Animal experiments and ethics statement
C57BL/6 wild-type male mice (six-week-old) were purchased from Dae Han Bio Link (Eumseoung, Republic of Korea) and bred and managed at the Hanyang Laboratory Animal Research Center. Mice were randomly assigned into the following four groups (Fig. 1A; n = 10 per group, n = 40 in total): normal control, 0.03% TAA (Sigma Aldrich, St. Louis, MO, USA) in drinking water for 19 weeks, TAA + 10 mg/kg/day OCA (APExBIO, Boston, MA, USA) for nine weeks, and TAA + 10 mg/kg/day 11β-HSD1 inhibitor (J2H Biotech, Suwon, Republic of Korea) for nine weeks. (13, 14). After 10 weeks of TAA administration, the treatment groups were administered TAA combined with OCA or 11β-HSD1 inhibitor for the next nine weeks. OCA and the 11β-HSD1 inhibitor were orally administered by gavage once a day (15). The study was approved by the Hanyang University Institutional Animal Care and Use Committee (HY-IACUC-21-0001). The research protocol was prepared, and the study was performed in compliance with their ethical guidelines.
Human natural killer cell isolation
Peripheral blood mononuclear cells (PBMCs) were isolated form healthy donor with density gradient in CPT tube (BD Biosciences, Cas.No 362753), centrifuged 1800G, RT, 20min. Freshly isolated NK cells using the Human Natural Killer Cell Isolation kit (Miltenyl Biotec, Cas.No 130-092-657). NK cells were cultured in NK MACS Medium human (Miltenyl Biotec, Cas.No 130-114-429) supplemented with 1% NK MACS Supplement human (Miltenyl Biotec), 5% human AB serum (Sigma Aldrich, Cas.No H3667), 500IU/ML IL-2 IS, premium grade (Milteny Biotec, Cas.No 130-097-744). Cells were maintained in a humidified atmosphere containing 5% CO2 and 95% air at 37°C incubator. Total RNA from NK cells obtained from healthy donor (n = 1) was isolated using Trizol Reagent (Invitrogen, Cas.NO 15596018) and cDNA was prepared with PrimeScript RT reagent Kit (TaKaRa; Otsu, Shiga, Japan), 2µg of total RNA was reverse transcribed. qRT-PCR amplification was performed using a LightCycler 480 (Roche Diagnostics, Mannheim, Germany) with LightCycler 480 SYBR Green I Master mix (Roche Diagnostics). All tests were performed in three replicated with 96 well plates and normalized to β-actin.
Study materials
The 11β-HSD1 inhibitor (J2H Biotech, Suwon, Republic of Korea) was synthesized, OCA (APExBIO, Boston, MA, USA) was purchased. The LX-2 and HepG2 cell lines were used for in vitro experiments.
Biochemical and histopathological analysis
Levels of serum ALT, AST, total bilirubin, and albumin were measured by chemistry analyzer AU480 (Beckman Coulter, Tokyo, Japan). H&E and Sirius Red staining were performed to assess inflammation and fibrosis. Inflammation and ballooning scores were determined using the MASLD activity score system for non-alcoholic steatohepatitis (16). The detailed procedures are described in the Supplementary Materials.
Western blotting
Total protein from the liver tissues was extracted with RIPA lysis buffer (GenDEPOT, Hanam, Republic of Korea), Protease inhibitor (GenDEPOT, Hanam, Republic of Korea) and Phosphatase inhibitor (GenDEPOT, Hanam, Republic of Korea). RIPA lysis buffer and protease inhibitor, phosphatase inhibitor was used in a ratio of 100:1. For electrophoresis, the isolated proteins (11 µg) were placed onto a 10% sodium dodecyl sulfate-polyacrylamide gel. The separated proteins were transferred onto nitrocellulose (NC) membranes (0.45 µm-pore size; Bio-Rad, Hercules, CA, USA), blocked with 1× EzBlock Chemi solution (ATTO, Tokyo, Japan) for 30 min, and incubated overnight at 4°C with the primary antibodies: Col1a1 (1:1000; Abcam, Cambridge, UK), FN-1 (1:1000; Abcam, Cambridge, UK), Vimentin (1:1000; Abcam, Cambridge, UK), IL6 (1:1000; Abcam, Cambridge, UK), NLRP3 (1:1000, Adipogen, Liestal, Switzerland), Hes1 (1:1000; Cell Signaling Technology, Massachusetts, USA), Spp1 (1:1000; Cell Signaling Technology, Massachusetts, USA), Sox9 (1:1000; Cell Signaling Technology, Massachusetts, USA), β-actin (1:1000; Santa Cruz Biotechnology, California, USA). After that, the membranes were incubated for 1 h at 25°C with a secondary HRP-conjugated anti-rabbit or mouse antibody (1:3000; Jackson Immunoresearch, West Grove, PA, USA). As a protein loading control, β-actin was used. The Dyne ECL STAR western blotting Detection Kit (Dyne Bio, Seongnam, Republic of Korea) was used to visualize positive protein bands, and the results were quantified using an image analyzer (Image Lab 3.0, Bio-Rad, Hercules, CA, USA).
LX-2 culture
The human HSC line LX-2 cells were cultured in high glucose DMEM (Gibco, Grand Island, NY, USA) with 2% FBS (Gibco, Grand Island, NY, USA) and 1% penicillin/streptomycin (Welgene, Gyeongsan, Republic of Korea) at 37°C incubator with 5% CO2. After cultured for 24 hours, LX-2 cells were treated with 20 ng/ml TGF-β (R&D Systems, Minneapolis, MN, USA), and 11β-HSD1 inhibitor (1 µM).
HepG2 culture
The human hepatocellular carcinoma cell line HepG2 cells were cultured in low glucose DMEM (Gibco, Grand Island, NY, USA) with 10% FBS and 1% penicillin/streptomycin at 37°C incubator with 5% CO2. After cultured for 24 hours, HepG2 cells were treated with 0.02 g/ml TAA and 11β-HSD1 inhibitor (1 µM).
RNA extraction and quantitative real-time polymerase chain reaction (qRT-PCR)
Total RNA was isolated from liver tissues, LX-2 cells and HepG2 cells using TRIzol reagent (Invitrogen Co., Carlsbad, CA, USA). 1 mL of TRIzol and 0.2 mL of Chloroform were added to liver tissue, and after centrifugation at 12000rpm, 15 min, 4 ℃, only the supernatant was separated. After that, 0.5 mL of Isopropanol per 1 mL of Trizol was added, incubated at 4℃ for 10 minutes, and centrifuged at 12000 rpm, 10 min, 4 ℃ to obtain an RNA pellet. The absorbance of RNA was measured using a Nanodrop ND-2000 spectrophotometer (Thermo Scientific Inc., DE, USA). High-purity RNA having a purity of 1.8 to 2.0 was obtained and used at absorbance A260/A280 (nm). The extracted RNA (2 µg) was synthesized into cDNA using PrimeScript™ RT Reagent Kit. qRT-PCR amplification was performed using a LightCycler 480 with LightCycler 480 SYBR Green I Master mix. All tests were performed in three replicated with 96 well plates and normalized to GAPDH and β-actin. The sequences of all primers are listed in Table S5 and S6.
RNA sequencing analysis
RNA sequencing was performed by Macrogen, Inc. (Seoul, Republic of Korea) on liver tissues from the groups of mice whose efficacy was evaluated: normal control (n = 3), TAA-treated group (n = 3), TAA + OCA group (n = 3), TAA + 11β-HSD1 inhibitor responder group (n = 3), and TAA + 11β-HSD1 inhibitor non-responder group (n = 3). 11β-HSD1 inhibitor group, the subgroup that showed alleviation of fibrosis (depending on the fibrosis score [≤ 4] and expression of fibrosis-related markers) was defined as the responder group, whereas the group that did not show alleviation of fibrosis was defined as the non-responder group. The detailed procedures are described in the Supplementary Materials.
Data analysis for RNA sequencing
FastQC was used to check the quality of the raw sequencing data (17). FASTX_Trimmer (18) and BBMap (19) were used to eliminate adapters and low-quality readings (< Q20). TopHat was used to map the trimmed reads to the reference genome (20). Gene expression levels were estimated using fragments per kilobase per million read values using Cufflinks (21). These values were adjusted by the quantile normalization method using EdgeR within R (R Development Core Team, 2016).
Immune profiling of mouse peripheral blood mononuclear cells (PBMC) by mass cytometry
The immunological mechanism of action of the 11β-HSD1 inhibitor was investigated by determining the change in the population of immune cell types from mouse PMBC samples using mass cytometry.(22) The isolated PBMCs were stained with lanthanide metal-tagged surface antibodies. The detailed procedures are described in the Table S7. The analysis groups for mass cytometry were: normal control (n = 5), TAA-treated (n = 6), and TAA + 11β-HSD1 inhibitor (n = 8). Few samples were excluded owing to the limited number of live cells. For data processing and visualization, Cytobank (https://www.cytobank.org) was used. Subsequently, the primary immune cell types and subtypes were manually gated using the gating strategy (Table S8). The t-distributed stochastic neighbor embedding (t-SNE) approach was applied to minimize dimensionality and show data at single-cell resolution. Automated clustering methods, such as the t-SNE and citrus methods, were used in addition to the manual gating approach in this study. Samples with less than 1000 cells were excluded from the analysis. When mass cytometry analysis was performed, 20% of the cells were unassigned, and the remaining were assigned as leukocytes.
Statistical analysis
Statistical analyses were performed using Statistical Package for GraphPad Prism 9.0 (GraphPad Software, Inc., San Diego, CA, USA). All data are expressed as mean ± standard deviation. Data were analyzed using the Mann–Whitney U test (if the general assumptions were met, e.g., not normally distributed according to Kolmogorov–Smirnov test), one-way analysis of variance for multiple comparisons, and Tukey’s test for post-hoc multiple comparisons (equal variances assumed). Statistical significance was set at p < 0.05.