International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Liver cirrhosis is a global health challenge, responsible for ~1.47 million deaths (2.4% of all deaths) in 2019, with the greatest burden occurring in Southeast Asia and the Western Pacific regions (1). Chronic hepatitis B virus (HBV) infection, affecting an estimated 296 million people worldwide, remains the leading cause of cirrhosis and hepatocellular carcinoma, accounting for ~331,000 cirrhosis-related deaths in 2019 and contributing to more than half of hepatocellular carcinoma cases in endemic areas (2). The most recent WHO estimates further indicate that HBV caused ~1.1 million deaths in 2022, mainly attributable to cirrhosis and hepatocellular carcinoma (3). Pathologically, HBV-associated cirrhosis is characterized by progressive fibrosis, distortion of hepatic architecture and sustained infiltration of diverse immune cell populations, including neutrophils, hepatic macrophages, T and B lymphocytes and natural killer cells, accompanied by a pro-inflammatory cytokine milieu rich in IL-1, IL-6, IL-17, IL-22, IL-35, TGF-β and TNF-α (4,5). Persistent hepatic inflammation promotes stellate cell activation and extracellular matrix deposition, perpetuating fibrogenesis and facilitating the transition to decompensated disease (6).
Gut-derived microbiota and their metabolites, together with nutrients and other signals, reach the liver via the portal vein, a process referred to as the gut-liver axis (7,8). Inflammatory cytokines and immune responses mediated by infiltrating cells are key contributors to the initiation and progression of liver fibrosis (9). In recent years, growing attention (10-12) has been directed toward the gut microbiota as a key regulator of liver physiology, particularly via the gut-liver axis. These complex microbial communities regulate both physiological and immunological functions of the host, influencing not only local intestinal processes but also systemic responses. Numerous microbiome-wide association studies have demonstrated a link between gut microbial dysbiosis and chronic diseases, including metabolic syndrome, inflammatory bowel disease and liver pathologies (13,14). Microbiota-derived metabolites and pathogen-associated molecular patterns directly influence hepatic immune responses and metabolic processes. In liver diseases such as non-alcoholic fatty liver disease (NAFLD), alcoholic liver disease and cirrhosis, consistent shifts in microbial composition have been documented (15,16). Notably, in cirrhosis, enrichment of taxa such as Fusobacterium, Veillonella and members of the Enterobacteriaceae family is associated with heightened systemic inflammation and greater disease severity, with several studies also indicating that these changes are more pronounced in decompensated patients (17,18).
Mechanistic studies have begun to clarify how microbial alterations influence liver health (7,19). The gut microbiota produces a broad repertoire of bioactive metabolites, including short-chain fatty acids (SCFAs), lipopolysaccharides (LPS), bile acids, choline derivatives, indole compounds, vitamins, lipids and niacin, which that regulate hepatic immune signaling and metabolic balance (19,20). Among these, SCFAs such as butyrate, acetate and propionate are particularly notable for their ability to strengthen epithelial tight junctions, enhance mucosal barrier integrity, suppress intestinal inflammation and limit overgrowth of pathogenic bacteria, in part via activation of free FA receptors 2 and 3(20). In experimental NAFLD models, enrichment of SCFA-producing taxa is associated with reduced hepatic lipid accumulation and attenuation of inflammatory responses (21,22). Other microbial products such as bile acids, tryptophan metabolites and LPS further modulate macrophage polarization, antimicrobial peptide production and fibrotic remodeling through receptors such as farnesoid X Receptor), TGR5(Takeda G-protein coupled receptor 5) and AhR(Aryl Hydrocarbon receptor), thereby linking gut dysbiosis to the pathophysiology of both cirrhosis and hepatocellular carcinoma (8,23,24).
In parallel with these metabolic and immunomodulatory functions, the gut microbiota is key for preserving intestinal homeostasis. It achieves this by maintaining the integrity of the mucus layer, sustaining the expression and spatial organization of tight junction proteins and coordinating mucosal immune responses. In cirrhosis, this equilibrium is disrupted, characterized by diminished tight junction protein expression, increased intestinal permeability and translocation of bacterial products, which collectively exacerbate hepatic inflammation, accelerate fibrogenesis and foster conditions that facilitate pathogenic biofilm formation, aberrant immune activation and chronic inflammation (25-27). Histopathological analyses of cirrhotic livers typically demonstrate dense infiltration by neutrophils, macrophages, T and B lymphocytes and dendritic cells, indicative of a sustained pro-inflammatory microenvironment (28,29). Within this immune landscape, CD4+ T helper (Th) cell subsets, including Th1, Th2 and Th17, and regulatory T cells serve a central role in orchestrating immune responses: They regulate CD8+ T cell expansion, promote B cell activation and modulate multiple immune effectors throughout disease progression (30,31). Although evidence links gut microbial dysbiosis to immune activation in cirrhosis, most studies are observational, and the precise mechanistic interplay between microbiota alterations, systemic inflammation and barrier dysfunction, particularly in HBV-associated cirrhosis, remains to be elucidated (32-34).
The present study aimed to systematically characterize the composition, diversity and predicted functions of the gut microbiota in patients with HBV-associated liver cirrhosis. By integrating microbial profiling with histological assessment of immune infiltration and intestinal barrier integrity, the present study aimed to elucidate the mechanistic underpinnings of the gut-liver axis in cirrhosis progression and provide a basis for microbiome-based diagnostic and therapeutic strategies.
A total of 31 individuals with HBV-associated liver cirrhosis (21 male, 10 female; median age, 48 years; age range, 28-65 years) were enrolled between April 2023 and April 2024, before receiving any therapeutic intervention. Inclusion criteria were as follows: age between 18 and 70 years; a confirmed diagnosis of liver cirrhosis based on histology or imaging as described above; chronic HBV infection; no prior receipt of antiviral therapy for HBV. Exclusion criteria included: hepatocellular carcinoma; human immunodeficiency virus infection; pregnancy or severe cardiac; pulmonary; renal diseases. Cirrhosis was diagnosed by liver biopsy or by concordant findings from at least two imaging modalities (ultrasound, computed tomography, or magnetic resonance imaging). Disease severity was assessed using the Child-Pugh classification (35), which evaluates serum total bilirubin and albumin, prothrombin time, ascites and hepatic encephalopathy, categorizing patients as class A (5-6 points), B (7-9 points) or C (≥10 points). The control group included 15 healthy volunteers (8 male, 7 female; median age, 46 years; age range, 30-58 years) with no history of physical or psychological disorder, confirmed through annual comprehensive health evaluations including blood, urine and fecal analyses, liver function and biochemical tests, hepatitis virus markers, chest radiography and abdominal ultrasound. All participants were recruited from the Third Affiliated Hospital of Sun Yat-Sen University (Guangzhou, China).
Fecal sample collection was performed using a TinyGene fecal collection box according to the manufacturer's instructions. Boxes were placed on ice and stored at -80˚C before further analysis.
A total of six normal liver tissue sections were from para-hemangioma sites of patients with hepatic hemangioma without hepatitis (three male, three female; median age, 45 years; age range, 32-60 years). Paired samples of liver cirrhosis were obtained from six separate patients with HBV-infected liver cirrhosis during operations (four male, 2 female; median age, 48 years; age range, 30-65 years) before any therapeutic intervention. Colonic mucosal specimens of 10 patients with untreated liver cirrhosis (6 male, 4 female; median age, 47 years; age range, 27-63 years) and 10 healthy volunteers (5 male, 5 female; median age, 45 years; age range, 27-57 years) were obtained from the Endoscopic Center of the Third Affiliated Hospital of Sun Yat-Sen University. All tissues were verified by histopathology. The acquisition of these tissues was approved by the Clinical Research Ethics Committee of the Third Affiliated Hospital of Sun Yat-Sen University (approval no. RG2023-033-01).
The 16S rRNA gene was amplified using primers as follows: Forward, 5'-GTGCCAGCMGCCGCGGTAA-3' and reverse, 5'-CCGTCAATTCMTTTGAGTTT-3' targeting the V5-V6 hyper-variable regions (M were degenerate bases, representing A or C) and sequenced using the Illumina, Inc. platform following the manufacturer's protocols. The library was constructed by using the two-step PCR amplification method with the following thermocycling conditions: Initial preamplification stage at 94˚C for 2 min and then 22 cycles at 94˚C for 30 sec, 55˚C for 30 sec, and 72˚C for 30 sec. The second step preamplification stage at 94˚C for 2 min and then 8 cycles at 94˚C for 30 sec, 55˚C for 30 sec, and 72˚C for 30 sec. QIAamp DNA Stool Mini Kit (51604, QIAGEN) and Phusion™ High-Fidelity DNA Polymerase (M0530L, New England Biolabs were used. The quality and integrity of the extracted DNA were verified using agarose gel electrophoresis and a Bioanalyzer 2100 (Agilent Technologies). Sequencing libraries were quantified using a Qubit fluorometer (Thermo Fisher Scientific) and the final loading concentration was adjusted to 4 nM. Paired-end sequencing (2x300 bp) was performed on an Illumina MiSeq platform using the MiSeq Reagent Kit v3 (600-cycl, MS-102-300, Illumin). Raw reads were processed using Trimmomatic v0.39 (usadellab.org/cms/?page=trimmomatic), FLASH v1.2.11 (ccb.jhu.edu/software/FLASH/), and Mothur v1.39.5 (mothur.org/). After demultiplexing, dereplication, and filtering, operational taxonomic units (OTUs) were clustered at 97% similarity using UCLUST v1.2.22q (drive5.com/usearch/) against the SILVA v128 (arb-silva.de/) and Greengenes v13_8 databases (greengenes.secondgenome.com). α-diversity (Chao1, ACE, Shannon, Simpson) was calculated using Mothur v1.39.5 and QIIME v1.9.1 (qiime.org/), while β-diversity (Jaccard, unweighted UniFrac, weighted UniFrac) was assessed and visualized by PCoA with FastTree v2.1.3 (microbesonline.org/fasttree/). Differentially abundant taxa were identified using MetaStats (metastats.cbcb.umd.edu/), and LEfSe (huttenhower.sph.harvard.edu/lefse/) was used to detect taxa differing significantly between groups. β-diversity clustering was evaluated by analysis of similarity (ANOSIM) and correlations between distance matrices were examined with the Mantel test.
The USEARCH v9.2.64 global alignment command (drive5.com/usearch/) was used to capture OTU representative sequences from the Greengenes database v13_8 (greengenes.secondgenome.com). Functional metagenome reconstruction was then performed using PICRUSt v1.1.4 (picrust.github.io/picrust/). The predicted metagenome functions were annotated against the Kyoto Encyclopedia of Genes and Genomes (KEGG) orthology (kegg.jp/).
Hematoxylin-eosin (H&E), Sirius red, immunohistochemical and immunofluorescence staining were performed as previously described (36-38). The cell index was determined by dividing the number of histological staining signal-positive cells by the total number of cells in ≥20 randomly selected fields of view (magnification, x200). The antibodies were as follows: Anti-myeloperoxidase (MPO; cat. no. A1374, Abclonal), -CD4 (cat. no. sc-19641), -CD19 (cat. no. sc-19650; both Santa Cruz Biotechnology, Inc.), -CD68 (cat. no. ab31630), -α-smooth muscle actin (cat. no. ab5694; both Abcam), -claudin-1 (cat. no. 13255S), -zonula occludens-1 (ZO-1; cat. no. 8193T) and -E-cadherin (cat. no. 14472S; all Cell Signaling Technology, Inc.).
DAO and endotoxin levels from peripheral superficial vein blood were examined with ELISA kits (cat. nos. JL-T0829 and JL52002D, respectively; both J&L Biological) to estimate intestinal permeability and bacterial translocation according to the manufacturer's instructions.
Data were analyzed using R software (version 3.4.1) and GraphPad Prism 6 (Dotmatics) software, including richness estimators, the Ace, Chao and Shannon index, a diversity estimator and rank dissimilarity and abundance distribution. Microbiome compositional dissimilarity was represented by PCoA plots, and quantified by unweighted or weighted UniFrac distance values. The significant separation of the microbiome composition was determined by ANOSIM and the significance differences in α and β diversity and taxonomy between groups were analyzed using Wilcox test. Continuous data are expressed as the mean ± SEM of ≥3 independent experimental repeats and analyzed by Student's two-tailed paired t-test or one- or two-way ANOVA followed by Bonferroni correction as described previously (39). P<0.05 was considered to indicate a statistically significant difference.
The present study compared the gut microbiota of healthy volunteers with that of patients with liver cirrhosis using Illumina MiSeq high-throughput sequencing of the 16S rRNA gene. In total, ~1,466,285 sequence reads of 16S rRNA genes, with an average length of ~410 bp, and 432 OTUs were obtained after sequencing and quality filtering (Fig. 1A). On average, healthy volunteers had more OTUs than patients with liver cirrhosis (371 vs. 362 OTUs, respectively; 301 core OTUs were shared). The average number of reads was not significantly different between healthy and cirrhosis group (31,643 vs. 31,988 reads, respectively).
To evaluate the alterations in the microbiota structure between healthy volunteers and patients with liver cirrhosis, α diversity was measured. Patients with liver cirrhosis had notably decreased microbial species richness and evenness in comparison with volunteers, based on the Chao and Shannon index and OTU rank abundance analysis (Fig. 1A). Furthermore, rank dissimilarity revealed that the between-group difference was greater than the within-group difference (Fig. 1B), and the Chao and Shannon indices analyzed using the Wilcoxon test showed that patients with liver cirrhosis had significantly lower gut microbial diversity than healthy volunteers (Fig. 1B). β diversity based on OTUs or phylogenetic analysis was calculated using Jaccard and unweighted and weighted UniFrac phylogenetic distance matrices; microbial composition of patients with liver cirrhosis was different from that of healthy volunteers (Fig. 1C). Core bacterial genus analysis using a heatmap demonstrated that the two groups had similar microbiota structures or communities, but different richness and abundance (Fig. 2A). The relative bacterial community richness and abundance at the family and genus levels were similar between groups (Fig. 2B and C).
LEfSe was conducted to determine the most relevant microbial taxa responsible for the differences between the groups (Fig. 3A). This analysis identified 29 taxa that were differentially abundant between the two groups. The microbiota of patients with liver cirrhosis was enriched in Fusobacterium (genus), Veillonellaceae (family), Lactobacillales (family), Negativicutes (class), Gammaproteobacteria (class), and Enterobacteriaceae (family), whereas healthy the microbiota of healthy volunteers was enriched in Clostridiales (order), Ruminococcaceae (family), Bacteroides (species), Subdoligranulum (genus) and Subdoligranulum (genus; Fig. 3A and B). Moreover, the differentially abundant features of the microbial taxa at the genus level verified that the abundance of Fusobacterium, Megamonas, Streptococcus, Lachnoclostridium, Veillonella and other bacteria was increased in the fecal samples of patients with liver cirrhosis (Fig. 4A).
Based on the differences in community richness and abundance, metagenome functional content based on the 16S rRNA gene sequences was predicted using PICRUSt. Predicted KEGG orthology pathways significantly enriched in liver cirrhosis included ‘starch and sucrose metabolism’, ‘butanoate metabolism’, ‘fructose and mannose metabolism’, ‘propanoate metabolism’ and ‘phosphotransferase system (PTS)’, indicating that these metabolic pathways participated in the progression of liver cirrhosis (Fig. 4B). Moreover, to verify the most relevant functional pathways based on the microbial differences between healthy volunteers and patients with liver cirrhosis, LEfSe analysis was performed. Functional composition of the total gut microbiota in patients with liver cirrhosis was primarily associated with ‘phosphotransferase system (PTS)’ and ‘membrane transport’ involved in environmental information processing, in contrast to ‘amino acid metabolism’ and ‘energy metabolism’ functions enriched in healthy volunteers (Fig. 5A-C). Collectively, these data demonstrated the gut microbiota profile differs between healthy volunteers and patients with liver cirrhosis, and that a specific microbial community, diversity, and associated metagenome function are present in those with liver cirrhosis.
The bacterial species with increased abundance in liver cirrhosis contribute to environmental information and metabolic pathway components, which may instigate inflammation of the host (17). Moreover, the gut microbiota participates in the preservation of tolerance and immunity of mucosal surfaces by regulating organic inflammatory responses (40). Based on these findings, the colonic mucosal specimens from patients with liver cirrhosis and healthy volunteers were analyzed, which demonstrated increased infiltration of inflammatory cells, including T lymphocytes (CD4), B lymphocytes (CD19), macrophages (CD68) and neutrophils (MPO), in the colonic tissues of patients with liver cirrhosis. However, there was no notable histological architectural alterations between the two groups based on H&E staining (Fig. 6A and B). As expected, upregulated T and B lymphocyte, macrophage and neutrophil infiltration, as well as excess collagen deposition, was found in the liver tissues of cirrhotic patients compared with those of healthy volunteers (Fig. 6C and D). On this basis, the present study investigated how gut microbial changes relate to barrier dysfunction and inflammatory responses in cirrhosis.
The functional composition of the total gut microbiota in patients with liver cirrhosis primarily involved ‘phosphotransferase system (PTS)’ and ‘membrane transport’. Changes in the gut microbiota modulate systemic microbe-derived metabolite levels by altering intestinal permeability and the gut barrier, especially tight junctions between epithelial cells (16). Claudin-1, ZO-1 and E-cadherin were downregulated in colonic samples from patients with cirrhosis whose liver tissues showed excess collagen deposition by Sirius red staining (Fig. 7A-C). Intestinal permeability, based on blood DAO determination, was significantly increased, and blood endotoxin detection showed enhanced bacterial translocation analysis in cirrhotic liver sections (Fig. 7D). In addition, the blood DAO and endotoxin levels showed a positive correlation with collagen deposition, as indicated by Sirius red staining of the liver tissue (Fig. 7E). The results demonstrated that the gut microbiota influences colonic and hepatic immune responses, intestinal permeability and the gut barrier, which would contribute to the progression of liver cirrhosis (Fig. 7F).
The gut microbiome serves a key role in immune development and function in the host, and in determining the host metabolic state. An increasing number of studies have suggested that the gut-derived microbiota and their components, metabolites, nutrients and other signals can be delivered to the liver via the portal vein circulation to serve as a bioreactor for autonomous metabolic and immunological regulation and regulate responses within the host environment (8,23,24). Here, patients with HBV-associated cirrhosis exhibit distinct gut microbial profiles compared with healthy individuals, characterized by decreased diversity, enrichment of pro-inflammatory taxa, functional shifts toward environmental information processing pathways, compromised intestinal barrier function and marked hepatic immune cell infiltration.
By comparing the gut microbiota between healthy volunteers and patients with liver cirrhosis using high-throughput sequencing of the 16S rRNA gene, the present study demonstrated that patients with liver cirrhosis had decreased microbial species richness, evenness, and diversity, although they had similar microbial structures and communities according to core bacterial genus analysis. This is supported by several studies (41,42) indicating a disordered gut microbial community and decreased diversity and species richness associated with cirrhosis. Decreased microbial diversity is now recognized as a feature of disease states, including inflammatory disorder, colorectal cancer, and gastric carcinoma (43-46). Furthermore, LEfSe analysis showed that the gut microbiota of patients with liver cirrhosis was enriched in Fusobacterium, Veillonellaceae, Lactobacillales, Negativicutes, Gammaproteobacteria, Enterobacteriaceae, whereas the proportion of phylum Bacteroides, Clostridiales, Ruminococcaceae, and Subdoligranulum was decreased compared with that of the healthy volunteers. These results align with a previous study that observed a marked loss of Bacteroides and significant increases in Veillonella, Enterobacteriaceae and Fusobacterium abundance in cirrhosis (47). This suggest that dysbiosis, or an unfavorable change in the composition of the microbiome in liver cirrhosis, is central to the pathophysiology of the onset, progression and development of complications of liver cirrhosis. Nonetheless, inconsistencies remain; for example, Chen et al (47) reported decreased Bacteroidetes and increased Proteobacteria and Fusobacteria, whereas Sarangi et al (48) observed no significant differences in microbial abundance. The differences in findings emphasize the need for more research to clarify the specific roles of microbial taxa in cirrhosis progression and treatment.
To explore functional consequences, KEGG orthology analysis was performed on metagenomic sequencing data and found that the total microbiota of patients with liver cirrhosis was predominantly enriched in ‘phosphotransferase system (PTS)’ and ‘membrane transport’, pathways involved in environmental information processing, whereas the microbiota of healthy controls showed enrichment in ‘amino acid metabolism’ and ‘energy metabolism’. Consistent with the present data, a previous study also reported enhanced transport- and metabolism-associated pathways, along with a marked loss of cell cycle-associated gene functions, in cirrhotic patients (7,47). Notably, the depletion of SCFA-producing taxa, such as Faecalibacterium, Eubacterium hallii, Ruminococcus and Agathobacter, has also been observed in cirrhotic patients (49). These butyrate-producing bacteria are key suppliers of energy for colonocytes and contribute to maintaining mucosal integrity. Their loss may compromise epithelial energy metabolism, attenuate butyrate-mediated anti-inflammatory signaling and disrupt the regulation of immune homeostasis. This depletion may aggravate the decrease in tight junction proteins and the increased intestinal permeability, thereby weakening barrier function and facilitating microbial translocation to the liver (22,40). These findings highlight the association between the gut microbiota and host metabolism in liver cirrhosis, with the microbial balance shifted toward a dysbiosis during the process of liver cirrhosis.
The activation of inflammatory cells to produce inflammatory cytokines and components is a key contributor to the initiation and development of liver cirrhosis (28,29). Increased T and B lymphocyte, macrophage and neutrophil infiltration was detected in patients with liver cirrhosis compared with healthy volunteers, and this was accompanied by excess collagen deposition in liver tissue. These data indicated that various inflammatory cells and responses are affected by gut-derived microbiota, and their components, metabolites or signals participate in the initiation and development of liver fibrosis.
The dysbiotic microbial community associated with liver cirrhosis is essential for the development and regulation of the immune and metabolism systems of the host (7,19,20). Changes in the gut microbiota modulate systemic microbe-derived metabolite levels or signals by altering intestinal permeability and the gut barrier, thus contributing to disease development (23).
A critical mechanistic link between the dysbiotic microbial community and the immune and metabolic systems of the host is the structural compromise of the intestinal epithelial barrier, which relies on tight junction proteins such as claudin-1 and ZO-1 and the adherens junction protein E-cadherin. To identify potential associations between tight junctions in colonic epithelial cells and microbial dysbiosis, the present study analyzed colonic sections and found that claudin-1, ZO-1 and E-cadherin levels were suppressed in colonic samples from cirrhotic patients; this was associated with enhanced intestinal permeability and bacterial translocation, suggesting that the tight junctions between colonic epithelial cells were damaged during liver fibrogenesis. In radiation-induced enteritis, gut dysbiosis disrupts the localization of claudin-1, occludin and ZO-1, weakening epithelial cohesion (50). Therapeutic modulation of the microbiota can mitigate such injury; for example, Yu-Ping-Feng-San (a traditional Chinese medicine decoction) treatment restores ZO-1, occludin and claudin-1 expression in LPS-induced barrier damage while decreasing inflammatory responses (51). At the molecular level, claudin-1 expression is regulated via the Piezo1/ROCK1/2 pathway, whereby Piezo1 activation by mechanical or inflammatory cues decreases claudin-1 protein expression and thereby impairs junctional integrity (52). Similarly, loss of SLC26A3 results in downregulation of ZO-1, occludin and E-cadherin alongside microbial imbalance, underscoring the reciprocal association between junctional protein expression and microbiota composition (53). In metabolic liver diseases such as NAFLD and NASH, breakdown of tight junctions allows endotoxin influx to the liver, promoting inflammatory and fibrotic changes (54). These data highlight that gut barrier impairment, mediated by altered junctional proteins in the context of dysbiosis, is a key driver of microbial translocation and hepatic injury in cirrhosis.
While the present findings provide insights into the gut microbiota-immune-barrier axis in HBV-associated cirrhosis, several limitations should be noted. First, the cross-sectional design precludes causal inference between microbiota dysregulation and inflammation or barrier dysfunction, and the contribution of HBV infection cannot be excluded; interventional approaches such as probiotics, fecal microbiota transplantation or other microbiota-targeted strategies are needed to test whether restoring microbial balance can ameliorate these changes. Second, the sample size may have limited the detection of subtle associations, and data on diet, medication and antiviral therapy, important microbiome modulators, were incomplete; these should be collected in future longitudinal and interventional studies. Finally, functional predictions based on 16S rRNA sequencing lack the resolution of shotgun metagenomics, metatranscriptomics or targeted metabolomics, which should be integrated to define microbiota-host interactions in HBV-related cirrhosis. To address these issues, future studies should adopt longitudinal designs with larger and well-characterized cohorts, integrate clinical and dietary information with multi-omics approaches, and evaluate interventional strategies such as probiotic or prebiotic supplementation, dietary modulation and fecal microbiota transplantation to determine whether restoring microbial balance can mitigate inflammation, reinforce barrier function and slow disease progression.
In conclusion, the gut microbiota profile in patients with HBV-associated liver cirrhosis differs markedly from that of healthy individuals, showing enrichment of taxa such as Fusobacterium, Veillonella and members of the Enterobacteriaceae family, alongside depletion of beneficial butyrate-producing genera including Faecalibacterium, Ruminococcus and Agathobacter. These compositional shifts, together with decreased overall diversity and altered functional potential, particularly enrichment in ‘phosphotransferase system (PTS)’ and ‘membrane transport’ and depletion of ‘amino acid metabolism’ and ‘energy metabolism’, are closely associated with impaired colonic and hepatic immune responses, increased intestinal permeability and compromised gut barrier integrity. Gut microbiota dysbiosis contributes to the pathophysiology of HBV-associated cirrhosis. Understanding these microbial alterations may provide a basis for microbiota-targeted strategies to mitigate hepatic inflammation and preserve barrier function in affected patients. Future longitudinal and interventional studies are warranted to establish causality and evaluate the therapeutic potential of microbiota-targeted interventions in HBV-associated cirrhosis.
Not applicable.
Funding: The present study was supported by the Natural Science Foundation of Guangdong Province (grant no. 2023A1515011204), the National Natural Science Foundation of China (grant no. 82170569) and the Science and Technology Planning Projects of Guangzhou City (grant nos. 2024A04J6565 and 2025A03J3193).
The data generated in the present study may be found in the ScienceDB under accession number 31253.11.sciencedb.28999 or at the following URL: doi.org/10.57760/sciencedb.28999.
KX and YZ designed the methodology and analyzed data. KX and YZ confirm the authenticity of all the raw data. ShT, XO and JL designed the methodology. SiT conceived the study and wrote and edited the manuscript. All authors have read and approved the final manuscript.
Informed consent was obtained from all participants before the initiation of the study. The present study protocol was approved by the Institute Research Ethics Committee of the Third Affiliated Hospitals of Sun Yat-sen University (Guangzhou, China; approval no. RG2023-033-01).
Not applicable.
The authors declare that they have no competing interests.
|
Huang DQ, Terrault NA, Tacke F, Gluud LL, Arrese M, Bugianesi E and Loomba R: Global epidemiology of cirrhosis-aetiology, trends and predictions. Nat Rev Gastroenterol Hepatol. 20:388–398. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Hsu YC, Huang DQ and Nguyen MH: Global burden of hepatitis B virus: Current status, missed opportunities and a call for action. Nat Rev Gastroenterol Hepatol. 20:524–537. 2023.PubMed/NCBI View Article : Google Scholar | |
|
World Health Organization. Hepatitis B. Fact sheet. Geneva, 2025. Available from: https://www.who.int/news-room/fact-sheets/detail/hepatitis-b. | |
|
Suh YG, Kim JK, Byun JS, Yi HS, Lee YS, Eun HS, Kim SY, Han KH, Lee KS, Duester G, et al: CD11b(+) Gr1(+) bone marrow cells ameliorate liver fibrosis by producing interleukin-10 in mice. Hepatology. 56:1902–1912. 2012.PubMed/NCBI View Article : Google Scholar | |
|
Kornek M, Popov Y, Libermann TA, Afdhal NH and Schuppan D: Human T cell microparticles circulate in blood of hepatitis patients and induce fibrolytic activation of hepatic stellate cells. Hepatology. 53:230–242. 2011.PubMed/NCBI View Article : Google Scholar | |
|
Hammerich L and Tacke F: Hepatic inflammatory responses in liver fibrosis. Nat Rev Gastroenterol Hepatol. 20:633–646. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Usami M, Miyoshi M and Yamashita H: Gut microbiota and host metabolism in liver cirrhosis. World J Gastroenterol. 21:11597–11608. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Levy M, Blacher E and Elinav E: Microbiome, metabolites and host immunity. Curr Opin Microbiol. 35:8–15. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Luedde T and Schwabe RF: NF-kappaB in the liver-linking injury, fibrosis and hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 8:108–118. 2011.PubMed/NCBI View Article : Google Scholar | |
|
Pabst O, Hornef MW, Schaap FG, Cerovic V, Clavel T and Bruns T: Gut-liver axis: Barriers and functional circuits. Nat Rev Gastroenterol Hepatol. 20:447–461. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Hov JR and Karlsen TH: The microbiota and the gut-liver axis in primary sclerosing cholangitis. Nat Rev Gastroenterol Hepatol. 20:135–154. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Anand S and Mande SS: Host-microbiome interactions: Gut-Liver axis and its connection with other organs. NPJ Biofilms Microbiomes. 8(89)2022.PubMed/NCBI View Article : Google Scholar | |
|
Lynch SV and Pedersen O: The human intestinal microbiome in health and disease. N Engl J Med. 375:2369–2379. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Lavelle A, Lennon G, Winter DC and O'Connell PR: Colonic biogeography in health and ulcerative colitis. Gut Microbes. 7:435–442. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Maslennikov R, Poluektova E, Zolnikova O, Sedova A, Kurbatova A, Shulpekova Y, Dzhakhaya N, Kardasheva S, Nadinskaia M, Bueverova E, et al: Gut microbiota and bacterial translocation in the pathogenesis of liver fibrosis. Int J Mol Sci. 24(16502)2023.PubMed/NCBI View Article : Google Scholar | |
|
Liu Y, Chen Z, Li C, Sun T, Luo X, Jiang B, Liu M, Wang Q, Li T, Cao J, et al: Associations between changes in the gut microbiota and liver cirrhosis: A systematic review and meta-analysis. BMC Gastroenterol. 25(16)2025.PubMed/NCBI View Article : Google Scholar | |
|
Kang EJ, Cha MG, Kwon GH, Han SH, Yoon SJ, Lee SK, Ahn ME, Won SM, Ahn EH and Suk KT: Akkermansia muciniphila improve cognitive dysfunction by regulating BDNF and serotonin pathway in gut-liver-brain axis. Microbiome. 12(181)2024.PubMed/NCBI View Article : Google Scholar | |
|
Sharma SP, Gupta H, Kwon GH, Lee SY, Song SH, Kim JS, Park JH, Kim MJ, Yang DH, Park H, et al: Gut microbiome and metabolome signatures in liver cirrhosis-related complications. Clin Mol Hepatol. 30:845–862. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Hackstein CP, Assmus LM, Welz M, Klein S, Schwandt T, Schultze J, Förster I, Gondorf F, Beyer M, Kroy D, et al: Gut microbial translocation corrupts myeloid cell function to control bacterial infection during liver cirrhosis. Gut. 66:507–518. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Qin N, Yang F, Li A, Prifti E, Chen Y, Shao L, Guo J, Le Chatelier E, Yao J, Wu L, et al: Alterations of the human gut microbiome in liver cirrhosis. Nature. 513:59–64. 2014.PubMed/NCBI View Article : Google Scholar | |
|
Shashni B, Tajika Y, Ikeda Y, Nishikawa Y and Nagasaki Y: Self-assembling polymer-based short chain fatty acid prodrugs ameliorate non-alcoholic steatohepatitis and liver fibrosis. Biomaterials. 295(122047)2023.PubMed/NCBI View Article : Google Scholar | |
|
Mao T, Zhang C, Yang S, Bi Y, Li M and Yu J: Semaglutide alters gut microbiota and improves NAFLD in db/db mice. Biochem Biophys Res Commun. 710(149882)2024.PubMed/NCBI View Article : Google Scholar | |
|
Spiljar M, Merkler D and Trajkovski M: The immune system bridges the gut microbiota with systemic energy homeostasis: Focus on TLRs, mucosal barrier, and SCFAs. Front Immunol. 8(1353)2017.PubMed/NCBI View Article : Google Scholar | |
|
Acharya C and Bajaj JS: Gut microbiota and complications of liver disease. Gastroenterol Clin North Am. 46:155–169. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Jandl B, Dighe S, Baumgartner M, Makristathis A, Gasche C and Muttenthaler M: Gastrointestinal biofilms: Endoscopic detection, disease relevance, and therapeutic strategies. Gastroenterology. 167:1098–1112.e5. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Wang J, He M, Yang M and An X: Gut microbiota as a key regulator of intestinal mucosal immunity. Life Sci. 345(122612)2024.PubMed/NCBI View Article : Google Scholar | |
|
Kuo CH, Wu LL, Chen HP, Yu J and Wu CY: Direct effects of alcohol on gut-epithelial barrier: Unraveling the disruption of physical and chemical barrier of the gut-epithelial barrier that compromises the host-microbiota interface upon alcohol exposure. J Gastroenterol Hepatol. 39:1247–1255. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Jung Y, Witek RP, Syn WK, Choi SS, Omenetti A, Premont R, Guy CD and Diehl AM: Signals from dying hepatocytes trigger growth of liver progenitors. Gut. 59:655–665. 2010.PubMed/NCBI View Article : Google Scholar | |
|
Seki E and Schwabe RF: Hepatic inflammation and fibrosis: Functional links and key pathways. Hepatology. 61:1066–1079. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Du YN, Tang XF, Xu L, Chen WD, Gao PJ and Han WQ: SGK1-FoxO1 signaling pathway mediates Th17/Treg imbalance and target organ inflammation in angiotensin II-induced hypertension. Front Physiol. 9(1581)2018.PubMed/NCBI View Article : Google Scholar | |
|
Muranski P and Restifo NP: Essentials of Th17 cell commitment and plasticity. Blood. 121:2402–2414. 2013.PubMed/NCBI View Article : Google Scholar | |
|
Liu R, Kang JD, Sartor RB, Sikaroodi M, Fagan A, Gavis EA, Zhou H, Hylemon PB, Herzog JW, Li X, et al: Neuroinflammation in murine cirrhosis is dependent on the gut microbiome and is attenuated by fecal transplant. Hepatology. 71:611–626. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Zhao B, Jin Y, Shi M, Yu L, Li G, Cai W, Lu Z and Wei C: Gut microbial dysbiosis is associated with metabolism and immune factors in liver fibrosis mice. Int J Biol Macromol. 258(129052)2024.PubMed/NCBI View Article : Google Scholar | |
|
Santiago A, Sanchez E, Clark A, Pozuelo M, Calvo M, Yañez F, Sarrabayrouse G, Perea L, Vidal S, Gallardo A, et al: Sequential changes in the mesenteric lymph node microbiome and immune response during cirrhosis induction in rats. mSystems. 4:e00278–18. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Durand F and Valla D: Assessment of the prognosis of cirrhosis: Child-Pugh versus MELD. J Hepatol. 42 (Suppl 1):S100–S107. 2005.PubMed/NCBI View Article : Google Scholar | |
|
Tan S, Chen X, Xu M, Huang X, Liu H, Jiang J, Lu Y, Peng X and Wu B: PGE2/EP4 receptor attenuated mucosal injury via β-arrestin1/Src/EGFR-mediated proliferation in portal hypertensive gastropathy. Br J Pharmacol. 174:848–866. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Tan S, Li L, Chen T, Chen X, Tao L, Lin X, Tao J, Huang X, Jiang J, Liu H and Wu B: β-Arrestin-1 protects against endoplasmic reticulum stress/p53-upregulated modulator of apoptosis-mediated apoptosis via repressing p-p65/inducible nitric oxide synthase in portal hypertensive gastropathy. Free Radic Biol Med. 87:69–83. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Tan S, Lu Y, Xu M, Huang X, Liu H, Jiang J and Wu B: β-Arrestin1 enhances liver fibrosis through autophagy-mediated Snail signaling. FASEB J. 33:2000–2016. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Xiao Y, Liu X, Xie K, Luo J, Zhang Y, Huang X, Luo J and Tan S: Mitochondrial dysfunction induced by HIF-1α under hypoxia contributes to the development of gastric mucosal lesions. Clin Transl Med. 14(e1653)2024.PubMed/NCBI View Article : Google Scholar | |
|
Wang R, Li B, Huang B, Li Y, Liu Q, Lyu Z, Chen R, Qian Q, Liang X, Pu X, et al: Gut microbiota-derived butyrate induces epigenetic and metabolic reprogramming in myeloid-derived suppressor cells to alleviate primary biliary cholangitis. Gastroenterology. 167:733–749.e3. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Solé C, Guilly S, Da Silva K, Llopis M, Le-Chatelier E, Huelin P, Carol M, Moreira R, Fabrellas N, De Prada G, et al: Alterations in gut microbiome in cirrhosis as assessed by quantitative metagenomics: relationship with acute-on-chronic liver failure and prognosis. Gastroenterology. 160:206–218.e13. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Shen Y, Wu SD, Chen Y, Li XY, Zhu Q, Nakayama K, Zhang WQ, Weng CZ, Zhang J, Wang HK, et al: Alterations in gut microbiome and metabolomics in chronic hepatitis B infection-associated liver disease and their impact on peripheral immune response. Gut Microbes. 15(2155018)2023.PubMed/NCBI View Article : Google Scholar | |
|
Hui W, Yu D, Cao Z and Zhao X: Butyrate inhibit collagen-induced arthritis via Treg/IL-10/Th17 axis. Int Immunopharmacol. 68:226–233. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Gevers D, Kugathasan S, Denson LA, Vazquez-Baeza Y, Van Treuren W, Ren B, Schwager E, Knights D, Song SJ, Yassour M, et al: The treatment-naive microbiome in new-onset Crohn's disease. Cell Host Microbe. 15:382–392. 2014.PubMed/NCBI View Article : Google Scholar | |
|
Lepage P, Häsler R, Spehlmann ME, Rehman A, Zvirbliene A, Begun A, Ott S, Kupcinskas L, Doré J, Raedler A and Schreiber S: Twin study indicates loss of interaction between microbiota and mucosa of patients with ulcerative colitis. Gastroenterology. 141:227–236. 2011.PubMed/NCBI View Article : Google Scholar | |
|
Ahn J, Sinha R, Pei Z, Dominianni C, Wu J, Shi J, Goedert JJ, Hayes RB and Yang L: Human gut microbiome and risk for colorectal cancer. J Natl Cancer Inst. 105:1907–1911. 2013.PubMed/NCBI View Article : Google Scholar | |
|
Chen Y, Yang F, Lu H, Wang B, Chen Y, Lei D, Wang Y, Zhu B and Li L: Characterization of fecal microbial communities in patients with liver cirrhosis. Hepatology. 54:562–572. 2011.PubMed/NCBI View Article : Google Scholar | |
|
Sarangi AN, Goel A, Singh A, Sasi A and Aggarwal R: Faecal bacterial microbiota in patients with cirrhosis and the effect of lactulose administration. BMC Gastroenterol. 17(125)2017.PubMed/NCBI View Article : Google Scholar | |
|
Qi P, Yang XX, Wang CK, Sang W, Zhang W and Bai Y: Analysis of gut and circulating microbiota characteristics in patients with liver cirrhosis and portal vein thrombosis. Front Microbiol. 16(1597145)2025.PubMed/NCBI View Article : Google Scholar | |
|
Jian Y, Zhang D, Liu M, Wang Y and Xu ZX: The impact of gut microbiota on radiation-induced enteritis. Front Cell Infect Microbiol. 11(586392)2021.PubMed/NCBI View Article : Google Scholar | |
|
Wang Y, Wang Y, Ma J, Li Y, Cao L, Zhu T, Hu H and Liu H: YuPingFengSan ameliorates LPS-induced acute lung injury and gut barrier dysfunction in mice. J Ethnopharmacol. 312(116452)2023.PubMed/NCBI View Article : Google Scholar | |
|
Jiang Y, Song J, Xu Y, Liu C, Qian W, Bai T and Hou X: Piezo1 regulates intestinal epithelial function by affecting the tight junction protein claudin-1 via the ROCK pathway. Life Sci. 275(119254)2021.PubMed/NCBI View Article : Google Scholar | |
|
Kumar A, Priyamvada S, Ge Y, Jayawardena D, Singhal M, Anbazhagan AN, Chatterjee I, Dayal A, Patel M, Zadeh K, et al: A novel role of SLC26A3 in the maintenance of intestinal epithelial barrier integrity. Gastroenterology. 160:1240–1255.e3. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Jadhav K and Cohen TS: Can you trust your gut? Implicating a disrupted intestinal microbiome in the progression of NAFLD/NASH. Front Endocrinol (Lausanne). 11(592157)2020.PubMed/NCBI View Article : Google Scholar |