1
|
Liu M, Lai M, Li D, Zhang R, Wang L, Peng W, Yang J, He W, Sheng Y, Xiao S, Nan A, Zeng X. Nucleus-localized circSLC39A5 suppresses hepatocellular carcinoma development by binding to STAT1 to regulate TDG transcription. Cancer Sci 2023; 114:3884-3899. [PMID: 37549641 PMCID: PMC10551608 DOI: 10.1111/cas.15906] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 08/09/2023] Open
Abstract
Accumulating evidence indicates that circular RNAs (circRNAs) are inextricably linked to cancer development. However, the function and mechanism of nucleus-localized circRNAs in hepatocellular carcinoma (HCC) still require investigation. Here, qRT-PCR and receiver-operating characteristic curve were used to detect the expression and diagnostic potential of circSLC39A5 for HCC. The biological function of circSLC39A5 in HCC was investigated in vitro and in vivo. Nucleoplasmic separation assay, fluorescence in situ hybridization, RNA pulldown, RNA immunoprecipitation, the HDOCK Server, the NucleicNet Webserver, crosslinking-immunoprecipitation, MG132 treatment, and chromatin immunoprecipitation were utilized to explore the potential molecular mechanism of circSLC39A5 in HCC. The results showed that circSLC39A5 was downregulated in both HCC tissues and plasma and was associated with satellite nodules and lymph node metastasis/vascular invasion. CircSLC39A5 was stably expressed in plasma samples under different storage conditions, showing good diagnostic potential for HCC (AUC = 0.915). CircSLC39A5 inhibited proliferation, migration, and invasion, facilitated the apoptosis of HCC cells, and was associated with low expression of Ki67 and CD34. Remarkably, circSLC39A5 is mainly localized in the nucleus and binds to the transcription factor signal transducer and activator of transcription 1 (STAT1), affecting its stabilization and expression. STAT1 binds to the promoter of thymine DNA glycosylase (TDG). Overexpression of circSLC39A5 elevates TDG expression and reverses the increase of proliferating cell nuclear antigen (PCNA) expression and the overactive cell proliferation caused by TDG silencing. Our findings uncovered a novel plasma circRNA, circSLC39A5, which may be a potential circulating diagnostic marker for HCC, and the mechanism by which nucleus-localized circSLC39A5 exerts a transcriptional regulatory role in HCC by affecting STAT1/TDG/PCNA provides new insights into the mechanism of circRNAs.
Collapse
Affiliation(s)
- Meiliang Liu
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
| | - Mingshuang Lai
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
| | - Deyuan Li
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
| | - Ruirui Zhang
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
- Department of Toxicology, School of Public HealthGuangxi Medical UniversityNanningChina
| | - Lijun Wang
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
| | - Wenyi Peng
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
- Department of Toxicology, School of Public HealthGuangxi Medical UniversityNanningChina
| | - Jialei Yang
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
| | - Wanting He
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
| | - Yonghong Sheng
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
| | - Suyang Xiao
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
| | - Aruo Nan
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
- Department of Toxicology, School of Public HealthGuangxi Medical UniversityNanningChina
| | - Xiaoyun Zeng
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent DiseasesGuangxi Medical UniversityNanningChina
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of EducationNanningChina
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency TumorNanningChina
| |
Collapse
|
2
|
Yang G, Liu R, Rezaei S, Liu X, Wan YJY. Uncovering the Gut-Liver Axis Biomarkers for Predicting Metabolic Burden in Mice. Nutrients 2023; 15:3406. [PMID: 37571345 PMCID: PMC10421148 DOI: 10.3390/nu15153406] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Western diet (WD) intake, aging, and inactivation of farnesoid X receptor (FXR) are risk factors for metabolic and chronic inflammation-related health issues ranging from metabolic dysfunction-associated steatotic liver disease (MASLD) to dementia. The progression of MASLD can be escalated when those risks are combined. Inactivation of FXR, the receptor for bile acid (BA), is cancer prone in both humans and mice. The current study used multi-omics including hepatic transcripts, liver, serum, and urine metabolites, hepatic BAs, as well as gut microbiota from mouse models to classify those risks using machine learning. A linear support vector machine with K-fold cross-validation was used for classification and feature selection. We have identified that increased urine sucrose alone achieved 91% accuracy in predicting WD intake. Hepatic lithocholic acid and serum pyruvate had 100% and 95% accuracy, respectively, to classify age. Urine metabolites (decreased creatinine and taurine as well as increased succinate) or increased gut bacteria (Dorea, Dehalobacterium, and Oscillospira) could predict FXR deactivation with greater than 90% accuracy. Human disease relevance is partly revealed using the metabolite-disease interaction network. Transcriptomics data were also compared with the human liver disease datasets. WD-reduced hepatic Cyp39a1 (cytochrome P450 family 39 subfamily a member 1) and increased Gramd1b (GRAM domain containing 1B) were also changed in human liver cancer and metabolic liver disease, respectively. Together, our data contribute to the identification of noninvasive biomarkers within the gut-liver axis to predict metabolic status.
Collapse
Affiliation(s)
- Guiyan Yang
- Department of Medical Pathology, Laboratory Medicine in Sacramento, University of California, Davis, CA 95817, USA;
| | - Rex Liu
- Department of Computer Science, University of California, Davis, CA 95616, USA; (R.L.); (S.R.); (X.L.)
| | - Shahbaz Rezaei
- Department of Computer Science, University of California, Davis, CA 95616, USA; (R.L.); (S.R.); (X.L.)
| | - Xin Liu
- Department of Computer Science, University of California, Davis, CA 95616, USA; (R.L.); (S.R.); (X.L.)
| | - Yu-Jui Yvonne Wan
- Department of Medical Pathology, Laboratory Medicine in Sacramento, University of California, Davis, CA 95817, USA;
| |
Collapse
|
3
|
Aranda S, Alcaine-Colet A, Ballaré C, Blanco E, Mocavini I, Sparavier A, Vizán P, Borràs E, Sabidó E, Di Croce L. Thymine DNA glycosylase regulates cell-cycle-driven p53 transcriptional control in pluripotent cells. Mol Cell 2023:S1097-2765(23)00517-8. [PMID: 37506700 DOI: 10.1016/j.molcel.2023.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/11/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023]
Abstract
Cell cycle progression is linked to transcriptome dynamics and variations in the response of pluripotent cells to differentiation cues, mostly through unknown determinants. Here, we characterized the cell-cycle-associated transcriptome and proteome of mouse embryonic stem cells (mESCs) in naive ground state. We found that the thymine DNA glycosylase (TDG) is a cell-cycle-regulated co-factor of the tumor suppressor p53. Furthermore, TDG and p53 co-bind ESC-specific cis-regulatory elements and thereby control transcription of p53-dependent genes during self-renewal. We determined that the dynamic expression of TDG is required to promote the cell-cycle-associated transcriptional heterogeneity. Moreover, we demonstrated that transient depletion of TDG influences cell fate decisions during the early differentiation of mESCs. Our findings reveal an unanticipated role of TDG in promoting molecular heterogeneity during the cell cycle and highlight the central role of protein dynamics for the temporal control of cell fate during development.
Collapse
Affiliation(s)
- Sergi Aranda
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain.
| | - Anna Alcaine-Colet
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Cecilia Ballaré
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Enrique Blanco
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Ivano Mocavini
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | | | - Pedro Vizán
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Blanquerna School of Health Science, Universitat Ramon Llull, Barcelona 08025, Spain
| | - Eva Borràs
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Eduard Sabidó
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Luciano Di Croce
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; ICREA, Pg. Lluis Companys 23, Barcelona 08010, Spain.
| |
Collapse
|
4
|
Hassan HM, Onabote O, Isovic M, Passos DT, Dick FA, Torchia J. Regulation of Chromatin Accessibility by the Farnesoid X Receptor Is Essential for Circadian and Bile Acid Homeostasis In Vivo. Cancers (Basel) 2022; 14:cancers14246191. [PMID: 36551676 PMCID: PMC9777377 DOI: 10.3390/cancers14246191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
The Farnesoid X Receptor (FXR) belongs to the nuclear receptor superfamily and is an essential bile acid (BA) receptor that regulates the expression of genes involved in the metabolism of BAs. FXR protects the liver from BA overload, which is a major etiology of hepatocellular carcinoma. Herein, we investigated the changes in gene expression and chromatin accessibility in hepatocytes by performing RNA-seq in combination with the Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC-seq) using a novel FXR knockout mouse model (Fxrex5Δ: Nr1h4ex5Δ/ex5Δ) generated through CRISPR/Cas9. Consistent with previous Fxr knockout models, we found that Fxrex5Δ mice develop late-onset HCC associated with increased serum and hepatic BAs. FXR deletion was associated with a dramatic loss of chromatin accessibility, primarily at promoter-associated transcription factor binding sites. Importantly, several genes involved in BA biosynthesis and circadian rhythm were downregulated following loss of FXR, also displayed reduced chromatin accessibility at their promoter regions. Altogether, these findings suggest that FXR helps to maintain a transcriptionally active state by regulating chromatin accessibility through its binding and recruitment of transcription factors and coactivators.
Collapse
Affiliation(s)
- Haider M. Hassan
- Department of Biochemistry, Western University, London, ON N6A 5C1, Canada
- Department of Oncology, London Regional Cancer Program and the Lawson Health Research Institute, London, ON N6A 5W9, Canada
| | - Oladapo Onabote
- Department of Biochemistry, Western University, London, ON N6A 5C1, Canada
- Department of Oncology, London Regional Cancer Program and the Lawson Health Research Institute, London, ON N6A 5W9, Canada
| | - Majdina Isovic
- Department of Oncology, London Regional Cancer Program and the Lawson Health Research Institute, London, ON N6A 5W9, Canada
| | - Daniel T. Passos
- Department of Oncology, London Regional Cancer Program and the Lawson Health Research Institute, London, ON N6A 5W9, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 5C1, Canada
| | - Frederick A. Dick
- Department of Oncology, London Regional Cancer Program and the Lawson Health Research Institute, London, ON N6A 5W9, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 5C1, Canada
| | - Joseph Torchia
- Department of Biochemistry, Western University, London, ON N6A 5C1, Canada
- Department of Oncology, London Regional Cancer Program and the Lawson Health Research Institute, London, ON N6A 5W9, Canada
- Correspondence: ; Tel.: +519-685-8692
| |
Collapse
|
5
|
Subramanian P, Gargani S, Palladini A, Chatzimike M, Grzybek M, Peitzsch M, Papanastasiou AD, Pyrina I, Ntafis V, Gercken B, Lesche M, Petzold A, Sinha A, Nati M, Thangapandi VR, Kourtzelis I, Andreadou M, Witt A, Dahl A, Burkhardt R, Haase R, Domingues AMDJ, Henry I, Zamboni N, Mirtschink P, Chung KJ, Hampe J, Coskun Ü, Kontoyiannis DL, Chavakis T. The RNA binding protein human antigen R is a gatekeeper of liver homeostasis. Hepatology 2022; 75:881-897. [PMID: 34519101 DOI: 10.1002/hep.32153] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS NAFLD is initiated by steatosis and can progress through fibrosis and cirrhosis to HCC. The RNA binding protein human antigen R (HuR) controls RNAs at the posttranscriptional level; hepatocyte HuR has been implicated in the regulation of diet-induced hepatic steatosis. The present study aimed to understand the role of hepatocyte HuR in NAFLD development and progression to fibrosis and HCC. APPROACH AND RESULTS Hepatocyte-specific, HuR-deficient mice and control HuR-sufficient mice were fed either a normal diet or an NAFLD-inducing diet. Hepatic lipid accumulation, inflammation, fibrosis, and HCC development were studied by histology, flow cytometry, quantitative PCR, and RNA sequencing. The liver lipidome was characterized by lipidomics analysis, and the HuR-RNA interactions in the liver were mapped by RNA immunoprecipitation sequencing. Hepatocyte-specific, HuR-deficient mice displayed spontaneous hepatic steatosis and fibrosis predisposition compared to control HuR-sufficient mice. On an NAFLD-inducing diet, hepatocyte-specific HuR deficiency resulted in exacerbated inflammation, fibrosis, and HCC-like tumor development. A multi-omic approach, including lipidomics, transcriptomics, and RNA immunoprecipitation sequencing revealed that HuR orchestrates a protective network of hepatic-metabolic and lipid homeostasis-maintaining pathways. Consistently, HuR-deficient livers accumulated, already at steady state, a triglyceride signature resembling that of NAFLD livers. Moreover, up-regulation of secreted phosphoprotein 1 expression mediated, at least partially, fibrosis development in hepatocyte-specific HuR deficiency on an NAFLD-inducing diet, as shown by experiments using antibody blockade of osteopontin. CONCLUSIONS HuR is a gatekeeper of liver homeostasis, preventing NAFLD-related fibrosis and HCC, suggesting that the HuR-dependent network could be exploited therapeutically.
Collapse
Affiliation(s)
- Pallavi Subramanian
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Sofia Gargani
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Centre "Alexander Fleming"VariGreece
| | - Alessandra Palladini
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of MedicineTechnische Universität DresdenDresdenGermany.,German Center for Diabetes ResearchNeuherbergGermany
| | - Margarita Chatzimike
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Centre "Alexander Fleming"VariGreece
| | - Michal Grzybek
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of MedicineTechnische Universität DresdenDresdenGermany.,German Center for Diabetes ResearchNeuherbergGermany
| | - Mirko Peitzsch
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Anastasios D Papanastasiou
- Department of Biomedical SciencesUniversity of West AtticaAthensGreece.,Histopathology UnitBiomedical Sciences Research Center "Alexander Fleming"VariGreece
| | - Iryna Pyrina
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Vasileios Ntafis
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Centre "Alexander Fleming"VariGreece
| | - Bettina Gercken
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Mathias Lesche
- DRESDEN-concept Genome CenterCenter for Molecular and Cellular BioengineeringTechnische Universität DresdenDresdenGermany
| | - Andreas Petzold
- DRESDEN-concept Genome CenterCenter for Molecular and Cellular BioengineeringTechnische Universität DresdenDresdenGermany
| | - Anupam Sinha
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Marina Nati
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Veera Raghavan Thangapandi
- Department of Internal Medicine IUniversity Hospital and Faculty of Medicine, Technische Universität DresdenDresdenGermany
| | - Ioannis Kourtzelis
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany.,National Center for Tumor DiseasesPartner Site Dresden, Dresden and German Cancer Research CenterHeidelbergGermany.,York Biomedical Research Institute, Hull York Medical SchoolUniversity of YorkYorkUK
| | - Margarita Andreadou
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Centre "Alexander Fleming"VariGreece
| | - Anke Witt
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Andreas Dahl
- DRESDEN-concept Genome CenterCenter for Molecular and Cellular BioengineeringTechnische Universität DresdenDresdenGermany
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Hospital RegensburgRegensburgGermany
| | - Robert Haase
- Scientific Computing FacilityMax Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | | | - Ian Henry
- Scientific Computing FacilityMax Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Nicola Zamboni
- Institute of Molecular Systems BiologyETH ZurichZurichSwitzerland
| | - Peter Mirtschink
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Kyoung-Jin Chung
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Jochen Hampe
- Department of Internal Medicine IUniversity Hospital and Faculty of Medicine, Technische Universität DresdenDresdenGermany
| | - Ünal Coskun
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of MedicineTechnische Universität DresdenDresdenGermany.,German Center for Diabetes ResearchNeuherbergGermany
| | - Dimitris L Kontoyiannis
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Centre "Alexander Fleming"VariGreece.,Department of Genetics, Development & Molecular Biology, School of BiologyAristotle University of ThessalonikiThessalonikiGreece
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany.,Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of MedicineTechnische Universität DresdenDresdenGermany.,German Center for Diabetes ResearchNeuherbergGermany.,National Center for Tumor DiseasesPartner Site Dresden, Dresden and German Cancer Research CenterHeidelbergGermany
| |
Collapse
|
6
|
Chen W, Zhang Q, Ding M, Yao J, Guo Y, Yan W, Yu S, Shen Q, Huang M, Zheng Y, Lin Y, Wang Y, Liu Z, Lu L. Alcohol triggered bile acid disequilibrium by suppressing BSEP to sustain hepatocellular carcinoma progression. Chem Biol Interact 2022; 356:109847. [PMID: 35149083 DOI: 10.1016/j.cbi.2022.109847] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 01/20/2022] [Accepted: 02/07/2022] [Indexed: 12/12/2022]
Abstract
Bile acids (BAs), the most important components of bile, attribute predominately to maintain metabolic homeostasis. In hepatocellular carcinoma (HCC) patients, the BAs homeostasis was seriously disturbed, especially in those patients with alcohol-intake history. However, whether alcohol consumption could promote HCC progression via influencing BAs homeostasis and the precise mechanism underlying are still unclear. In our study, by collecting HCC specimens from both alcohol-drinkers (n = 15) and non-alcohol drinkers (n = 22), we found that compared to non-alcohol intake HCC patients, BAs homeostasis was disturbed in HCC patients who drank alcohol. Furthermore, ethanol treatment was also found to promote HCC progression by markedly activating oncogenes (RAS, MYC, MET, and HER2), while remarkably suppressing tumor suppressor genes (BRCA2 and APC). We evaluated 14 key functional genes that maintain the homeostasis of BAs and found that either in alcohol-intake HCC patients (n = 15), or in ethanol-treated mice, BSEP, rate-limiting transporter governing excreting BAs from liver into bile duct, was remarkably decreased when exposed to alcohol. Moreover, by screening for changes in the epigenetic landscape of liver cancer cells exposed to alcohol, we strikingly found that histone methyltransferases (RBBP-5, Suv39h1, ASH2L, and SET7/9) were increased, and KMT3B, KMT4, and KMT7 gene expression was also elevated, while histone demethyltransferases (JARID1a, JARID1b, JARID1c) were decreased. In summary, we found that alcohol could trigger BAs disequilibrium to initiate and promote HCC progression. Our study provided a novel and supplementary mechanism to determine the important role of alcohol-intake in HCC development regarding from the perspective of BAs homeostasis.
Collapse
Affiliation(s)
- Wenbo Chen
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Qisong Zhang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; Medical College of Guangxi University, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Ming Ding
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Jingjing Yao
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Yajuan Guo
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Wenxin Yan
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Shaofang Yu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Qinghong Shen
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Min Huang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Yaqiu Zheng
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Yuefang Lin
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Ying Wang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Zhongqiu Liu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, SAR, China.
| | - Linlin Lu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, SAR, China.
| |
Collapse
|
7
|
The Role of Thymine DNA Glycosylase in Transcription, Active DNA Demethylation, and Cancer. Cancers (Basel) 2022; 14:cancers14030765. [PMID: 35159032 PMCID: PMC8833622 DOI: 10.3390/cancers14030765] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Thymine DNA Glycosylase (TDG) is a DNA repair protein that plays an important role in gene regulation. Recent studies have shown that TDG interacts with various transcription factors to activate target genes. TDG also functions in a pathway known as active DNA demethylation, which removes 5-mC from DNA and replaces it with unmethylated cytosine. In this review, we summarize the various functions of TDG in gene regulation as well as the physiological relevance of TDG in cancer. Abstract DNA methylation is an essential covalent modification that is required for growth and development. Once considered to be a relatively stable epigenetic mark, many studies have established that DNA methylation is dynamic. The 5-methylcytosine (5-mC) mark can be removed through active DNA demethylation in which 5-mC is converted to an unmodified cytosine through an oxidative pathway coupled to base excision repair (BER). The BER enzyme Thymine DNA Glycosylase (TDG) plays a key role in active DNA demethylation by excising intermediates of 5-mC generated by this process. TDG acts as a key player in transcriptional regulation through its interactions with various nuclear receptors and transcription factors, in addition to its involvement in classical BER and active DNA demethylation, which serve to protect the stability of the genome and epigenome, respectively. Recent animal studies have identified a connection between the loss of Tdg and the onset of tumorigenesis. In this review, we summarize the recent findings on TDG’s function as a transcriptional regulator as well as the physiological relevance of TDG and active DNA demethylation in cancer.
Collapse
|
8
|
Wu J, Nagy LE, Wang L. The long and the small collide: LncRNAs and small heterodimer partner (SHP) in liver disease. Mol Cell Endocrinol 2021; 528:111262. [PMID: 33781837 PMCID: PMC8087644 DOI: 10.1016/j.mce.2021.111262] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 02/08/2023]
Abstract
Long non-coding RNAs (lncRNAs) are a large and diverse class of RNA molecules that are transcribed but not translated into proteins, with a length of more than 200 nucleotides. LncRNAs are involved in gene expression and regulation. The abnormal expression of lncRNAs is associated with disease pathogenesis. Small heterodimer partner (SHP, NR0B2) is a unique orphan nuclear receptor that plays a pivotal role in many biological processes by acting as a transcriptional repressor. In this review, we present the critical roles of SHP and summarize recent findings demonstrating the regulation between lncRNAs and SHP in liver disease.
Collapse
Affiliation(s)
- Jianguo Wu
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA.
| | - Laura E Nagy
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Gastroenterology and Hepatology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Li Wang
- Independent Researcher, Tucson, AZ, USA
| |
Collapse
|
9
|
Bile Acids and Microbiota: Multifaceted and Versatile Regulators of the Liver-Gut Axis. Int J Mol Sci 2021; 22:ijms22031397. [PMID: 33573273 PMCID: PMC7866539 DOI: 10.3390/ijms22031397] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/25/2021] [Accepted: 01/28/2021] [Indexed: 12/12/2022] Open
Abstract
After their synthesis from cholesterol in hepatic tissues, bile acids (BAs) are secreted into the intestinal lumen. Most BAs are subsequently re-absorbed in the terminal ileum and are transported back for recycling to the liver. Some of them, however, reach the colon and change their physicochemical properties upon modification by gut bacteria, and vice versa, BAs also shape the composition and function of the intestinal microbiota. This mutual interplay of both BAs and gut microbiota regulates many physiological processes, including the lipid, carbohydrate and energy metabolism of the host. Emerging evidence also implies an important role of this enterohepatic BA circuit in shaping mucosal colonization resistance as well as local and distant immune responses, tissue physiology and carcinogenesis. Subsequently, disrupted interactions of gut bacteria and BAs are associated with many disorders as diverse as Clostridioides difficile or Salmonella Typhimurium infection, inflammatory bowel disease, type 1 diabetes, asthma, metabolic syndrome, obesity, Parkinson’s disease, schizophrenia and epilepsy. As we cannot address all of these interesting underlying pathophysiologic mechanisms here, we summarize the current knowledge about the physiologic and pathogenic interplay of local site microbiota and the enterohepatic BA metabolism using a few selected examples of liver and gut diseases.
Collapse
|
10
|
Hassan HM, Isovic M, Underhill MT, Torchia J. TDG is a novel tumor suppressor of liver malignancies. Mol Cell Oncol 2020; 7:1768819. [PMID: 32944627 PMCID: PMC7469547 DOI: 10.1080/23723556.2020.1768819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In a recent publication, we demonstrated that conditional deletion of the gene encoding thymine DNA glycosylase (TDG) leads to a late onset of hepatocellular carcinoma (HCC). TDG loss causes disruption in active DNA demethylation in the liver and dysregulation of the farnesoid X receptor and small heterodimer partner (FXR-SHP) regulatory cascade. This leads to a loss of bile acid and glucose homeostasis, which predisposes mice to HCC.
Collapse
Affiliation(s)
- Haider M Hassan
- Department of Biochemistry, Western University, London, Ontario, Canada.,Department of Oncology, The London Regional Cancer Program and The Lawson Health Research Institute, London, Ontario, Canada
| | - Majdina Isovic
- Department of Biochemistry, Western University, London, Ontario, Canada.,Department of Oncology, The London Regional Cancer Program and The Lawson Health Research Institute, London, Ontario, Canada
| | - Michael Tully Underhill
- Department of Cellular and Physiological Sciences and the Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Joseph Torchia
- Department of Biochemistry, Western University, London, Ontario, Canada.,Department of Oncology, The London Regional Cancer Program and The Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|