Background

Primary sclerosing cholangitis (PSC) is a chronic cholestatic liver disorder characterized by inflammation and bile duct narrowing, which results in the accumulation of bile acids (BAs) in the liver, leading to hepatic damage, progressive liver fibrosis, cirrhosis, and even liver cancer [1, 2]. Over the last several decades, extensive efforts have been made to identify the mechanisms underlying cholestatic liver injury [3,4,5]. However, no effective therapy has been developed due to the complexity of disease pathogenesis. Liver transplantation remains the only life-extending treatment for end-stage PSC patients [4]. It has been well-accepted that dysregulation of BA homeostasis and inflammation are the major driving forces of the disease progression of PSC [6]. In addition, dysbiosis and intestinal barrier dysfunction also have been reported as key contributors to cholestatic liver injury [7,8,9]. Therefore, an effective therapeutic agent for PSC must be able to modulate bile acid metabolism, inflammatory response, and the gut microbiome.

Berberine (BBR), an isoquinoline alkaloid isolated from the rhizome of the herb Coptis chinensis and Berberis vulgaris, is one of the most commonly used plant medicines in China and Asia with various biological activities [10,1, 4]. Currently, there is no effective medication for improving transplant-free survival in these cases. Liver transplantation is the only definitive treatment for PSC, though it carries a high risk of disease recurrence [2, 29, 30]. Recent studies suggest that targeting pathways, such as BA synthesis and transport, hepatic inflammation, mitochondrial respiration, oxidative stress, intestinal inflammation, and gut microbiota, could provide new therapeutic strategies for cholestatic liver diseases [26, 27, 31,32,33,34,35].

BBR has long been used in Asia as an anti-bacterial medicine. Clinical and preclinical studies highlight its potential in treating metabolic diseases by modulating various molecular targets, including transcription factors, cell survival/proliferative proteins, enzymes, metastatic/invasion molecules, growth factors, platelet activation, inflammatory cytokines, apoptotic proteins, protein kinases, receptors, and the others [14, 36]. There are a considerable number of studies demonstrating that BBR has preventive or therapeutic effects on various liver diseases, such as hepatitis, MAFLD, and liver fibrosis [11, 37,38,39,40,41,42]. However, its impact on cholestatic liver injury remains unexplored. To elucidate the therapeutic effect and potential mechanisms of BBR on cholestatic liver injury, we conducted a series of analyses, including histological imaging, biochemical analysis, molecular biology, and RNA-seq transcriptome analysis. Using bioinformatic tools, we identified differentially expressed genes regulated by BBR followed by GO, KEGG pathway, and functional category analysis. The findings of this study strongly suggest that BBR is potentially effective in treating cholestatic liver injury. The major mechanisms underlying BBR's beneficial effects include reducing bile duct injury and hepatic fibrosis, alleviating hepatic inflammation and ER stress, restoring BA homeostasis, and improving intestinal barrier function as well as modulating gut microbiome.

PSC is an inflammatory liver disease often associated with severe cholestatic liver injury [43]. BBR is known for its potent anti-inflammatory activities in liver disease [16, 17]. Key pathways, such as NF-κB signaling pathway, MAPK pathways, and oxidative phosphorylation, are involved in inflammation-driven cholestatic liver injury [8, 23, 44, 45]. Our RNA-seq gene analysis and pathway profiling showed that BBR significantly reduced inflammation in Mdr2−/− mice. This reduction is achieved through BBR's ability to inhibit inflammatory macrophage infiltration in the liver, which is evident from the decreased expression of various chemokines, cytokines, and cell surface adhesion molecules (Fig. 4, Additional file 1: Fig. S6). Additionally, BBR modulates NF-κB signaling, the MAPK signaling pathway, and oxidative phosphorylation (Additional file 1: Fig. S7–S9).

CCL2/MCP-1 chemokines, produced by fibroblasts, activated cholangiocytes, resident macrophages, and endothelial cells, play a crucial role in the inflammatory response. A recent study suggests that targeting the CCR2/CCL2 axis can limit monocyte recruitment and reduce fibrosis and cholestasis, offering a potential treatment approach for PSC [46]. In line with these findings, our study demonstrates that BBR suppresses the hepatic expression of CCR2 and CCL2. Furthermore, the ER stress response, a key factor in inflammation and metabolic disorders, is significantly modulated by BBR [24, 47, 48]. Disruptions in ER homeostasis activate the UPR, leading to inflammation and cell injury. The IRE1, protein kinase RNA-like ER kinase (PERK), and ATF6 pathways are the three major branches of the UPR [24]. BBR has been previously shown to inhibit HIV protease inhibitor-induced ER stress in macrophages and inhibit free fatty acid and LPS-induced inflammation via modulating the PERK-ATF4-CHOP signaling pathway in macrophages and hepatocytes [17, 18]. Consistently, our current study found that BBR significantly reduced ER stress in Mdr2−/− mice, particularly inhibiting the PERK-ATF4-CHOP pathway (Fig. 5 and Additional file 1: Fig. S10).

BA homeostasis is crucial in managing cholestatic liver diseases [3, 49]. BAs are synthesized in hepatocytes and immediately secreted into bile through the bile duct. The majority of BAs are reabsorbed in the terminal ileum and transported back to the liver through portal vein. The enterohepatic circulation of BA is an important physiological process, making the synthesis and transport of BAs vital targets for cholestatic liver injury [50, 51]. Our studies show that BBR can restore BA homeostasis by modulating key enzymes, nuclear receptors, and hepatic transporters involved in BA synthesis and transport (Fig. 6 and Additional file 1: Fig. S11). Elevated serum BA levels, particularly total, primary, conjugated BAs, including TCA, were common in PSC patients [52,53,54]. The current study showed that BBR treatment in Mdr2−/− mice significantly reduced these BA levels in serum, liver, and small intestine while increasing fecal BA output without causing diarrhea (Fig. 7 and Additional file 1: Fig. S12). This suggests BBR's role as a differential BA transport inhibitor, indicated by reduced Ntcp and Asbt expression (Fig. 6). Furthermore, BBR has been shown to influence key regulators of BA homeostasis significantly. In our study, BBR increased the expression of Fxrα, a crucial regulator in BA homeostasis, and increased the expression of Shp, which represses Cyp7a1 by inhibiting LRH-1 activity [55, 56]. However, the RNA-seq data showed BBR had no significant effects on LRH-1, but upregulated both Cyp7a1 and Cyp27a1 levels in the liver (Fig. 6). These results suggest the potential compensatory mechanisms to counteract the inhibition of BA up taking in Mdr2−/− mice with BBR treatment. Although FXR agonists and ASBT inhibitors have been tested in clinical trials for various liver diseases, the potential to treat PSC remains uncertain. Our previous studies reported that increased primary conjugated BA is responsible for cholestatic liver injury and liver fibrosis via activating sphingosine-1 phosphate receptor 2 (S1PR2), which can upregulate lncRNA H19 in Mdr2−/− mice [57,58,59,60]. Our recent study showed that BBR reduced the expression of H19 in a MASH mouse model [16]. Consistently, in this study, our results showed that H19 was inhibited by BBR treatment in Mdr2−/− mice.

Recent clinical studies have established a link between disrupted intestinal barrier function, bacterial translocation, and the progression of cholestatic liver diseases, such as PSC and PBS [61]. Specifically, in Mdr2−/− mice, impairment in intestinal barrier function has been observed, including diminished tight junction protein expression, reduced mucus layers, increased permeability, and enhanced bacterial translocation [9]. Our previous study has reported that ER stress-induced activation of CHOP leads to disruption of intestinal barrier function, bacterial translocation, activation of inflammation, and eventually results in fibrosis in the liver [7]. In line with these findings, our current study demonstrates that BBR effectively decreased CHOP expression in both the liver and intestine (Fig. 5d and Additional file 1: Fig. S13b), suggesting its potential to mitigate these pathological processes. Moreover, recent studies reported that H19 exacerbates intestinal barrier dysfunction by inhibiting autophagy and impairing goblet and Paneth cell functions [62, 63]. Consistent with this, our results show that BBR significantly inhibits H19, correlating with restored epithelial barrier function as evidenced by increased expression of mucin-2 and ZO-1 (Figs. 8e-f & Additional file 1: Fig. S13b).

A previous study using hamsters found that orally administered BBR predominantly accumulates in the gut rather than in circulation, significantly affecting both gut and circulatory metabolites despite low serum levels [64]. Our study aligns with these findings, showing that BBR concentrations are highest in the stomach, intestine, and colon and relatively lower in the liver, kidney, heart, lung, brain, and spleen. This suggests that BBR mitigates cholestatic liver injury by modulating the gut-liver axis (Additional file 1: Fig. S14). It is well established that the gut microbiota regulates BA composition and levels, particularly in PSC. BBR has been reported to have antidiabetic effects by modulating the gut microbiome [10, 15]. Our current study further indicates that BBR alters the gut microbiota composition in Mdr2−/− mice. Specifically, BBR increased the relative abundance of Bacteroidetes and decreased that of Firmicutes (Additional file 1: Fig. S15), which is significant as bacteria in Firmicutes are known for high bile salt hydrolase (BSH) activity, promoting BA deconjugation and fecal excretion [65, 66]. This alteration in microbiota composition aligns with the interplay between intestinal microbiota and BAs, where each influence the other.

Conclusion

In summary, our study sheds light on the potential mechanisms by which BBR attenuates cholestatic liver injury in a PSC mouse model. As illustrated in Fig. 9, BBR can directly or indirectly target various liver cells, including hepatocytes, macrophages, stellate cells, and cholangiocytes, modulating multiple pathways related to bile duct injury, fibrosis, inflammation, ER stress, and BA metabolism and transport in the gut-liver-axis. Furthermore, BBR enhances intestinal barrier function and reduces bacterial translocation, while also restoring BA homeostasis and gut microbiota. These findings suggest that BBR has potential as a pharmacological treatment for cholestatic liver injury such as PSC.

Fig. 9
figure 9

Schematic Representation of BBR’s Potential Mechanisms in Alleviating Cholestatic Liver Injury. This diagram illustrates the proposed molecular and cellular mechanisms through which BBR mitigates cholestatic liver injury in a mouse model of sclerosing cholangitis. It visually summarizes the pathways and interactions influenced by BBR treatment, highlighting its multifaceted role in addressing liver disease pathology

Materials and methods

Reagents

Berberine chloride hydrate (BBR) was purchased from Sigma (St. Louis, MO, USA, Cat #14050). Common laboratory chemicals were purchased from Sigma Aldrich (St. Louis, MO, USA). All antibodies used in this study are listed in Additional file 2: Table S1.

Animal experiments

FVB Mdr2−/− mice (100 days old, both sexes, n = 9–12) were originally obtained from Dr. Gianfranco Alpini (Texas A&M HSC College of Medicine). Mdr2−/− mouse (C57/BL6 background) is a kind gift from Dr. Daniel Goldenberg at the Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel. Mice were randomly divided into the vehicle control group and BBR group. Mice were treated with BBR (50 mg/kg) or vehicle (0.5% carboxyl methyl cellulose sodium solution) by intragastric administration once daily for 8 weeks. All mice were housed in a 12 h light/12 h dark cycle with a controlled room temperature between 21 and 23 °C and free access to water. All the experimental procedures were performed according to protocols approved by the Richmond VA Medical Center and Virginia Commonwealth University Institutional Animal Care and Use Committee. All animal experiments were performed in accordance with institutional guidelines for ethical animal studies. At the end of the experiment, mice were weighed and anesthetized by exposure to inhaled isoflurane. The blood was collected by cardiac puncture. The serum was collected and stored at − 80 °C for later analysis. After euthanasia, the liver and small intestine were collected for histological analysis, RNA profiling, and Western blot analysis. Fecal samples were collected for 16S rRNA gene sequencing to measure the gut microbiome.

RNA sequencing (RNAseq) and bioinformatic analysis

Total liver RNA was isolated using Chemagic Prepito®-D Nucleic Acid Extractor (PerkinElmer, Waltham, MA, USA) with a Prepito RNA kit (PerkinElmer, USA). The RNAseq with ribosomal RNA (rRNA) depletion was done by Genewiz Company using the Illumina Hiseq® X platform (Genewiz Co., South Plainfield, NJ, USA). Sequencing reads were trimmed and filtered using bbduk to remove adapters and low-quality reads. Reads from mouse samples were mapped to Ensembl GRCm38 transcripts annotation (release 82), using RSEM. Gene expression data normalization and differential expression analysis were performed using the R package edgeR. Significantly up- or downregulated genes were determined as fold change ≥ 2 and p-value < 0.05. Hierarchical clustering was performed to show distinguishable mRNA expression profiles among the samples (Heatmap was plotted by http://www.bioinformatics.com.cn, an online platform for data analysis and visualization). The volcano graph and heatmaps were created to visualize significantly dysregulated mRNAs using GraphPad Prism (version 8; GraphPad Software Inc., San Diego, CA, USA). Gene Ontology (GO) analysis was used to investigate three functionality domains: biological process (BP), cellular component (CC), and molecular function (MF) using DAVID (Database for Annotation, Visualization, and Integrated Discovery) v6.8 (https://david.ncifcrf.gov/). Pathway analysis was performed to functionally analyze and map genes to Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways (https://pathview.uncc.edu/).

Serum biochemical analysis and hepatic hydroxyproline content measurement

The serum levels of ALP, AST and ALT, total triglyceride (TG), total cholesterol (TC), very-low-density lipoprotein (VLDL), and ALB were determined using the Alfa Wassermann Vet ACE Axcel® System with commercially available assay kits (Alfa Wassermann diagnostic technologies, NJ, USA). To quantify liver fibrosis, hepatic hydroxyproline was measured using the Hydroxyproline Assay kit (Sigma Aldrich, MO, USA) according to the manufacturer's instructions.

Histological and immunohistochemical staining

Liver tissues were processed for hematoxylin and eosin (H&E) staining and immunohistochemistry (IHC) staining for CK-19 and Ki67 at the Mouse Model Core at the VCU Massey Cancer Center (Richmond, VA, USA). Picro Sirius Red Staining was performed using the commercial Kit (Abcam, USA) with the paraffin-embedded tissue sections according to the manufacturer's instructions. Small intestine tissues were processed for H&E staining. Alcian blue staining was performed using the Alcian blue Stain Kit (Abcam, USA). Immunofluorescence staining of ZO-1 was performed with the paraffin-embedded tissue sections according to the manufacturer's instructions. All the stained slides were scanned using a Vectra Polaris Automated Quantitative Pathology Imaging System (Akoya Biosciences, MA, USA), and the images were captured using Phenochart software (Akoya Biosciences, MA, USA).

Bile acid (BA) analysis

The serum, liver tissues, intestine contents, and colon feces were processed for BA analysis, as described previously [16]. The composition and levels of BAs in serum, liver, intestine and fecal samples were measured using a Shimadzu liquid chromatography/tandem mass spectrometric (LC–MS/MS) 8600 system as described previously [16]. Data were collected and processed using Lab Solutions software.

Tissue distribution of BBR

Mdr2−/− mice were treated with BBR (50 mg/kg) by intragastric administration after a 12-h fast. Blood, heart, lung, liver, kidney, brain, spleen, stomach, intestine, colon, and feces were collected after 3, 6, 9, and 12 h of BBR treatment, respectively. The contents of BBR in serum and tissues were analyzed using LC–MS/MS.

A reliable LC–MS/MS method was developed and validated to quantify BBR, using L-tetrahydropalmatine as the internal standard (IS). To quantify BBR in the serum, serum samples and IS were incubated with acetonitrile/methanol/water l (1/1, v/v) in a 1.5 mL vial. For quantification of BBR in the spleen, lung, kidney, heart, stomach contents, intestine contents, and feces, tissue samples were incubated with acetonitrile/methanol (1/1, v/v) in a 2 mL vial with beads. The homogenized samples and IS were mixed with acetonitrile/methanol/water (1/1, v/v) in a 1.5 mL vial. After centrifugation at 12,000 × g for 2 min at room temperature, the supernatant was filtered through 0.2 µm PTFE membrane, and 2 µL aliquots were injected into the LC–MS/MS system. The analyte was separated on a C18 reverse phase column and analyzed in the multiple reaction monitoring (MRM) mode using ESI with positive ionization, m/z 335.9 → 320.1 for BBR and m/z 355.9 → 192.2 for IS. Mobile phase A was 0.05% acetic acid in water, while mobile phase B was acetonitrile. The gradient was optimized at 30% to 75% B in 2 min and then maintained 75% B for 0.5 min. The column was equilibrated with 30% B for 1.5 min. Data were collected and processed using Lab Solutions software.

Quantitative RT-PCR

Total liver RNA was isolated using Chemagic Prepito®-D Nucleic Acid Extractor (PerkinElmer, USA) with Prepito RNA kit (PerkinElmer, USA). cDNA synthesis and Quantitative RT-PCR analysis of relative mRNA expression levels of target genes were previously described [16]. Primer sequences will be provided upon request.

Immunoblotting analysis

Total proteins were prepared using cold RIPA buffer. Nuclear proteins were isolated, as previously described. Protein concentration was measured using the Bio-Rad Protein Assay reagent. Proteins were resolved on 10% SDS-PAGE and transferred to nitrocellulose membranes (Thermo, Waltham, MA, USA). 5% milk was used to block the background. The target proteins were probed with the specific primary antibodies and detected using HRP-conjugated secondary antibodies and ECL reagents (Thermo, USA). Images were captured using the Bio-Rad Gel Doc XR + Imaging System (Hercules, CA, USA). The density of immunoblotted bands was analyzed using BioRad Image Lab computer software and normalized with histone 3 or β-Actin.

FITC-DEXTRAN permeability and bacterial translocation assay

FITC-Dextran solution (100 mg/mL) was prepared in PBS. FITC-Dextran was administered to mice by oral gavage (600 mg/kg) and blood samples were taken after 4 h. The serum concentration of FITC-dextran was measured using Victor Multilabel Plate Counter (PerkinElmer, Waltham, MA) with an excitation wavelength of 490 nm and an emission wavelength of 530 nm. Blood and mesenteric lymph nodes (MLNs) were harvested in sterile conditions. Blood and homogenized MLNs were diluted in series and plated on Blood Agar Plates. After 72 h incubation at 37 °C in aerobic conditions, colony-forming units (CFUs) were counted and calculated.

Microbiota analysis

Fecal samples of Mdr2−/− mice treated with BBR 50 mg/kg or 100 mg/kg for 8 weeks were collected for 16S rRNA gene sequencing. Extraction, library preparation, sequencing, and analysis were performed at Rutgers Center for Microbiome Analysis Core, New Jersey Institute for Food, Nutrition and Health. All DNA samples were quantified using the Qubit 1 × dsDNA HS assay kit (Thermo Fisher Scientific), which measured DNA concentration based on the fluorescence intensity of a fluorescent dye binding to double-stranded DNA. DNA integrity was assessed using agarose gel electrophoresis.

Statistical analysis

Data are expressed as the mean ± SEM from at least three independent experiments. The student's t-test was used to analyze the difference between the two groups by GraphPad Prism (version 8; GraphPad Software Inc., San Diego, CA). A p-value < 0.05 was considered statistically significant.