We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research ArticleOpen Accesscc iconby iconnc iconnd icon

Ferrostatin-1 improves prognosis and regulates gut microbiota of steatotic liver transplantation recipients in rats

    Mengshu Yuan

    Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, PR China

    ,
    Yuxin Wang

    Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, PR China

    ,
    Xiaorong Tian

    Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, PR China

    ,
    Weiping Zheng

    Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin, 300192, PR China

    NHC Key Laboratory of Critical Care Medicine, Tianjin, 300192, PR China

    ,
    Huaiwen Zuo

    Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, PR China

    ,
    Xinru Zhang

    Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, PR China

    &
    Hongli Song

    *Author for correspondence:

    E-mail Address: songhl@tmu.edu.cn

    Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin, 300192, PR China

    Tianjin Key Laboratory of Organ Transplantation, Tianjin, PR China

    Published Online:https://doi.org/10.2217/fmb-2023-0133

    Abstract

    Aims: To investigate the effects of Ferrostatin-1 (Fer-1) on improving the prognosis of liver transplant recipients with steatotic liver grafts and regulating gut microbiota in rats. Methods: We obtained steatotic liver grafts and established a liver transplantation model. Recipients were divided into sham, liver transplantation and Fer-1 treatment groups, which were assessed 1 and 7 days after surgery (n = 6). Results & conclusion: Fer-1 promotes recovery of the histological structure and function of steatotic liver grafts and the intestinal tract, and improves inflammatory responses of recipients following liver transplantation. Fer-1 reduces gut microbiota pathogenicity, and lowers iron absorption and improves fat metabolism of recipients, thereby protecting steatotic liver grafts.

    Graphical abstract

    Liver transplantation (LT) remains the only therapeutic option for acute liver failure, end-stage liver disease and malignant liver cancer [1]. However, increasing imbalance between donor liver shortage and liver transplant candidates forces transplant centers to utilize extended-criteria donors, including donation after circulatory death and steatotic livers [2]. With the rising prevalence of obesity, metabolic syndrome and type 2 diabetes, the incidence rate of nonalcoholic fatty liver disease (NAFLD) is increasing, affecting approximately 25% of the population. The increasing prevalence of NAFLD has multiple impacts on liver transplant donors, with early fatty liver disease accounting for approximately 25% of liver transplant donors. The United Network for Organ Sharing database confirms that NAFLD has reduced the liver donor pool, and overall utilization of liver grafts is predicted to decrease from 78 to 44% by 2030 [3]. Although liver transplantation technology is maturing, the abandonment rate due to severe fatty liver disease remains high [4]. Therefore, optimizing the utilization rate of severe steatotic liver grafts in liver transplantation is crucial.

    Steatotic liver transplantation ischemia reperfusion (IR) can damage recipient liver and extrahepatic organs, especially the heart, lungs and intestines. These organ injuries are also the main factors leading to poor prognosis following liver transplantation, especially intestinal injuries, which can increase perioperative complications of liver transplantation, leading to increased perioperative mortality [3]. Although ischemic preconditioning, preservation solutions and machine perfusion had encouraging preclinical data, they still failed to make a significant difference in clinical practice [5].

    Protecting intestinal mucosal barrier function is an important element of perioperative management of liver transplantation [6,7]. Intestinal symbiotic bacteria and their products can reach the liver through the portal vein, thereby regulating liver innate and adaptive immunity, causing liver inflammation and promoting liver fibrosis [8–10]. Some specific bacterial populations are associated with the occurrence of bloodstream infections before and after transplantation [11]. LT significantly affects the gut microbiota [12,13]. Moreover, ecological imbalance following LT seems to be related to the colonization of multidrug-resistant bacteria [14].

    Recent research indicates that progression from simple fat to fatty hepatitis to liver fibrosis is closely related to increased lipid peroxidation, and is caused by accumulation of fatty acids and large amounts of iron in liver cells [15–17]. Iron overload is another risk factor for liver ischemia-reperfusion injury (IRI) [18]. Our recent research showed that regulating ferroptosis in steatotic liver grafts can improve IRI in steatotic liver transplantations [19,20].

    Therefore, the present study aimed to establish a rat model of liver transplantation with severe steatotic liver grafts. Using 16S rRNA amplicon sequencing and bioinformatics algorithms to identify key subgroups of the gut microbiota, we explored the effects of ferroptosis inhibitor Ferrostatin-1 (Fer-1) on improving the prognosis of LT recipients with severe steatotic liver grafts in rats. We also assessed changes in the gut microbiota, providing research directions for expanding the donor liver pool and improving the prognosis of recipients.

    Materials & methods

    Establishment of animal & severe steatotic liver graft models

    Specific pathogen-free grade rats (China Food and Drug Administration, Beijing, China) were used. We obtained severe steatosis liver grafts following a high-fat diet for 12 weeks (final body weight 450–500 g). Hematoxylin and eosin staining showed that the area of mixed macrovesicular steatosis was more than 60% under the microscope, indicating that a model of severe steatotic liver was established successfully [13]; liver transplantation recipients were male Sprague–Dawley rats fed on a normal basal diet until 22–24 weeks of age (final body weight 450–500 g). All animal experiments followed guidance for the care and use of experimental animals, and the study was reviewed and approved by the Animal Ethics Committee of Nankai University (ethics no. 2021-SYDWLL-000331).

    Establishment of a rat liver transplantation model

    Nonarterialized orthotopic liver transplantation was performed in rats using the double-cuff technique [21]. Recipients were divided into three groups: sham operation (sham group), steatotic liver transplantation (LT group) and Fer-1 group. The Fer-1 group received intraperitoneal injection (10 mg/kg/day) 24 h before surgery (MCE, Shanghai, China) [19]. A total of 24 recipients (six per group) were used for survival analysis; 35 rats were used for biochemical analysis (blood, liver and intestinal tissue: five in the sham operation group and five in the other experimental groups at postoperative day 1 (POD 1) and postoperative day 7 (POD 7).

    Constructing an injury model using IAR20 cells & steatotic IAR20 cells

    Lipopolysaccharide (LPS; 10 μg/ml) was administered to rat hepatocyte IAR20 cells for 24 h to establish an IAR20 cell injury model. Cells were divided into 1) NC (normal IAR20 cells); 2) NL (10 μg/ml LPS to stimulate normal IAR20 cells for 24 h); and 3) NF (10 μg/ml LPS plus 2 μM Fer-1) groups.

    Palmitic acid (Sigma-Aldrich, MO, USA; 100 μM) and 200 μM oleic acid (Sigma) were used to stimulate IAR20 cells for 24 h to obtain adipose IAR20 cells. These were divided into 1) FC (adipose degeneration IAR20 cells); 2) FL (10 μg/ml LPS to stimulate adipose degeneration IAR20 cells for 24 h); and 3) FF (10 μg/ml LPS plus 2 μM Fer-1) groups. The activity of IAR20 cells and levels of reactive oxygen species (ROS) and ferroptosis-related proteins were measured.

    Constructing an injury model of intestinal epithelial cells

    LPS (10 μg/ml) was applied to growing rat intestinal epithelial IEC-6 cells and human colon adenocarcinoma Caco-2 cells for 24 h to establish injury models of human and rat intestinal epithelial cells. They were divided into 1) NC (normal intestinal epithelial cells); 2) NL (10 μg/ml LPS the stimulate normal intestinal epithelial cells for 24 h); and 3) NF (10 μg/ml LPS plus 2 μM Fer-1) groups.

    Liver function test

    Collected sera were analyzed by a fully Cobas 800 automated biochemical analyzer (Roche, Basel, Switzerland) to assess liver function by measuring alanine aminotransferase (ALT) and aspartate aminotransferase (AST) and alkaline phosphatase (ALP) enzyme activities, and total bilirubin (TBil) and total cholesterol levels. Very-low-density lipoprotein (VLDL), low-density lipoprotein (LDL) and high-density lipoprotein (HDL) contents were also measured. All operations were performed according to the manufacturer's instructions.

    Histopathology

    Liver and intestinal tissues were fixed in 4% formalin then embedded in paraffin. Slices 4 μm thick were then stained with hematoxylin and eosin.

    Immunohistochemistry

    After sectioning tissue specimens, paraffin was dewaxed with xylene and samples were dehydrated using a graded alcohol series. Antigen repair was performed by heating in ethylene diamine tetraacetic acid (pH 9.0) in a microwave oven for 15 min. Tissue slides were then incubated in hydrogen peroxide for 30 min to block endogenous peroxidase activity, and further blocked with 5% normal goat serum. Slides were incubated overnight at 4 °C with antibody-recognizing glutathione peroxidase 4 (GPX4; 1:500), cyclooxygenase-2 (COX2; 1:500), myeloperoxidase (MPO; 1:300) or CD68 (1:500). Slides and biotinylated secondary antibody were then incubated at room temperature for 60 min, and streptavidin-conjugated horseradish peroxidase was added and incubated at room temperature for 30 min. Finally, slides were stained with diaminobenzidine and restained with hematoxylin.

    Transmission electron microscopy

    Fresh liver and ileocecal intestinal tissue samples were cut into 1 × 1 × 2 mm3 pieces and observed using an HT7800 transmission electron microscope (Hitachi, Tokyo, Japan).

    ELISA

    Serum levels of diamine oxidase (DAO), d-lactic acid (D-LA) and LPS were measured using ELISA kits (Multisciences Biotechnology Co., Hangzhou, China) according to the manufacturer's instructions.

    Western blotting

    Radioimmunoprecipitation assay and phenylmethylsulphonyl fluoride were used to extract proteins from tissues and cells. For detailed experimental methods, please refer to our previous study [12]. ImageJ 7.0 software (NIH, MD, USA) was used to analyze grayscale values and calculate relative protein expression levels.

    Flow cytometry

    Collected cells were subjected to extracellular staining (CD3, CD4; BioLegend, CA, USA). A FACS Canto II instrument (BD Biosciences, CA, USA) was used for cell identification and data were analyzed using FlowJo software (ThreeStar, OR, USA).

    16S rRNA sequencing

    Sequencing of fecal 16S rRNA was performed by Metware Biotechnology Co. Ltd. (Wuhan, China). Fresh fecal samples (∼0.2 g) from rats in each group were place in a sealed, sterile, frozen storage tube and stored in liquid nitrogen. According to the characteristics of the V3–V4 region of the 16S rDNA gene of bacteria, a small fragment library was constructed and paired-end sequenced using an Illumina NovaSeq sequencing platform. First, original data were spliced and filtered to obtain valid data (clean data). Operational taxonomic unit (OTU) clustering was then employed and based on the OTU clustering results, representative sequences of each OTU were annotated to obtain corresponding species information and abundance distribution based on species. Furthermore, OTUs were analyzed based on abundance, alpha-diversity, Venn plots and petal plots to obtain information on species richness and evenness within samples, and to identify common and unique OTUs among different samples. In addition, multiple sequence alignment was performed on OTUs and phylogenetic trees were constructed. Through dimensionality reduction analyses such as Shannon, Simpson, chao1, abundance-based coverage estimator (ACE), principal co-ordinates analysis (PCoA), principal component analysis (PCA) and non-metric multi-dimensional scaling (NMDS) sample cluster trees were generated and differences in community structure among different samples or groups were investigated. In order to further explore differences in community structure among grouped samples, statistical analysis methods such as T-test, similarity percentage (Simper) and line discriminant analysis effect size (LEfSe) were used to assess the significance of differences in species composition and community structure of grouped samples. Annotation results for amplified genes were assessed against GreenGenes (16S), FAPROTAX and KEGG functional databases to perform functional prediction analysis of microbial communities in samples.

    Statistical analysis

    SPSS 13.0 (SPSS GmbH, Munich, Germany) and GraphPad 8.0 (GraphPad Software Company, CA, USA) were used for statistical analysis. Results are expressed as mean ± standard deviation. One-way analysis of variance (ANOVA) was employed to test statistical significance. Results are expressed as a percentage (%), and a Chi-squared test was used to assess statistical significance. Kaplan–Meier survival curves and log bank tests (Mantel–Cox) were used for survival analysis (p < 0.05 was considered statistically significant).

    Results

    Fer-1 inhibits ferroptosis in transplanted steatotic liver & recipient intestinal tissues, & prolongs recipient survival rate

    Morphological changes of severe steatotic grafts were typically yellow with a greasy texture, blunt edges, and tight and smooth capsules (Figure 1A), indicating that the donor liver model with severe steatosis was successfully established. Using normal SD rats as recipients, we successfully performed liver transplantation on transplanted livers with severe steatosis (Figure 1B). Survival curves show that the median survival (30 days) of the Fer-1 group was significantly higher than that of the LT group (16 days; p < 0.05). Therefore, inhibiting ferroptosis could prolong the survival of liver transplant recipients with severe steatotic liver grafts (Figure 1C).

    Figure 1. Changes in the morphology of severe steatotic liver grafts and ferroptosis in the liver and intestines after transplantation.

    (A) Severe steatotic liver grafts. (B) Liver transplantation with severe steatotic liver grafts. (C) Survival time after steatotic liver transplantation. (D) Immunohistochemistry of liver tissue GPX4 (×200) and electron microscopy showing ferroptosis in severe steatotic liver grafts after transplantation. The red arrow indicates mitochondrial outer membrane rupture. (E) Immunohistochemistry of intestinal tissue GPX4 (×200) and electron microscopy showing iron death in intestinal tissue after transplantation. The red arrow indicates mitochondrial outer membrane rupture.

    POD 1: Postoperative day 1; POD 7: Postoperative day 7.

    Immunohistochemical results of liver tissue analyses showed that at POD 1, the level of GPX4 was significantly decreased in the LT group. Application of ferroptosis inhibitor Fer-1 increased GPX4 levels (Figure 1D). Electron microscopy results showed that mitochondria of steatotic liver grafts were swollen with prominent mitochondrial cristae, and no rupture of the outer membrane was observed. In the LT group, the intracellular structure was disordered, the mitochondrial volume decreased and cristae were significantly fewer. At POD 7 there was significant mitochondrial injury (indicated by a red arrow). However, in the Fer-1 group more normal mitochondria remained visible (Figure 1D). This indicates significant ferroptosis in liver tissue after liver transplantation from severe steatotic liver grafts, which was more severe at POD 7. Application of ferroptosis inhibitor Fer-1 could significantly reduce ferroptosis in liver cells after transplantation.

    Immunohistochemical results for intestinal tissues showed that at POD 1, GPX4 levels in the LT group were significantly decreased, while in the Fer-1 group they were increased (Figure 1E). Electron microscopy observations showed that in the sham group, microvilli were neatly arranged. In the LT group, intestinal microvilli became shorter and atrophied, with obvious mitochondrial swelling and rupture of the adventitia (indicated by a red arrow). In the Fer-1 group, microvilli of the intestinal wall were arranged more neatly, with more normal mitochondria visible (Figure 1E). At POD 7, significant mitochondrial outer membrane rupture remained evident in the LT group. In the Fer-1 group, GPX4 levels were increased, with more normal mitochondria visible. This indicates ferroptosis in both liver and intestinal epithelial cells after severe steatotic liver transplantation, and there were no significant differences in intestinal epithelial cells between POD 1 and POD 7. After application of Fer-1, ferroptosis in liver and intestinal epithelial cells was decreased.

    Fer-1 can improve structural & functional changes in transplanted livers & the recipient intestinal tract

    Liver histology

    In the sham group mixed steatosis was observed. At POD 1, steatosis remained evident in the LT group, accompanied by disorder of the hepatic cord, severe congestion of hepatic sinuses, and obvious cell necrosis and infiltration of inflammatory cells. In the Fer-1 group there was less infiltration of inflammatory cells than in the LT group. At POD 7 the LT group showed significant liver tissue necrosis; in the Fer-1 group mild steatosis of hepatocytes was observed, and some hepatic cords (near the portal vein) were arranged neatly (Figure 2A).

    Figure 2. Histological and functional changes in transplanted liver and recipient intestines.

    (A) Histopathology of severe steatotic transplanted liver (hematoxylin and eosin [H&E] staining, ×200). (B) Liver function. (C) Recipient intestinal histopathology (H&E staining, ×200). (D) Intestinal permeability of recipients.

    ALP: Alkaline phosphatase; ALT: Alanine aminotransferase; AST: Aspartate aminotransferase; DAO: Diamine oxidase; D-LA: D-lactic acid; HDL: High-density lipoprotein; LDL: Low-density lipoprotein; LPS: Lipopolysaccharide; POD 1: Postoperative day 1; POD 7: Postoperative day 7; TBil: Total bilirubin; TC: Total cholesterol; VLDL: Very low-density lipoprotein.

    Liver function

    At POD 7 the LT group had the highest ALT, AST, TBiL, ALP and TC values (Figure 2B). In the LT group, VLDL was significantly increased at POD 1 but decreased at POD 7 to a level similar to that of the Fer-1 group. There were no differences between groups for LDL and HDL at POD 1. However, at POD 7 LDL was significantly increased in LT, followed by Fer-1 (p < 0.05). These results indicate that inhibiting ferroptosis can improve donor liver histology, liver function and lipid metabolism in steatosis.

    Intestinal histology

    Intestinal villi of rats in the sham group were arranged regularly. In the LT group, at POD 1 intestinal villi were significantly damaged, with fracture and epithelial loss visible; at POD 7 significant intestinal villi fracture remained evident. In the Fer-1 group, severe villi epithelial loss was observed at POD 1; at POD 7 intestinal villi were regularly arranged with slight villi epithelial loss (Figure 2C).

    Intestinal function

    At POD 1 LT had the highest plasma levels of D-LA, DAO and LPS, followed by Fer-1 (p < 0.05). At POD 7 only D-LA in the LT group continued to increase. In the Fer-1 group, D-LA, DAO and LPS levels were lower than at POD 1 (p < 0.05; Figure 2D). These results suggest that inhibiting ferroptosis could improve recipient intestinal histology and permeability.

    Effects of Fer-1 on injured hepatocytes & intestinal epithelial cells in vitro

    In the IAR20 cell injury model, the same concentration of LPS stimulated normal and steatosis IAR20 cells, and cell activity in the FL group was lowest (p < 0.05) (Figure 3A). Intracellular lipid ROS content in the FL group was highest, followed by the FF group (Figure 3B). COX2 protein content was highest, while GPX4 protein content was lowest in FL and NL groups (Figure 3C). These results suggest that LPS can aggravate ferroptosis in steatosis IAR20 cells, while Fer-1 can alleviate ferroptosis in IAR20 cells induced by LPS to some extent.

    Figure 3. Lipopolysaccharide damages IAR20 and intestinal epithelial cells.

    (A) Normal and steatosis IAR20 cell activity. (B) Lipid reactive oxygen species (ROS) content in normal and steatosis IAR20 cells. (C) Expression of COX2 and GPX4 in normal and steatosis IAR20 cells. (D) Intestinal epithelial cell activity in each group. (E) Lipid ROS content in IEC-6 cells. (F) Lipid ROS content in Caco-2 cells.

    *p < 0.05; **p < 0.01.

    FC: Adipose degeneration IAR20 cells; FF: 10 μg/ml LPS plus 2 μM Fer-1; FL: 10 μg/ml LPS to stimulate adipose degeneration IAR20 cells for 24 h; LPS: Lipopolysaccharide; NC: Normal IAR20 cells; NF: 10 μg/ml LPS plus 2 μM Fer-1; NL: 10 μg/ml LPS to stimulate normal IAR20 cells for 24 h.

    In the intestinal epithelial cell injury model, cell activity in the NL group was lowest (Figure 3D). Intracellular lipid ROS content in the NL group was highest (Figure 3E–F). In addition, COX2 protein levels in the NL group were significantly increased, while GPX4 protein levels were decreased (p < 0.05; Supplementary Figure 1). These results suggest that LPS can cause ferroptosis in intestinal epithelial cells, while Fer-1 can alleviate ferroptosis in intestinal epithelial cells caused by LPS to a certain extent.

    Fer-1 reduces inflammatory responses of fatty liver transplantation & recipients

    Neutrophil (marker = MPO) and Kupffer cells (marker = CD68) play a key role in the occurrence and development of nonalcohol fatty liver disease and liver IR [19]. Changes in T-cell subsets are closely related to transplant rejection and the survival of transplant recipients. Therefore, we explored changes in liver MPO and CD68 levels, blood and liver T-cell subsets in each group.

    Immunohistochemical results for liver tissues revealed a small number of MPO- and CD68-positive cells in the sham group, indicating inflammatory damage in the steatotic liver graft itself (Figure 4A & B). Levels of MPO and CD68 in the LT group were highest, especially at POD 7 (Figure 4A & B).

    Figure 4. Inhibiting the effects of ferroptosis on recipient inflammatory responses.

    (A) Myeloperoxidase immunohistochemistry of liver tissue (×200). (B) Immunohistochemistry of liver tissue CD68 (×200). (C) Flow cytometry of blood T-cell subsets. (D) Flow cytometry of T-cell subsets in liver grafts.

    POD 1: Postoperative day 1; POD 7: Postoperative day 7.

    Flow cytometry of blood T-cell subsets showed that the proportion of CD8+ T cells in the LT group was increased significantly, especially at POD 7. Fer-1 could reduce the proportion of CD8+ T cells and increase the proportion of CD4+ T cells, and its effect at POD 7 was significantly higher than at POD 1 (Figure 4C). In liver grafts, the proportion of CD8+ T cells in the LT group was increased significantly. Compared with the LT group, the proportion of CD8+ T cells in the Fer-1 group was decreased, while the proportion of CD4+ T cells was significantly increased (Figure 4D).

    This suggests that Fer-1 could reduce the infiltration of neutrophils and Kupffer cells in severe steatotic liver grafts. In addition, it could improve inflammatory responses of recipients after liver transplantation by regulating the ratio of CD4+ T cells to CD8+ T cells. As time progressed the effects of Fer-1 became more obvious.

    Fer-1 alters the gut microbiota composition of transplant recipients

    Gut microbiota sequencing depth

    In vitro experiments showed that Fer-1 could only improve damaged hepatocytes and intestinal epithelial cells to a certain extent, but could not completely restore the activity of damaged cells, reducing the content of lipid ROS and COX2 proteins in cells. In addition, changes in the liver and intestinal functions of recipients can have a significant impact on the composition and function of the gut microbiota. Therefore, we collected and sequenced fecal samples. Species accumulation boxplots, rarefaction and rank abundance curves and goods coverage index reflected observed species, amount of sequencing data and sequencing depth were enough to cover all species in the sample (Figure 5A–D).

    Figure 5. Changes in the gut microbiota of recipients after severe steatotic liver transplantation.

    (A) Species accumulation boxplot. The flat position of this box chart indicates that species in this environment will not significantly increase with increasing sample size, hence sampling was sufficient for data analysis. (B) Rarefaction curve. (C) Rank abundance curve. (D) Good coverage index reflecting adequate sequencing depth. (E) Species abundance at the family level. (F) Species abundance at the phylum level.

    POD 1: Postoperative day 1; POD 7: Postoperative day 7.

    Dominant bacteria in the intestines of transplant recipient rats in each group

    At the family level Peptostreptococcaceae, Bacteroidaceae, Clostridiaceae, Muribaculaceae, Enterobacteriaceae and Lactobacillaceae were the most abundant microbiota (Figure 5E). At the phylum level Firmicutes, Bacteroidota, Proteobacteria, Verrucomicrobiota, Fusobacteriota and unidentified bacteria had the highest abundance in each group. At POD 7 in the LT group the Firmicutes/Bacteroidota ratio was significantly decreased (Figure 5F).

    Alpha-diversity

    Alpha-diversity clustering analysis showed that steatotic liver transplantation significantly disrupted the number and diversity of gut microbiota species. ACE, Shannon, Simpson and Chao1 indices all showed that species diversity within the LT group was greatest (Supplementary Figure 2A–D), and the number of OTUs in the LT group was largest, especially at POD 7 (Supplementary Figure 2E).

    Beta-diversity

    To explore microbial diversity between groups, unweighted UniFrac and weighted UniFrac algorithms were used for beta-diversity analysis (Supplementary Figure 3A & B). In non-NMDS, PCA and PCoA analyses there were significant differences in spatial indices between any two groups, and the LT group had the largest difference from the sham group at POD 7, while the Fer-1 group had the smallest difference from the sham group at POD 7 (Supplementary Figure 3C–F).

    Diversity of bacterial populations in each group

    Simper (similarity percentage) was used to decompose the Bray-Curtis difference index to quantify the contribution of each species to differences between two groups. The results revealed the top ten species and their abundance at the family level that contribute to differences between two groups.

    In the LT group at POD 1, compared with the sham group, the abundances of Enterobacteriaceae and Staphylococcaceae were higher; the abundances of Akkermansiaceae, Muribaculaceae, Lactobacillaceae and Lachnospiraceae were decreased significantly (p < 0.05; Supplementary Figures 4 & 5A). At POD 7 the abundances of Bacteroidaceae and Fusobacteriaceae were increased significantly while the abundances of Peptostreptococcaceae, Enterobacteriaceae, Staphylococcaceae and Lactobacillaceae were decreased significantly (Supplementary Figure 4 & 5B).

    In the Fer-1 group at POD 1, compared with the sham group, the abundances of Clostridiaceae and Oscillospiraceae increased were increased while the abundances of Peptostreptococcaceae and Akkermansiaceae were decreased (Supplementary Figures 4 & 5C). At POD 7 the abundance of Peptostreptococcaceae was increased while the abundance of Clostridiaceae was decreased (Supplementary Figures 4 & 5D). Compared with the LT group, the abundances of Clostridiaceae and Muribaculaceae were increased in the Fer-1 group at POD 1, while the abundances of Peptostreptococcaceae, Enterobacteriaceae, Bacteroidaceae and Staphylococcaceae were lower in the Fer-1 group, and differences were significant (p < 0.05; Supplementary Figures 4 & 5E). At POD 7 the abundances of Peptostreptococcaceae, Muribaculaceae and Prevotellaceae were increased while the abundances of Bacteroidaceae, Fusobacteriaceae and Enterobacteriaceae were decreased (Supplementary Figures 4 & 5F).

    Fer-1 causes functional changes in the gut microbiota of transplant recipients

    Bugbase functional prediction analysis

    See Supplementary Figure 6A–I. Based on the GreenGenes (16S) database, microbial communities are classified according to seven phenotypes: Gram-positive, Gram-negative, biofilm-forming, pathogenic potential, mobile element-containing, oxygen-utilizing (aerobic, anaerobic and facultatively anaerobic) and oxidative stress-tolerant. Gram-positive were most abundant in the sham group. At POD 7 the number of Gram-positive bacteria in the LT group was lowest, while the number of Gram-negative bacteria was significantly increased. In addition, in the LT group at POD 1, the abundance of bacteria with pathogenic potential was highest, and it was decreased at POD 7. Facultative anaerobic bacteria were most abundant in the LT group at POD1 and lowest in the Fer-1 group at POD 7. This suggests that the number of harmful stimuli received by the transplanted liver from the intestinal tract was increased, and Fer-1 could decrease pathogenic bacteria and obesity-causing bacteria.

    FAPRITAX function prediction

    See Figure 6A & Supplementary Figure 7. In the LT group the chemoheterotrophy function was significantly reduced, especially for aerobic chemoheterotrophy. In addition, human pathogens including those causing diarrhea were increased significantly in the LT group but decreased significantly in the Fer-1 group at POD 7. Dark iron oxidation iron respiration of gut microbiota functions were highest in the sham group and the Fer-1 group at POD 1, while at POD 7 they were significantly increased in the LT group (Figure 6B & Supplementary Figure 7). In addition, gut microbiota function in the LT group at POD 1 and the sham group displayed the greatest difference (Figure 6C). This indicates that liver transplantation with fatty liver could significantly affect the functions of the gut microbiota.

    Figure 6. Functional prediction of the gut microbiota in each group.

    (A) FAPROTAX function prediction. (B) FAPROTAX function prediction related to iron metabolism. (C) Annotated Principal Component Analysis (PCA) results based on operational taxonomic unit (OTU) FAPROTAX function. (D) OTU-based clustering heatmap for each group of PICRUST functional annotations. (E) Annotated PCA results based on OTU PICRUST function.

    PICRUSt functional prediction

    Gut microbiota Brite Hierarchies and Cellular Processes were highest in the sham group, while human diseases and environmental information-processing pathways were significantly enhanced in the LT group at POD 1 (Figure 6D). As with FAPRITAX functional prediction, functional differences in the gut microbiota were greatest between the LT group at POD 1 and the sham group (Figure 6E).

    Gut microbiota may influence the recovery of recipients after liver transplantation

    We also conducted T-tests between groups based on quantitative results from PICRUSt functional annotation. In the LT group at POD 1, compared with the sham group, pathways related to bacterial growth such as cell growth were decreased, while bacterial invasion such as Yersinia infection, bacterial invasion of epiphytic cells and LPS biosynthesis proteins were increased (Supplementary Figure 8A). Pathways related to energy metabolism such as carbon hydrate and fat were significantly decreased, and functions related to iron metabolism (biosynthesis of siderophores and nonribosomal peptides) were increased. At POD 7 bacterial invasiveness of the LT group was further enhanced (two-component systems, ABC transporters and the bacterial secretion system; Supplementary Figure 8B).

    In the Fer-1 group at POD 1, compared with the sham and LT groups, pathways related to bacterial growth and reproduction ability such as cell growth were enhanced, bacterial invasion ability was decreased, and carbon fixation and fat metabolism ability were significantly enhanced. Compared with the sham group, the ferroptosis function in the Fer-1 group was enhanced (Supplementary Figures 9 & 10A). At POD 7 the metabolism of carbohydrates and fats in the Fer-1 group was significantly enhanced, and metabolic activity of the gut microbiota associated with ferroptosis (terpenoid backbone biosynthesis) was also enhanced (Supplementary Figure 10B). Thus, Fer-1 could decrease the invasiveness of the gut microbiota, enhance the metabolism of energy-related substances and stimulate ferroptosis-related pathways.

    Discussion

    Steatotic livers are often used in liver transplantations. Such organs are more prone to IRI, which affects liver regeneration, leading to poor prognosis after liver transplantation [19,20]. Therefore, the use of severe steatotic liver grafts is currently avoided in clinical practice [3]. Therefore, it is very important to develop methods for improving severe steatotic liver graft function after transplantation. The present work investigated the role of ferroptosis inhibitor Fer-1 in liver transplantation and postoperative intestinal injury in recipients, and revealed the role of inflammation and changes in gut microbiota under the influence of Fer-1.

    Apoptosis, necrosis, ferroptosis and other kinds of cell deaths exist in steatotic liver transplantation [22]. These cell deaths have considerable overlap and crosstalk. However, since apoptosis requires energy, the chronic ATP depletion in steatosis liver my lead to failure to induce apoptosis [22]. Necrosis in the liver has been long considered an unregulated process [23]. Indeed, iron overload in steatosis livers suggests an important role of ferroptosis [19,20].

    We observed ferroptosis in both transplanted liver and intestine after liver transplantation with severe steatotic liver grafts. Following liver transplantation, ferroptosis of transplanted livers was most obvious at POD 7, with more mitochondria swelling, cristae disappearance and outer membrane rupture than at POD 1. There were no significant differences in the degree of ferroptosis between POD 1 and POD 7 in intestinal epithelial cells of recipients. However, Fer-1 could improve ferroptosis in the transplanted liver and intestinal epithelium of recipients, and the effect became more evident over time.

    In addition to directly inhibiting ferroptosis, we found that Fer-1 had a repair effect on the structure and function of the transplanted liver and the recipient intestinal tract. After liver transplantation, significant hepatocyte steatosis, congestion and inflammatory cell infiltration could still be seen at POD 7. However, Fer-1 brought about transplanted liver recovery at POD 7, and lipid droplet and inflammatory cell infiltration were significantly decreased. Liver function tests showed that Fer-1 could significantly improve liver function and lipid metabolism in rats following steatotic liver transplantation. Due to clamping of the portal vein and inferior vena cava during liver transplantation with severe steatotic grafts, mesenteric arteries become congested, resulting in structural changes in the intestinal wall and pathophysiological changes including vascular congestion, edema and decreased or relaxed tight connections [13,24]. We found that plasma D-LA, DAO and LPS levels were significantly increased after liver transplantation, indicating an increase in intestinal permeability. However, under the action of Fer-1, levels were decreased significantly. This indicates that Fer-1 could improve the function of steatotic liver transplantation while also reducing intestinal mucosal damage and intestinal permeability.

    However, in vitro experiments showed that Fer-1 did not significantly improve the activity of rat hepatocyte IAR20 and intestinal epithelial cells (IEC-6 and Caco-2), intracellular lipid ROS content, or ferroptosis-related protein levels induced by LPS. This is significantly different from the apparent role of Fer-1 in our previous in vivo experiments, hence we continued to explore whether other factors were involved. To this end we investigated the inflammatory responses of transplant recipients.

    The proportion of CD8+ T and CD4+ T cells in transplant recipients is closely related to the clinical outcome of organ transplantation [25]. Neutrophils and Kupffer cells play important roles in the development of IRI during steatotic liver transplantation [26]. Therefore, we tested inflammatory cells associated with IRI and T-cell subsets associated with recipient prognosis. We found that after liver transplantation, there was a significant inflammatory response in the recipient. The number of neutrophils, Kupffer cells and CD8+ T cells was significantly increased, while Fer-1 significantly reduced the number of proinflammatory cells and the CD8+ T-cell ratio, especially at POD 7, thereby reducing IRI injury and inflammatory responses in steatotic liver transplantation. This indicates that Fer-1 could also inhibit inflammatory reactions and improve the prognosis of recipients.

    Considering changes in portal vein pressure, severe damage to recipient liver and intestinal mucosa, and changes in immune function, we suspected that interactions between the liver and the gut microbiota had also altered. Based on the study that gut microbiota metabolites enhance GPX4 expression and suppress ferroptosis in intestinal I/R injury [27] and the therapeutic role of gut microbiota in NAFLD [28], we hypothesize that the gut microbiota may also play a role in the occurrence of ferroptosis in steatotic liver grafts.

    Therefore, we explored changes in the gut microbiota after transplantation and detected significant changes in Firmicutes/Bacteroides, suggesting changes in the composition of the gut microbiota caused by liver damage and Fer-1. As the time after transplantation increased, the increase in Peptostreptococcaceae and Enterobacteriaceae became more significant. Members of the Peptostreptococcaceae family can secrete over 20 known toxins and enzymes that may affect homeostasis of the gut microbiota and induce pathogenesis of the host gut, leading to the development of colitis and tumors [29]. In addition, pathogenic bacteria Enterococcus faecalis, Streptococcus bovis, Bacteroides fragilis, Escherichia coli and Fusobacterium spp. may be involved in liver and intestinal mucosal damage after transplantation. LPS, a typical product of Gram-negative bacteria, is a marker of systemic inflammation and prognosis that is negatively correlated with survival [30,31]. After liver transplantation the number of Gram-negative bacteria significantly increased, while under the effect of Fer-1 the abundance of Gram-negative bacteria decreased. Erysipelotrichaceae and facultative anaerobic bacteria are considered ‘bad’ bacteria that cause obesity and promote diabetes [32], and they were more abundant after liver transplantation with steatosis. Thus, after steatotic liver transplantation there was a significant increase in pathogenic bacteria and obesity-causing bacteria, with increased bacterial invasiveness and an increase in harmful stimuli received by the transplanted liver from the gut.

    In the Fer-1 group, some bacterial populations involved in receptor energy metabolism, especially fat metabolism, were increased significantly. Oscillospiraceae members are related to regulating body lipid metabolism and promoting lipid lowering and weight loss [33]. Oscillospiraceae members may use host polysaccharides as a growth substrate to reduce chronic inflammation caused by obesity, hence a decrease in the abundance of Oscillospiraceae may lead to mild inflammation and hyperglycemia [33]. Muribaculaceae members have beneficial effects on intestinal ecological disorders through immune regulation and controlling intestinal homeostasis [34,35]. Prevotellaceae is an important bacterial group associated with degradation of polysaccharides and the formation of short-chain fatty acids [36,37]. These bacterial groups related to fat metabolism were increased to varying degrees in the Fer-1 group. Thus, changes in the gut microbiota caused by Fer-1 were mainly related to recipient lipid metabolism.

    Lactobacillus, a major class of bacteria in the intestinal tract of humans and animals, is widely used as a probiotic. Lactobacilli have immunomodulatory effects and promote intestinal host defences through interactions with the immune system [37]. After application of Fer-1, the abundance of Lachnospiraceae and Clostridiaceae increased significantly. These are important butyrate-producing bacteria [38] that can significantly improve carbohydrate and lipid metabolism in recipients. Butyrate has been shown to protect the intestinal epithelial barrier, regulate energy metabolism and inflammation, and influence tumor growth and development [39]. In addition, butyrate can also improve obesity, inhibit insulin resistance and reduce cholesterol synthesis [39–41]. This indicates that Fer-1 increases butyrate-producing bacteria and indirectly participates in recipient lipid metabolism.

    Lactobacillus members also detect intestinal iron levels and reduce host iron absorption [42]. Lactobacillus was increased significantly in the LT group at POD 1, but decreased significantly in the LT group at POD 7. However, after application of Fer-1, it increased significantly over time. Pasteurellaceae showed the strongest negative correlation with serum ferritin levels, consistent with their dependence on the use of iron in host transferrin for growth and survival [42]. Pasteurellaceae was significantly increased in the Fer-1 group at POD 1. This indicates that Fer-1 promoted the utilization of iron in gut microbiota and reduced iron absorption of recipients. This also explains why Fer-1 enhanced the ferroptosis-related pathways of gut microbiota.

    Our previous study demonstrated that that bone marrow mesenchymal stem cells exhibited significant protective effects on steatotic liver grafts [19,20], as well as the ability to modulate the gut microbiota of recipients [13]. It was evident from our findings that the effectiveness of Fer-1 in promoting recovery of steatotic liver grafts was comparatively weaker than that of bone marrow mesenchymal stem cells. Furthermore, the functional changes observed in the gut microbiota with the presence of Fer-1 primarily involve potential pathogenic bacteria and iron absorption. This indicates that the recovery benefits of Fer-1 for steatotic liver grafts are limited in their scope. Moreover, our study focused solely on the changes in gut microbiota and did not investigate alterations in the metabolites of the gut microbiota or conduct experiments to abolish the gut microbiota and validate our results. In future studies, we will focus on the molecular effects of Fer-1 in steatotic liver transplantation and gut microbiota metabolism changes under the effect of Fer-1.

    Conclusion

    Fer-1 directly inhibits ferroptosis, improves inflammatory responses of recipients after liver transplantation, and promotes recovery of the histological structure and function of transplanted liver and recipient intestinal tracts. Fer-1 reduces the abundance of intestinal pathogenic bacteria and obesity-causing bacteria in recipients and reduces the pathogenicity of the gut microbiota. It also protects transplanted liver with steatosis by improving lipid metabolism and reducing iron absorption of recipients.

    Summary points
    • Fer-1 promotes recovery of histological structure and function of steatotic liver grafts and the intestinal tract.

    • Fer-1 improves inflammatory responses of recipients after liver transplantation.

    • Fer-1 alters the gut microbiota composition of the recipient.

    • Fer-1 significantly decreased pathogenic bacteria such as Enterobacteriaceae and Peptostreptococcaceae, increased beneficial Lachnospiraceae and Clostridiaceae bacteria, increased Lactobacillus, and Pasteurellaceae decreased iron absorption of recipients.

    Supplementary data

    To view the supplementary data that accompany this paper please visit the journal website at: www.futuremedicine.com/doi/suppl/10.2217/fmb-2023-0133

    Author contributions

    HL Song, MS Yuan conceived the study. HL Song and MS Yuan designed the research. YX Wang, XR Tian, WP Zheng, HW Zuo and XR Zhang assisted in mouse experiments. MS Yuan, YX Wang, XR Tian, WP Zheng, HW Zuo and XR Zhang performed the experiments. MS Yuan and WP Zheng analyzed the data. HL Song, WP Zheng supervised the study. MS Yuan wrote the paper. WP Zheng and HL Song contributed to revision. All authors contributed to the article and approved the submitted version.

    Acknowledgments

    We thank Key Laboratory of Emergency and Care Medicine of Ministry of Health and Tianjin Key Laboratory of Organ Transplantation for allowing this work to progress in their laboratories. The manuscript was edited for proper English language, grammar, punctuation, spelling, and overall style by one or more of the highly qualified native English-speaking editors at ELIXIGEN.

    Financial disclosure

    The work was supported by the National Natural Science Foundation of China (nos. 82070639, 81670574, 81441022, and 81270528). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    Competing interests disclosure

    The authors have no competing interests or relevant affiliations with any organization or entity with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

    Writing disclosure

    No writing assistance was utilized in the production of this manuscript.

    Ethical conduct of research

    The authors state that the study was reviewed and approved by the Animal Ethics Committee of Nankai University (ethics no. 2021-SYDWLL-000331) and have followed the principles outlined in the animal experimental investigations.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Schlegel A, van Reeven M, Croome K et al. A multicentre outcome analysis to define global benchmarks for donation after circulatory death liver transplantation. J. Hepatol. 76(2), 371–382 (2022).
    • 2. Goldaracena N, Cullen JM, Kim D-S, Ekser B, Halazun KJ. Expanding the donor pool for liver transplantation with marginal donors. Int. J. Surg. 82S, 30–35 (2020). • The current status of liver transplantation using marginal donor grafts and efforts to optimize usage are discussed with attention to elderly donors, steatotic livers, donors after circulatory death and split liver grafts.
    • 3. Yang X, Lu D, Wang R et al. Single-cell profiling reveals distinct immune phenotypes that contribute to ischaemia-reperfusion injury after steatotic liver transplantation. Cell Prolif. 54(10), e13116 (2021).
    • 4. Hann A, Osei-Bordom D-C, Neil DAH, Ronca V, Warner S, Perera MTPR. The human immune response to cadaveric and living donor liver allografts. Front. Immunol. 11, 1227 (2020).
    • 5. Patrono D, De Stefano N, Vissio E et al. How to preserve steatotic liver grafts for transplantation. J. Clin. Med. 12(12), 3982 (2023).
    • 6. Ancona G, Alagna L, Lombardi A et al. The interplay between gut microbiota and the immune system in liver transplant recipients and its role in infections. Infect. Immun. 89(11), e0037621 (2021). •• Provides an overview of the current understanding of the interplay between the gut microbiota and the immune system in liver transplant recipients and the role of the former in infections.
    • 7. Liu Y, Méric G, Havulinna AS et al. Early prediction of incident liver disease using conventional risk factors and gut-microbiome-augmented gradient boosting. Cell Metab. 34(5), 719–730 (2022).
    • 8. Watt KD. The evolving role of the microbiome in liver failure and liver transplantation. Liver Transpl. 22(Suppl. 1), 58–61 (2016).
    • 9. Wu Z-W, Ling Z-X, Lu H-F et al. Changes of gut bacteria and immune parameters in liver transplant recipients. Hepatobiliary Pancreat. Dis. Int. 11(1), 40–50 (2012).
    • 10. Ianiro G, Segal JP, Mullish BH et al. Fecal microbiota transplantation in gastrointestinal and extraintestinal disorders. Future Microbiol. 15, 1173–1183 (2020).
    • 11. Kelly D, Conway S, Aminov R. Commensal gut bacteria: mechanisms of immune modulation. Trends Immunol. 26(6), 326–333 (2005).
    • 12. Bajaj JS, Fagan A, Sikaroodi M et al. Liver transplant modulates gut microbial dysbiosis and cognitive function in cirrhosis. Liver Transpl. 23(7), 907–914 (2017).
    • 13. Yuan M, Lin L, Cao H et al. Intestinal microbiota participates in the protective effect of HO-1/BMMSCs on liver transplantation with steatotic liver grafts in rats. Front. Microbiol. 13, 905567 (2022).
    • 14. Annavajhala MK, Gomez-Simmonds A, Macesic N et al. Colonizing multidrug-resistant bacteria and the longitudinal evolution of the intestinal microbiome after liver transplantation. Nat. Commun. 10(1), 4715 (2019).
    • 15. Zhang H, Zhang E, Hu H. Role of ferroptosis in non-alcoholic fatty liver disease and its implications for therapeutic strategies. Biomedicines 9(11), 1660 (2021). • Describes further directions for the treatment of non-alcoholic fatty liver disease and the selection of corresponding drugs that target ferroptosis.
    • 16. Chen J, Li X, Ge C, Min J, Wang F. The multifaceted role of ferroptosis in liver disease. Cell Death Differ. 29(3), 467–480 (2022).
    • 17. Nelson JE, Wilson L, Brunt EM et al. Relationship between the pattern of hepatic iron deposition and histological severity in nonalcoholic fatty liver disease. Hepatology 53(2), 448–457 (2011).
    • 18. Chen Y, Fan H, Wang S, Tang G, Zhai C, Shen L. Ferroptosis: a novel therapeutic target for ischemia-reperfusion injury. Front. Cell. Dev. Biol. 9, 688605 (2021).
    • 19. Li X, Wu L, Tian X et al. miR-29a-3p in exosomes from heme oxygenase-1 modified bone marrow mesenchymal stem cells alleviates steatotic liver ischemia-reperfusion injury in rats by suppressing ferroptosis via iron responsive element binding protein 2. Oxid. Med. Cell Longev. 2022, 6520789 (2022).
    • 20. Wu L, Tian X, Zuo H et al. miR-124-3p delivered by exosomes from heme oxygenase-1 modified bone marrow mesenchymal stem cells inhibits ferroptosis to attenuate ischemia-reperfusion injury in steatotic grafts. J. Nanobiotechnol. 20(1), 196 (2022).
    • 21. Cao H, Wu L, Tian X et al. HO-1/BMMSC perfusion using a normothermic machine perfusion system reduces the acute rejection of DCD liver transplantation by regulating NKT cell co-inhibitory receptors in rats. Stem Cell Res. Ther. 12(1), 587 (2021).
    • 22. Neri A-A, Dontas IA, Iliopoulos DC, Karatzas T. Pathophysiological changes during ischemia-reperfusion injury in rodent hepatic steatosis. In Vivo 34(3), 953–964 (2020).
    • 23. Guicciardi ME, Malhi H, Mott JL, Gores GJ. Apoptosis and necrosis in the liver. Compr. Physiol. 3(2), 977– 1010 (2013).
    • 24. Mu J, Chen Q, Zhu L et al. Influence of gut microbiota and intestinal barrier on enterogenic infection after liver transplantation. Curr. Med. Res. Opin. 35(2), 241–248 (2019).
    • 25. Li S-P, Zhang J-M, Chen X-J et al. Characteristics of changes in double positive CD4CD8 T cells in liver transplantation. Int. Immunopharmacol. 110, 109028 (2022).
    • 26. Schuster S, Cabrera D, Arrese M, Feldstein AE. Triggering and resolution of inflammation in NASH. Nat. Rev. Gastroenterol. Hepatol. 15(6), 349–364 (2018).
    • 27. Deng F, Zhao B-C, Yang X et al. The gut microbiota metabolite capsiate promotes Gpx4 expression by activating TRPV1 to inhibit intestinal ischemia reperfusion-induced ferroptosis. Gut Microbes 13(1), 1–21 (2021).
    • 28. Tarantino G, Finelli C. Systematic review on intervention with prebiotics/probiotics in patients with obesity-related nonalcoholic fatty liver disease. Future Microbiol 10(5), 889–902 (2015).
    • 29. Liu B, Pan X, Liu Z et al. Fecal microbiota as a noninvasive biomarker to predict the tissue iron accumulation in intestine epithelial cells and liver. FASEB J. 34(2), 3006–3020 (2020).
    • 30. Liang D, Leung RK-K, Guan W, Au WW. Involvement of gut microbiome in human health and disease: brief overview, knowledge gaps and research opportunities. Gut Pathog. 10, 3 (2018).
    • 31. Rossi E, Longo F, Barbagallo M et al. Glucose availability enhances lipopolysaccharide production and immunogenicity in the opportunistic pathogen Acinetobacter baumannii. Future Microbiol. 11(3), 335–349 (2016).
    • 32. Lang S, Schnabl B. Microbiota and fatty liver disease-the known, the unknown, and the future. Cell Host Microbe 28(2), 233–244 (2020).
    • 33. Zhang F, Chen D, Zhang L et al. Diaphragma juglandis extracts modifies the gut microbiota during prevention of type 2 diabetes in rats. J. Ethnopharmacol. 283, 114484 (2022).
    • 34. Liang H, Song H, Zhang X et al. Metformin attenuated sepsis-related liver injury by modulating gut microbiota. Emerg. Microbes. Infect. 11(1), 815–828 (2022).
    • 35. Yuan G, Tan M, Chen X. Punicic acid ameliorates obesity and liver steatosis by regulating gut microbiota composition in mice. Food Funct. 12(17), 7897–7908 (2021). • Describes the increase in the population of Anaerotruncus, Faecalibaculim and Mucispirillum, and the decrease in the population of Lactobacillus, Roseburia and Oscillibacter at the genus level induced by high-fat diet.
    • 36. Ren Z, Fan H, Gu S et al. Expression analysis and the roles of the sec1 gene in regulating the composition of mouse gut microbiota. Genes (Basel) 13(10), 1858 (2022).
    • 37. Bajaj JS, Khoruts A. Microbiota changes and intestinal microbiota transplantation in liver diseases and cirrhosis. J. Hepatol. 72(5), 1003–1027 (2020).
    • 38. Koh A, De Vadder F, Kovatcheva-Datchary P, Bäckhed F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell 165(6), 1332–1345 (2016). • Shows how short-chain fatty acids are produced by microbes in the gut and their functions.
    • 39. Liu P, Wang Y, Yang G et al. The role of short-chain fatty acids in intestinal barrier function, inflammation, oxidative stress, and colonic carcinogenesis. Pharmacol. Res. 165, 105420 (2021).
    • 40. Salvi PS, Cowles RA. Butyrate and the intestinal epithelium: modulation of proliferation and inflammation in homeostasis and disease. Cells 10(7), (2021).
    • 41. Das NK, Schwartz AJ, Barthel G et al. Microbial metabolite signaling is required for systemic iron homeostasis. Cell Metab. 31(1), 115–130 (2020).
    • 42. Mayneris-Perxachs J, Cardellini M, Hoyles L et al. Iron status influences non-alcoholic fatty liver disease in obesity through the gut microbiome. Microbiome 9(1), 104 (2021). •• Identifies a significant interplay among the gut microbiome, iron status and liver fat accumulation is revealed, with potential significance for target therapies.