
CYP8B1 is a prognostic biomarker with important functional implications in hepatocellular carcinoma
- Authors:
- Published online on: June 27, 2025 https://doi.org/10.3892/or.2025.8937
- Article Number: 104
-
Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Hepatocellular carcinoma (HCC) is the sixth most commonly diagnosed cancer globally (4.7%) and ranks as the third leading cause of cancer-associated mortality (8.3%) in 2020 (1). Although early-stage HCC can potentially be treated with surgical resection, liver transplantation or ablation, the majority of patients are diagnosed with inoperable disease, resulting in a ~18% 5-year relative survival rate (2,3). In recent years, despite advancements in HCC treatments, including loco-regional therapies, targeted drugs and immunotherapy (4,5), overall survival outcomes remain unsatisfactory. Therefore, understanding the mechanism underlying HCC occurrence and progression and identifying effective targets are key for improving treatment strategies.
Cytochrome P450 (CYP) enzymes constitute a superfamily of monooxygenases notable for their extensive substrate diversity, surpassing all other enzyme families in this regard (6). Liver CYP enzymes serve a crucial role in the oxidative metabolism of endogenous compounds and xenobiotics, including drugs, carcinogens and toxins. Typically, aging is associated with a decline in CYP enzyme activity, which impacts the metabolism and clearance of CYP substrates (7). Bile acids (BAs), the end products of cholesterol catabolism, serve as amphiphilic emulsifiers, aiding lipid absorption in the intestine and facilitating the biliary excretion of cholesterol and phospholipids (8,9). Numerous CYP enzymes are involved in BA metabolism. Primary BAs are synthesized in the liver through the classical and alternative pathways. In the classical pathway, cholesterol is converted to 7α-hydroxycholesterol by cholesterol 7α-hydroxylase (CYP7A1), the rate-limiting enzyme, and subsequently to cholic acid by sterol 12α-hydroxylase, also known as cytochrome P450 8B1 (CYP8B1). In the alternative pathway, sterol 27-hydroxylase (CYP27A1) and oxysterol 7α-hydroxylase (CYP7B1) metabolize cholesterol into chenodeoxycholic acid (10). Beyond BA production, CYP8B1 also regulates lipid and glucose metabolism (11). CYP8B1 expression is influenced by various factors, including nutritional status and metabolic signaling (12). In metabolic diseases such as diabetes and obesity, CYP8B1 expression is increased, indicating its potential role in pathological states (13). Moreover, CYP8B1 activity is associated with key biological processes, including cholesterol metabolism and lipid absorption, making it a valuable biomarker for investigating metabolic syndrome and associated diseases (14). The formation and progression of tumors is a complex process influenced by the interplay of internal and external factors. The tumor microenvironment, genetic variation, lifestyle choices and environmental influences all contribute to tumorigenesis. Within this multifactorial framework, the development of HCC and other cancers is strongly associated with disruptions in BA metabolism. Alterations in BA composition and metabolism may facilitate tumor cell proliferation and metastasis, particularly in organs such as the liver, which are associated with BA metabolism (15). A comprehensive understanding of the multifactorial mechanisms underlying tumorigenesis is key for developing new therapeutic strategies. CYP8B1 has been implicated in various pathological conditions. For example, in patients with ulcerative colitis, Chen et al (10) demonstrated that the CYP8B1-cholic acid metabolic axis suppresses the renewal of leucine-rich repeat-containing G-protein coupled receptor 5) intestinal stem cells by inhibiting peroxisome proliferator-activated receptor α-mediated fatty acid oxidation. This inhibition slows epithelial barrier repair and exacerbates enteritis progression (10). Esophageal squamous cell carcinoma (ESCC), one of the most prevalent cancers worldwide with a poor prognosis and limited therapeutic targets, is also associated with CYP8B1. Liu et al (15) confirmed that CYP8B1 contributes to ESCC-associated malignancy and serves as a potential prognostic factor. Additionally, CYP8B1 expression is associated with the severity of non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH), suggesting its potential as a biomarker for NASH (16). In bariatric surgery, hepatic CYP8B1 is a critical target for regulating glycolipid metabolism. CYP8B1 controls the proportion of 12α hydroxylated BAs in the BA pool; changes in this ratio significantly alter gut microbiota composition, intestinal fat absorption and various metabolic signaling pathways, underscoring its central role in systemic metabolic regulation (17). Although some studies (18,19) have highlighted the association between CYP8B1 and HCC, its precise mechanisms and prognostic value in HCC remain insufficiently understood.
The present study aimed to explore the mechanisms through which CYP8B1 influences HCC progression.
Materials and methods
Clinical samples
HCC and adjacent normal tissues (distance, >5cm) were collected from five patients (age range from 51 to 73, five patients were all males) with HCC from September to November 2024 in The First People's Hospital of Anqing (Anqing, China) during routine surgery. The present study was approved by the Research Ethics Committee of The First People's Hospital of Anqing (approval no. 20240077), and written informed consent was obtained from all participants prior to participation.
Gene expression profiles
Gene expression profiles for datasets GSE84402, GSE26538 and GSE141090 were retrieved from the Gene Expression Omnibus (GEO, ncbi.nlm.nih.gov.cn). The Xiantao Academic website (20) was used for generating Venn diagrams and analyzing the association between immune infiltration and CYP8B1 expression. The association between CYP8B1 expression and immune cell infiltration was analyzed based on ssGSEA algorithm provided in R package (version 4.2.1; cran.r-project.org/). The expression of CYP8B1 and the content of immune cells was obtained from The Cancer Genome Atlas (TCGA) database [TCGA-liver hepatocellular carcinoma (LIHC, the URL was: portal.gdc.cancer.gov). The association between the expression of CYP8B1 and the content of immune cells was analyzed by Spearman's test and to further show the relationship between CYP8B1 and immune infiltration. The content of the immune cells can indicate the degree of immune infiltration. The survival and expression analyses of CYP8B1 were performed using CYP8B1 expression data and clinical information from the UALCAN (ualcan.path.uab.edu) (21) and Gene Expression Profiling Interactive Analysis (GEPIA) platform (gepia.cancer-pku.cn) (22). Cox regression analysis and Kaplan-Meier plots for TCGA datasets was performed using RStudio software (version 4.2.1) to investigate the association between CYP8B1 expression and cancer prognosis, including overall survival in LIHC subclinical groups. Additionally, the UALCAN and GEPIA database were also used to determine the association between CYP8B1 expression and overall survival. The heatmap of positively and negatively associated genes with CYP8B1 in LIHC was analyzed by UALCAN database to show the top 24 associated genes. Furthermore, gene analysis of positively and negatively associated genes with CYP8B1 in LIHC were performed using the Metascape website (metascape.org) to show the pathway and biological progress of these genes (23). Gene expression of CYP8B1 in HCC cell lines was performed using the Depmap website (depmap.org/portal/).
Cell culture
HCC cell lines [Huh7 (cat. no. SCSP-526) and Hep3b (cat. no. SCSP-5045)] were obtained from Shanghai Institutes of the Chinese Academy of Sciences. The cells were maintained in high-glucose DMEM (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin and 100 µg/ml streptomycin (complete medium) at 37°C in a humidified incubator with 5% CO2.
Overexpression of CYP8B1 in HCC cell lines
The recombinant CYP8B1 plasmid was constructed by cloning into the pcDNA3.0 (Shanghai GeneChem Co., Ltd.) vector and transfected into HCC cells using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), following the manufacturer's protocols. A total of 2 µg/well overexpressed CYP8B1 plasmid (oe-CYP8B1) and empty vector (negative control, NC) was transfected into HCC cells in 6-plate well for 6 h for duration in at 37°C, the medium was replaced with complete medium. After 48 h, the cells were collected for subsequent experimentation.
Cell Counting Kit-8 (CCK-8) assay
The CCK-8 assay were performed to assess cell proliferation as previously described (20). Briefly, transfected 5000 HCC cells were seeded into every well of 96-well plates. Cell proliferation was measured using CCK-8 (Nanjing KeyGen Biotech Co., Ltd.) according to the manufacturer's instructions after incubation for 1, 2, 3 and 4 days. All the experiments were performed in triplicate.
EdU assay
EdU incorporation assay was used to measure cell proliferation rate as previously described (20). The proliferation rate was calculated following the manufacturer's instructions using the BeyoClick™ EdU-555 EdU kit (Beyotime Institute of Biotechnology). Each EdU incorporation experiment was performed ≥3 times.
TUNEL assay
Cell apoptosis was assessed using the TUNEL assay kit (Nanjing KeyGen Biotech Co., Ltd.) according to the manufacturer's instructions, as previously described (24). The experiments were repeated ≥3 times.
Western blotting analysis
Protein samples from patient tissue and tumor cells) were lysed in RIPA buffer supplemented with fresh protease and phosphatase inhibitor cocktails (Beyotime Biotech Co., Ltd.). The protein concentration was determined using the BCA assay (Pierce; Thermo Fisher Scientific, Inc.). Equal amounts of proteins (20 µg/lane) were separated by10% SDS-PAGE and transferred onto PVDF membranes. The membranes were blocked with 3% BSA (Beyotime Biotech Co., Ltd.) in 10 mM Tris-HCl (pH 7.4) containing 0.05% Tween-20 to prevent non-specific binding. Membranes were incubated with primary antibody at 4°C for 12 h, followed by incubation with a corresponding peroxidase-conjugated secondary antibody (Abcam; Table SI) at room temperature for 2 h. Immunoreactive bands were visualized using SuperSignal West Pico Chemiluminescent Substrate (Pierce; Thermo Fisher Scientific, Inc.) and band density was quantified using a Versadoc Imaging System Model 3000. Each experiment was conducted ≥3 times.
Reverse transcription-quantitative (RT-q)PCR
Total RNA was extracted from HCC cell lines using TRIzol (Invitrogen; Thermo Fisher Scientific, Inc.) following the manufacturer's instructions. RT-qPCR was performed as previously described (25). Briefly, 1 µg total RNA was reverse-transcribed using random primers and Primescript reverse transcriptase (Vazyme Biotech Co., Ltd.). RT protocol was as follows: 37°C for 15 min, 85°C for 5 sec, 4°C forever. qPCR for the target genes was conducted using the SYBR-Green qPCR kit (Vazyme Biotech Co., Ltd.) on a fluorescent temperature cycler (ABI 7500 Real-Time PCR system; Thermo Fisher Scientific, Inc.). The primers were as follows: CYP8B1: Forward, 5′-ACCTGAGCTTGTTCGGCTAC-3′ and reverse, 5′-CGGAGAGCATCTTGTGAAAG-3′ and β-actin: Forward, 5′-ATCGTGCGTGACATTAAGGAGAAC-3′ and reverse, 5′-AGGAAGGAAGGCTGGAAGAGTG-3′. The thermocycling conditions were as follows: Initial denaturation at 95°C for 5 min, followed by 45 cycles of amplification at 95°C for 15 sec and annealing at 60°C for 1 min. Each experiment was performed ≥3 three times. Relative levels of mRNA expression were obtained via the 2−ΔΔCq method (26).
Statistical analysis
All statistical analyses were performed using SPSS 18.0 software (SPSS Inc.). Data are presented mean ± SD, and independent experiments repeats three times. Differences between categorical variables were analyzed using the independent sample unpaired or paired Student's t-test. P<0.05 was considered to indicate a statistically significant difference.
Results
CYP8B1 expression is downregulated in HCC
HCC gene expression profiles from GEO datasets, including data from human, mice and rat studies, were used to identify differentially expressed genes (DEGs) using a Venn diagram. The analysis revealed one up- (CDK1) and four downregulated DEGs [CYP8B1, MFSD2A (major facilitator super family domain containing 2a), VIPR1 (Vasoactive intestinal peptide receptor 1), THRSP (Thyroid hormone-responsive protein)] in HCC compared with normal tissues (Fig. 1A-C). Except for CYP8B1, these DEGs have been studied in HCC (27–29); therefore, the present study investigated CYP8B1. In TCGA database, CYP8B1 expression was significantly lower in HCC tumor tissues compared with normal tissues, as shown on the UALCAN (Fig. 1D) and the GEPIA platform (Fig. 1E). Consistently, GTEx and TCGA databases demonstrated reduced CYP8B1 expression in HCC compared with normal tissue (Fig. 1F). In the GSE84402 dataset, CYP8B1 levels were significantly lower in HCC tissues than in corresponding normal tissue (Fig. 1G). Additionally, tumor and adjacent normal tissue samples were collected from five patients with HCC. Immunoblotting analysis showed that CYP8B1 expression was significantly lower in tumor tissues compared with adjacent normal tissues (Fig. 1H and I).
Prognostic value of CYP8B1 in HCC
To evaluate the prognostic significance of CYP8B1 in HCC, UALCAN and GEPIA databases were used. Patients with HCC with low CYP8B1 expression exhibited worse overall survival outcomes (Fig. 2A and B). Decreased CYP8B1 expression was associated with poorer OS in specific clinical subgroups, including those with AFP (α-fetoprotein) levels >400 ng/ml, individuals aged ≥60 years, male patients, patients taller than 170 cm, Asian patients and patients with residual tumor (R0; Fig. 2C-H).
Association of CYP8B1 with immune cell infiltration
Given the key role of tumor-infiltrating lymphocytes in cancer progression and their impact on patient prognosis (30), along with the potential oncogenic role of CYP8B1 in HCC, the relationship between CYP8B1 expression and the degree of immune infiltration in HCC was investigated. We detected the association between CYP8B1 expression and different immune cells content in TCGA database and found that CYP8B1 expression was positively associated with content of the following immune cells such as T helper cell (Th17) cells, neutrophils, central memory T cell) cells, Tregs (Regulatory T cells), DC (Dendritic cells), Tgd (T γΔ) cells and eosinophils. Conversely, CYP8B1 expression showed a significant negative correlation with content of the following immune cells such as Th1 cells, active Dendritic cells), B cells (Bone marrow-dependent lymphocyte), interdigitating Dendritic cells), Th (T helper cells) and T (Thymus-dependent lymphocyte) cells, macrophages and effective memory T cell), Th2 (T helper cell 2), TFH (Follicular helper T cell) and NK (Nature killer) CD56 bright cells. (Fig. 3A-G). Furthermore, we selected 17 types immune cells which was significantly associated with expression of CYP8B1 and found that the low and high expression of CYP8B1 has significant differences in all the 16 immune cells except for eosinophils (Fig. 3H).
Detection of CYP8B1 overexpression efficiency
To investigate the role of CYP8B1 in HCC, its expression levels were searched in public datasets (depmap.org/portal/). CYP8B1 expression was high in HepG2 and low in Huh7 and Hep3B cells (Table SII). To study the functional effects, CYP8B1 was overexpressed in Hep3B and Huh7 cells by transfection with a CYP8B1-overexpression plasmid. Following transfection, both mRNA (Fig. 4A and C) and protein levels (Fig. 4B and D) of CYP8B1 were significantly increased in the two HCC cell lines.
CYP8B1 overexpression inhibits cell proliferation and promotes apoptosis
To evaluate the effect of CYP8B1 on cell proliferation, CCK-8 (Fig. 5A) and EdU assays (Fig. 5B and C) were performed in cells overexpressing CYP8B1. Both assays showed that CYP8B1 overexpression significantly decreased the proliferation of Huh7 and Hep3b cells (Fig. 5D-F).
To investigate the effect of CYP8B1 on apoptosis, TUNEL assay was performed. CYP8B1 overexpression markedly increased the apoptosis rate in Huh7 (Fig. 6A and B) and Hep3b cells (Fig. 6C and D).
Mechanisms of CYP8B1 in regulating HCC cell function
To explore the mechanisms of CYP8B1 in HCC, the top 24 genes positively and negatively correlated with CYP8B1 expression in LIHC were identified using the UALCAN platform (Fig. 7A and B) and submitted to the Metascape platform for pathway enrichment analysis (Fig. 7C and D). This revealed two key pathways potentially regulated by CYP8B1: ‘Regulation of PLK activity at G2/M transition’ and ‘intrinsic pathway for apoptosis’ (Fig. 7C). As CYP8B1 overexpression inhibited cell proliferation and promoted apoptosis, the present study identified genes from PubMed involved in the G2/M transition and apoptosis. YWHAZ (Tyrosine 3/tryptophan 5 monooxygenase activation protein ζ) downregulation induces G2/M transition and apoptosis (23,24). Expression of YWHAZ was negatively associated with CYP8B1 in LIHC (Fig. 7A). Immunoblot assay was used to clarify the expression of YWHAZ after CYP8B1 overexpression, as well as the expression of downstream factors of YWHAZ, including Cyclin B1, CDK1, Bax and Bcl2 (Fig. 7E and F) in Huh7 and Hep3b cells. YWHAZ was significantly downregulated following CYP8B1 overexpression. Downstream factors of YWHAZ, including Cyclin B1 and CDK1 which was involved in the G2/M cell cycle was significantly downregulated. Apoptosis-related genes Bax was upregulated and Bcl2 was downregulated after CYP8B1 overexpression. The results suggested that CYP8B1 mediates G2/M arrest via the YWHAZ/CyclinB1/CDK1 axis and apoptosis primarily via the YWHAZ/Bax/Bcl2 axis.
Discussion
CYP8B1 plays a key role in the conversion of cholesterol into BAs. It facilitates the hydroxylation of the steroid ring at the C12 position, leading to the production of the BA cholic acid (31). Due to its importance in cholesterol homeostasis and lipid metabolism, CYP8B1 is as a key target for managing metabolic diseases, including NAFLD and type 2 diabetes (32,33). CYP8B1 has been implicated in tumor growth and apoptosis. For example, kaempferol has been shown to upregulate CYP8B1, thereby attenuating colorectal cancer progression (34). The present study identified CYP8B1 as a prognostic biomarker for HCC based on analysis from GEO datasets, with low expression correlating with poor prognosis. The tumor microenvironment, especially the immune microenvironment, serves a vital role in the malignant progression and metastasis of cancer (35). The present findings further demonstrated an association between CYP8B1 expression and immune cell infiltration in the liver cancer microenvironment. Additionally, CYP8B1 overexpression inhibited proliferation and promoted apoptosis in HCC cells. Through analysis of CYP8B1 negatively correlated genes in LIHC, the present study identified YWHAZ as a potential downstream factor regulated by CYP8B1.
YWHAZ, a member of the 14-3-3protein family, is a critical hub protein involved in numerous signal transduction pathways and plays a key role in tumor progression (36). YWHAZ regulates the G2/M cell cycle transition, thereby promoting cell proliferation in various types of cancer such as gastric and colorectal cancer (37,38). Silencing YWHAZ has been shown to increase apoptosis by modulating key apoptotic markers such as Bax and Bcl2 (24). CylinB1 and CDK1 are the downstream factors of YWHAZ that regulate G2/M transition (37), and Bax and Bcl2 are the downstream factors of YWHAZ involved in cell apoptosis (39). Consistent with the present findings, CYP8B1 is negatively associated with YWHAZ and G2/M cell cycle. YWHAZ may serve as a critical downstream effector of CYP8B1. Subsequent validation by immunoblotting confirmed this hypothesis.
The number of clinical samples in the present study was small; future studies require more samples to prove the present conclusions. Secondly, the present study was based on in vitro experiments and lacked validation in animal experiments. Thirdly, the application of this gene in the clinical treatment of HCC has not been explored.
In summary, by analyzing gene expression profiles from humans, mice and rats, the present study identified CYP8B1 as a significantly dysregulated gene in HCC. Bioinformatics and experimental analyses demonstrated that low CYP8B1 expression correlates with poor prognosis in HCC. Overexpression of CYP8B1 inhibited proliferation and promoted apoptosis in HCC cell lines. Mechanistically, CYP8B1 may regulate cell proliferation and apoptosis, at least in part, via YWHAZ. CYP8B1 could be a biomarker for prognosis of HCC. The present study provides evidence that CYP8B1 could serve as a promising therapeutic target for HCC.
Supplementary Material
Supporting Data
Acknowledgements
Not applicable.
Funding
The present study was supported by Anqing Science and Technology Bureau Project (grant no. 2023Z2007).
Availability of data and materials
The data generated in the present study may be requested from the corresponding author.
Authors' contributions
CL and FL designed the study and wrote the manuscript. CL, FL, QD and QP performed the experiments. ZK supplied the clinical samples. FL, QD, QP and ZK analyzed the data. FL, DQ, ZK QP and CL confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.
Ethics approval and consent to participate
Ethics approval was permitted by the Research Ethics Committee of The First People's Hospital of Anqing (approval no. 20240077; Anqing, China) and written informed consent was obtained from the patients.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI | |
Bi F, Qiu Y, Wu Z, Liu S, Zuo D, Huang Z, Li B, Yuan Y, Niu Y and Qiu J: METTL9-SLC7A11 axis promotes hepatocellular carcinoma progression through ferroptosis inhibition. Cell Death Discov. 9:4282023. View Article : Google Scholar : PubMed/NCBI | |
Yang C, Zhang H, Zhang L, Zhu AX, Bernards R, Qin W and Wang C: Evolving therapeutic landscape of advanced hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 20:203–222. 2023. View Article : Google Scholar : PubMed/NCBI | |
Shokoohian B, Negahdari B, Aboulkheyr Es H, Abedi-Valugerdi M, Baghaei K, Agarwal T, Maiti TK, Hassan M, Najimi M and Vosough M: Advanced therapeutic modalities in hepatocellular carcinoma: Novel insights. J Cell Mol Med. 25:8602–8614. 2021. View Article : Google Scholar : PubMed/NCBI | |
Kim HJ, Lee SH, Shim HJ, Bang HJ, Cho SH, Chung IJ, Hwang EC, Hwang JE and Bae WK: Hepatic arterial infusion chemotherapy versus systemic therapy for advanced hepatocellular carcinoma: A systematic review and meta-analysis. Front Oncol. 13:12652402023. View Article : Google Scholar : PubMed/NCBI | |
Sheikholeslami B, Tootoonchi Z, Lavasani H, Hosseinzadeh Ardakani Y and Rouini M: Investigation of MDMA inhibitory effect on cytochromeP450 3A4 in isolated perfused rat liver model using tramadol. Adv Pharm Bull. 11:530–536. 2021. View Article : Google Scholar : PubMed/NCBI | |
Haduch A, Bromek E, Kuban W, Basińska-Ziobroń A, Danek PJ, Alenina N, Bader M and Daniel WA: The effect of brain serotonin deficit (TPH2-KO) on the expression and activity of liver cytochrome P450 enzymes in aging male Dark Agouti rats. Pharmacol Rep. 75:1522–1532. 2023. View Article : Google Scholar : PubMed/NCBI | |
Chiang JYL and Ferrell JM: Discovery of farnesoid X receptor and its role in bile acid metabolism. Mol Cell Endocrinol. 548:1116182022. View Article : Google Scholar : PubMed/NCBI | |
Shulpekova Y, Shirokova E, Zharkova M, Tkachenko P, Tikhonov I, Stepanov A, Sinitsyna A, Izotov A, Butkova T, Shulpekova N, et al: A recent ten-year perspective: Bile acid metabolism and signaling. Molecules. 27:19832022. View Article : Google Scholar : PubMed/NCBI | |
Chen L, Jiao T, Liu W, Luo Y, Wang J, Guo X, Tong X, Lin Z, Sun C, Wang K, et al: Hepatic cytochrome P450 8B1 and cholic acid potentiate intestinal epithelial injury in colitis by suppressing intestinal stem cell renewal. Cell Stem Cell. 29:1366–1381.e9. 2022. View Article : Google Scholar : PubMed/NCBI | |
Kaur A, Patankar JV, de Haan W, Ruddle P, Wijesekara N, Groen AK, Verchere CB, Singaraja RR and Hayden MR: Loss of Cyp8b1 improves glucose homeostasis by increasing GLP-1. Diabetes. 64:1168–1179. 2015. View Article : Google Scholar : PubMed/NCBI | |
Pozzo L, Vornoli A, Coppola I, Croce CM, Giorgetti L, Gervasi PG and Longo V: Effect of HFD/STZ on expression of genes involved in lipid, cholesterol and glucose metabolism in rats. Life Sci. 166:149–156. 2016. View Article : Google Scholar : PubMed/NCBI | |
Pathak P and Chiang JYL: Sterol 12α-hydroxylase aggravates dyslipidemia by activating the ceramide/mTORC1/SREBP-1C pathway via FGF21 and FGF15. Gene Expr. 19:161–173. 2019. View Article : Google Scholar : PubMed/NCBI | |
Hoogerland JA, Lei Y, Wolters JC, de Boer JF, Bos T, Bleeker A, Mulder NL, van Dijk TH, Kuivenhoven JA, Rajas F, et al: Glucose-6-phosphate regulates hepatic bile acid synthesis in mice. Hepatology. 70:2171–2184. 2019. View Article : Google Scholar : PubMed/NCBI | |
Liu X, Hong R, Du P, Yang D, He M, Wu Q, Li L, Wang Y, Chen J, Min Q, et al: The metabolic genomic atlas reveals potential drivers and clinically relevant insights into the etiology of esophageal squamous cell carcinoma. Theranostics. 12:6160–6178. 2022. View Article : Google Scholar : PubMed/NCBI | |
Xu F, Yu Z, Liu Y, Du T, Yu L, Tian F, Chen W and Zhai Q: A high-fat, high-cholesterol diet promotes intestinal inflammation by exacerbating gut microbiome dysbiosis and bile acid disorders in cholecystectomy. Nutrients. 15:38292023. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Tu J, Shi L, Fang Z, Fan M, Zhang J, Ding L, Chen Y, Wang Y, Zhang E, et al: CYP8B1 downregulation mediates the metabolic effects of vertical sleeve gastrectomy in mice. Hepatology. 79:1005–1018. 2024. View Article : Google Scholar : PubMed/NCBI | |
Wang X, Liao X, Yang C, Huang K, Yu T, Yu L, Han C, Zhu G, Zeng X, Liu Z, et al: Identification of prognostic biomarkers for patients with hepatocellular carcinoma after hepatectomy. Oncol Rep. 41:1586–1602. 2019.PubMed/NCBI | |
Zhang R, Huang M, Wang H, Wu S, Yao J, Ge Y, Lu Y and Hu Q: Identification of potential biomarkers from hepatocellular carcinoma with MT1 deletion. Pathol Oncol Res. 27:5975272021. View Article : Google Scholar : PubMed/NCBI | |
Vivian J, Rao AA, Nothaft FA, Ketchum C, Armstrong J, Novak A, Pfeil J, Narkizian J, Deran AD, Musselman-Brown A, et al: Toil enables reproducible, open source, big biomedical data analyses. Nat Biotechnol. 35:314–316. 2017. View Article : Google Scholar : PubMed/NCBI | |
Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce-Rodriguez I, Chakravarthi BVSK and Varambally S: UALCAN: A portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia. 19:649–658. 2017. View Article : Google Scholar : PubMed/NCBI | |
Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45:W98–W102. 2017. View Article : Google Scholar : PubMed/NCBI | |
Li Y, Li R, Guo S, Li Y, Wang Y, Wen X, Lan T and Gong K: Bioinformatics-based identification of lipid- and immune-related biomarkers in abdominal aortic aneurysms. Heliyon. 9:e136222023. View Article : Google Scholar : PubMed/NCBI | |
Li F, Liu CS, Wu P, Ling AS, Pan Q and Li XN: CCT4 suppression inhibits tumor growth in hepatocellular carcinoma by interacting with Cdc20. Chin Med J (Engl). 134:2721–2729. 2021. View Article : Google Scholar : PubMed/NCBI | |
Liu C, Li F, Li X, Cao M, Feng G, Yuan X and Shi X: WIPI2 depletion inhibits the growth of hepatocellular carcinoma cells through the AMPK signaling pathway. Oncol Rep. 43:1467–1478. 2020.PubMed/NCBI | |
Wang W, Chen H, Gao W, Wang S, Wu K, Lu C, Luo X, Li L and Yu C: Girdin interaction with vimentin induces EMT and promotes the growth and metastasis of pancreatic ductal adenocarcinoma. Oncol Rep. 44:637–649. 2020. View Article : Google Scholar : PubMed/NCBI | |
Xing S, Kan J, Su A, Liu QD, Wang K, Cai X and Dong J: The prognostic value of major facilitator superfamily domain-containing protein 2A in patients with hepatocellular carcinoma. Aging (Albany NY). 11:8474–8483. 2019. View Article : Google Scholar : PubMed/NCBI | |
Fu Y, Liu S, Rodrigues RM, Han Y, Guo C, Zhu Z, He Y, Mackowiak B, Feng D, Gao B, et al: Activation of VIPR1 suppresses hepatocellular carcinoma progression by regulating arginine and pyrimidine metabolism. Int J Biol Sci. 18:4341–4356. 2022. View Article : Google Scholar : PubMed/NCBI | |
Hu Q, Ma X, Li C, Zhou C, Chen J and Gu X: Downregulation of THRSP promotes hepatocellular carcinoma progression by triggering ZEB1 transcription in an ERK-dependent manner. J Cancer. 12:4247–4256. 2021. View Article : Google Scholar : PubMed/NCBI | |
Liu F, Zeng G, Zhou S, He X, Sun N, Zhu X and Hu A: Blocking Tim-3 or/and PD-1 reverses dysfunction of tumor-infiltrating lymphocytes in HBV-related hepatocellular carcinoma. Bull Cancer. 105:493–501. 2018. View Article : Google Scholar : PubMed/NCBI | |
Liu J, Carlson HA and Scott EE: The structure and characterization of human cytochrome P450 8B1 supports future drug design for nonalcoholic fatty liver disease and diabetes. J Biol Chem. 298:1023442022. View Article : Google Scholar : PubMed/NCBI | |
Patankar JV, Wong CK, Morampudi V, Gibson WT, Vallance B, Ioannou GN and Hayden MR: Genetic ablation of Cyp8b1 preserves host metabolic function by repressing steatohepatitis and altering gut microbiota composition. Am J Physiol Endocrinol Metab. 314:E418–E432. 2018. View Article : Google Scholar : PubMed/NCBI | |
Mao Y, Zhao Y, Luo S, Chen H, Liu X, Wu T, Ding G, Liu X, Sheng J, Meng Y and Huang H: Advanced paternal age increased metabolic risks in mice offspring. Biochim Biophys Acta Mol Basis Dis. 1868:1663552022. View Article : Google Scholar : PubMed/NCBI | |
Li X, Khan I, Huang G, Lu Y, Wang L, Liu Y, Lu L, Hsiao WLW and Liu Z: Kaempferol acts on bile acid signaling and gut microbiota to attenuate the tumor burden in ApcMin/+ mice. Eur J Pharmacol. 918:1747732022. View Article : Google Scholar : PubMed/NCBI | |
Mou P, Ge QH, Sheng R, Zhu TF, Liu Y and Ding K: Research progress on the immune microenvironment and immunotherapy in gastric cancer. Front Immunol. 14:12911172023. View Article : Google Scholar : PubMed/NCBI | |
Gan Y, Ye F and He XX: The role of YWHAZ in cancer: A maze of opportunities and challenges. J Cancer. 11:2252–2264. 2020. View Article : Google Scholar : PubMed/NCBI | |
Sheng N, Yan L, Wu K, You W, Gong J, Hu L, Tan G, Chen H and Wang Z: TRIP13 promotes tumor growth and is associated with poor prognosis in colorectal cancer. Cell Death Dis. 9:4022018. View Article : Google Scholar : PubMed/NCBI | |
Ma J, Chen W, Wang K, Tian K, Li Q, Zhao T, Zhang L, Wang L, Wu Z and Zhang J: Identification of the different roles and potential mechanisms of T isoforms in the tumor recurrence and cell cycle of chordomas. Onco Targets Ther. 12:11777–11791. 2019. View Article : Google Scholar : PubMed/NCBI | |
Guo F, Jiao D, Sui GQ, Sun LN, Gao YJ, Fu QF and Jin CX: Anticancer effect of YWHAZ silencing via inducing apoptosis and autophagy in gastric cancer cells. Neoplasma. 65:693–700. 2018. View Article : Google Scholar : PubMed/NCBI |