liver diseases.
Nakamoto and colleagues found that Klebsiella pneumoniae is present in the gut microbiota of patients with PSC.[16 ] They showed in a mouse model that K. pneumoniae prompts human intestinal epithelial cells to create pores, resulting in the disruption
of the intestinal epithelial barrier and triggering dysbiosis. After introducing the
microbiota from PSC patients into germ-free (GF) mice, K. pneumoniae , Proteus mirabilis , and Enterococcus faecium were successfully cultured and isolated from the mesenteric lymph nodes of these
mice. Together, these three bacterial species contribute to the advancement of hepatobiliary
disorders through the TH17 immune response. Importantly, K.
pneumoniae is pivotal in mediating the impact of intestinal epithelial damage on the activation
of the liver's TH17 response.
In recent years, the issue of liver damage due to fungal translocation has received
considerable attention. β-glucans, the primary components of fungal cell wall polysaccharides,
is a critical pathogen-associated molecular pattern that interacts with various pathogen
recognition receptors in the body, including Dectin-1, complement receptor 3, and
TLR4, which are crucial for initiating innate immune responses.[17 ]
[18 ] Yang et al[19 ] have shown that when the intestinal barrier is compromised, β-glucans, particularly
those produced by Candida parapsilosis , can enter the liver via the bloodstream. In the liver, β-glucans interacts with
the dectin-1 on Kupffer cells, leading to an increase in the expression and secretion
of IL-1β, ultimately causing hepatocyte damage in mice. However, Candida albicans is the predominant species in the human gut mycobiome, rather than C. parapsilosis . Therefore, the compositional changes identified in the article may not substantiate
a causal relationship with liver disease progression. Additionally, the structural
differences in β-glucans derived from various fungi could influence the immunogenicity
of β-glucans, thereby exhibiting different effects on the liver. Although a direct
causal link between β-glucans and AILDs remains to be established, existing mechanistic
evidence suggests a potentially relevant immunological axis: β-glucans binding to
dectin-1 on Kupffer cells activates the Syk/NF-κB signaling cascade, leading to IL-1β
and IL-6 production—cytokines known to promote Th17 cell differentiation. This is
particularly relevant in AIH and PSC, where aberrant Th17 responses have been implicated
in pathogenesis.
Research has indicated that additional fungal metabolites may also be transferred
to the liver through a damaged intestinal barrier. Candidalysin, a peptide toxin secreted
by C. albicans , may directly induce hepatocyte death through various mechanisms, including the activation
of the MAPK/c-Fos/MAP kinase phosphatase 1 signaling pathway.[20 ] Moreover, specific fungi located in the intestinal lining can transmit signals to
the liver through the movement of antigen-reactive cells, including Th17 cells, which
could also trigger abnormal responses in the liver.[21 ] Evidence suggests that patients with PSC and choledochal candidiasis experience
more severe cholangitis, accompanied by elevated C-reactive protein and serum bilirubin
levels, whereas patients without candidiasis do not experience these conditions.[22 ]
Therefore, while the contribution of fungi to the pathogenesis of AILDs remains hypothetical,
these findings collectively support a conceptual framework wherein fungal components—by
activating innate immune receptors and modulating the Th17 axis—may amplify hepatic
autoimmunity in genetically susceptible hosts. Further studies are warranted to validate
this hypothesis in autoimmune settings.
Within-Host Evolution of Gut Pathobionts
Numerous research efforts indicate that distinct strains of microorganisms can adapt
and evolve throughout an individual's life span as a response to the evolution of
their host. Similar to other populations, the evolutionary processes affecting intestinal
microbes predominantly involve migration, mutation, genetic drift, natural selection,
and recombination. Each day, the gut microbiota generates billions of new mutations,
contributing to the diversification and adaptation of symbiotic microbes. Importantly,
findings from mutation accumulation experiments suggest that the majority of these
mutations are detrimental. Current understanding posits that horizontal gene transfer
(HGT), clonal interference, and selective sweeps significantly influence the mutation
process; however, the exact mechanisms remain inadequately comprehended.[23 ]
Yi et al[24 ] performed a series of in vivo experimental evolution studies alongside comparative
genomics research in mice, demonstrating that the intestinal pathogen Enterococcus gallinarum evolved into distinct strains, each with different ecological niches and levels of
pathogenicity due to host-driven evolution. One particular strain has adapted to thrive
in the intestinal lumen and is highly vulnerable to immune responses, whereas another
strain is better equipped for mucosa colonization and possesses the ability to evade
immune detection and clearance. Additionally, the strain adapted to the mucosa shows
an increased capacity for transepithelial transport, which aids its movement to the
mesenteric lymph nodes and liver. This strain exhibits a higher rate of metastasis
and survival in these sites, leading to more severe inflammation in the intestines
and liver. In contrast to typical pathogenic bacteria, which the immune system can
quickly eliminate, these bacteria tend to remain somewhat concealed within organs
for a brief duration, managing to avoid detection by the immune system. Nonetheless,
their prolonged presence may result in pathological consequences, such as the onset
of autoimmune diseases. This situation offers some insight into why certain individuals
harboring potentially harmful bacteria do not develop illnesses, even as their risk
for disease escalates with aging. Beyond E. gallinarum , similar situations might also be observed with other intestinal pathogens. Yaffe
and Relman[25 ] established a technique that utilizes high-throughput chromosome conformation capture
along with a probabilistic noise model to monitor evolution. Leveraging advanced technologies
to investigate the evolution and adaptive transformations of other potential pathogens,
along with their interactions with the host immune system, will enhance our understanding
of the gut microbiota's influence on the host's autoimmune disorders.
Gut Microbial Metabolites
A growing body of evidence indicates a strong correlation between alterations in the
metabolites produced by the gut microbiota and the development of various immune-related
inflammatory diseases. To date, only a restricted variety of microbial metabolites
have been identified; however, these metabolites are highly diverse and their functions
have been the subject of extensive investigation ([Table 2 ]). These metabolites may affect the functioning of various immune cells, such as
T cells, B cells, dendritic cells (DCs), macrophages, among others, implicating them
in the development of immune-mediated inflammatory diseases.
Table 2
Gut flora metabolites and their main effects
Source
Metabolites
Receptors
Functions
Dietary components
SCFAs
GRP41
GRP43
GRP109a
Promote T cells differentiation[131 ]
[132 ]
Regulate cytokines production (e.gIL-10,IL-22)[34 ]
[35 ]
[133 ]
Promote B cells activation and antibody secretion[134 ]
Regulate Neutrophil activity and function[135 ]
Stimulate dendrite elongation in DCs[136 ]
inhibit the activation of BMDCs via suppressing the LPS-mediated expression of co-stimulatory
molecules and the production of cytokines [137 ];
Affect macrophage activity and metabolism, regulate the polarization of macrophages[138 ]
Produced by the host
and modified by gut bacteria
Synthesized de novo by gut microbiota
Secondary
bile acids
LPS
BACCs
FXR
TGR5
TLR4
mTOR
A metabolic homeostat for bile acid, glucose and lipid metabolism in the liver[40 ]
Inhibit the differentiation of TH17 cells, while enhance the differentiation of Treg
cells[139 ]
Modulate NLRP3 Inflammasome activation to regulate macrophage inflammation[140 ]
Promote ISC renewal and drive regeneration in response to injury[141 ]
Control the accumulation of CXCR6 + NKT cells in the liver by regulating the level
of CXCL16 on LSEC[142 ]
Stimulates the formation of NETs[59 ]
Induce the secretion of inflammatory factors (e.g., TNF-α,IL-6, IL-1β) and the production
of GM-CSF[51 ]
Downregulates the expression of the TGF-β pseudo-receptor Bambi[143 ]
Promote anabolism(e.g., protein translation) and inhibit catabolism(e.g.autophagy)[144 ]
Stimulate peripheral blood mononuclear cells, resulting in a significant increase
in IFN-γ production[145 ]
Synergistically reducing the number of CSCs and enhancing HCC chemotherapy sensitivity[146 ]
Abbreviations: BACCs, branched-chain amino acids; BMDCs, bone-marrow-derived DCs;
CAR, constitutive androstane receptor; CSCs, cancer stem cells; FXR, farnesoid X receptor;
GM-CSF, granulocyte–macrophage colony-stimulating factor; HCC, hepatocellular carcinoma;
IFN-γ, interferon-γ; ILCs, innate lymphoid cells; ISC, intestinal stem cell; LSEC,
liver sinusoidal endothelial cells; NETs, neutrophil extracellular traps; PXR, pregnane
X receptor; TGR5, Takeda G protein-coupled receptor 5; VDR, vitamin D receptor.
Short-Chain Fatty Acids
Carbohydrates that are not fully digested may undergo fermentation by gut microbiota,
particularly by species such as Faecalibacterium prausnitzii , Blautia , Bifidobacterium , and Bacteroidetes , leading to the production of SCFAs ([Fig. 2 ]). Various SCFAs are distributed in distinct regions of the intestine: acetate and
propionate can be found in both the small and large intestines, whereas butyrate is
predominantly located in the colon and cecum. SCFAs play several roles, such as fostering
the production of antimicrobial substances, regulating the turnover of epithelial
cells, and preserving the integrity of the epithelial barrier. For instance, butyrate
enhances the expression of TJs and stabilizes the levels of hypoxia-inducible factor,
thereby strengthening the intestinal barrier.[26 ]
[27 ] Additionally, SCFAs influence a variety of immune cells—including T cells, B cells,
and macrophages—via the G protein-coupled receptors (GPCRs) signaling pathway.[28 ] Furthermore, SCFAs provide a protective effect in the intestine by hindering the
establishment of pathogenic bacteria, such as C. albicans , through the acidification of the gut environment.[29 ] These fatty acids also exhibit immunomodulatory effects on organs outside the intestine.
Among them, butyrate is particularly influential in the gut–liver axis, where it can
trigger the production of IL-18 by Kupffer cells and hepatocytes in a manner dependent
on the GPR109A receptor, and IL-18 has been shown to be linked with mitochondrial
function and the maturation of natural killer cells in the liver.[30 ] The proliferation and growth of liver cells are reliant on the biosynthesis of membrane
phospholipids, and the metabolites from gut microbiota travel through the gut–liver
axis, significantly aiding in lipid biosynthesis.
Yin et al[31 ] therefore investigated the role of gut microbiota in promoting liver regeneration
through the biosynthesis of hepatic membrane phospholipids, with a particular focus
on the link between SCFAs in gut microbiota and liver lipid metabolism. They used
C57Bl/6J wild-type mice to perform a 70% partial hepatectomy (PHx) experiment, with
some mice treated with broad-spectrum antibiotics before surgery to induce intestinal
dysbosis, while using GF mice and oligo-mouse-microbiota (OMM) for comparison. It
was found that the antibiotic-treated mice showed a decrease in α diversity of the
gut microbiota, an increase in Proteobacteria in particular, and a decrease in SCFAs-producing
flora, accompanied by a decrease in the expression of stearoyl-CoA desaturase 1 (SCD1),
which catalyzes the formation of monounsaturated fatty acids. SCD1 was found to be
SCFAs-inducibly expressed in vitro and in vivo and is necessary for human liver cancer
cell proliferation. In addition, SCD1 was also found to be associated with liver regeneration
in human patient tissue biopsies. These mice had delayed liver regeneration and impaired
hepatocyte proliferation after hepatectomy. In contrast, GF mice had impaired liver
regeneration after hepatectomy, whereas the OMM partially recovered their liver regeneration
capacity. This study for the first time revealed that SCFAs have the function of promoting
liver regeneration, providing new ideas for further research on the treatment of liver
disease with the gut microbiota.
SCFAs can enter cells through three main pathways: simple diffusion, carrier-mediated
transport involving the transporters SMCT1 (SLC5A8) and MCT1 (SLC16A1), and activation
of GPCRs. These SCFAs have the ability to modulate immune cell functions by inhibiting
histone deacetylase (HDAC) and/or activating GPRs such as GPR41, GPR43, and GPR109A.[32 ]
[33 ] Both GPR41 and GPR43 interact with acetate, propionate, and butyrate, whereas GPR109A
is primarily activated by butyrate. SCFAs can influence the differentiation of naive
T cells and suppress the release of proinflammatory cytokines through HDAC inhibition,
potentially prompting intestinal B cells to generate IgA. The stimulation of GPR41
has been shown to promote the production of IL-22 by CD4+ T cells and innate lymphoid
cells. In contrast, the activation of GPR43 leads to the production of IL-10 by Th1
cells and alters the migration and activities of neutrophils during inflammatory responses.[34 ]
[35 ]
[36 ] Regarding GPR109A activation, research indicates that it inhibits cyclic adenosine
monophosphate production and encourages colonic DCs and macrophages to release IL-10[Fig.2 ]
[37 ]
Fig. 2 Cellular entry and immunoregulatory mechanisms of short-chain fatty acids. Created in BioRender. ShiHui, W. (2025)
https://BioRender.com/i21t449
.
Secondary Bile Acids
Primary bile acids (BAs) are synthesized primarily in the liver and subsequently excreted
into the intestines. Intestinal bacteria facilitate the conversion of primary BAs
into secondary BAs, including deoxycholic acid (DCA) and lithocholic acid (LCA), through
a series of enzymatic processes, including dehydrogenation, 7α-dehydroxylation, and
exoisomerisation. As well as promoting digestion and having an antibacterial effect,
secondary BAs also play an important role in immune regulation.[38 ] To date, the BA receptors that are well documented and closely associated with hepatobiliary
diseases are primarily farnesoid X receptor (FXR) and TGR5.
FXR serves as a transcription factor whose activation is crucial for restoring homeostasis
in both the intestinal and gut vascular barriers.[39 ] Furthermore, it is believed that processes such as energy regulation, autophagy,
inflammatory responses, and fibrosis are significantly associated with FXR.[40 ] Similarly, it regulates the transcription of key BA synthesis enzyme genes Cyp7a1
and Cyp8b1 through the endocrine pathway of fibroblast growth factor 15/19 in the
ileum, in response to postprandial or abnormal gut–liver BA flow.[41 ] Leveraging these functions, researchers have found that numerous medications can
mitigate different models of liver damage through the activation of the FXR signaling
pathway.[42 ]
[43 ]
[44 ] In recent years, there has been a growing interest in the use of FXR agonists for
the treatment of PBC. Initial investigations indicated that LCA can enhance the levels
of apoptosis-related proteins, including cleaved poly (ADP-ribose) polymerase (PARP)
and cleaved caspase-3, leading to hepatocyte apoptosis in murine models.[45 ] Building on this, Lu et al[46 ] noted that in mice with cholestatic liver injury induced by LCA and pretreated with
obeticholic acid (OCA), OCA not only alleviated cholestasis by downregulating BA export
transporters but also reduced LCA-induced hepatocyte apoptosis by lowering the levels
of apoptosis-related proteins such as cleaved caspase-3, cleaved caspase-8, and cleaved
PARP. This finding offers fresh insight into the role of FXR agonists in combating
cholestatic liver injury. In addition, Tang et al innovatively revealed using a mouse
model that hepatocyte FGF4 might be a direct downstream target of hepatic FXR, which
signals to transcriptionally control Cyp7a1 and Cyp8b1 expression through a heretofore
uncharacterized intracellular FGFR4-LRH-1 signaling node within the liver.[47 ] However, these findings necessitate further validation to determine whether the
beneficial effects of FGF4 and related pathways on BA metabolism observed in mice
can be replicated in humans and their safety.
TGR5 functions as a receptor for bile salts that is coupled to G-proteins. Initial
research indicated that TGR5 not only induces nitric oxide production in rat liver
endothelial cells and decreases the cytokine gene induction in LPS-activated rat Kupffer
cells by inhibiting the NF-κB signaling pathway, thus producing anti-inflammatory
effects, but it also safeguards the liver from BAs overload consequences following
PHx by regulating bile hydrophobicity and cytokine release while facilitating liver
regeneration.[48 ] Given this evidence, it raises the question of whether changes in TGR5 expression
within bile duct epithelial cells (BECs) correlate with the development of cholestatic
diseases. Reich et al[49 ] found that patients diagnosed with PSC and Abcb4 −/− mice (which serve as a common
model for sclerosing cholangitis) exhibited decreased expression of TGR5 in BECs.
This reduction was noted early in the disease trajectory, showed specificity to cell
types, and was particular to PSC as well as Abcb4 −/− BECs and the extrahepatic bile
ducts of Abcb4 −/− mice; it was not encountered in other hepatic conditions such as
PBC, nonalcoholic steatohepatitis, drug-induced liver injury, or viral hepatitis.
Additional studies have shown that the reduction of TGR5 correlates with intensified
damage in BECs. Mice lacking TGR5 demonstrate greater bile duct injury, while enhancing
TGR5 expression in Abcb4 −/− models can mitigate PSC and enhance BEC activation along
with the inflammatory phenotype.
Lipopolysaccharide
Endogenous LPS have been recognized as a contributing factor in the exacerbation of
liver injury across various models. Under typical physiological circumstances, the
intestinal barrier effectively inhibits LPS from penetrating the bloodstream. Nevertheless,
factors such as systemic inflammation, intestinal dysbiosis, or disruption of TJs
compromise this barrier's integrity, increasing permeability and allowing LPS to enter
circulation. Once in the bloodstream, LPS interacts with TLR4 through CD 14 to trigger
the host immune response, which is a crucial mediator of both adaptive and innate
immune reactions to LPS. This activation stimulates the release of numerous inflammatory
mediators, including TNF-α,as well as the production of granulocyte–macrophage colony-stimulating
factor, ultimately resulting in irreversible liver damage.[50 ]
[51 ] Significant evidence underscores the importance of TLR4 signaling, particularly
the TLR4/NF-κB pathway, in various liver pathologies, making this axis a valuable
target for numerous pharmacological interventions aimed at mitigating liver damage.[52 ]
[53 ]
[54 ] In addition, previous studies have indicated that both LPS and soluble CD14 levels
are elevated in AILDs, with soluble CD14 being linked to a poorer prognosis.[55 ] Furthermore, research by Zhang et al[56 ] revealed that the stimulator of interferon genes (STING) signaling pathway in Kupffer
cells plays a role in LPS-induced liver injury. Their study demonstrated that LPS
stimulation increased mitochondrial reactive oxygen species (ROS) production dependent
on dynamin-related protein 1, which facilitated the release of mtDNA into the cytoplasm
and subsequent activation of STING signaling in Kupffer cells. Additionally, their
experiments confirmed that the absence of STING afforded protection to liver function,
reduced the systemic inflammatory response, and decreased mortality in LPS-treated
mice, while the administration of a STING agonist yielded the opposite outcome. By
modulating the activation of STING, it is possible to balance the immune response,
reduce inflammation, and promote tissue repair. However, further research is needed
to clarify the precise mechanisms of STING signaling in AILDs and to develop targeted
therapies that can selectively modulate its activity without exacerbating liver injury.
The integration of STING regulation with other immunoregulatory pathways, such as
the IL-33/ST2 axis[57 ] and autophagy,[58 ] may provide a comprehensive approach for treating AILDs and improving patient prognosis.
In recent years, research have indicated that neutrophils represent a significant
target for intestinal-derived LPS-induced liver damage. Liu et al[59 ] attempted to reveal one of the possible mechanisms in their study. They found that
in patients with chronic alcoholism, intestinal bacteria and LPS can stimulate the
formation of neutrophil extracellular traps (related to tumor immunity in hepatocellular
carcinoma [HCC]) through the TLR4 pathway to promote alcoholic liver fibrosis and
play a significant role in the development of alcoholic liver cirrhosis and subsequent
HCC.
Clinical and Laboratory Evidence for the Association between the Gut Microbiota and
Autoimmune Liver Diseases
Clinical and Laboratory Evidence for the Association between the Gut Microbiota and
Autoimmune Liver Diseases
Autoimmune Hepatitis
The current diagnosis of AIH relies on the detection of circulating autoantibodies,
increased levels of IgG and gamma globulins, and specific histological alterations.
The standard initial treatment approach is the administration of corticosteroids,
either alone or in combination with azathioprine. Nevertheless, patients who either
do not receive a prompt diagnosis or fail to respond to first-line therapies often
experience a notable rise in liver-related morbidity and mortality.[60 ] Recent studies have investigated the role of gut microbiota in the pathogenesis
of AIH, offering new prospects for its early diagnosis and treatment. In their earlier
work, Yuksel et al[61 ] created an innovative AIH mouse model utilizing HLA-DR3 transgenic mice in a nonobese
diabetic mouse background, where intestinal dysbiosis was observed in cases of experimental
AIH. This investigation revealed an intensified Th1 immune response alongside a reduced
frequency of regulatory T cells in the livers of immunized mice. Additionally, research
conducted by Manfredo Vieira et al[62 ] identified a pathogenic bacterium, E. gallinarum , within the gut flora. When the intestinal barrier is breached, these bacteria may
move into the systemic circulation of hosts susceptible to autoimmune conditions,
potentially contributing to the development of autoimmunity. It is conceivable that
these translocated bacteria not only affect the differentiation of helper T cells
but may also exert direct effects on colonized tissues, including the liver. Enterococcus gallinarum has been detected in liver biopsies from patients diagnosed with AIH and cirrhosis,
conditions characterized by significant intestinal barrier damage. The translocation
of E. gallinarum stimulates the activation of the AhR signaling pathway in the liver, subsequently
leading to autoimmune liver injury. This process occurs through the induction of pathogenic
TH17 cells, the generation of RNA and double-stranded DNA autoantibodies, as well
as an inflammatory response.
Primary Biliary Cholangitis
PBC is marked by nonsuppurative granulomatous and lymphocytic inflammation affecting
the small bile ducts within the liver. The challenge of early diagnosis arises from
the initial symptoms being masked by PBC and the absence of specific biomarkers. Numerous
studies focusing on the compositional analysis of gut microbiota indicate that changes
in the community composition of gut microbiota correlate with the progression of PBC.
Lv et al[63 ] employed techniques such as 16S rRNA gene metagenomic sequencing, ultra-performance
liquid chromatography-tandem mass spectrometry for small molecule detection, and liquid
chip assays for serum cytokines to investigate alterations in the gut microbiota of
early-stage PBC patients who did not have other significant health issues. Their findings
revealed that, in comparison to healthy subjects, PBC patients exhibited a reduced
relative abundance of potentially beneficial bacteria (such as Acidobacteria , Lachnobacterium sp., and Bacteroides eggerthii ), while there was an increase in the presence of pathogenic bacteria (including Actinobacillus pleuropneumoniae , Klebsiella , Neisseria , etc.). Furthermore, they noted that most of the modified intestinal bacteria were
linked to liver damage indicators, serum inflammatory cytokines, and metabolic irregularities.
Importantly, alterations in the metabolite profiles of blood, urine, and feces were
observed in PBC patients, suggesting that changes in intestinal bacteria could indirectly
affect metabolism and cholestasis. In a study conducted by Tang et al,[64 ] a comparative analysis was performed on fecal samples from 79 untreated PBC patients
and 114 healthy controls. This research revealed a decrease in the populations of
Bacteroides , Faecalibacterium , Sutterella , and Oscillospira spp., alongside an increase in Veillonella , Clostridium , Lactobacillus , Streptococcus , Pseudomonas , Klebsiella , and an unidentified genus within the Enterobacteriaceae family (Enterobacteriaceae ) among PBC patients.
The treatment of PBC has traditionally relied on therapies centered around BAs. In
a recent study, Li et al[65 ] demonstrated that the administration of colestipol resulted in a significant decrease
in BA levels in the bloodstream, altered the composition of BAs, and lowered the hydrophobic
index of these acids. Furthermore, the intervention brought about changes in the intestinal
microbiota, notably an increase in bacteria that produce SCFAs among patients who
showed a more favorable response to the treatment, alongside a rise in the levels
of conditionally pathogenic K. pneumoniae in those with poorer responses. These microbiota changes correlated with enhancements
in the clinical symptoms of the patients, indicating that BA sequestrants may benefit
individuals with PBC by influencing the intestinal microbiome and its metabolic products.
Collectively, these findings underscore the potential significance of intestinal microorganisms
in both the diagnosis and management of PBC.
Primary Sclerosing Cholangitis
The long-term prognosis for PSC patients is unfavorable, with up to 40% of patients
requiring liver transplantation in the later stages and 20 to 30% developing bile
duct cancer.[66 ] Additionally, there is a considerable risk of developing colorectal cancer.[67 ] Although ursodeoxycholic acid (UDCA) has been extensively utilized in the treatment
of chronic cholestatic liver diseases, with demonstrated efficacy in improving biochemical
markers of cholestasis, its ability to prevent the progression of PSC remains limited
in most patients. Most patients with PSC also have inflammatory bowel disease (IBD),
which makes PSC a model disease for studying the gut–liver axis, suggesting that intestinal
microorganisms may contribute to the occurrence of PSC.[68 ] In a study conducted by Tedesco et al,[69 ] a comparison was made between a multidrug resistance gene 2 knockout (Mdr2 −/− )
mouse model and an FVB/NJ mouse model. The results demonstrated that the Mdr2 −/−
mice exhibited increased intestinal permeability, which resulted in the translocation
of intestinal flora to the liver, particularly Lactobacillus gasseri . Furthermore, the researchers discovered that γδ T cells extracted from the livers
of patients with PSC were capable of producing IL-17, whereas cells from hepatitis
C patients were not. This indicates that the activation of γδ TCR+ cells in the liver
and the production of IL-17 mediated by translocated L. gasseri may be a contributing factor in the development of cholestatic liver diseases such
as PSC. At present, researchers are directing their attention to the intestinal fungal
community. Lemoinne et al[70 ] discovered that the intestinal fungal flora of PSC patients is dysbiotic, exhibiting
a relative increase in biodiversity and alterations in community composition. Additionally,
an increase in the abundance of Exophiala spp. and a decrease in the abundance of Saccharomyces cerevisiae were observed. Exophiala spp. has been demonstrated to be capable of causing infections in immunocompromised
hosts, whereas S. cerevisiae has anti-inflammatory properties and has been shown to reduce the recurrence rate
in patients with IBD.[71 ] Based on these findings, Rühlemann et al[72 ] observed an increased abundance of Candida and Clostridium butyricum in German PSC patients, whereas Exophiala spp. was not detected in the German samples. Further research is necessary to ascertain
if these inconsistencies stem from variations in methodology, including the choice
of primer sets, tools for data analysis, and the depth of sampling. It can be concluded
that individuals with PSC and biliary candidiasis experience more severe cholangitis,
alongside elevated levels of C-reactive protein and serum bilirubin, in contrast to
those without a Candida infection.[22 ] Moreover, biliary candidiasis is linked to decreased survival rates among patients
suffering from PSC.[73 ]
IgG4-Related Hepatobiliary Diseases
IgG4-related hepatobiliary disorders are immune-mediated conditions that are closely
associated with IgG4-RD. This group primarily encompasses IgG4-related sclerosing
cholangitis (IgG4-SC), IgG4-related hepatopathy, and IgG4-related cholangitis. The
subtle onset, gradual progression, and infrequent emergence of severe clinical symptoms
or acute organ failure in these conditions often result in many patients having some
level of irreversible organ dysfunction by the time they receive a diagnosis.[74 ]
While extensive research has been conducted on intestinal dysbiosis in AIH, PBC, and
PSC, the involvement of gut microbiota in IgG4-related hepatobiliary diseases remains
uncertain. Liu et al[75 ] compared the stool composition of patients diagnosed with IgG4-SC to that of those
with PSC and healthy controls. Their integrated multiomics analyses, incorporating
16S rRNA gene amplicon sequencing along with untargeted metabolomics, revealed key
characteristics of the fecal microbiome in IgG4-SC, including reduced intraspecific
diversity and altered microbiome structure. These differences were significant when
comparing IgG4-SC patients to both PSC and healthy controls. Moreover, the research
identified 58 metabolites exhibiting varying abundances in IgG4-SC in relation to
healthy controls. Notably, specific intestinal microbial metabolites, particularly
succinic acid and L-palmitoylcarnitine, showed significant elevation in patients with
IgG4-SC. Prior investigations have indicated that alterations in succinic acid signaling
are linked to the activation and functionality of immune cells,[76 ] whereas L-palmitoylcarnitine is also believed to influence inflammatory activation.[77 ] Importantly, several studies have indicated that microbiome-derived succinic acid
can stimulate type II immune responses,[78 ] which aligns with the observed type II immunity skewing in IgG4-SC. In conclusion,
despite certain limitations of this study (including being a single-center study with
a small sample size), it provides evidence suggesting that the alterations in intestinal
microbiota and metabolic processes in IgG4-SC may play a role in the disease's inflammatory
mechanisms.
The Potential Value of Gut Microbiota in the Diagnosis and Treatment of Autoimmune
Liver Diseases
The Potential Value of Gut Microbiota in the Diagnosis and Treatment of Autoimmune
Liver Diseases
The Possibility of Specific Flora as Diagnostic Markers
Accumulating evidence suggests that gut microbiota profiles are associated with a
variety of diseases, including HCC,[79 ] IBD,[80 ] colorectal cancer, and Parkinson's disease. Similarly, microbial alterations observed
in AILDs have been proposed as potential early indicators, although their diagnostic
value remains to be validated ([Table 3 ]).
Table 3
Altered gut microbiota associated with autoimmune liver diseases
Models
Disease
Specific flora
Patients (N )
Controls
References
Human
Human
Human
Human
Human
Human/Mice
Human
Human
AIH
AIH
PBC
PBC
PBC
PSC
PSC
IgG4-SC
Enrichment of Veillonella , Klebsiella , Streptococcus , and
Lactobacillus;
Depletion of e.g., Clostridiales , Oscillospira , Coprococcus
Depletion of Bifidobacterium , Faecalibacterium ;
Enrichment of Veillonella , Lactobacillus , Streptococcus
Enrichment of Enterobacteriaceae .g, Veillonella , Klebsiella ;
Depletion of Faecalibacterium and Sutterella , both increased after UDCA treatment
Enrichment of Lactobacillus , Enterococcus ;
Depletion of Faecalibacterium , Anaerostipes
Enrichment of e.g., Streptococcus , Veillonella , Haemophilus
Enrichment of Klebsiella pneumoniae ,
Proteus mirabilis , Enterococcus faecium
Enrichment of Enterococcus , Fusobacterium , Lactobacillus ;
Depletion of Faecalibacterium , Ruminococcaceae , Roseburia
Enrichment of Streptococcus ;
Depletion of Blautia , Lachnospiraceae ND3007
119
72
79
76
60
17 PSC
GF-mice
23 PSC + UC
11 PSC only
13 IgG4-SC
15 PSC
132 HC
95 HC
99 PBC
81 UC
114 HC
23 HC
60 HC
13 HC
23 UC
22 CD
68 HC
15 HC
[81 ]
[85 ]
[64 ]
[86 ]
[63 ]
[16 ]
[88 ]
[75 ]
Yiran et al[81 ] conducted an analysis of the gut microbiota in patients with AIH compared with healthy
individuals. The results indicated that, in comparison to the control group, the abundance
of several bacterial species was significantly reduced in AIH patients, including
Clostridiales , RF 39, Ruminococcaceae , Rikenellaceae , Oscillospira , Parabacteroides , and Coprococcus . Conversely, the gut microbiota of AIH patients exhibited an increased abundance
of Veillonella , Klebsiella , Streptococcus , and Lactobacillus compared with the control group. Interestingly, earlier studies have also reported
that altered abundances of Veillonella and Lactobacillus in patients with PBC and PSC, suggesting a potential association with disease-related
microbial dysbiosis.[82 ]
[83 ]
[84 ] Of note, Veillonella dispar positively correlated with serum level of AST and liver inflammation was the most
strongly disease-associated taxa in this study. Based on the above, they subsequently
applied multivariable stepwise logistic regression analysis to determine the most
obvious classified flora of AIH and controls, the authors ultimately suggested that
a microbial signature comprising Veillonella , Lactobacillus , Oscillospira , and Clostridiales exhibited promising discriminatory power for distinguishing AIH patients from controls
within their study cohort. However, it is important to emphasize that the findings
are based on correlational analysis and do not imply causality. In addition, the only
study to date that directly compares different ALIDs was conducted by Liwinski et al,[85 ] they sequenced stool samples from AIH patients and control groups (including healthy
controls, PBC controls, and ulcerative colitis (UC) controls). Similarly, they also
observed significant changes in the relative abundance of Veillonella and Lactobacillus , as well as Bifidobacterium , and Faecalibacterium in the AIH group, where a significant reduction in the number of Bifidobacterium was observed in patients, which was associated with the AIH Bifidobacterium was significantly reduced in the patients, which was related to the degree of AIH
remission.
In the aforementioned study by Lv et al,[63 ] receiver operating characteristic curve analysis suggested that Streptococcus sp. and Veillonella sp. may may have potential discriminatory value in differentiating patients with
PBC from healthy controls. Furthermore, Tang et al[64 ] reported that several genera, including Enterobacteriaceae (unclassified genus), Klebsiella , Veillonella , and Streptococcus , were significantly enriched in PBC patients, with Enterobacteriaceae showing the strongest association. In contrast, Faecalibacterium —a well-known butyrate-producing genus with anti-inflammatory properties—was significantly
reduced in PBC compared with healthy controls. These findings suggest a microbial
imbalance in PBC, characterized by an overrepresentation of potentially pathogenic
bacteria and depletion of beneficial taxa such as Faecalibacterium , although causality has yet to be established. Building on these foundation, Furukawa et al[86 ] analyzed fecal samples from 76 Japanese patients with PBC, of whom 73 had been treated
with UDCA for more than 1 year. Using 16S rRNA gene sequencing, they compared the
gut microbiota profiles of PBC patients with those of healthy controls. The study
revealed that, compared with healthy individuals, the relative abundance of the order
Clostridiales , including butyrate-producing genera such as Faecalibacterium , Roseburia , and Anaerostipes —was significantly reduced in PBC patients. In contrast, members of the order Lactobacillales , including Lactobacillus , Enterococcus , and Streptococcus , were markedly enriched. Notably, the abundance of Faecalibacterium was significantly lower in UDCA nonresponders compared with responders, as defined
by the Nara criteria, suggesting a potential association between reduced Faecalibacterium levels and poor treatment response. Additionally, Zhang et al[87 ] conducted a two-sample bidirectional Mendelian randomization study to assess the
causal relationship between gut microbiota composition and PBC. The analysis revealed
that a higher abundance of certain taxa, such as Ruminococcaceae, Peptostreptococcaceae,
Christensenellaceae R7 group, Anaerofilum, Ruminococcaceae UCG-013, and Holdemania,
was causally associated with a lower risk of PBC. Among these, Ruminococcaceae is
known to include butyrate-producing genera, suggesting a potential protective role
through maintaining intestinal barrier integrity and reducing systemic immune activation.
Conversely, taxa such as Selenomonadales, Bifidobacteriales, Lachnospiraceae UCG-004,
Oscillospira, and Eubacterium nodatum group were positively associated with increased
PBC risk. These findings highlight specific microbial taxa that may play divergent
roles in the pathogenesis of PBC and warrant further mechanistic investigation.
Sabino et al[88 ] demonstrated that intestinal dysbiosis in patients with PSC is independent of coexisting
IBD. After controlling for potential confounders including antibiotic and probiotic
use, UDCA treatment, liver cirrhosis, and liver transplantation, the genera Enterococcus , Lactobacillus , and Fusobacterium were significantly enriched in PSC patients regardless of IBD status or treatment.
These findings suggest that PSC is characterized by a distinct gut microbial signature
that is not secondary to IBD or medication exposure. As previously stated, Nakamoto
et al[16 ] identified an enrichment of K. pneumoniae , P. mirabilis , and E. faecium in the gut microbiota of PSC patients. These bacteria were shown to impair intestinal
barrier integrity and induce hepatic Th17 responses in gnotobiotic mouse models, supporting
their potential pathogenic role in PSC progression. Among them, K. pneumoniae exhibited the strongest capacity to trigger Th17-mediated inflammation. These findings
suggest that tracking the abundance of these pathobionts, particularly K. pneumoniae , may provide insights into PSC pathogenesis and hold potential value for future diagnostic
strategies.
In the study by Liu et al,[75 ] although a substantial overlap in gut microbial alterations was observed between
patients with IgG4-SC and PSC, three taxa exhibited relatively higher specificity
in IgG4-SC: enrichment of Streptococcus and depletion of Blautia and Lachnospiraceae _ND3007_group. These distinct microbial signatures may provide potential clues for
differentiating IgG4-SC from PSC based on microbiome analysis.
It remains unclear whether the observed reduction in gut microbiota diversity in patients
with AILDs is primarily driven by the disease process itself or by therapeutic interventions,
such as UDCA, antibiotics, or immunosuppressants. Most existing studies rely on 16S
rRNA gene sequencing, which, although useful for community-level profiling, lacks
the resolution to distinguish microbial taxa at the species or strain level. To address
these limitations, future research should incorporate higher-resolution approaches
such as whole-genome shotgun metagenomics,[55 ] metatranscriptomics, and culture-based techniques, which allow more accurate identification
of functionally relevant strains. Additionally, a more comprehensive stratification
of disease status is essential. In particular, IBD, commonly coexisting with PSC,
represents a major confounding factor in gut microbiome analyses. The influence of
IBD activity and its treatments on microbial diversity and metabolic output should
be carefully controlled in future studies.
Application of Probiotics
The administration of particular probiotics has the potential to restore the equilibrium
of the gut microbiota and mitigate the inflammatory response in autoimmune disorders
([Fig. 3 ]). For example, supplementation with Bifidobacterium has been demonstrated to balance
the Treg/Th17/Th1 ratio, thereby preventing excessive activation of CD4+ lymphocytes.[89 ] Zhang et al[90 ] found that Bifidobacterium animalis ssp. lactis 420 (B420) can significantly alleviate s100-induced experimental AIH
(EAH) by regulating the RIP3 signaling pathway and cytokine profile of hepatic macrophages
to inhibit Th17 cell differentiation. B420 also strengthens the intestinal barrier
by upregulating TJ proteins. In addition, B420 alters the composition of the mouse
gut microbiota, which is characterized by a decrease in Bacteroides , Ruminococcus , and an increase in Lactobacillus , Alistipes , and Rikenella at the genus level. Ma et al[91 ] selected 50 patients with active AIH who had not received any drug intervention
and established an experimental mouse model of AIH to analyze the effect of prednisone
combined with Lactobacillus treatment. The study found that compared with patients treated with prednisone alone,
those treated with Lactobacillus-prednisone had significantly higher levels of Bacteroides fragilis , Clostridium , Clostridium
leptum , and Bifidobacterium in their stools and a more significant reduction in the relative levels of alanine
aminotransferase (ALT), aspartate aminotransferase (AST), smooth muscle antibody,
antinuclear antibody, IgG, and so on in the serum of patients. Further studies in
the EAH mouse model showed that Lactobacillus
reuteri improved the therapeutic effect of prednisone, and the two may regulate the Tfh response
in EAH mice through the TLR4/MyD88/NF-κB pathway. The findings indicate that combining
Lactobacillus reuteri supplementation with prednisone may offer therapeutic benefits in managing AIH.
Fig. 3 Multifunctional mechanisms of probiotics in promoting host health. Created in BioRender. ShiHui, W. (2025)
https://BioRender.com/t81t864
.
Furthermore, Wu et al[92 ] conducted an animal experiment and a randomized controlled clinical trial to evaluate
the effect of Lactobacillus
acidophilus on patients with cholestasis. The results of the animal experiment showed that L. acidophilus significantly alleviated liver damage in mice by activating intestinal FXR signaling
to inhibit the synthesis of BAs in the liver, increasing unbound BAs by accumulating
bile salt hydrolase, and promoting the excretion of BAs. In addition, a randomized
controlled clinical trial found that L. acidophilus supplementation can promote recovery of liver function in patients, providing another
treatment option for cholestatic liver disease. A study of patients with PBC also
revealed that probiotic supplementation, including Lactobacillus , may assist in reducing symptoms and improving liver function indicators. Moreover,
it has been demonstrated that metabolites of probiotics, such as indolepropionate,
can mitigate the immune response of the liver by reducing endotoxin levels.[93 ]
Akkermansia muciniphila is regarded as a “new generation of probiotics” that exerts a beneficial influence
in a multitude of ailments, including diabetes, obesity, cancer, and metabolic syndrome.[94 ] Xia et al[95 ] observed in a mouse model of acetaminophen-induced liver injury that A. muciniphila alleviated oxidative stress in the liver by regulating the reduced glutathione/oxidized
glutathione (GSH/GSSG) balance and enhancing superoxide dismutase activity. Additionally,
it has been demonstrated to diminish the generation of proinflammatory cytokines,
inhibit the infiltration of macrophages and neutrophils in the liver, and mitigate
hepatocyte apoptosis by activating the phosphoinositide 3-kinase (PI3K)/Akt signaling
pathway. Nevertheless, the majority of the research is based on animal models, and
it remains to be seen whether the same effect can be observed in liver damage induced
by other factors and whether the results can be replicated in humans. Further animal
experiments and clinical studies are necessary to verify these hypotheses. Moreover,
the utilization of its regulation of the gut microbiota and its metabolites and the
enhancement of intestinal barrier integrity to treat AILDs-related increased intestinal
permeability represents a further potential avenue of investigation.
As discussed in earlier sections, several genera traditionally regarded as probiotics,
such as Lactobacillus , Bifidobacterium , and Enterococcus , have been reported to be enriched in patients with AILDs. This raises potential
concerns about their indiscriminate use in therapeutic settings. Although specific
probiotic strains have demonstrated immunomodulatory and hepatoprotective effects
in animal models or pilot clinical trials, their applicability to patients with preexisting
dysbiosis requires careful consideration. Notably, emerging evidence also points to
potential risks of probiotic-related bacteremia,[96 ] and that probiotic intake may affect the evolution of resident intestinal microorganisms,
potentially altering their genomic content, metabolic functions, or resistance patterns
over time.[97 ] Moreover, emerging evidence from the broader probiotic safety literature indicates
that probiotic strains can harbor antibiotic resistance genes and act as reservoirs
for HGT.[98 ]
[99 ]
[100 ] Common probiotic species such as Lactobacillus and Bifidobacterium have been found to carry mobile resistance determinants like tet, erm, and van genes.
Some of these genes are plasmid-borne and have been shown to be transferable to pathogenic
or commensal bacteria in both in vitro and in vivo studies. These findings underscore
the need for strain-level genomic screening and resistome profiling prior to the use
of probiotics as adjunctive therapy in AILDs, particularly in patients with compromised
gut barriers or immunological dysfunction.
Probiotics can reduce the level of endotoxin in the body by inhibiting the production
of endotoxin by lysogenic bacteria, while inhibiting apoptosis through the PI3K–AKT
pathway. In BA metabolism, probiotics can reduce BA synthesis through the FXR pathway,
increase BSH activity to increase unconjugated BAs and promote their excretion. Furthermore,
probiotics display anti-inflammatory properties by enhancing the expression of TJ
proteins, strengthening the intestinal barrier's integrity, decreasing the synthesis
of proinflammatory substances, and elevating the concentrations of anti-inflammatory
factors. Probiotics also have immunomodulatory effects, affecting the activity of
various immune cells, including neutrophils, Th17 cells, Th1 cells, Treg cells, and
macrophages. Probiotics can regulate the composition of the intestinal microbiota,
maintain intestinal health, and at the same time inhibit the harmful effects of oxidative
stress by increasing the expression of superoxide dismutase and increasing the GSH/GSSG
ratio ([Fig. 2 ]).
Fecal Microbiota Transplantation
Transferring fecal microbiota from a healthy donor into a patient's intestine is becoming
a promising therapy for autoimmune diseases linked to microbial factors. Fecal microbiota
transplantation (FMT) is most commonly associated with the treatment of recurrent
Clostridioides difficile infection.[101 ] Furthermore, studies have indicated that, in addition to gastrointestinal disorders
such as IBD and Helicobacter pylori infection,[102 ] seemingly nongastrointestinal disorders may also serve as potential therapeutic
targets for FMT, for example non-alcoholic steatohepatitis, hepatic encephalopathy,
depression, etc.[103 ] Given the above, restoring the gut microbiota through FMT to improve the condition
of patients with PSC seems like a good idea. A pilot clinical trial conducted by Allegretti
et al[104 ] investigated the effects of FMT in patients with PSC and concurrent IBD. Of the
14 enrolled participants, 10 completed the FMT protocol, which consisted of six weekly
administrations. The study observed a significant increase in gut microbial α-diversity
following treatment. Additionally, 3 out of 10 patients (30%) experienced a ≥50% reduction
in serum alkaline phosphatase levels, indicating a biochemical response. Although
reductions in serum transaminases (ALT and AST) were reported in some individuals,
detailed stratified data were not provided. Importantly, no serious adverse events
related to FMT were observed during the study. Ma et al[105 ] investigated the therapeutic effects of FMT in a mouse model of EAH induced by hepatic
antigen S100. The study demonstrated that FMT significantly reduced serum levels of
ALT, AST, and total bilirubin, and alleviated histological liver inflammation. Mechanistically,
FMT reversed the elevation of proinflammatory cytokine IL-21 and promoted the expression
of anti-inflammatory cytokines IL-10 and TGF-β. Additionally, FMT restored the balance
between follicular helper T (Tfh) cells and follicular regulatory T (Tfr) cells, potentially
via downregulation of the TLR/MyD88 signaling pathway.
Although FMT shows promising immunomodulatory effects in preclinical studies of ALIDs,
several potential risks should be considered. These include donor-derived pathogen
transmission, unpredictable immune responses due to host–microbiota interactions,
immunological imbalance from unintended microbial shifts, and the lack of standardized
protocols for donor screening and microbial composition. The evidence to date remains
limited, especially for PBC and IgG4-SC, and further large-scale, controlled clinical
trials are warranted to validate its safety and efficacy in AILDs.
Intervention with Microbial Metabolites
Myeloid-derived suppressor cells (MDSCs) have been shown to have potent immunosuppressive
functions by upregulating inducible nitric oxide synthase, ROS, arginase 1, and prostaglandin
E2. A 2018 study by Zhang et al[106 ] showed that MDSC accumulation in PBC patients was negatively correlated with disease
severity. On this basis, Wang et al[107 ] recently found that gut microbial butyrate promotes the differentiation and function
of MDSCs by inhibiting HDAC3, enhancing their immunosuppressive capacity and thus
ameliorating the condition of PBC, providing a new potential target for the treatment
of PBC.
Recent studies have highlighted the potential of combining Quercetin and A. muciniphila as a therapeutic strategy for immune-related liver diseases. Juárez-Fernández et
al[108 ] divided 21-day-old rats into a control group and a high-fat diet group and fed them
a control diet and a high-fat diet for 6 weeks. Then, quercetin and/or A.
muciniphila were added to the control group diet with/without quercetin for 3 weeks. The results
showed that A.
muciniphila and quercetin could significantly increase the level of total BAs in plasma, among
which the concentration of primary BAs increased the most. In addition, the synbiotic
combination of A.
muciniphila and quercetin increased the ratio of unconjugated to conjugated BAs. Specifically,
plasma concentrations of all unconjugated primary BA species increased after supplementation
with this synbiotic, with the most significant increases in cholic acid (CA), β-murucic
acid (βMCA), and α-murucic acid (αMCA). CA has been shown to be negatively correlated
with TLR2 expression, which may be related to protection against inflammation. In
addition, the increase in MCA levels means that the toxicity and hydrophobicity of
the BA pool is reduced, creating a healthier hydrophilic BA pool. Beyond BA modulation,
quercetin also exerts anti-inflammatory effects through Nrf2/HO-1 pathway activation
and NF-κB suppression.[109 ] Simultaneously, A.
muciniphila has been shown to enhance intestinal barrier integrity and modulate immune responses
by shaping γδT17 cell activity and macrophage polarization.[110 ] More strikingly, a recent study revealed that the combination of A.
muciniphila and inosine restored the Treg/Th17/Th1 balance and upregulated key immunoregulatory
markers—CD39, CD73, and the adenosine A2A receptor—within the gut–liver axis, offering
protection against alcohol-induced liver injury.[111 ] However, the current evidence supporting the use of A.
muciniphila and quercetin in AILDs remains preliminary and largely extrapolated from non-AILD
models. Future studies should incorporate AILD-specific animal models and patient-derived
data to validate these effects. Moreover, attention should be given to the complex
immunopathology of AILDs, potential strain- and dose-specific effects, and safety
profiles before clinical application. Integration of multiomics approaches and personalized
intervention strategies may further enhance the translational potential of this synbiotic
combination.
Other Therapeutic Strategies Based on Intestinal Microecological Modulation
Other Therapeutic Strategies Based on Intestinal Microecological Modulation
Antibiotics
It has been demonstrated that the administration of antibiotics to eradicate E. faecium can mitigate the progression of extraintestinal autoimmune diseases.[62 ] A pilot clinical trial has also indicated a potential therapeutic effect of metronidazole
and vancomycin in the treatment of PSC.[112 ] Furthermore, a systematic review and meta-analysis were performed by Shah et al[113 ] explore the efficacy of antibiotics in treating PSC, both with and without IBD.
Their results indicate that vancomycin could be the most promising antimicrobial option
for managing PSC. The researchers aim to utilize more selective antibiotics to avoid
targeting other symbiotic flora and to develop new mechanisms of action to prevent
antibiotic resistance, with the objective of achieving long-term stable medication.
However, the optimal antibiotic drug, dosage, regimen, and potential long-term side
effects remain largely unknown. Even if targeted eradication of pathogenic bacteria
is achieved, it is possible that unintended consequences may arise through indirect
changes to the microbial ecological balance.
Monocytes
In a study conducted by Kunzmann et al,[114 ] it was observed that, in comparison to patients diagnosed with PBC and healthy individuals,
patients with PSC had a noticeably greater quantity of CD4+ T cells that produced
IL-17A in the peripheral blood. Additionally, patients with PSC-associated cirrhosis
exhibited augmented numbers of CD14hiCD16int and CD14loCD16hi monocyte–macrophages
in the vicinity of bile ducts within the liver, when compared with individuals with
alcoholic cirrhosis. The study revealed that monocytes from patients with PSC exhibited
heightened production of IL-1β and IL-6 following stimulation with C. albicans and Enterococcus faecalis (which may translocate in the context of increased intestinal barrier permeability).
These two cytokines are known to drive the differentiation of Th17 cells. The results
indicate that monocytes may serve as a functional conduit between proinflammatory
microbiota and T cells and may contribute to the pathogenesis of PSC. Accordingly,
future research should consider the potential of monocytes as a therapeutic target
for PSC.
Vaccination
The feasibility of vaccination against pathogenic bacteria for microbiologically induced
immune diseases is still under investigation. This is due to the fact that the same
pathogenic bacteria may play a beneficial role in normal or other disease conditions,
as well as easily remove or cause ecological disorders in the microbiota. However,
intramuscular vaccination against the pathogenic bacterium Enterococcus avium in animal models has been demonstrated to be safe and effective in preventing translocation
to internal organs and systemic autoimmunity.[62 ]
Conclusions
The complex interplay between the gut microbiota and the host immune system plays
a pivotal role in the pathogenesis of AILDs by modulating intestinal barrier integrity,
immune homeostasis, and the production of bioactive metabolites. Despite substantial
advances in delineating the association between intestinal microecology and AILDs,
current research remains limited by several constraints. The high interindividual
variability and complexity of the gut microbial ecosystem present significant challenges
for its therapeutic exploitation. Moreover, most existing studies concentrate on individual
microbial taxa or specific metabolites, offering only a fragmented view of the dynamic
equilibrium between the microbiome and host immunity.
Further investigations are needed to elucidate the specific mechanisms through which
the gut microbiota contributes to the onset and progression of distinct AILD subtypes
and to determine whether these microbial alterations are causal or consequential.
Translational strategies, such as probiotic and prebiotic supplementation, FMT, and
microbial metabolite modulation have shown preliminary potential in both experimental
and clinical settings. However, their clinical implementation is still hindered by
uncertainties regarding safety, efficacy, and durability of effect. Rigorous, large-scale
clinical trials are essential to validate these approaches and to optimize microbiota-based
interventions in the management of AILDs.