2,020
Views
0
CrossRef citations to date
0
Altmetric
Research Paper

Phocaeicola vulgatus alleviates diet-induced metabolic dysfunction-associated steatotic liver disease progression by downregulating histone acetylation level via 3-HPAA

, , , , , , , , , , , , & ORCID Icon show all
Article: 2309683 | Received 26 Sep 2023, Accepted 19 Jan 2024, Published online: 05 Feb 2024

ABSTRACT

Diet-induced metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent metabolic disorder with limited effective interventions available. A novel approach to address this issue is through gut microbiota-based therapy. In our study, we utilized multi-omics analysis to identify Phocaeicola vulgatus (P. vulgatus) as a potential probiotic for the treatment of MASLD. Our findings from murine models clearly illustrate that the supplementation of P. vulgatus mitigates the development of MASLD. This beneficial effect is partly attributed to the metabolite 3-Hydroxyphenylacetic acid (3-HPAA) produced by P. vulgatus, which reduces the acetylation levels of H3K27 and downregulates the transcription of Squalene Epoxidase (SQLE), a rate-limiting enzyme in steroid biosynthesis that promotes lipid accumulation in liver cells. This study underscores the significant role of P. vulgatus in the development of MASLD and the critical importance of its metabolite 3-HPAA in regulating lipid homeostasis. These findings offer a promising avenue for early intervention therapy in the context of MASLD.

Introduction

Diet-induced metabolic dysfunction-associated steatotic liver disease (MASLD) is a chronic metabolic disease characterized by hepatic steatosis without a history of significant alcohol consumption. It is associated with hepatocellular fat deposition, inflammation, and varying degrees of fibrosis, which can progress to cirrhosis and hepatocellular carcinoma.Citation1 The global prevalence of MASLD is estimated to be 24%, and even higher in some regions, making it the most common liver disease worldwide.Citation2 Despite the high prevalence and increasing impact on global health, there are few approved treatments for MASLD currently.Citation3 Lifestyle changes, including dietary modification and exercise, remain the cornerstone of MASLD management.Citation4,Citation5

In recent years, an increasing number of studies have shown a close relationship between gut microbiota and the development of MASLD.Citation6,Citation7 Gut microbiota interacts with liver through the gut-liver axis, involving specific metabolites such as bile acids (BAs)Citation8 and short-chain fatty acids (SCFAs).Citation9 Animal model-based studies have shown that probiotics, prebiotics, and synbiotics can regulate gut microbiota homeostasis and further effectively improve MASLD, including liver fat content reduction, liver inflammation, and fibrosis alleviation.Citation10–12 In recently published meta-analyses of randomized clinical trials (RCTs) involving probiotic or synbiotic treatments for individuals with MASLD/NASH, microbial therapies have shown a significant association with improved levels of alanine amino transferase (ALT) and aspartate transaminase (AST).Citation13–15

Probiotics may affect the progression of metabolic diseases in multiple ways. Among them, they can even directly affect gene transcription in liver cells through metabolites via the gut-liver axis. For example, supplementation of prebiotics and synbiotics regulates the expression of genes involved in beta-oxidation and fat synthesis, including peroxisome proliferator-activated receptor alpha (PPAR-α), sterol regulatory element-binding protein 1c (SREBP-1c) and malic enzyme (ME).Citation16 However, due to the complexity of the gut microbiome and the interaction among different bacterial groups, there is a relative scarcity of research related to the interplay among gut microbiota, metabolites, and gene transcription in the liver.

To deeper understand the relationship between gut microbiota and MASLD, we established murine models and carried out the longitudinal 16S rRNA gene sequencing at different phases as well as liver transcriptome. Multi-omics analysis showed Phocaeicola vulgatus (P. vulgatus, recently reclassified from Bacteroides vulgatus Citation17) significantly correlated with the progression of MASLD, which might be a potential probiotic for MASLD. Then, the protective role of P. vulgatus for MASLD was determined through murine model. We revealed that P. vulgatus attenuated the development of MASLD through the production of 3-hydroxyphenylacetic acid (3-HPAA), which is the metabolites of P. vulgatus. This metabolite was further discovered to reduce the acetylation status of histones, consequently resulting in decreased expression of squalene epoxidase (SQLE), which effectively inhibits fat accumulation. In this study, we focused on different stages of MASLD and demonstrated that P. vulgatus and its metabolite 3-HPAA could serve as early interventions for MASLD. This finding provided a novel approach to mitigate the development of the disease.

Result

Dynamic alterations of general indicators and gut microbiota during MASLD development

We established a murine model of MASLD using a high-fat, high-carbohydrate (HFHC) diet.Citation18,Citation19 Mice were sacrificed at the fourth, eighth, and sixteenth weeks respectively.Citation20 Their serum, liver tissues, intestinal tissues, and feces samples were collected to investigate the dynamic alterations of general indicators and gut microbiota (). The general indicators included weight gain, hematoxylin-eosin staining (H&E score), as well as the content of ALT, AST, and triglyceride (TG) in the serum. At the fourth week of modeling, the mice in the HFHC group were already heavier than those in the Chow group (), as well as the TG content in the serum (). Moreover, the HFHC group exhibited increased levels of ALT and AST, which are the indicators of liver damage, as compared to the Chow group (). These trends became more pronounced over time. To understand the conditions in liver, H&E and Oil Red O staining assays showed that steatosis had already appeared at the fourth week. As the disease progresses, liver cells showed more ballooning and were even filled with lipid droplets at the sixteenth week (). Sirius red staining reveal that liver tissue also exhibited fibrosis at this time (Supplementary Fig S1). The quantified scores of H&E staining and Oil Red O staining are shown in Supplementary Fig S2. The results indicated that both pathological and functional changes occurred during the early stages of MASLD, highlighting the importance of early intervention in managing this disease. Subsequently, we collected feces from mice at these three end points and performed 16S rRNA gene sequencing. Microbial richness was initially decreased in the fourth week but showed a slight rebound trend afterward based on ɑ-diversity indexes (Chao1, Shannon, and Simpson) (Supplementary Fig S3). Gut microbiota compositional discrimination was observed between mice in the HFHC and the Chow group through principal coordinates analysis (PCoA), utilizing both unweighted and weighted UniFrac (Bray-curtis) distances. It could be found that the gut microbiota structure of these two groups was already distinct at the fourth week, and the differences always present during modeling time (, Supplementary Fig S4; PERMANOVA, p < .05). At the phylum level, the top eleven most abundant gut taxa (including “others”) are listed at three time points. Among all the time points, the abundance of Firmicutes was significantly higher than that of Bacteroidota in the HFHC group, while in the Chow group, the abundance of both phyla was similar. In detail, at the fourth week, Campilobacterota and Proteobacteria were enriched in the HFHC group compared with Chow group, while Actinobacteriota was depleted (). At the eighth week, the tendencies of Proteobacteria, Campilobacterota, and Actinobacteriota were similar to that at the fourth week. Whereas at the sixteenth week, Proteobacteria was depleted in HFHC group compared with Chow group, which was opposite to the previous trend. This suggested that in the late stage of MASLD, the structure of gut microbiota might not fully represent the overall microbial composition during the progression of MASLD. To better show the trend of microbial changes, heatmap and cluster analysis of the 16S rRNA gene sequencing data at three endpoints were performed. Heatmap showed that one cluster of the microbiota was clearly separated from others, which had the characteristic of a significant decrease in content as the disease progressed (, Cluster 1). These gut microbiotas were screened out for subsequent analysis (Supplementary Table S1). Interestingly, we found that there was a sharp change in the abundance of gut microbiota at the eighth week, which suggested interventions targeting the gut microbiota may have a specific temporal window.

Figure 1. Dynamic alterations of liver pathological change and gut microbiota during MASLD development. (a) Schematic diagram of the HFHC diet-induced MASLD model. (b-e) body weight gain, serum TG levels, ALT levels, and AST levels of two groups of mice at three time points. (f, g) H&E staining images and oil red O staining images of liver sections. The arrow represents the typical ballooning hepatocytes in the 16w HFHC group. Scale bar 100 μm. (h-i) PCoA clustering plot of 16S rRNA gene sequencing data and relative abundance of microbial phyla at three time points. (j) OTU clustering heatmap based on the progression of MASLD modeling. The ‘Ward.D’ method was used for clustering, and the ‘Euclidean’ method was used for distance calculation. Cluster 1, marked by blue frame, were selected further analysis. Data are presented as mean±SEM. *p < .05, **p < .01.

Figure 1. Dynamic alterations of liver pathological change and gut microbiota during MASLD development. (a) Schematic diagram of the HFHC diet-induced MASLD model. (b-e) body weight gain, serum TG levels, ALT levels, and AST levels of two groups of mice at three time points. (f, g) H&E staining images and oil red O staining images of liver sections. The arrow represents the typical ballooning hepatocytes in the 16w HFHC group. Scale bar 100 μm. (h-i) PCoA clustering plot of 16S rRNA gene sequencing data and relative abundance of microbial phyla at three time points. (j) OTU clustering heatmap based on the progression of MASLD modeling. The ‘Ward.D’ method was used for clustering, and the ‘Euclidean’ method was used for distance calculation. Cluster 1, marked by blue frame, were selected further analysis. Data are presented as mean±SEM. *p < .05, **p < .01.

Multi-omics analysis provides evidence for the involvement of phocaeicola vulgatus in the development of MASLD

In addition to 16S rRNA gene sequencing above, a series of transcriptome sequencing of liver tissues was also conducted. Through PCoA analysis, it was found that the differences in liver cell transcriptome appeared slightly later than the differences in 16s rRNA gene sequencing (). Similar to alterations in the microbiota, transcriptome clustering analysis revealed two series of genes that showed gradual changes over time as the disease progressed (). Gene lists in these two clusters were shown in the Supplementary Table S2 , S3. Interestingly, a sharp change was also observed from the eighth week in transcriptome data. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed the decreased genes were mainly concentrated in the cGMP-PKG pathway (, Supplementary Fig S5). In contrast, upregulated genes were mainly concentrated in metabolic pathways (, Supplementary Fig S6), including heat shock protein family D (Hsp60) member 1 (HSPD1), nicotinamide phosphoribosyltransferase (NAMPT), apolipoprotein A4 (APOA4), and other genes which played crucial roles in MASLD development.Citation21–23 To further explore the relationship between gene transcription and gut microbiota structure, we specifically focused on the down-regulated microbiota (Cluster 1) and the up-regulated genes (Cluster B). Employing a Spearman correlation analysis and utilizing the ‘Ward.d2’ clustering method, we identified strongly correlated microbiota-gene pairs, comprising 31 bacterial taxa at the genus level and 238 genes (). Subsequently, through stringent criteria (r < −0.6 and correlation test p < .05), we refined our selection to 27 bacterial taxa at the genus level. Detailed information on their correlation with gene expression, along with the correlation test results, is available in Supplementary Table S4. Additionally, general indicators (including weight gain, ALT and TG content, H&E staining score) were linked to the microbiota content using a Redundancy (RDA) analysis (). It can be found that the blue-labeled microbiota, including Parabacteroides, Helicobacter, Akkermansia, Bacteroides, Bifidobacterium and Dubosiella, were negatively correlated with general indicators. Two genera of microbiota: Parabacteroides and Bacteroides were identified by overlapping the results of Spearman analysis and the RDA analysis (Supplementary Fig S7a), and their strongly correlated genes were marked in red in Supplementary Table S4. A quantitative reverse transcription PCR (RT-qPCR) analysis was conducted at the species level to evaluate the colonization of these two genera. Seven bacterial species belonging to these genera were analyzed, including P. vulgatus, which was historically classified under the genus Bacteroides but has recently been reclassified as a member of the Phocaeicola genus (not yet reflected in the 16S rRNA analysis annotations). In these seven bacterial species, P. vulgatus emerged as the most abundant bacterium (Supplementary Fig S7b). Interestingly, when examined the changes in the abundance of different species within Parabacteroides and Bacteroides at three time points, we observed that only P. vulgatus and B. acidifaciens showed consistently lower abundances compared to the Chow group across all three time points during HFHC modeling, beginning at the fourth week. Furthermore, the abundance of P. vulgatus in the Chow group remained relatively stable over time (Supplementary Fig S8). We speculated that P. vulgatus might confer an important protective role against MASLD in the early stage.

Figure 2. Dynamic alterations of liver transcriptome and multiomics analysis. (a-c) PCA plot of RNA sequencing data at three time points. (d) Heatmap of RNA clustering analysis based on the progression of disease. Clustering was performed using the ‘Median’ method and the ‘Manhattan’ distance calculation method. The ‘cluster B’, highligted by the red frame, was used for further analysis. (e) KEGG pathway analysis of the down-regulated genes. (f) KEGG pathway analysis of the up-regulated genes. (g) Spearman correlation heatmap of the up-regulated genes’ cluster (cluster B) and the down-regulated bacteria’s cluster (cluster 1). Clustering was performed using the ‘Ward.D2’ method and the ‘Manhattan’ distance calculation method. (h) RDA plot showing the correlation between the abundance of bacteria and general indicators. Permutation test showed that the RDA model was statistically significant (p < .05).

Figure 2. Dynamic alterations of liver transcriptome and multiomics analysis. (a-c) PCA plot of RNA sequencing data at three time points. (d) Heatmap of RNA clustering analysis based on the progression of disease. Clustering was performed using the ‘Median’ method and the ‘Manhattan’ distance calculation method. The ‘cluster B’, highligted by the red frame, was used for further analysis. (e) KEGG pathway analysis of the down-regulated genes. (f) KEGG pathway analysis of the up-regulated genes. (g) Spearman correlation heatmap of the up-regulated genes’ cluster (cluster B) and the down-regulated bacteria’s cluster (cluster 1). Clustering was performed using the ‘Ward.D2’ method and the ‘Manhattan’ distance calculation method. (h) RDA plot showing the correlation between the abundance of bacteria and general indicators. Permutation test showed that the RDA model was statistically significant (p < .05).

P. vulgatus confers a protective role against MASLD

To verify the function of P. vulgatus on MASLD, an eight-week study on antibiotic-depleted ApoE−/− mice was conducted initially. ApoE−/− mice are genetically engineered mice that lack a functional copy of the apolipoprotein E (ApoE) and are commonly used in research related to metabolic disorders such as MASLD.Citation24,Citation25 The choice of ApoE−/− mice in our study is further supported by their characteristic lipid profile, where the absence of functional ApoE leads to alterations in cholesterol distribution, mimicking aspects of dyslipidemia observed in MASLD patients.Citation26 This unique physiological trait makes ApoE−/− mice a suitable model for investigating the interplay between P. vulgatus and MASLD. Mice were given HFHC diet for six weeks after two weeks of antibiotic clearance (Supplementary Fig S9). Meanwhile, the reinforced clostridial agar (RCM) group was given the RCM medium as negative control. The group that received P. vulgatus (1 × 109 cfu/100 μL) was referred to as the P. vul group (, Supplementary Fig S10). Mice in P. vul group showed lower TG levels compared to the RCM group at the end of the modeling time (, Supplementary Fig S11). Hepatic ballooning degree were reduced in the mice fed with P. vulgatus according to H&E staining (). Oil Red O staining also showed a significant reduction in hepatic fat deposition in P. vul group (). In our previous study, we used this model to investigate the therapeutic effect of Akkermansia muciniphila on MASLD, and we found that in this model, P. vulgatus had more stable performance (Supplementary Fig S12).

Figure 3. P. vulgatus alleviated disease progression in MASLD. (a) Schematic diagram of HFHC-induced APOE−/− mice orally gavaged with P. vulgatus. (b-e) body weight, TG levels, ALT levels, and AST levels of two groups of mice at the end of the modeling time. (f, g) H&E staining and oil red O staining of liver sections at the end of the modeling period. Scale bar 100 μm, 200 μm. (h) Schematic diagram of HFHC-induced C57BL/6 mice orally gavaged with P. vulgatus. (i-l) body weight, TG levels, ALT levels, and AST levels of four groups of mice at the end of the modeling time. (m) H&E staining of liver sections from four groups of mice at the end of the modeling period. (n) Nile red staining, oil red O staining and Sirius red staining of liver sections. Scale bar 100 μm. Data are presented as mean±SEM. *p < .05, **p < .01, ns, no significance.

Figure 3. P. vulgatus alleviated disease progression in MASLD. (a) Schematic diagram of HFHC-induced APOE−/− mice orally gavaged with P. vulgatus. (b-e) body weight, TG levels, ALT levels, and AST levels of two groups of mice at the end of the modeling time. (f, g) H&E staining and oil red O staining of liver sections at the end of the modeling period. Scale bar 100 μm, 200 μm. (h) Schematic diagram of HFHC-induced C57BL/6 mice orally gavaged with P. vulgatus. (i-l) body weight, TG levels, ALT levels, and AST levels of four groups of mice at the end of the modeling time. (m) H&E staining of liver sections from four groups of mice at the end of the modeling period. (n) Nile red staining, oil red O staining and Sirius red staining of liver sections. Scale bar 100 μm. Data are presented as mean±SEM. *p < .05, **p < .01, ns, no significance.

For further investigation of the specific therapeutic effect of P. vulgatus on MASLD, a sixteen-week HFHC modeling study on C57BL/6 mice were conducted. Given the significant alteration of the transcriptome and microbiota at the eighth week, we divided the mice into four groups: the TP group received P. vulgatus throughout the modeling period, the F8w group received for the first eight weeks, the B8w group received for the next eight weeks, and the RCM group served as a negative control (). It was found that the weight gain in the TP, F8w and B8w groups was significantly lower, especially in the F8w group (). Similar results were also found in the serum TG (), ALT and AST levels (). The pathological staining including H&E and oil red O demonstrated that the F8w group had the least fat deposition and collagen fiber deposition among these four groups ( and Supplementary Fig S13). It could be concluded that treatment with P. vulgatus confers a protective role against MASLD, and early treatment may improve some aspects of whole-body metabolism.

P. vulgatus relieves MASLD via three-hydroxyphenylacetic acid

It is well known that the gut microbiota generally influence the development of disease through its metabolites.Citation27 To further investigate the functional metabolite of gut microbiota, the feces from mice in Chow and HFHC group were subjected to metabolome analysis. It can be observed that in the HFHC group, the content of short chain fatty acids (SCFAs) was significantly reduced, while the content of amino acids was significantly increased (). The volcano plot shows ten substances with decreased levels in the HFHC group (, Supplementary Fig S14). Among these metabolites, 3-HPAA is the intestinal metabolite of quercetin, which has been widely reported to be inversely associated with the progression of MASLD.Citation28,Citation29 Its content was found to be reduced in the HFHC group (). However, it was unclear whether the metabolites of P. vulgatus play a role in reducing fat accumulation. Therefore, fecal samples were collected from mice after oral gavage of P. vulgatus, then dissolved with PBS to form fecal supernatant. The fecal supernatant was applied to the human normal liver cell line MIHA and the mouse normal liver cell line AML12 in vitro (). The fat deposition in liver cells was reduced after treatment with the supernatant of P. vul group, despite being subjected to high-fat modeling (, Supplementary Fig S15-S16). In order to find out whether it was 3-HPAA in the metabolite that played a role, in vitro experiments were conducted and showed that compared to other substances such as quercetin, γ-aminobutyric acid (GABA) and pentadecanoic acid (C15:0), 3-HPAA could inhibit lipid deposition in liver cells more significantly (, Supplementary Fig S17). At the same time, in in vitro experiments, after adding 3-HPAA, the transcription of key genes that contributed to fatty acid metabolism in the liver cell lines MIHA and AML12 was also reduced (Supplementary Fig S18). To confirm that 3-HPAA is indeed a metabolite of P. vulgatus, we introduced quercetin into the culture medium of P. vulgatus and analyzed its metabolites with high performance liquid chromatography (HPLC) (). After 24 hours of cultivation, we observed a decrease in the quercetin content and a significant increase in its metabolite, 3-HPAA (). In in vivo experiments, after oral gavage of P. vulgatus, the content of 3-HPAA in feces of mice also increased significantly (). In conclusion, we demonstrated that P. vulgatus confers a protective role against MASLD through the metabolite 3-HPAA.

Figure 4. P. vulgatus confers a protective role against MASLD through the metabolite 3-HPAA. (a) Distribution of metabolite content of different types in feces between the high-fat diet group and the normal diet group. (b) Volcano plot of substances with significant differences between the two groups. (c) Bar graph of the content of 3-HPAA. (d) Schematic diagram of treatment with fecal supernatant on liver cell lines. (e) Oil red O and nile red staining of AML12 treated with fecal supernatant. PAOA: high-fat modeling (palmitic acid and oleic acid); RCM: the fecal supernatant of mice in the RCM group based on high-fat modelling; P. vul: the fecal supernatant of mice in the P. vul group based on high-fat modeling. Scale bar 50 μm. (f) Oil red O and nile red staining of cell lines treated with different candidate metabolites. C15:0: pentadecanoic acid. Scale bar 50 μm. (g) Schematic diagram of HPLC detection of P. vulgatus metabolites and the standard curve of 3-HPAA. HPLC peaks and statistics of quercetin(h) and 3-HPAA(i) in these two groups of bacterial culture medium. (j) HPLC peaks and statistics of 3-HPAA in the feces of two groups of mice. The red box represents the characteristic peak. Data are presented as mean±SEM. *p < .05, **p < .01.

Figure 4. P. vulgatus confers a protective role against MASLD through the metabolite 3-HPAA. (a) Distribution of metabolite content of different types in feces between the high-fat diet group and the normal diet group. (b) Volcano plot of substances with significant differences between the two groups. (c) Bar graph of the content of 3-HPAA. (d) Schematic diagram of treatment with fecal supernatant on liver cell lines. (e) Oil red O and nile red staining of AML12 treated with fecal supernatant. PAOA: high-fat modeling (palmitic acid and oleic acid); RCM: the fecal supernatant of mice in the RCM group based on high-fat modelling; P. vul: the fecal supernatant of mice in the P. vul group based on high-fat modeling. Scale bar 50 μm. (f) Oil red O and nile red staining of cell lines treated with different candidate metabolites. C15:0: pentadecanoic acid. Scale bar 50 μm. (g) Schematic diagram of HPLC detection of P. vulgatus metabolites and the standard curve of 3-HPAA. HPLC peaks and statistics of quercetin(h) and 3-HPAA(i) in these two groups of bacterial culture medium. (j) HPLC peaks and statistics of 3-HPAA in the feces of two groups of mice. The red box represents the characteristic peak. Data are presented as mean±SEM. *p < .05, **p < .01.

3-HPAA alleviated the progression of MASLD

To further explore the function of 3-HPAA in alleviating the progression of MASLD, 3-HPAA was added into the culture medium of liver cell lines at various concentrations (, Supplementary Fig S19). In the condition of high fat modeling, addition of 3-HPAA significantly reduced the degree of hepatocytic fat deposition in a dose-dependent manner up to 200 μM (, Supplementary Fig S20), whereas this effect was not observed with the treatment of quercetin (). At the same time, the transcription levels of the lipid metabolism-related genes were significantly decreased (), which indicated 3-HPAA, rather than quercetin, could attenuate the progression of the fat deposition. Mice which were fed with the HFHC diet were administered 3-HPAA for sixteen weeks to conduct in vivo experiments (). The intake of 3-HPAA during the modeling process did not affect the food intake of mice (Supplementary Fig S21). After administering 3-HPAA to mice, we found that the 3-HPAA content in the mouse serum increased (Supplementary Fig S22). After sixteen weeks of oral administration, the body weight gain and serum TG levels in mice fed with 3-HPAA were significantly lower than those fed with PBS (). In addition, hepatic injury in 3-HPAA group was also obviously reduced (). H&E, Oil red O and Sirius red staining showed that 3-HPAA treatment significantly reduced the ballooning of liver cell, declined lipid deposition and reduced liver fibrosis (, Supplementary Fig S23). Results above confirmed that 3-HPAA inhibited the progression of MASLD and regulated the transcription of lipid metabolism-related genes.

Figure 5. 3-HPAA alleviated the progression of MASLD. (a) Schematic diagram of the effect of 3-HPAA on liver cells in vitro. Oil red O staining is shown in (b-c). Scale bar 50 μm. (d-e) transcriptional effects of different concentrations of 3-HPAA on genes related to lipid metabolism in two cell lines. (f) Schematic diagram of 3-HPAA gavage in mice. (g-j) weight gain, TG levels, ALT levels and AST levels of two groups of mice after gavage. (k) H&E staining images of two groups of mice after gavage. Oil red O staining images(l) of four groups of mice after modeling and Nile red staining(m). Scale bar 200 μm. Data are presented as mean±SEM. *p < .05, **p < .01.

Figure 5. 3-HPAA alleviated the progression of MASLD. (a) Schematic diagram of the effect of 3-HPAA on liver cells in vitro. Oil red O staining is shown in (b-c). Scale bar 50 μm. (d-e) transcriptional effects of different concentrations of 3-HPAA on genes related to lipid metabolism in two cell lines. (f) Schematic diagram of 3-HPAA gavage in mice. (g-j) weight gain, TG levels, ALT levels and AST levels of two groups of mice after gavage. (k) H&E staining images of two groups of mice after gavage. Oil red O staining images(l) of four groups of mice after modeling and Nile red staining(m). Scale bar 200 μm. Data are presented as mean±SEM. *p < .05, **p < .01.

3-HPAA promotes transcription of SQLE through affecting histone acetylation

Three-hydroxyphenylacetic acid is a type of phenolic acid that can easily penetrated the lipid bilayer, then directly affect the epigenetic modifications of genes, thereby affecting their transcription.Citation30,Citation31 Therefore, we speculated that 3-HPAA might have an epigenetic regulatory effect on genes related to fat metabolism, and one important form of such epigenetic modification is histone acetylation.Citation32 To validate this hypothesis, the acetylation levels of vital histones were measured in hepatic cell lines after treated with 3-HPAA in vitro. It was found that the acetylation levels of H3K27 and H3K9 were obviously decreased after the treatment of 3-HPAA, which could be rescued by the histone acetylation activator CTPB (, Supplementary Fig S24). Furthermore, Oil Red O staining demonstrated that CTPB could reverse the inhibition of lipid accumulation caused by 3-HPAA (, Supplementary Fig S25–27). In mice fed with HFHC diet, we found that oral gavage of P. vulgatus and 3-HPAA both significantly reduced H3K27 acetylation (). To identify the critical downstream target genes of 3-HPAA, we screened out genes which highly correlated with P. vulgatus (|r| > 0.4,p<.05, Supplementary Table S5). Analysis of KEGG implied that apart from the overall metabolic pathway, the steroid biosynthesis pathway was particularly significant. Among genes in this pathway, the transcription of the squalene epoxidase (SQLE) and NAD(P) dependent steroid dehydrogenase-like (NSDHL) both decreased with the addition of 3-HPAA, and these changes were both rescued by CTPB (). Previous study has demonstrated that SQLE acts as a connection between steroid synthesis and lipid metabolism, thus accelerates the progression of MASLD.Citation33 In light of this, our study sought to investigate the impact of 3-HPAA on SQLE expression levels. Encouragingly, results showed that treatment with 3-HPAA significantly decreased the expression of SQLE in cells, and this effect could be reversed by CTPB ( and Supplementary Fig S28). In Oil Red O staining, knocking down SQLE led to a reduction in hepatic fat deposition, while its overexpression promoted its deposition, providing compelling evidence for the promoting effect of SQLE on MASLD ( and Supplementary Fig S29). Furthermore, in in vivo experiment, gavage of P. vulgatus and 3-HPAA both markedly reduced the expression of SQLE in the liver (). The regulation of histone acetylation is achieved through the combined action of acetyltransferases (HATs) and deacetylases (HDACs).Citation34 To confirm that 3-HPAA is indeed capable of reducing HAT activity to modulate histone acetylation, we measured HAT enzyme activity. Results showed that HAT activity in cells significantly decreased after treatment with 3-HPAA and increased with CTPB (). To explore the direct binding between 3-HPAA and HAT, AutoDock Vina was employed to perform docking simulations and calculate the interaction between 3-HPAA and E1A binding protein P300 (EP300), which is the key protein of HAT.Citation35,Citation36 There were multiple groups of residues used to form interactions between the receptor protein EP300 and ligand 3-HPAA, such as the hydrogen bond formed by TRP1466 of EP300 and 3-HPAA. With these interaction forces, the binding energy of protein-ligand complex was −5.9 kcal/mol, which shows a good performance ( and Supplementary Table S6). Overall, these results suggested that 3-HPAA, the metabolism of P. vulgatus, affected the transcription levels of downstream genes such as SQLE by inhibiting the activity of HAT and reducing the level of H3K27 acetylation ().

Figure 6. 3-HPAA affects the transcription of downstream genes by histone acetylase. (a) Western blot analysis of histone acetylation in two liver cell lines treated with 3-HPAA and the HAT inhibitor CTPB. (b) Oil red O staining of two liver cell lines treated with 3-HPAA and CTPB under the high-fat molding by PA and OA. Scale bar 50 μm. (c) Immunohistochemical staining of H3K27ac in liver tissue sections from four groups of mice with time-divided gavage of P. vulgatus. scale bar 200 μm. (d) The KEGG of genes strongly related with P. vulgatus. (e) The transcriptional changes of genes related to cholesterol biosynthesis pathway were investigated in AML12 cell line after treatment with 3-HPAA and CTPB. (f) Western blot of the expression of SQLE in two liver cell lines treated with 3-HPAA and the HAT inhibitor CTPB. (g) Oil red O staining on two liver cell lines treated with siRNA and overexpression of SQLE under high-fat conditions. The scale bar is 50 μm. (h) Immunofluorescence staining of SQLE in liver tissue sections from four groups of mice with time-divided gavage of P. vulgatus. Scale bar 50 μm. (i) Activity assay of HAT inhibition by 3-HPAA. (j) The protein-ligand interaction figure was generated by PyMOL. The EP300 protein is represented as a slate cartoon model, ligand is shown as a cyan stick, and their binding sites are shown as magentas stick structures. Nonpolar hydrogen atoms are omitted. The hydrogen bond, ionic interactions, and hydrophobic interactions are depicted as yellow, magentas and green dashed lines, respectively. PA: palmitic acid; OA: oleic acid. Data are presented as mean±SEM. *p < .05, **p < .01.ns, no significance.

Figure 6. 3-HPAA affects the transcription of downstream genes by histone acetylase. (a) Western blot analysis of histone acetylation in two liver cell lines treated with 3-HPAA and the HAT inhibitor CTPB. (b) Oil red O staining of two liver cell lines treated with 3-HPAA and CTPB under the high-fat molding by PA and OA. Scale bar 50 μm. (c) Immunohistochemical staining of H3K27ac in liver tissue sections from four groups of mice with time-divided gavage of P. vulgatus. scale bar 200 μm. (d) The KEGG of genes strongly related with P. vulgatus. (e) The transcriptional changes of genes related to cholesterol biosynthesis pathway were investigated in AML12 cell line after treatment with 3-HPAA and CTPB. (f) Western blot of the expression of SQLE in two liver cell lines treated with 3-HPAA and the HAT inhibitor CTPB. (g) Oil red O staining on two liver cell lines treated with siRNA and overexpression of SQLE under high-fat conditions. The scale bar is 50 μm. (h) Immunofluorescence staining of SQLE in liver tissue sections from four groups of mice with time-divided gavage of P. vulgatus. Scale bar 50 μm. (i) Activity assay of HAT inhibition by 3-HPAA. (j) The protein-ligand interaction figure was generated by PyMOL. The EP300 protein is represented as a slate cartoon model, ligand is shown as a cyan stick, and their binding sites are shown as magentas stick structures. Nonpolar hydrogen atoms are omitted. The hydrogen bond, ionic interactions, and hydrophobic interactions are depicted as yellow, magentas and green dashed lines, respectively. PA: palmitic acid; OA: oleic acid. Data are presented as mean±SEM. *p < .05, **p < .01.ns, no significance.

Figure 7. The mechanism diagram of P. vulgatus alleviating MASLD. P. vulgatus converts quercetin into 3-HPAA in the intestine. 3-HPAA inhibits HAT in liver cells, resulting a decreased the acetylation level of H3K27, and further leading the inhibition of SQLE transcription, ultimately slowing down intracellular lipid deposition.

Figure 7. The mechanism diagram of P. vulgatus alleviating MASLD. P. vulgatus converts quercetin into 3-HPAA in the intestine. 3-HPAA inhibits HAT in liver cells, resulting a decreased the acetylation level of H3K27, and further leading the inhibition of SQLE transcription, ultimately slowing down intracellular lipid deposition.

Discussion

Metabolic dysfunction-associated steatotic liver disease is a highly prevalent metabolic disorder and the incidence is likely to increase with further social development. Previous study reported the gut microbiota represent an environmental factor associated with the development of MASLD. However, how the specific microbiota is involved in the progression of MASLD has not been studied thoroughly. Here, we have identified the crucial role of the microbiota P. vulgatus in the progression of MASLD, especially in early stage. Furthermore, we have demonstrated that this bacterium reduced fat accumulation by down regulating the expression of SQLE through its metabolite 3-HPAA, which exerted its effect by inhibiting HAT enzyme activity and reducing acetylation levels of H3K27. These results revealed the interplay among gut microbiota, metabolites, and gene transcription in the liver.

In this study, a multi-omics approach was used to explore the functional microbiota. The integrational analysis of transcriptomics and 16S rRNA gene sequencing data allowed for a more comprehensive biological interpretation of the microbiota. Through correlation analysis, we identified several bacteria associated with MASLD development, including Parabacteroides, Dubosiella, Akkermansia, Phocaeicola. Among which, the Akkermansia muciniphila has received considerable attention in recent research, particularly for its potential application in MASLD.Citation37,Citation38 Yet, the role of other selected bacteria remains unclear and deserve further investigation. P. vulgatus is a genus of gram-negative bacteria, which can break down the complex carbohydrates in human, such as glycans, to generate energy and other essential nutrients.Citation39 Some studies have shown that Phocaeicola can alleviate atherosclerosisCitation40 and colitis,Citation41 but its underlying mechanism remains obscure. As one of the potential next-generation probiotics, P. vulgatus seems has both positive and negative effects. In most studies, it is beneficial for maintaining intestinal barrier, reducing DSS-induced colitis,Citation41–43 and preventing atherosclerosis.Citation40 However, some studies have shown that P. vulgatus may positively associated with metabolic disease, such as type 2 diabetes.Citation44–46 These disparities may arise from variations in study populations, methodologies, and sequencing techniques. Gut microbiota comprising numerous species with distinct interactions further complicates the picture. In addition, as found in our study, changes in microbiota are not monotonic in disease, and different sampling time points during the development of the disease may also lead to different research results. Given these discrepancies, further research is imperative. Future investigations should delve into the interaction among microbiota and consider broader gut microbiota factors. This nuanced approach promises a better understanding of the role of P. vulgatus in metabolic health and potential therapeutic applications.

As for the exploration of the specific mechanisms of P. vulgatus, we further demonstrated that it act through its metabolites 3-HPAA. There is already a large body of literature on the therapeutic effects of bacteria metabolites on MASLD, such as butyrateCitation47 and bile acids.Citation48 3-HPAA was one of the metabolites of quercetin, which abundant in human dietary.Citation28 One study has found that the Parabacteroides was positively correlated with 3-HPAA, and that the microbial metabolite 3-HPAA can protect cardiac dysfunction by activating NRF2.Citation49 In addition, studies have shown that 3-HPAA can protect the liver from oxidative damage.Citation50–52 However, there is little literature to verify the exact relationship between bacterial strains and 3-HPAA metabolism, and there is no study on the alleviation effect of 3-HPAA in MASLD. In our study, we identified 3-HPAA as a metabolite of P. vulgatus through HPLC and found that it can prevent the progression of MASLD. Our data suggested that 3-HPAA might serve as a candidate drug for subsequent intervention in MASLD, with great application potential.

Histone acetylation is an epigenetic modification that regulates gene transcription and is closely related to the development of MASLD.Citation53 Acetylation can relax the structure of histone proteins, making DNA more accessible for transcription.Citation54 Previous studies have reported acetylation of histone promoted the expression of genes involved in fatty acid synthesis in hepatocytes, such as acyl-CoA synthase 5 (ACSL5)Citation55 and the fatty acid translocase CD36.Citation56 Overexpression of these genes leads to the accumulation of fatty acids in hepatocytes. The acetylation of H3K27 is one of the common types of histone acetylation.Citation57 Studies have shown that the gene expression influenced by the acetylation of H3K27 is closely related to the progression of MASLD.Citation58 Our study has uncovered a novel function of 3-HPAA in regulating histone acetylation, filling a gap in our understanding of the biological effects of 3-HPAA. As the downstream of 3-HPAA, Sqle is a key enzyme in the cholesterol biosynthesis pathway, involved in the oxidation of squalene.Citation59 In this study, we showed SQLE could also be regulated by histone acetylation.

Several limitations exist in the present study. Firstly, the use of antibiotics to deplete the gut microbiota may not entirely replicate a germ-free environment in the mice. Subsequent experiments with germ-free mice would provide a more accurate model. Secondly, our analysis relied on time-series heatmaps to depict changes in the gut microbiota of Chow group mice over time. While insightful, this approach may not fully capture nuanced temporal trends in microbial composition. Future research could explore more sophisticated methods to elucidate dynamic shifts in gut microbiota over time. In addition, to study the specific metabolites of the target bacterium, a relatively single bacterial specie was involved in the intervention. We also focused on a specific bacterium, P. vulgatus, to produce 3-HPAA, without considering the potential contribution of other microbial groups to 3-HPAA metabolism. It is essential to recognize that the gut microbiota is a complex ecosystem with multiple species capable of metabolizing various compounds. Regarding metabolite analysis, this study did not comprehensively encompass all gut metabolites, including those associated with quercetin, such as 3,4-dihydroxyphenylacetic acid. In terms of application of our findings, the starting point of this experiment was the modeling in mice, the experimental results have not been validated in the human. The situation in the human body is more complex, and more follow-up experiments are needed.

In summary, this study provided novel evidence that P. vulgatus confers a protective role against MASLD through the production of its metabolite 3-HPAA, which, in turn, reduces the acetylation of histones in liver cells. This reduction in histone acetylation affects the transcription of genes involved in lipid metabolism, such as SQLE. These findings highlight the relationship between gut microbiota and gene transcription in the liver.

Materials and methods

Animals and diets

All animals were kept in specific pathogen-free (SPF) conditions with a constant temperature of 20–22°C and a 12-hour light-dark cycle. Six mice were housed per cage. Autoclaved corn cob bedding was used as the substrate. Bedding was replaced and cages were cleaned twice a week.

Male C57BL/6J mice (six weeks old) were purchased from Charles River Laboratories and bred in the Laboratory Animal Center of Zhejiang University (Zhejiang, China). To establish the murine MASLD model, mice were subjected to a two-week course of antibiotics at 6-weeks old (ampicillin 1 g/L, gentamicin 1 g/L, metronidazole 1 g/L, and vancomycin 0.5 g/L)Citation60,Citation61. After antibiotic cleaning, mice were fed chow diet and high-fat, high-calorie fructose (HFHC) diet which was reported to induce murine MASLD.Citation62 HFHC diet (TrophicDiet, Nantong, China, Ltd, China) contains 5.56 kcal/g, the Chow diet (TrophicDiet, Nantong, China, Ltd, China) contains 4.11 kcal/g. The nutrient composition of the ingredients and their proportions in the diet are shown in . Drinking water was supplemented with fructose (55%) 23.1 g/L and sucrose (45%) 18.9 g/L. Mice were sacrificed and sampled at weeks 0, 4, 8, and 16, respectively.Citation20

Table 1. Ingredients table of HFHC diet.

In intervention experiments, mice were fed with P. vulgatus (for varying durations) or 3-HPAA after antibiotic clearance and then fed with HFHC diet until week 16. Mice were maintained under a 12-hour light/12-hour dark cycle. Body weight and food intake of each group of mice were measured weekly. At the end of the experiment, fecal samples were collected from each group of mice for bacterial 16S rRNA gene sequencing. Mice were fasted for 12 hours, and blood was collected from the retroorbital sinus. Body and visceral weights were recorded. Liver was rapidly excised, weighed, and divided into three parts, one part was fixed in 4% paraformaldehyde for histological staining, another part was snap-frozen in liquid nitrogen for frozen sectioning, and the last part was stored at −80°C for further experiments. Mice were euthanized by cervical dislocation.

Eight-week-old male ApoE−/− mice were purchased from the Nanjing GemPharmatech Company (Nanjing, China) and subjected to the same antibiotic cocktail described above, along with a HFHC diet for six weeks.Citation24,Citation63 In intervention experiments, mice were fed with P. vulgatus for six weeks. All subsequent operations were the same as for C57BL/6J mice.

All animal protocols were approved by Zhejiang University Experimental Animal Ethics Committee and used the ARRIVE1reporting guidelines.Citation64

Additional methods are provided in supplementary file.

Authors’ contributions

SX Jin and P Chen were involved in study design, performed experiments and drafted the manuscript; J Yang performed original draft and review & editing; DG Li, QM Xia, XL Liu and YF Tong performed animal experiments; WH Yu performed histological evaluation; YY Zhang performed original draft and review & editing; YL Li, YX Li, GQ Chen performed review & editing; XX Fan, H Lin designed, supervised the study and revised the manuscript.

Data availability material

The datasets analyzed in our study are openly available in figshare at 10.6084/m9.figshare.23622366.

Supplemental material

Table S3 Up regulated genes.docx

Download MS Word (118.9 KB)

Table S5 Genes strongly relate with P vulgatus.docx

Download MS Word (21.7 KB)

Supplementary materials clean.docx

Download MS Word (5.2 MB)

Table S1 Enriched microbial communities.docx

Download MS Word (35.3 KB)

Table S2 Down regulated genes.docx

Download MS Word (96.6 KB)

Table S7 Primers used in this study.docx

Download MS Word (15 KB)

Table S4 Strong related gene bacterium pairs clean.docx

Download MS Word (147.2 KB)

Table S6 Binding site of 3HPAA and EP300.docx

Download MS Word (13.4 KB)

Acknowledgments

We thank Xiaoli Hong and Chao Bi from the Core Facilities, Zhejiang University School of Medicine for their technical support. We also thank Wei Liu and Jiaqing Wang from Zhejiang Academy of Agricultural Sciences for their technical support.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Supplementary material

Supplemental data for this article can be accessed online at https://doi.org/10.1080/19490976.2024.2309683

Additional information

Funding

This work was supported by the National Natural Science Foundation of China [81874059 and 82102105]; Natural Science Foundation of Zhejiang Province [LQ22H160017, LQ21H030012]; Medical and Health Research Project of Zhejiang Province [2019325792].

References

  • Papatheodoridi M, Cholongitas E. Diagnosis of Non-alcoholic Fatty liver disease (NAFLD): Current concepts. Curr Pharm Des. 2018;24(38):4574–19. doi:10.2174/1381612825666190117102111. PubMed PMID: 30652642.
  • Younossi Z, Anstee QM, Marietti M, Hardy T, Henry L, Eslam M, George J, Bugianesi E. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nat Rev Gastroenterol Hepatol. 2018 Jan;15(1):11–20. doi:10.1038/nrgastro.2017.109. PubMed PMID: 28930295.
  • Muthiah MD, Sanyal AJ. Current management of non-alcoholic steatohepatitis. Liver Int. 2020 Feb;40 Suppl 1(Suppl 1):89–95. doi:10.1111/liv.14355. PubMed PMID: 32077609; PubMed Central PMCID: PMCPMC7654093.
  • Raza S, Rajak S, Upadhyay A, Tewari A, Sinha, RA. Current treatment paradigms and emerging therapies for NAFLD/NASH. Front Biosci. 2021 Jan 1;26(2):206–237. doi:10.2741/4892. PubMed PMID: 33049668; PubMed Central PMCID: PMCPMC7116261.
  • Powell EE, Wong VW, Rinella M. Non-alcoholic fatty liver disease. Lancet. 2021 Jun 5;397(10290):2212–2224. doi:10.1016/S0140-6736(20)32511-3. PubMed PMID: 33894145.
  • Kim BS, Jeon YS, Chun J. Current status and future promise of the human microbiome. Pediatr Gastroenterol Hepatol Nutr. 2013 Jun;16(2):71–9. doi:10.5223/pghn.2013.16.2.71. PubMed PMID: 24010110; PubMed Central PMCID: PMCPMC3760697.
  • Le Roy T, Llopis M, Lepage P, Bruneau A, Rabot S, Bevilacqua C, Martin P, Philippe C, Walker F, Bado A. et al. Intestinal microbiota determines development of non-alcoholic fatty liver disease in mice. Gut. 2013 Dec;62(12):1787–94. doi:10.1136/gutjnl-2012-303816. PubMed PMID: 23197411.
  • Jiao N, Baker SS, Chapa-Rodriguez A, Liu W, Nugent CA, Tsompana M, Mastrandrea L, Buck MJ, Baker RD, Genco RJ. et al. Suppressed hepatic bile acid signalling despite elevated production of primary and secondary bile acids in NAFLD. Gut. 2018 Oct;67(10):1881–1891. doi:10.1136/gutjnl-2017-314307. PubMed PMID: 28774887.
  • Zhang S, Zhao J, Xie F, He H, Johnston LJ, Dai X, Wu C, Ma X. Dietary fiber-derived short-chain fatty acids: a potential therapeutic target to alleviate obesity-related nonalcoholic fatty liver disease. Obes Rev. 2021 Nov;22(11):e13316. doi:10.1111/obr.13316. PubMed PMID: 34279051.
  • Li C, Nie SP, Zhu KX, Ding Q, Li C, Xiong T, Xie M-Y. Lactobacillus plantarum NCU116 improves liver function, oxidative stress and lipid metabolism in rats with high fat diet induced non-alcoholic fatty liver disease. Food Funct. 2014 Dec;5(12):3216–3223. doi:10.1039/c4fo00549j. PubMed PMID: 25317840.
  • Carpi RZ, Barbalho SM, Sloan KP, Laurindo LF, Gonzaga HF, Grippa PC, Zutin TLM, Girio RJS, Repetti CSF, Detregiachi CRP. et al. The effects of probiotics, prebiotics and synbiotics in Non-Alcoholic Fat liver disease (NAFLD) and Non-Alcoholic Steatohepatitis (NASH): A systematic review. IJMS. 2022 Aug 8;23(15):8805. doi:10.3390/ijms23158805. PubMed PMID: 35955942; PubMed Central PMCID: PMCPMC9369010.
  • Hong Y, Sheng L, Zhong J, Tao X, Zhu W, Ma J, Yan J, Zhao A, Zheng X, Wu G. et al. Desulfovibrio vulgaris, a potent acetic acid-producing bacterium, attenuates nonalcoholic fatty liver disease in mice. Gut Microbes. 2021 Jan-Dec;13(1):1–20. doi:10.1080/19490976.2021.1930874. PubMed PMID: 34125646; PubMed Central PMCID: PMCPMC8205104.
  • Loman BR, Hernandez-Saavedra D, An R, Rector, RS. Prebiotic and probiotic treatment of nonalcoholic fatty liver disease: a systematic review and meta-analysis. Nutr Rev. 2018 Nov 1;76(11):822–839. doi:10.1093/nutrit/nuy031. PubMed PMID: 30113661.
  • Koutnikova H, Genser B, Monteiro-Sepulveda M, Faurie J-M, Rizkalla S, Schrezenmeir J, Clément K. Impact of bacterial probiotics on obesity, diabetes and non-alcoholic fatty liver disease related variables: a systematic review and meta-analysis of randomised controlled trials. BMJ Open. 2019 Mar 30;9(3):e017995. doi:10.1136/bmjopen-2017-017995. PubMed PMID: 30928918; PubMed Central PMCID: PMCPMC6475231.
  • Sharpton SR, Maraj B, Harding-Theobald E, Vittinghoff E, Terrault NA. Gut microbiome–targeted therapies in nonalcoholic fatty liver disease: a systematic review, meta-analysis, and meta-regression. The American Journal Of Clinical Nutrition. 2019 Jul 1;110(1):139–149. doi:10.1093/ajcn/nqz042. PubMed PMID: 31124558; PubMed Central PMCID: PMCPMC6599739.
  • Alves CC, Waitzberg DL, de Andrade LS, dos Santos Aguiar L, Reis MB, Guanabara CC, Júnior OA, Ribeiro DA, Sala P. Prebiotic and synbiotic modifications of beta oxidation and lipogenic gene expression after experimental hypercholesterolemia in rat liver. Front Microbiol. 2017;8:2010. doi:10.3389/fmicb.2017.02010. PubMed PMID: 29089934; PubMed Central PMCID: PMCPMC5650986.
  • Garcia-Lopez M, Meier-Kolthoff JP, Tindall BJ, Gronow S, Woyke T, Kyrpides NC, Hahnke RL, Göker M. Analysis of 1,000 type-strain genomes improves taxonomic classification of bacteroidetes. Front Microbiol. 2019;10:2083. doi:10.3389/fmicb.2019.02083. PubMed PMID: 31608019; PubMed Central PMCID: PMCPMC6767994.
  • Kohli R, Kirby M, Xanthakos SA, Softic S, Feldstein AE, Saxena V, Tang PH, Miles L, Miles MV, Balistreri WF. et al. High-fructose, medium chain trans fat diet induces liver fibrosis and elevates plasma coenzyme Q9 in a novel murine model of obesity and nonalcoholic steatohepatitis. Hepatology. 2010 Sep;52(3):934–44. doi:10.1002/hep.23797. PubMed PMID: 20607689; PubMed Central PMCID: PMCPMC2932817.
  • Jahn D, Kircher S, Hermanns HM, Geier A. Animal models of NAFLD from a hepatologist’s point of view. Biochim Biophys Acta Mol Basis Dis. 2019 May 1;1865(5):943–953. doi:10.1016/j.bbadis.2018.06.023. PubMed PMID: 29990551.
  • Krishnan A, Abdullah TS, Mounajjed T, Hartono S, McConico A, White T, LeBrasseur N, Lanza I, Nair S, Gores G. et al. A longitudinal study of whole body, tissue, and cellular physiology in a mouse model of fibrosing NASH with high fidelity to the human condition. Am J Physiol Gastrointest Liver Physiol. 2017 Jun 1;312(6):G666–G680. doi:10.1152/ajpgi.00213.2016. PubMed PMID: 28232454; PubMed Central PMCID: PMCPMC6146305.
  • Albadawy R, Agwa SHA, Khairy E, Saad M, El Touchy N, Othman M, Matboli M. Clinical Significance of HSPD1/MMP14/ITGB1/miR-6881-5P/Lnc-SPARCL1-1:2 RNA Panel in NAFLD/NASH Diagnosis: Egyptian Pilot Study. Biomedicines. 2021 Sep 17;9(9):1248. doi:10.3390/biomedicines9091248. PubMed PMID: 34572434; PubMed Central PMCID: PMCPMC8472260.
  • Wang LF, Wang XN, Huang CC, Hu L, Xiao Y-F, Guan X-H, Qian Y-S, Deng K-Y, Xin H-B. Inhibition of NAMPT aggravates high fat diet-induced hepatic steatosis in mice through regulating Sirt1/AMPKα/SREBP1 signaling pathway. Lipids Health Dis. 2017 Apr 27;16(1):82. doi:10.1186/s12944-017-0464-z. PubMed PMID: 28449683; PubMed Central PMCID: PMCPMC5408374.
  • Cheng C, Liu XH, He J, Gao J, Zhou J-T, Fan J-N, Jin X, Zhang J, Chang L, Xiong Z. et al. Apolipoprotein A4 restricts diet-induced hepatic steatosis via SREBF1-mediated lipogenesis and enhances IRS-PI3K-Akt signaling. Mol Nutr Food Res. 2022 Sep;66(18):e2101034. doi:10.1002/mnfr.202101034. PubMed PMID: 35909347.
  • Camargo FN, Matos SL, Araujo LCC, Carvalho, CR, Amaral, AG, Camporez, JP. Western diet-fed ApoE knockout male mice as an experimental model of Non-Alcoholic Steatohepatitis. Curr Issues Mol Biol. 2022 Oct 8;44(10):4692–4703. doi:10.3390/cimb44100320. PubMed PMID: 36286035; PubMed Central PMCID: PMCPMC9600038.
  • Martinez-Clemente M, Ferre N, Titos E, Horrillo R, González-Périz A, Morán-Salvador E, López-Vicario C, Miquel R, Arroyo VD, Funk C. et al. Disruption of the 12/15-lipoxygenase gene (Alox15) protects hyperlipidemic mice from nonalcoholic fatty liver disease. Hepatology. 2010 Dec;52(6):1980–91. doi:10.1002/hep.23928. PubMed PMID: 20967760.
  • Kuipers F, Jong MC, Lin Y, Eck M, Havinga R, Bloks V, Verkade HJ, Hofker MH, Moshage H, Berkel TJ. et al. Impaired secretion of very low density lipoprotein-triglycerides by apolipoprotein E- deficient mouse hepatocytes. J Clin Invest. 1997 Dec 1;100(11):2915–22. doi:10.1172/JCI119841. PubMed PMID: 9389759; PubMed Central PMCID: PMCPMC508499.
  • Debnath N, Kumar R, Kumar A, Mehta, PK, Yadav, AK. Gut-microbiota derived bioactive metabolites and their functions in host physiology. Biotechnol Genet Eng Rev. 2021 Oct;37(2):105–153. doi:10.1080/02648725.2021.1989847. PubMed PMID: 34678130.
  • Dias P, Pourova J, Voprsalova M, Nejmanová I, Mladěnka P. 3-hydroxyphenylacetic acid: a blood pressure-reducing flavonoid metabolite. Nutrients. 2022 Jan 13;14(2):328. doi:10.3390/nu14020328. PubMed PMID: 35057508; PubMed Central PMCID: PMCPMC8781193.
  • Porras D, Nistal E, Martinez-Florez S, Pisonero-Vaquero S, Olcoz JL, Jover R, González-Gallego J, García-Mediavilla MV, Sánchez-Campos S. Protective effect of quercetin on high-fat diet-induced non-alcoholic fatty liver disease in mice is mediated by modulating intestinal microbiota imbalance and related gut-liver axis activation. Free Radic Biol Med. 2017 Jan;102:188–202. doi:10.1016/j.freeradbiomed.2016.11.037. PubMed PMID: 27890642.
  • Jang YG, Ko EB, Choi KC. Gallic acid, a phenolic acid, hinders the progression of prostate cancer by inhibition of histone deacetylase 1 and 2 expression. J Nutr Biochem. 2020 Oct;84:108444. doi:10.1016/j.jnutbio.2020.108444. PubMed PMID: 32615369.
  • Lee J, Song JH, Chung MY, Lee J-H, Nam T-G, Park JH, Hwang J-T, Choi H-K. 3,4-dihydroxytoluene, a metabolite of rutin, suppresses the progression of nonalcoholic fatty liver disease in mice by inhibiting p300 histone acetyltransferase activity. Acta Pharmacol Sin. 2021 Sep;42(9):1449–1460. doi:10.1038/s41401-020-00571-7. PubMed PMID: 33303988; PubMed Central PMCID: PMCPMC8379200.
  • Grunstein M. Histone acetylation in chromatin structure and transcription. Nature. 1997 Sep 25;389(6649):349–52. doi:10.1038/38664. PubMed PMID: 9311776.
  • Liu D, Wong CC, Fu L, Chen H, Zhao L, Li C, Zhou Y, Zhang Y, Xu W, Yang Y. et al. Squalene epoxidase drives NAFLD-induced hepatocellular carcinoma and is a pharmaceutical target. Sci Transl Med. 2018 Apr 18;10(437):eaap9840. doi:10.1126/scitranslmed.aap9840. PubMed PMID: 29669855.
  • Roth SY, Denu JM, Allis CD. Histone acetyltransferases. Annu Rev Biochem. 2001;70(1):81–120. doi:10.1146/annurev.biochem.70.1.81. PubMed PMID: 11395403.
  • Eberhardt J, Santos-Martins D, Tillack AF, Forli S. AutoDock Vina 1.2.0: new docking methods, expanded force field, and Python Bindings. J Chem Inf Model. 2021 Aug 23;61(8):3891–3898. doi:10.1021/acs.jcim.1c00203. PubMed PMID: 34278794.
  • Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem. 2010 Jan 30;31(2):455–61. doi:10.1002/jcc.21334. PubMed PMID: 19499576; PubMed Central PMCID: PMCPMC3041641.
  • Yu Y, Lu J, Sun L, Lyu X, Chang X-Y, Mi X, Hu M-G, Wu C, Chen X. Akkermansia muciniphila: a potential novel mechanism of nuciferine to improve hyperlipidemia. Biomed Pharmacother. 2021 Jan;133:111014. doi:10.1016/j.biopha.2020.111014. PubMed PMID: 33246225.
  • Juarez-Fernandez M, Porras D, Petrov P, Román-Sagüillo S, García-Mediavilla MV, Soluyanova P, Martínez-Flórez S, González-Gallego J, Nistal E, Jover R. et al. The synbiotic combination of akkermansia muciniphila and quercetin ameliorates early obesity and NAFLD through gut microbiota reshaping and bile acid metabolism modulation. Antioxid (Basel). 2021 Dec 15;10(12):2001. doi:10.3390/antiox10122001. PubMed PMID: 34943104; PubMed Central PMCID: PMCPMC8698339.
  • Martens EC, Chiang HC, Gordon JI. Mucosal glycan foraging enhances fitness and transmission of a saccharolytic human gut bacterial symbiont. Cell Host & Microbe. 2008 Nov 13;4(5):447–57. doi:10.1016/j.chom.2008.09.007. PubMed PMID: 18996345; PubMed Central PMCID: PMCPMC2605320.
  • Yoshida N, Emoto T, Yamashita T, Watanabe H, Hayashi T, Tabata T, Hoshi N, Hatano N, Ozawa G, Sasaki N. et al. Bacteroides vulgatus and Bacteroides dorei Reduce Gut Microbial Lipopolysaccharide Production and Inhibit Atherosclerosis. Circulation. 2018 Nov 27;138(22):2486–2498. doi:10.1161/CIRCULATIONAHA.118.033714. PubMed PMID: 30571343.
  • Liu L, Xu M, Lan R, Hu D, Li X, Qiao L, Zhang S, Lin X, Yang J, Ren Z. et al. Bacteroides vulgatus attenuates experimental mice colitis through modulating gut microbiota and immune responses. Front Immunol. 2022;13:1036196. doi:10.3389/fimmu.2022.1036196. PubMed PMID: 36531989; PubMed Central PMCID: PMCPMC9750758.
  • Wang C, Xiao Y, Yu L, Tian F, Zhao J, Zhang H, Chen W, Zhai Q. Protective effects of different bacteroides vulgatus strains against lipopolysaccharide-induced acute intestinal injury, and their underlying functional genes. J Adv Res. 2022 Feb;36:27–37. doi:10.1016/j.jare.2021.06.012. PubMed PMID: 35127162; PubMed Central PMCID: PMCPMC8799915.
  • Waidmann M, Bechtold O, Frick JS, Lehr H-A, Schubert S, Dobrindt U, Loeffler J, Bohn E, Autenrieth IB. Bacteroides vulgatus protects against Escherichia coli-induced colitis in gnotobiotic interleukin-2-deficient mice. Gastroenterology. 2003 Jul;125(1):162–177. doi:10.1016/s0016-5085(03)00672-3. PubMed PMID: 12851881.
  • Debedat J, Le Roy T, Voland L, Belda E, Alili R, Adriouch S, Bel Lassen P, Kasahara K, Hutchison E, Genser L. et al. The human gut microbiota contributes to type-2 diabetes non-resolution 5-years after roux-en-Y gastric bypass. Gut Microbes. 2022 Jan-Dec;14(1):2050635. doi:10.1080/19490976.2022.2050635. PubMed PMID: 35435140; PubMed Central PMCID: PMCPMC9037437.
  • Vieira-Silva S, Falony G, Belda E, Nielsen T, Aron-Wisnewsky J, Chakaroun R, Forslund SK, Assmann K, Valles-Colomer M, Nguyen TTD. et al. Statin therapy is associated with lower prevalence of gut microbiota dysbiosis. Nature. 2020 May;581(7808):310–315. doi:10.1038/s41586-020-2269-x. PubMed PMID: 32433607.
  • Leite AZ, Rodrigues NC, Gonzaga MI, Paiolo JCC, de Souza CA, Stefanutto NAV, Omori WP, Pinheiro DG, Brisotti JL, Matheucci Junior E. et al. Detection of increased plasma interleukin-6 levels and prevalence of Prevotella copri and Bacteroides vulgatus in the feces of type 2 diabetes patients. Front Immunol. 2017;8:1107. doi:10.3389/fimmu.2017.01107. PubMed PMID: 28966614; PubMed Central PMCID: PMCPMC5605568.
  • Coppola S, Avagliano C, Calignano A, Berni Canani R. The protective role of butyrate against obesity and obesity-related diseases. Molecules. 2021 Jan 28;26(3):682. doi:10.3390/molecules26030682. PubMed PMID: 33525625; PubMed Central PMCID: PMCPMC7865491.
  • Gottlieb A, Canbay A. Why bile acids are so important in Non-Alcoholic Fatty liver disease (NAFLD) progression. Cells. 2019 Oct 30;8(11):1358. doi:10.3390/cells8111358. PubMed PMID: 31671697; PubMed Central PMCID: PMCPMC6912605.
  • Zhou Q, Deng J, Pan X, Meng D, Zhu Y, Bai Y, Shi C, Duan Y, Wang T, Li X. et al. Gut microbiome mediates the protective effects of exercise after myocardial infarction. Microbiome. 2022 May 31;10(1):82. doi:10.1186/s40168-022-01271-6. PubMed PMID: 35637497; PubMed Central PMCID: PMCPMC9153113.
  • Liu Y, Myojin T, Li K, Kurita A, Seto M, Motoyama A, Liu X, Satoh A, Munemasa S, Murata Y. et al. A Major intestinal catabolite of quercetin glycosides, 3-hydroxyphenylacetic acid, protects the hepatocytes from the Acetaldehyde-Induced Cytotoxicity through the enhancement of the total Aldehyde dehydrogenase activity. Int J Mol Sci. 2022 Feb 3;23(3):1762. doi:10.3390/ijms23031762. PubMed PMID: 35163684; PubMed Central PMCID: PMCPMC8836260.
  • Murota K, Nakamura Y, Uehara M. Flavonoid metabolism: the interaction of metabolites and gut microbiota. Biosci Biotechnol Biochem. 2018 Apr;82(4):600–610. doi:10.1080/09168451.2018.1444467. PubMed PMID: 29504827.
  • Li K, Wu H, Kidawara M, Lin Y, Satoh A, Zhang G, Munemasa S, Murata Y, Nakamura T, Nakamura Y. et al. The microbiota catabolites of quercetin glycosides concertedly enhance the resistance against acetaldehyde-induced oxidative stress. Free Radic Res. 2022 Sep-Oct;56(9–10):607–616. doi:10.1080/10715762.2022.2159820. PubMed PMID: 36576903.
  • Fu S, Yu M, Tan Y, Liu D. Role of histone deacetylase on nonalcoholic fatty liver disease. Expert Rev Gastroenterol Hepatol. 2021 Apr;15(4):353–361. doi:10.1080/17474124.2021.1854089. PubMed PMID: 33213187.
  • Shen Y, Wei W, Zhou DX. Histone acetylation enzymes coordinate metabolism and gene expression. Trends Plant Sci. 2015 Oct;20(10):614–621. doi:10.1016/j.tplants.2015.07.005. PubMed PMID: 26440431.
  • Hou T, Tian Y, Cao Z, Zhang J, Feng T, Tao W, Sun H, Wen H, Lu X, Zhu Q. et al. Cytoplasmic SIRT6-mediated ACSL5 deacetylation impedes nonalcoholic fatty liver disease by facilitating hepatic fatty acid oxidation. Mol Cell. 2022 Nov 3;82(21):4099–4115.e9. doi:10.1016/j.molcel.2022.09.018. PubMed PMID: 36208627.
  • Zhou B, Jia L, Zhang Z, Xiang L, Yuan Y, Zheng P, Liu B, Ren X, Bian H, Xie L. et al. The Nuclear Orphan Receptor NR2F6 promotes hepatic steatosis through upregulation of fatty acid transporter CD36. Adv Sci (Weinh). 2020 Nov;7(21):2002273. doi:10.1002/advs.202002273. PubMed PMID: 33173745; PubMed Central PMCID: PMCPMC7610302.
  • Kimura H. Histone modifications for human epigenome analysis. J Hum Genet. 2013 Jul;58(7):439–45. doi:10.1038/jhg.2013.66. PubMed PMID: 23739122.
  • Ma J, You D, Chen S, Fang N, Yi X, Wang Y, Lu X, Li X, Zhu M, Xue M. et al. Epigenetic association study uncovered H3K27 acetylation enhancers and dysregulated genes in high-fat-diet-induced nonalcoholic fatty liver disease in rats. Epigenomics. 2022 Dec;14(23):1523–1540. doi:10.2217/epi-2022-0362. PubMed PMID: 36851897.
  • Li C, Wang Y, Liu D, Wong CC, Coker OO, Zhang X, Liu C, Zhou Y, Liu Y, Kang W. et al. Squalene epoxidase drives cancer cell proliferation and promotes gut dysbiosis to accelerate colorectal carcinogenesis. Gut. 2022 Nov;71(11):2253–2265. doi:10.1136/gutjnl-2021-325851. PubMed PMID: 35232776; PubMed Central PMCID: PMCPMC9554078.
  • Muller PA, Matheis F, Schneeberger M, Kerner Z, Jové V, Mucida D. Microbiota-modulated CART + enteric neurons autonomously regulate blood glucose. Sci. 2020 Oct 16;370(6514):314–321. doi:10.1126/science.abd6176. PubMed PMID: 32855216; PubMed Central PMCID: PMCPMC7886298.
  • Vicentini FA, Keenan CM, Wallace LE, Woods C, Cavin J-B, Flockton AR, Macklin WB, Belkind-Gerson J, Hirota SA, Sharkey KA. et al. Intestinal microbiota shapes gut physiology and regulates enteric neurons and glia. Microbiome. 2021 Oct 26;9(1):210. doi:10.1186/s40168-021-01165-z. PubMed PMID: 34702353; PubMed Central PMCID: PMCPMC8549243.
  • Win S, Min RWM, Zhang J, Kanel G, Wanken B, Chen Y, Li M, Wang Y, Suzuki A, Aung FWM. et al. Hepatic mitochondrial SAB deletion or knockdown alleviates diet-induced metabolic syndrome, steatohepatitis, and hepatic fibrosis. Hepatology. 2021 Dec;74(6):3127–3145. doi:10.1002/hep.32083. PubMed PMID: 34331779; PubMed Central PMCID: PMCPMC8639630.
  • Ferre N, Martinez-Clemente M, Lopez-Parra M, González-Périz A, Horrillo R, Planagumà A, Camps J, Joven J, Tres A, Guardiola F. et al. Increased susceptibility to exacerbated liver injury in hypercholesterolemic ApoE-deficient mice: potential involvement of oxysterols. Am J Physiol Gastrointest Liver Physiol. 2009 Mar;296(3):G553–62. doi:10.1152/ajpgi.00547.2007. PubMed PMID: 19136384.
  • Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLoS Biol. 2010 Jun 29;8(6):e1000412. doi:10.1371/journal.pbio.1000412. PubMed PMID: 20613859; PubMed Central PMCID: PMCPMC2893951.