1
|
Zhao X, Wang C, Zhao L, Tian Z. HBV DNA polymerase regulates tumor cell glycogen to enhance the malignancy of HCC cells. Hepatol Commun 2024; 8:e0387. [PMID: 38358372 PMCID: PMC10871796 DOI: 10.1097/hc9.0000000000000387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/16/2023] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND The essential function of HBV DNA polymerase (HBV-DNA-Pol) is to initiate viral replication by reverse transcription; however, the role of HBV-DNA-Pol in HBV-associated HCC has not been clarified. Glycogen phosphorylase L (PYGL) is a critical regulator of glycogenolysis and is involved in tumorigenesis, including HCC. However, it is unknown whether HBV-DNA-Pol regulates PYGL to contribute to HCC tumorigenesis. METHODS Bioinformatic analysis, real-time quantitative PCR, western blotting, and oncology functional assays were performed to determine the contribution of HBV-DNA-Pol and PYGL to HCC development and glycolysis. The mechanisms of co-immunoprecipitation and ubiquitination were employed to ascertain how HBV-DNA-Pol upregulated PYGL. RESULTS Overexpression of HBV-DNA-Pol enhanced HCC progression in vitro and in vivo. Mechanistically, HBV-DNA-Pol interacted with PYGL and increased PYGL protein levels by inhibiting PYGL ubiquitination, which was mediated by the E3 ligase TRIM21. HBV-DNA-Pol competitively impaired the binding of PYGL to TRIM21 due to its stronger binding affinity to TRIM21, suppressing the ubiquitination of PYGL. Moreover, HBV-DNA-Pol promoted glycogen decomposition by upregulating PYGL, which led to an increased flow of glucose into glycolysis, thereby promoting HCC development. CONCLUSIONS Our study reveals a novel mechanism by which HBV-DNA-Pol promotes HCC by controlling glycogen metabolism in HCC, establishing a direct link between HBV-DNA-Pol and the Warburg effect, thereby providing novel targets for HCC treatment and drug development.
Collapse
Affiliation(s)
- Xiaoqing Zhao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunqing Wang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Laboratory Medicine, Jinan, China
| | - Liqing Zhao
- Department of Pediatrics, Zaozhuang Municipal Hospital, Zaozhuang, China
| | - Zhongzheng Tian
- Shandong Agricultural Technology Extending Center, Jinan, China
| |
Collapse
|
2
|
Kaffe E, Tisi A, Magkrioti C, Aidinis V, Mehal WZ, Flavell RA, Maccarrone M. Bioactive signalling lipids as drivers of chronic liver diseases. J Hepatol 2024; 80:140-154. [PMID: 37741346 DOI: 10.1016/j.jhep.2023.08.029] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/25/2023]
Abstract
Lipids are important in multiple cellular functions, with most having structural or energy storage roles. However, a small fraction of lipids exert bioactive roles through binding to G protein-coupled receptors and induce a plethora of processes including cell proliferation, differentiation, growth, migration, apoptosis, senescence and survival. Bioactive signalling lipids are potent modulators of metabolism and energy homeostasis, inflammation, tissue repair and malignant transformation. All these events are involved in the initiation and progression of chronic liver diseases. In this review, we focus specifically on the roles of bioactive lipids derived from phospholipids (lyso-phospholipids) and poly-unsaturated fatty acids (eicosanoids, pro-resolving lipid mediators and endocannabinoids) in prevalent chronic liver diseases (alcohol-associated liver disease, non-alcoholic fatty liver disease, viral hepatitis and hepatocellular carcinoma). We discuss the balance between pathogenic and beneficial bioactive lipids as well as potential therapeutic targets related to the agonism or antagonism of their receptors.
Collapse
Affiliation(s)
- Eleanna Kaffe
- Department of Immunobiology, Yale University School of Medicine, 06511, New Haven, CT, USA.
| | - Annamaria Tisi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | | | - Vassilis Aidinis
- Biomedical Sciences Research Center Alexander Fleming, 16672, Athens, Greece
| | - Wajahat Z Mehal
- Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, CT, 06520, USA; Veterans Affairs Medical Center, West Haven, CT, 06516, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, 06511, New Haven, CT, USA; Howard Hughes Medical Institute, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy; Laboratory of Lipid Neurochemistry, European Center for Brain Research (CERC), Santa Lucia Foundation IRCCS, 00143 Rome, Italy.
| |
Collapse
|
3
|
Ahmed EA, El-Derany MO, Anwar AM, Saied EM, Magdeldin S. Metabolomics and Lipidomics Screening Reveal Reprogrammed Signaling Pathways toward Cancer Development in Non-Alcoholic Steatohepatitis. Int J Mol Sci 2022; 24:ijms24010210. [PMID: 36613653 PMCID: PMC9820351 DOI: 10.3390/ijms24010210] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/09/2022] [Accepted: 11/19/2022] [Indexed: 12/24/2022] Open
Abstract
With the rising incidence of hepatocellular carcinoma (HCC) from non-alcoholic steatohepatitis (NASH), identifying new metabolic readouts that function in metabolic pathway perpetuation is still a demand. The study aimed to compare the metabolic signature between NASH and NASH-HCC patients to explore novel reprogrammed metabolic pathways that might modulate cancer progression in NASH patients. NASH and NASH-HCC patients were recruited and screened for metabolomics, and isotope-labeled lipidomics were targeted and profiled using the EXION-LCTM system equipped with a Triple-TOFTM 5600+ system. Results demonstrated significantly (p ≤ 0.05) higher levels of triacylglycerol, AFP, AST, and cancer antigen 19-9 in NASH-HCC than in NASH patients, while prothrombin time, platelet count, and total leukocyte count were decreased significantly (p ≤ 0.05). Serum metabolic profiling showed a panel of twenty metabolites with 10% FDR and p ≤ 0.05 in both targeted and non-targeted analysis that could segregate NASH-HCC from NASH patients. Pathway analysis revealed that the metabolites are implicated in the down-regulation of necroptosis, amino acid metabolism, and regulation of lipid metabolism by PPAR-α, biogenic amine synthesis, fatty acid metabolism, and the mTOR signaling pathway. Cholesterol metabolism, DNA repair, methylation pathway, bile acid, and salts metabolism were significantly upregulated in NASH-HCC compared to the NASH group. Metabolite-protein interactions network analysis clarified a set of well-known protein encoding genes that play crucial roles in cancer, including PEMT, IL4I1, BAAT, TAT, CDKAL1, NNMT, PNP, NOS1, and AHCYL. Taken together, reliable metabolite fingerprints are presented and illustrated in a detailed map for the most predominant reprogrammed metabolic pathways that target HCC development from NASH.
Collapse
Affiliation(s)
- Eman A. Ahmed
- Proteomics and Metabolomics Research Program, Department of Basic Research, Children’s Cancer Hospital 57357, Cairo 11441, Egypt
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Marwa O. El-Derany
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Ali Mostafa Anwar
- Proteomics and Metabolomics Research Program, Department of Basic Research, Children’s Cancer Hospital 57357, Cairo 11441, Egypt
| | - Essa M. Saied
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
- Institute for Chemistry, Humboldt Universität zu Berlin, Brook-Taylor-Str. 2, 12489 Berlin, Germany
| | - Sameh Magdeldin
- Proteomics and Metabolomics Research Program, Department of Basic Research, Children’s Cancer Hospital 57357, Cairo 11441, Egypt
- Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
- Correspondence: ; Tel.: +20-(10)-64962210
| |
Collapse
|
4
|
U MRA, Shen EYL, Cartlidge C, Alkhatib A, Thursz MR, Waked I, Gomaa AI, Holmes E, Sharma R, Taylor-Robinson SD. Optimized Systematic Review Tool: Application to Candidate Biomarkers for the Diagnosis of Hepatocellular Carcinoma. Cancer Epidemiol Biomarkers Prev 2022; 31:1261-1274. [PMID: 35545293 DOI: 10.1158/1055-9965.epi-21-0687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/17/2021] [Accepted: 05/09/2022] [Indexed: 12/24/2022] Open
Abstract
This review aims to develop an appropriate review tool for systematically collating metabolites that are dysregulated in disease and applies the method to identify novel diagnostic biomarkers for hepatocellular carcinoma (HCC). Studies that analyzed metabolites in blood or urine samples where HCC was compared with comparison groups (healthy, precirrhotic liver disease, cirrhosis) were eligible. Tumor tissue was included to help differentiate primary and secondary biomarkers. Searches were conducted on Medline and EMBASE. A bespoke "risk of bias" tool for metabolomic studies was developed adjusting for analytic quality. Discriminant metabolites for each sample type were ranked using a weighted score accounting for the direction and extent of change and the risk of bias of the reporting publication. A total of 84 eligible studies were included in the review (54 blood, 9 urine, and 15 tissue), with six studying multiple sample types. High-ranking metabolites, based on their weighted score, comprised energy metabolites, bile acids, acylcarnitines, and lysophosphocholines. This new review tool addresses an unmet need for incorporating quality of study design and analysis to overcome the gaps in standardization of reporting of metabolomic data. Validation studies, standardized study designs, and publications meeting minimal reporting standards are crucial for advancing the field beyond exploratory studies.
Collapse
Affiliation(s)
- Mei Ran Abellona U
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Eric Yi-Liang Shen
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- Department of Radiation Oncology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | | | - Alzhraa Alkhatib
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- National Liver Unit, Menoufiya University, Shbeen El Kom, Egypt
| | - Mark R Thursz
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Imam Waked
- National Liver Unit, Menoufiya University, Shbeen El Kom, Egypt
| | - Asmaa I Gomaa
- National Liver Unit, Menoufiya University, Shbeen El Kom, Egypt
| | - Elaine Holmes
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- Health Futures Institute, Murdoch University, Perth WA, Australia
| | - Rohini Sharma
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Simon D Taylor-Robinson
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| |
Collapse
|
5
|
Liu G, Sun BY, Sun J, Zhou PY, Guan RY, Zhou C, Yang ZF, Wang ZT, Zhou J, Fan J, Yi Y, Qiu SJ. BRG1 regulates lipid metabolism in hepatocellular carcinoma through the PIK3AP1/PI3K/AKT pathway by mediating GLMP expression. Dig Liver Dis 2022; 54:692-700. [PMID: 34158256 DOI: 10.1016/j.dld.2021.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Brahma-related gene 1 (BRG1) is essential for embryogenesis and cellular metabolism. A deficiency of BRG1 in vivo decreases lipid droplets, but the molecular mechanism underlying its role in lipid metabolism associated with hepatocellular carcinoma (HCC) remains unknown. AIMS We aimed to determine the role of BRG1 in lipid metabolism in HCC. METHODS We assessed the differential expression of BRG1 in HCC and adjacent non-tumorous tissues using tissue microarrays. We stained lipid droplets in HCC cells with Bodipy fluorescence and Oil Red O, and verified BRG1 binding to the promoter region of glycosylated lysosomal membrane protein (GLMP) using chromatin immunoprecipitation. RESULTS The expression of GLMP, a potential lipid metabolism regulator, was suppressed by BRG1 via transcriptional activity. Knockdown of BRG1 decreased lipid droplets, increased GLMP expression and altered the phosphoinositide-3-kinase adaptor protein 1 (PIK3AP1)/phosphatidylinositol-3 kinase (PI3K)/protein kinase B (AKT) pathway in HCC, which further GLMP knockdown partially restored. Thus, GLMP knockdown increased lipid droplets and differentially altered the PI3K/AKT pathway. CONCLUSIONS Downregulating BRG1 decreased lipid droplet deposition in HCC cells by upregulating GLMP and altering the PI3K/AKT pathway. Both BRG1 and GLMP might serve as therapeutic targets for disorders associated with dysregulated lipid metabolism, such as NAFLD and NAFLD-associated HCC.
Collapse
Affiliation(s)
- Gao Liu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Biomedical Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, People's Republic of China
| | - Bao-Ye Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Biomedical Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, People's Republic of China
| | - Jian Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Biomedical Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, People's Republic of China
| | - Pei-Yun Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Biomedical Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, People's Republic of China
| | - Ruo-Yu Guan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Biomedical Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, People's Republic of China
| | - Cheng Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Biomedical Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, People's Republic of China
| | - Zhang-Fu Yang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Biomedical Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, People's Republic of China
| | - Zhu-Tao Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Biomedical Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, People's Republic of China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Biomedical Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, People's Republic of China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Biomedical Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, People's Republic of China
| | - Yong Yi
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Biomedical Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, People's Republic of China.
| | - Shuang-Jian Qiu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Biomedical Research Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, People's Republic of China.
| |
Collapse
|
6
|
Paul B, Lewinska M, Andersen JB. Lipid alterations in chronic liver disease and liver cancer. JHEP Rep 2022; 4:100479. [PMID: 35469167 PMCID: PMC9034302 DOI: 10.1016/j.jhepr.2022.100479] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 03/01/2022] [Accepted: 03/07/2022] [Indexed: 02/06/2023] Open
Abstract
Lipids are a complex and diverse group of molecules with crucial roles in many physiological processes, as well as in the onset, progression, and maintenance of cancers. Fatty acids and cholesterol are the building blocks of lipids, orchestrating these crucial metabolic processes. In the liver, lipid alterations are prevalent as a cause and consequence of chronic hepatitis B and C virus infections, alcoholic hepatitis, and non-alcoholic fatty liver disease and steatohepatitis. Recent developments in lipidomics have also revealed that dynamic changes in triacylglycerols, phospholipids, sphingolipids, ceramides, fatty acids, and cholesterol are involved in the development and progression of primary liver cancer. Accordingly, the transcriptional landscape of lipid metabolism suggests a carcinogenic role of increasing fatty acids and sterol synthesis. However, limited mechanistic insights into the complex nature of the hepatic lipidome have so far hindered the development of effective therapies.
Collapse
|
7
|
Wang KX, Du GH, Qin XM, Gao L. 1H-NMR-based metabolomics reveals the biomarker panel and molecular mechanism of hepatocellular carcinoma progression. Anal Bioanal Chem 2022; 414:1525-1537. [PMID: 35024914 DOI: 10.1007/s00216-021-03768-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 11/30/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most extensive and most deadly cancers in the world. Biomarkers for early diagnosis of HCC are still lacking, and noninvasive and effective biomarkers are urgently needed. Metabolomics is committed to studying the changes of metabolites under stimulation, and provides a new approach for discovery of potential biomarkers. In the current work, 1H nuclear magnetic resonance (NMR) metabolomics approach was utilized to explore the potential biomarkers in HCC progression, and the biomarker panel was evaluated by receiver operating characteristic (ROC) curve analyses. Our results revealed that a biomarker panel consisting of hippurate, creatinine, putrescine, choline, and taurine might be involved in HCC progression. Functional pathway analysis showed that taurine and hypotaurine metabolism is markedly involved in the occurrence and development of HCC. Furthermore, our results indicated that the TPA activity and the level and expression of PKM2 were gradually increased in HCC progression. This research provides a scientific basis for screening potential biomarkers of HCC.
Collapse
Affiliation(s)
- Ke-Xin Wang
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China
| | - Guan-Hua Du
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China.
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China.
| | - Li Gao
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China.
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China.
| |
Collapse
|
8
|
Akkiz H, Carr BI, Guerra V, Donghia R, Yalçın K, Karaoğullarından U, Altıntaş E, Özakyol A, Şimşek H, Balaban HY, Balkan A, Uyanıkoğlu A, Ekin N, Delik A. Plasma lipids, tumor parameters and survival in HCC patients with HBV and HCV. JOURNAL OF TRANSLATIONAL SCIENCE 2021; 7:10.15761/jts.1000421. [PMID: 34457356 PMCID: PMC8389344 DOI: 10.15761/jts.1000421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION AND AIMS Hepatocellular carcinoma (HCC) is a consequence of chronic liver disease, particularly from hepatitis B or C and increasingly from obesity and metabolic syndrome. Since lipids are an important component of cell membranes and are involved in cell signaling and tumor cell growth, we wished to evaluate the relationship between HCC patient plasma lipids and maximum tumor diameter and other indices of HCC human biology. METHODS We examined prospectively-collected data from a multi-institutional collaborative Turkish HCC working group, from predominantly HBV-based patients, for plasma lipid profiles, consisting of triglycerides, total cholesterol, LDL-cholesterol (LDL) and HDL-cholesterol (HDL) and compared these with the associated patient maximum tumor diameter (MTD), portal vein thrombosis, alpha-fetoprotein (AFP) and also with patient survival. RESULTS We found that both low HDL (p=0.0002) and high LDL (p=0.003) levels were significantly associated with increased MTD, as well as in a final multiple linear regression model on MTD. The combination of low HDL combined with high HDL levels were significant in a regression model on MTD, PVT and an HCC Aggressiveness Index (Odds Ratio 12.91 compared to an Odds Ratio of 1 for the reference). Furthermore, in a Cox regression model on death, the HDL plus LDL combination had a significantly higher Hazard Ratio than the reference category. CONCLUSIONS Low plasma HDL, high plasma LDL and especially the combination, were significantly related to more aggressive HCC phenotype and the combination was significantly related to a higher Hazard Ratio for death.
Collapse
Affiliation(s)
- H Akkiz
- Çukurova University, Adana, Turkey
| | - BI Carr
- İnonu University, Malatya, Turkey
| | - V Guerra
- National Institute of Gastroenterology, S. de Bellis Research hospital, Castellana Grotte (BA), Italy
| | - R Donghia
- National Institute of Gastroenterology, S. de Bellis Research hospital, Castellana Grotte (BA), Italy
| | - K Yalçın
- Dikle University, Diyarbakır, Turkey
| | | | | | - A Özakyol
- Eskişehir Osmangazi University, Eskişehir, Turkey
| | - H Şimşek
- Hacettepe University, Ankara, Turkey
| | | | - A Balkan
- Gaziantep University, Gazientep, Turkey
| | | | - N Ekin
- Dikle University, Diyarbakır, Turkey
| | - A Delik
- Çukurova University, Adana, Turkey
| |
Collapse
|
9
|
Zhou Y, Guo Y, Zhu Y, Sun Y, Li W, Li Z, Wei L. Dual PPARγ/ɑ agonist oroxyloside suppresses cell cycle progression by glycolipid metabolism switch-mediated increase of reactive oxygen species levels. Free Radic Biol Med 2021; 167:205-217. [PMID: 33713839 DOI: 10.1016/j.freeradbiomed.2021.02.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/26/2021] [Accepted: 02/24/2021] [Indexed: 12/15/2022]
Abstract
Cancer cells prefers to rely on aerobic glycolysis than pyruvate oxidation to meet the high demand of energy for rapidly proliferation. Peroxisome proliferator-activated receptors (PPARs) are a kind of important ligand-inducible transcription factors and play crucial roles in glucose and lipid metabolism. Careful designing of novel agonists for PPARs, may show improvement with the side effects and also increase the therapeutic value for cancer and other metabolic disorder diseases. Compared with normal human liver cells, lower expression or acitivity of PPARs is observed in hepatocellular carcinoma (HCC). In this study, we show that oroxyloside (OAG) is a new dual agonist of PPARγ/ɑ, and inhibits cell proliferation of HCC based on metabolic switch. Via both PPAR-dependent and PPAR-independent regulations on glycolipid metabolic enzymes, OAG shuts down the catabolism of glucose and promotes fatty acids oxidation to generate acetyl-CoA for TCA cycle and oxidative phosphorylation. The metabolic switch induced by OAG results in a marked increase of reactive oxygen species (ROS) levels, leading to rapid dephosphorylation of RB and cell-cycle arrest in G1 phase. Pyruvate dehydrogenase kinase 4 (PDK4) and β-Oxidation are required for the suppression of cell cycle progression by OAG. Together, our findings provide a new drug candidate and a viable therapeutic strategy for HCC based on metabolic reprogram.
Collapse
Affiliation(s)
- Yuxin Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, PR China
| | - Yongjian Guo
- School of Biopharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, PR China
| | - Yejin Zhu
- School of Medicine & Holistic Integrative Medcine, Nanjing University of Chinese Medicine, 138 Xianlin Rd, Nanjing, PR China
| | - Yuening Sun
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, PR China
| | - Wei Li
- Research Center of Basic Medical College, Nanjing University of Chinese Medicine, 138 Xianlin Rd, Nanjing, PR China
| | - Zhiyu Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, PR China
| | - Libin Wei
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, PR China.
| |
Collapse
|
10
|
A Lipidomic Signature Complements Stemness Features Acquisition in Liver Cancer Cells. Int J Mol Sci 2020; 21:ijms21228452. [PMID: 33182805 PMCID: PMC7709039 DOI: 10.3390/ijms21228452] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/16/2022] Open
Abstract
Lipid catabolism and anabolism changes play a role in stemness acquisition by cancer cells, and cancer stem cells (CSCs) are particularly dependent on the activity of the enzymes involved in these processes. Lipidomic changes could play a role in CSCs’ ability to cause disease relapse and chemoresistance. The exploration of lipid composition and metabolism changes in CSCs in the context of hepatocellular cancer (HCC) is still incomplete and their lipidomic scenario continues to be elusive. We aimed to evaluate through high-throughput mass spectrometry (MS)-based lipidomics the levels of the members of the six major classes of sphingolipids and phospholipids in two HCC cell lines (HepG2 and Huh-7) silenced for the expression of histone variant macroH2A1 (favoring stemness acquisition), or silenced for the expression of focal adhesion tyrosine kinase (FAK) (hindering aggressiveness and stemness). Transcriptomic changes were evaluated by RNA sequencing as well. We found definite lipidomic and transcriptomic changes in the HCC lines upon knockdown (KD) of macroH2A1 or FAK, in line with the acquisition or loss of stemness features. In particular, macroH2A1 KD increased total sphingomyelin (SM) levels and decreased total lysophosphatidylcholine (LPC) levels, while FAK KD decreased total phosphatidylcholine (PC) levels. In conclusion, in HCC cell lines knocked down for specific signaling/epigenetic processes driving opposite stemness potential, we defined a lipidomic signature that hallmarks hepatic CSCs to be exploited for therapeutic strategies.
Collapse
|
11
|
Tang X, Benesch MGK, Brindley DN. Role of the autotaxin-lysophosphatidate axis in the development of resistance to cancer therapy. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158716. [PMID: 32305571 DOI: 10.1016/j.bbalip.2020.158716] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/31/2020] [Accepted: 04/09/2020] [Indexed: 12/17/2022]
Abstract
Autotaxin (ATX) is a secreted enzyme that hydrolyzes lysophosphatidylcholine to produce lysophosphatidate (LPA), which signals through six G-protein coupled receptors (GPCRs). Signaling through LPA is terminated by its degradation by a family of three lipid phosphate phosphatases (LPPs). LPP1 also attenuates signaling downstream of the activation of LPA receptors and some other GPCRs. The ATX-LPA axis mediates a plethora of activities such as cell proliferation, survival, migration, angiogenesis and inflammation, which perform an important role in facilitating wound healing. This wound healing response is hijacked by cancers where there is decreased expression of LPP1 and LPP3 and increased expression of ATX. This maladaptive regulation of LPA signaling also causes chronic inflammation, which has been recognized as one of the hallmarks in cancer. The increased LPA signaling promotes cell survival and migration and attenuates apoptosis, which stimulates tumor growth and metastasis. The wound healing functions of increased LPA signaling also protect cancer cells from effects of chemotherapy and radiotherapy. In this review, we will summarize knowledge of the ATX-LPA axis and its role in the development of resistance to chemotherapy and radiotherapy. We will also offer insights for developing strategies of targeting ATX-LPA axis as a novel part of cancer treatment. This article is part of a Special Issue entitled Lysophospholipids and their receptors: New data and new insights into their function edited by Susan Smyth, Viswanathan Natarajan and Colleen McMullen.
Collapse
Affiliation(s)
- Xiaoyun Tang
- Department of Biochemistry, University of Alberta, Edmonton T6G 2S2, Canada; Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton T6G 2S2, Canada
| | - Matthew G K Benesch
- Department of Biochemistry, University of Alberta, Edmonton T6G 2S2, Canada; Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton T6G 2S2, Canada; Discipline of Surgery, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador A1B 3V6, Canada
| | - David N Brindley
- Department of Biochemistry, University of Alberta, Edmonton T6G 2S2, Canada; Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton T6G 2S2, Canada.
| |
Collapse
|
12
|
Citral Induced Apoptosis through Modulation of Key Genes Involved in Fatty Acid Biosynthesis in Human Prostate Cancer Cells: In Silico and In Vitro Study. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6040727. [PMID: 32258129 PMCID: PMC7103989 DOI: 10.1155/2020/6040727] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/31/2019] [Accepted: 11/21/2019] [Indexed: 12/14/2022]
Abstract
The isomers of citral (cis-citral and trans-citral) were isolated from the Cymbopogon citratus (DC.) Stapf oil demonstrates many therapeutic properties including anticancer properties. However, the effects of citral on suppressing human prostate cancer and its underlying molecular mechanism have yet to be elucidated. The citral was isolated from lemongrass oil using various spectroscopic analyses, such as electron ionized mass spectrometry (EI-MS) and nuclear magnetic resonance (NMR) spectroscopy respectively. We carried out 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay to evaluate the cell viability of citral in prostate cancer cells (PC-3 and PC3M). Furthermore, to confirm that PC3 undergoes apoptosis by inhibiting lipogenesis, we used several detection methods including flow cytometry, DNA fragmentation, Hoechst staining, PI staining, oil staining, qPCR, and Western blotting. Citral impaired the clonogenic property of the cancer cells and altered the morphology of cancer cells. Molecular interaction studies and the PASS biological program predicted that citral isomers tend to interact with proteins involved in lipogenesis and the apoptosis pathway. Furthermore, citral suppressed lipogenesis of prostate cancer cells through the activation of AMPK phosphorylation and downregulation of fatty acid synthase (FASN), acetyl coA carboxylase (ACC), 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGR), and sterol regulatory element-binding protein (SREBP1) and apoptosis of PC3 cells by upregulating BAX and downregulating Bcl-2 expression. In addition, in silico studies such as ADMET predicted that citral can be used as a safe potent drug for the treatment of prostate cancer. Our results indicate that citral may serve as a potential candidate against human prostate cancer and warrants in vivo studies.
Collapse
|
13
|
Buechler C, Aslanidis C. Role of lipids in pathophysiology, diagnosis and therapy of hepatocellular carcinoma. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158658. [PMID: 32058031 DOI: 10.1016/j.bbalip.2020.158658] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/05/2019] [Accepted: 02/06/2020] [Indexed: 12/15/2022]
Abstract
Hepatocellular carcinoma (HCC) is an aggressive and widespread cancer. Patients with liver cirrhosis of different aetiologies are at a risk to develop HCC. It is important to know that in approximately 20% of cases primary liver tumors arise in a non-cirrhotic liver. Lipid metabolism is variable in patients with chronic liver diseases, and lipid metabolites involved therein do play a role in the development of HCC. Of note, lipid composition of carcinogenic tissues differs from non-affected liver tissues. High cholesterol and low ceramide levels in the tumors protect the cells from oxidative stress and apoptosis, and do also promote cell proliferation. So far, detailed characterization of the mechanisms by which lipids enable the development of HCC has received little attention. Evaluation of the complex roles of lipids in HCC is needed to better understand the pathophysiology of HCC, the later being of paramount importance for the development of urgently needed therapeutic interventions. Disturbed hepatic lipid homeostasis has systemic consequences and lipid species may emerge as promising biomarkers for early diagnosis of HCC. The challenge is to distinguish lipids specifically related to HCC from changes simply related to the underlying liver disease. This review article discusses aberrant lipid metabolism in patients with HCC.
Collapse
Affiliation(s)
- Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, Regensburg, Germany.
| | - Charalampos Aslanidis
- Institute for Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, Regensburg, Germany
| |
Collapse
|
14
|
Shi Y, Fan Y, Hu Y, Jing J, Wang C, Wu Y, Geng Q, Dong X, Li E, Dong D. α-Mangostin suppresses the de novo lipogenesis and enhances the chemotherapeutic response to gemcitabine in gallbladder carcinoma cells via targeting the AMPK/SREBP1 cascades. J Cell Mol Med 2019; 24:760-771. [PMID: 31762191 PMCID: PMC6933365 DOI: 10.1111/jcmm.14785] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 09/24/2019] [Accepted: 10/04/2019] [Indexed: 02/06/2023] Open
Abstract
High rates of de novo lipid synthesis have been discovered in certain kinds of tumours, including gallbladder cancer (GBC). Unlike several other tumours, GBC is highly insensitive to standard adjuvant therapy, which makes its treatment even more challenging. Although several potential targets and signalling pathways underlying GBC chemoresistance have been revealed, the precise mechanisms are still elusive. In this study, we found that α‐Mangostin, as a dietary xanthone, repressed the proliferation and clone formation ability, induced cell cycle arrest and the apoptosis, and suppressed de novo lipogenesis of gallbladder cancer cells. The underlying mechanisms might involve the activation of AMPK and, therefore, the suppression of SREBP1 nuclear translocation to blunt de novo lipogenesis. Furthermore, SREBP1 silencing by siRNA or α‐mangostin enhanced the sensitivity of gemcitabine in gallbladder cancer cells. In vivo studies also displayed that MA or gemcitabine administration to nude mice harbouring NOZ tumours can reduce tumour growth, and moreover, MA administration can significantly potentiate gemcitabine‐induced inhibition of tumour growth. Corroborating in vitro findings, tumours from mice treated with MA or gemcitabine alone showed decreased levels of proliferation with reduced Ki‐67 expression and elevated apoptosis confirmed by TUNEL staining, furthermore, the proliferation inhibition and apoptosis up‐regulation were obviously observed in MA combined with gemcitabine treatment group. Therefore, inhibiting de novo lipogenesis via targeting the AMPK/SREBP1 signalling by MA might provide insights into a potential strategy for sensitizing GBC cells to chemotherapy.
Collapse
Affiliation(s)
- Yu Shi
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yangwei Fan
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuan Hu
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiayu Jing
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chuying Wang
- Department of Gastroenterology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yinying Wu
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qianqian Geng
- Department of Nuclear Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xuyuan Dong
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Enxiao Li
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Danfeng Dong
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
15
|
Deregulated Lysophosphatidic Acid Metabolism and Signaling in Liver Cancer. Cancers (Basel) 2019; 11:cancers11111626. [PMID: 31652837 PMCID: PMC6893780 DOI: 10.3390/cancers11111626] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/18/2019] [Accepted: 10/20/2019] [Indexed: 02/06/2023] Open
Abstract
Liver cancer is one of the leading causes of death worldwide due to late diagnosis and scarcity of treatment options. The major risk factor for liver cancer is cirrhosis with the underlying causes of cirrhosis being viral infection (hepatitis B or C), metabolic deregulation (Non-alcoholic fatty liver disease (NAFLD) in the presence of obesity and diabetes), alcohol or cholestatic disorders. Lysophosphatidic acid (LPA) is a bioactive phospholipid with numerous effects, most of them compatible with the hallmarks of cancer (proliferation, migration, invasion, survival, evasion of apoptosis, deregulated metabolism, neoangiogenesis, etc.). Autotaxin (ATX) is the enzyme responsible for the bulk of extracellular LPA production, and together with LPA signaling is involved in chronic inflammatory diseases, fibrosis and cancer. This review discusses the most important findings and the mechanisms related to ATX/LPA/LPAR involvement on metabolic, viral and cholestatic liver disorders and their progression to liver cancer in the context of human patients and mouse models. It focuses on the role of ATX/LPA in NAFLD development and its progression to liver cancer as NAFLD has an increasing incidence which is associated with the increasing incidence of liver cancer. Bearing in mind that adipose tissue accounts for the largest amount of LPA production, many studies have implicated LPA in adipose tissue metabolism and inflammation, liver steatosis, insulin resistance, glucose intolerance and lipogenesis. At the same time, LPA and ATX play crucial roles in fibrotic diseases. Given that hepatocellular carcinoma (HCC) is usually developed on the background of liver fibrosis, therapies that both delay the progression of fibrosis and prevent its development to malignancy would be very promising. Therefore, ATX/LPA signaling appears as an attractive therapeutic target as evidenced by the fact that it is involved in both liver fibrosis progression and liver cancer development.
Collapse
|
16
|
Lu D, Yang F, Lin Z, Zhuo J, Liu P, Cen B, Lian Z, Xie H, Zheng S, Xu X. A prognostic fingerprint in liver transplantation for hepatocellular carcinoma based on plasma metabolomics profiling. Eur J Surg Oncol 2019; 45:2347-2352. [PMID: 31331801 DOI: 10.1016/j.ejso.2019.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 06/24/2019] [Accepted: 07/02/2019] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Tumor recurrence is a major cause of post-transplant mortality in liver transplantation for hepatocellular carcinoma (HCC). This study aimed to explore an effective noninvasive approach to accurately predict post-transplant tumor recurrence. MATERIALS AND METHODS Metabolomics profiling was performed on pre-operative plasma from 122 HCC patients undergoing liver transplantation, 52 healthy controls (HC) and 25 liver cirrhosis (LC) patients. RESULTS Five prognostic metabolites were identified by univariate analysis (P < 0.01), including phosphatidylcholine (PC) (16:0/P-18:1), PC(18:2/OH-16:0), PC(o-16:0/20:4), nutriacholic acid and 2-oxo-4-methylthiobutanoic acid. In the HCC group, PC(o-16:0/20:4), nutriacholic acid and 2-oxo-4-methylthiobutanoic acid were decreased, while PC(18:2/OH-16:0) was elevated compared with the LC group (e < 0.05). PC(16:0/P-18:1) was associated with tumor size, vascular invasion, and neutrophil-lymphocyte ratio (NLR; P < 0.05). Moreover, PC(18:2/OH-16:0) was also related to tumor number and NLR (P < 0.05). Multivariate cox regression showed that PC(16:0/P-18:1), PC(18:2/OH-16:0), nutriacholic acid and alpha-fetoprotein (AFP) were independent risk factors for tumor recurrence (P < 0.01). A prognostic fingerprint was established as a nomogram, which divided the patients into low risk (n = 45), moderate risk (n = 48) and highrisk groups (n = 29) with discriminated prognosis (P < 0.001). In patients fulfilling the Hangzhou criteria, the fingerprint/nomogram could also successfully stratify the patients into two groups with different recurrence risk (P < 0.05). CONCLUSIONS The established pre-operative plasma fingerprint/nomogram is efficient in the prediction of recurrence risk, which could facilitate candidate selection in liver transplantation for HCC.
Collapse
Affiliation(s)
- Di Lu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China; Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China
| | - Fan Yang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China; Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China
| | - Zuyuan Lin
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China; Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China
| | - Jianyong Zhuo
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China; Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China
| | - Peng Liu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China; Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China
| | - Beini Cen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China; Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China
| | - Zhengxing Lian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China; Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China; Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China; Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China; Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Hangzhou, 310000, China
| | - Xiao Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China; Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, 310000, China.
| |
Collapse
|
17
|
Interventional Radiologic Therapies for Hepatocellular Carcinoma: From Where We Began to Where We Are Going. MOLECULAR AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/978-3-030-21540-8_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
18
|
Han J, Qin WX, Li ZL, Xu AJ, Xing H, Wu H, Zhang H, Wang MD, Li C, Liang L, Quan B, Yan WT, Shen F, Wu MC, Yang T. Tissue and serum metabolite profiling reveals potential biomarkers of human hepatocellular carcinoma. Clin Chim Acta 2018; 488:68-75. [PMID: 30389456 DOI: 10.1016/j.cca.2018.10.039] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/10/2018] [Accepted: 10/29/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND AIMS Metabolomics serves as an important tool in distinguishing changes in metabolic pathways and the diagnosis of human disease. Hepatocellular carcinoma (HCC) is a malignance present of heterogeneous metabolic disorder and lack of effective biomarker for surveillance and diagnosis. In this study, we searched for potential metabolite biomarkers of HCC using tissue and serum metabolomics approach. METHODS A total of 30 pairs of matched liver tissue samples from HCC patients and 90 serum samples (30 HCC patients, 30 liver cirrhosis patients, and 30 healthy individuals) were assessed. Metabolomics was performed through ultra performance liquid chromatography-mass spectrometry in conjunction with multivariate and univariate statistical analyses. RESULTS A total of six differential metabolites including chenodeoxycholic acid (CDCA), glycocholic acid (GCA), LPC20:5, LPE18:0, succinyladenosine and uridine were present in HCC tissue and serum samples. CDCA, LPC20:5, succinyladenosine and uridine were used to construct a diagnostic model based on logistic regression. The four-metabolite panel discriminated HCC from liver cirrhosis with an AUC score of 0.938, sensitivity of 93.3% and specificity of 86.7%. For all HCC and cirrhosis patients, the diagnostic accuracy increased to 96.7% and 90.0%, respectively. CONCLUSION The combination of CDCA, LPC20:5, succinyladenosine and uridine can be used as a biomarker panel to improve HCC sensitivity and specificity. This panel significantly benefits HCC diagnostics and reveals new insight into HCC pathogenesis.
Collapse
Affiliation(s)
- Jun Han
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Wen-Xing Qin
- Department of Clinical Oncology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China; State Key Laboratory of New Drug & Pharmaceutial Process, Shanghai Institute of Pharmaceutial Industry, Shanghai 200437, China
| | - Zhen-Li Li
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Ai-Jing Xu
- Department of Infectious Disease, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Hao Xing
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Han Wu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Han Zhang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Ming-da Wang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Chao Li
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Lei Liang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Bing Quan
- Department of Clinical Medicine, Second Military Medical University, Shanghai 200433, China
| | - Wen-Tao Yan
- Department of Clinical Medicine, Second Military Medical University, Shanghai 200433, China
| | - Feng Shen
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Meng-Chao Wu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China.
| | - Tian Yang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China.
| |
Collapse
|
19
|
Jin GZ, Zhang Y, Cong WM, Wu X, Wang X, Wu S, Wang S, Zhou W, Yuan S, Gao H, Yu G, Yang W. Phosphoglucomutase 1 inhibits hepatocellular carcinoma progression by regulating glucose trafficking. PLoS Biol 2018; 16:e2006483. [PMID: 30335765 PMCID: PMC6193743 DOI: 10.1371/journal.pbio.2006483] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 09/14/2018] [Indexed: 12/13/2022] Open
Abstract
Glycogen metabolism commonly altered in cancer is just beginning to be understood. Phosphoglucomutase 1 (PGM1), the first enzyme in glycogenesis that catalyzes the reversible conversion between glucose 1-phosphate (G-1-P) and glucose 6-phosphate (G-6-P), participates in both the breakdown and synthesis of glycogen. Here, we show that PGM1 is down-regulated in hepatocellular carcinoma (HCC), which is associated with the malignancy and poor prognosis of HCC. Decreased PGM1 expression obstructed glycogenesis pathway, which leads to the increased flow of glucose into glycolysis, thereby promoting tumor cell proliferation and HCC development. The loss of forkhead box protein J2 (FOXJ2), at least partly due to low genomic copy number in HCC, releases cellular nucleic acid-binding protein (CNBP), a nucleic acid chaperon, to bind to and promote G-quadruplex formation in PGM1 promoter and therefore decreases PGM1 expression. In addition, integrated analyses of PGM1 and FOXJ2 expression provide a better prediction for the malignance and prognosis of HCC. This study establishes a tumor-suppressive role of PGM1 by regulating glucose trafficking and uncovers a novel regulatory mechanism of PGM1 expression. Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer in adults. Sorafenib is the only clinically approved systemic drug for patients with advanced HCC. Identification of novel targets and biomarkers will provide new therapeutic strategies for advanced HCC and better prognostic prediction. Phosphoglucomutase (PGM) is an evolutionarily conserved enzyme that regulates one of the most important pathways in glucose metabolis—catalyzing the bidirectional interconversion of glucose 1-phosphate (G-1-P) and glucose 6-phosphate (G-6-P). In this study, we identify PGM1 as a metabolic tumor suppressor. Its expression allocates more glucose to glycogenesis, which reduces the glycolytic intermediates for biosynthesis, thereby impairing HCC progression. We delineate the mechanism of PGM1 down-regulation in HCC, finding that forkhead box protein J2 (FOXJ2) loss releases cellular nucleic acid-binding protein (CNBP) to bind to and modify the DNA structure of PGM1 promoter, thereby inhibiting PGM1 expression. Immunohistochemical analyses of human HCC tumors indicate that low FOXJ2 and PGM1 expression correlates with the malignancy and poor progression of human HCC. These results also suggest that the activation of residual PGM1 may impair HCC development through switching glycolysis to glycogenesis.
Collapse
Affiliation(s)
- Guang-Zhi Jin
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Yajuan Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Wen-Ming Cong
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Xueyuan Wu
- Department of Radiation Oncology, First Affiliated Hospital of Wenzhou Medical College, Wenzhou, Zhejiang, China
| | - Xiongjun Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Siyang Wu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Siyao Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Weiping Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Shengxian Yuan
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Hong Gao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Guanzhen Yu
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- * E-mail: (GY); (WY)
| | - Weiwei Yang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- * E-mail: (GY); (WY)
| |
Collapse
|
20
|
Carr BI, Giannelli G, Guerra V, Giannini EG, Farinati F, Rapaccini GL, Marco MD, Zoli M, Caturelli E, Masotto A, Virdone R, Sacco R, Trevisani F. Plasma cholesterol and lipoprotein levels in relation to tumor aggressiveness and survival in HCC patients. Int J Biol Markers 2018; 33:423-431. [PMID: 29874983 DOI: 10.1177/1724600818776838] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS: Hepatocellular carcinoma is associated with several chronic liver diseases, especially chronic hepatitis B virus, hepatitis C virus, and alcoholism. It is increasingly appreciated that obesity/metabolic syndrome is also associated with chronic liver disease and subsequent hepatocellular carcinoma. METHODS: We retrospectively investigated the serum lipid profiles in a large hepatocellular carcinoma cohort, associated predominantly with the hepatitis B virus, hepatitis C virus, alcohol or nonalcoholic steatohepatitis. The cohort was examined both as a whole, as well as stratified by etiology. RESULTS: We found significant associations between parameters of hepatocellular carcinoma biology such as maximum tumor diameter, portal vein thrombosis, tumor multifocality or alpha-fetoprotein levels and individual lipid components, including total cholesterol, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, triglycerides and body mass index. In a final multiple linear regression model considering all lipid variables together, only high-density lipoprotein cholesterol was significantly associated with the tumor Tumor Aggressiveness Index. High-density lipoprotein cholesterol was found to have a statistically higher hazard ratio for death than low high-density lipoprotein cholesterol levels (Cox). On examination by etiological group, alpha-fetoprotein levels were significantly higher in patients with hepatitis C virus compared to those with alcohol or nonalcoholic steatohepatitis, but maximum tumor diameter, tumor multifocality and portal vein thrombosis were similar across etiological groups. Nonalcoholic steatohepatitis patients had significantly less cirrhosis than other groups and hepatitis B virus patients had significantly higher cholesterol and low-density lipoprotein cholesterol levels than hepatitis C virus patients. CONCLUSIONS: This is the first report, to our knowledge, of a relationship between serum lipid parameters and indices of hepatocellular carcinoma growth, invasion and aggressiveness, as well as with survival.
Collapse
Affiliation(s)
- Brian I Carr
- 1 Izmir Biomedicine and Genome Institute (iBG), Dokuz Eylul University, Izmir, Turkey
| | - Gianluigi Giannelli
- 2 National Institute of Digestive Diseases, IRCCS Saverio de Bellis, Castellana Grotte, Italy
| | - Vito Guerra
- 2 National Institute of Digestive Diseases, IRCCS Saverio de Bellis, Castellana Grotte, Italy
| | - Edoardo G Giannini
- 3 Department of Internal Medicine, Gastroenterology Unit, University of Genoa, Italy
| | - Fabio Farinati
- 4 Department of Surgical Science and Gastroenterology, Gastroenterology Unit, University of Padua, Italy
| | | | - Maria Di Marco
- 6 Division of Medicine, Azienda Ospedaliera Bolognini, Seriate, Italy
| | - Marco Zoli
- 7 Department of Medical and Surgical Science, Internal Medicine Unit, Alma Mater Studiorum, University of Bologna, Italy
| | | | - Alberto Masotto
- 9 Gastroenterology Unit, Ospedale Sacro Cuore Don Calabria, Negrar, Italy
| | - Roberto Virdone
- 10 Division of Internal Medicine 2, Ospedali Riuniti Villa Sofia Cervello, Palermo, Italy
| | - Rodolfo Sacco
- 11 Unita Operativa Medicina interna 2, Ospedali Riuniti, Cervello, Palermo, Italy and Metabolic Diseases, Pisa University Hospital, Pisa, Italy
| | - Franco Trevisani
- 12 Department of Medical Surgical Sciences, Medical Semiotics Unit, Alma Mater Studiorum, University of Bologna, Italy
| |
Collapse
|
21
|
Liu G, Hou G, Li L, Li Y, Zhou W, Liu L. Potential diagnostic and prognostic marker dimethylglycine dehydrogenase (DMGDH) suppresses hepatocellular carcinoma metastasis in vitro and in vivo. Oncotarget 2018; 7:32607-16. [PMID: 27119355 PMCID: PMC5078037 DOI: 10.18632/oncotarget.8927] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/31/2016] [Indexed: 12/23/2022] Open
Abstract
Key metabolic enzymes regulatethe fluxes of small compounds to provide the basal substrates for cellular architecture and energy. Some of them are reported to be important carcinogenesis- and metastasis-related genes. In our work, we performed RNA-seq for50 pairs of normal-tumor of hepatocellular carcinoma (HCC) samples and found that the expression of dimethylglycine dehydrogenase (DMGDH) is decreased in HCC. The analysis of protein levels with Western blotting and immunohistochemistry also conformed our findings. It is proven to be a valuable biomarker for both diagnosis and prognosis in three independent datasets. Furthermore, we revealed that DMGDH suppresses migration, invasion and metastasis both in vitro and in vivo. By utilizing gene expression microarray for DMGDH, we identified several possible pathways altered in a DMGDH over-expressing cell line. Among these pathways, we noted that the phosphorylation of Akt-308/473 was significantly suppressed when DMGDH was over-expressed. In summary, our work reveals that DMGDH is a potential valuable biomarker for both diagnosis and prognosisfor HCC, and DMGDH gene expression suppresses metastasis through the Akt signaling pathway.
Collapse
Affiliation(s)
- Gang Liu
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Guojun Hou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Liang Li
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, National Center for Liver Cancer, Shanghai, China
| | - Yixue Li
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Weiping Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Lei Liu
- Institute of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Grinchuk OV, Yenamandra SP, Iyer R, Singh M, Lee HK, Lim KH, Chow PK, Kuznetsov VA. Tumor-adjacent tissue co-expression profile analysis reveals pro-oncogenic ribosomal gene signature for prognosis of resectable hepatocellular carcinoma. Mol Oncol 2017; 12:89-113. [PMID: 29117471 PMCID: PMC5748488 DOI: 10.1002/1878-0261.12153] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/03/2017] [Accepted: 10/16/2017] [Indexed: 12/18/2022] Open
Abstract
Currently, molecular markers are not used when determining the prognosis and treatment strategy for patients with hepatocellular carcinoma (HCC). In the present study, we proposed that the identification of common pro‐oncogenic pathways in primary tumors (PT) and adjacent non‐malignant tissues (AT) typically used to predict HCC patient risks may result in HCC biomarker discovery. We examined the genome‐wide mRNA expression profiles of paired PT and AT samples from 321 HCC patients. The workflow integrated differentially expressed gene selection, gene ontology enrichment, computational classification, survival predictions, image analysis and experimental validation methods. We developed a 24‐ribosomal gene‐based HCC classifier (RGC), which is prognostically significant in both PT and AT. The RGC gene overexpression in PT was associated with a poor prognosis in the training (hazard ratio = 8.2, P = 9.4 × 10−6) and cross‐cohort validation (hazard ratio = 2.63, P = 0.004) datasets. The multivariate survival analysis demonstrated the significant and independent prognostic value of the RGC. The RGC displayed a significant prognostic value in AT of the training (hazard ratio = 5.0, P = 0.03) and cross‐validation (hazard ratio = 1.9, P = 0.03) HCC groups, confirming the accuracy and robustness of the RGC. Our experimental and bioinformatics analyses suggested a key role for c‐MYC in the pro‐oncogenic pattern of ribosomal biogenesis co‐regulation in PT and AT. Microarray, quantitative RT‐PCR and quantitative immunohistochemical studies of the PT showed that DKK1 in PT is the perspective biomarker for poor HCC outcomes. The common co‐transcriptional pattern of ribosome biogenesis genes in PT and AT from HCC patients suggests a new scalable prognostic system, as supported by the model of tumor‐like metabolic redirection/assimilation in non‐malignant AT. The RGC, comprising 24 ribosomal genes, is introduced as a robust and reproducible prognostic model for stratifying HCC patient risks. The adjacent non‐malignant liver tissue alone, or in combination with HCC tissue biopsy, could be an important target for developing predictive and monitoring strategies, as well as evidence‐based therapeutic interventions, that aim to reduce the risk of post‐surgery relapse in HCC patients.
Collapse
Affiliation(s)
| | | | | | - Malay Singh
- Bioinformatics InstituteSingapore
- Department of Computer ScienceSchool of ComputingNational University of SingaporeSingapore
| | - Hwee Kuan Lee
- Bioinformatics InstituteSingapore
- Department of Computer ScienceSchool of ComputingNational University of SingaporeSingapore
| | - Kiat Hon Lim
- Division of Surgical OncologyNational Cancer CentreSingaporeSingapore
| | - Pierce Kah‐Hoe Chow
- Division of Surgical OncologyNational Cancer CentreSingaporeSingapore
- Office of Clinical SciencesDuke‐NUS Graduate Medical SchoolSingaporeSingapore
- Department of HPB and Transplantation SurgerySingapore General HospitalSingapore
| | | |
Collapse
|
23
|
Xu M, Liu Z, Wang C, Yao B, Zheng X. EDG2 enhanced the progression of hepatocellular carcinoma by LPA/PI3K/AKT/ mTOR signaling. Oncotarget 2017; 8:66154-66168. [PMID: 29029500 PMCID: PMC5630400 DOI: 10.18632/oncotarget.19825] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/28/2017] [Indexed: 12/12/2022] Open
Abstract
HCC is the leading type of the malignant liver tumors with the unsatisfied prognosis. Liver resection has been considered as the predominant curative therapy, however, the post-surgical prognostic evaluation remains an urgent problem and the mechanism of HCC metastases has not been understood completely. EDG2 has been found to accelerate tumor progression through mediating different cell pathways, however, it remains unclear about the role of EDG2 on hepatocarcinogenesis. Here, EDG2 expression was found increased notably in HCC tissues by immunohistochemistry compared with adjacent liver tissues and comparison of survival curves revealed that EDG2 upregulation in HCC tissues was associated with the worse prognosis after liver resection. The positive correlation between EDG2 up-regulation and EMT was observed in HCC samples. Furthermore, EDG2 over-expression in HCC cells brought the typical EMT characteristics including up-regulation of Vimentin, Fibronectin and N-cadherin, suppression of E-cadherin, and enhanced cell migration and invasion capacities. Knockdown of EDG2 reversed the EMT phenotype in HCC cells. The in vivo experiments also identified the oncogenic role of EDG2 on HCC growth. The mechanistic studies elucidated that EDG2 enhanced mTOR phosphorylation via PI3K/AKT signaling and consequently induced EMT of HCC cells. Moreover, EDG2 was found to promote cell viability and proliferation of HCC cell through PI3K/AKT/mTOR/Skp2/p27Kip1 signaling. Taken together, the data here demonstrated EDG2 was a potential predictor for HCC patients receiving liver resection and accelerated HCC progression via regulating EMT driven by PI3K/AKT/mTOR signaling.
Collapse
Affiliation(s)
- Meng Xu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zhikui Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Cong Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Bowen Yao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xin Zheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
24
|
Zuckerman V, Sokolov E, Swet JH, Ahrens WA, Showlater V, Iannitti DA, Mckillop IH. Expression and function of lysophosphatidic acid receptors (LPARs) 1 and 3 in human hepatic cancer progenitor cells. Oncotarget 2016; 7:2951-67. [PMID: 26701886 PMCID: PMC4823083 DOI: 10.18632/oncotarget.6696] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 11/16/2015] [Indexed: 01/26/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary cancer of the liver and is characterized by rapid tumor expansion and metastasis. Lysophosphatidic acid (LPA) signaling, via LPA receptors 1–6 (LPARs1–6), regulates diverse cell functions including motility, migration, and proliferation, yet the role of LPARs in hepatic tumor pathology is poorly understood. We sought to determine the expression and function of endothelial differentiation gene (EDG) LPARs (LPAR1–3) in human HCC and complimentary in vitro models. Human HCC were characterized by significantly elevated LPAR1/LPAR3 expression in the microenvironment between the tumor and non-tumor liver (NTL), a finding mirrored in human SKHep1 cells. Analysis of human tissue and human hepatic tumor cells in vitro revealed cells that express LPAR3 (HCC-NTL margin in vivo and SKHep1 in vitro) also express cancer stem cell markers in the absence of hepatocyte markers. Treatment of SKHep1 cells with exogenous LPA led to significantly increased cell motility but not proliferation. Using pharmacological agents and cells transfected to knock-down LPAR1 or LPAR3 demonstrated LPA-dependent cell migration occurs via an LPAR3-Gi-ERK-pathway independent of LPAR1. These data suggest cells that stain positive for both LPAR3 and cancer stem cell markers are distinct from the tumor mass per se, and may mediate tumor invasiveness/expansion via LPA-LPAR3 signaling.
Collapse
Affiliation(s)
| | - Eugene Sokolov
- Department of Surgery, Carolinas Medical Center, Charlotte, NC, USA 28203
| | - Jacob H Swet
- Department of Surgery, Carolinas Medical Center, Charlotte, NC, USA 28203
| | - William A Ahrens
- Department of Pathology, Carolinas Medical Center, Charlotte, NC, USA 28203
| | - Victor Showlater
- Department of Surgery, Carolinas Medical Center, Charlotte, NC, USA 28203
| | - David A Iannitti
- Department of Surgery, Carolinas Medical Center, Charlotte, NC, USA 28203
| | - Iain H Mckillop
- Department of Surgery, Carolinas Medical Center, Charlotte, NC, USA 28203
| |
Collapse
|
25
|
Xiao R, Zhang X, Rong Z, Xiu B, Yang X, Wang C, Hao W, Zhang Q, Liu Z, Duan C, Zhao K, Guo X, Fan Y, Zhao Y, Johnson H, Huang Y, Feng X, Xu X, Zhang H, Wang S. Non-invasive detection of hepatocellular carcinoma serum metabolic profile through surface-enhanced Raman spectroscopy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:2475-2484. [PMID: 27520725 DOI: 10.1016/j.nano.2016.07.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 07/14/2016] [Accepted: 07/23/2016] [Indexed: 12/18/2022]
Abstract
The present study aims to identify distinctive Raman spectrum metabolic peaks to predict hepatocellular carcinoma (HCC). We performed a label-free, non-invasive surface-enhanced Raman spectroscopy (SERS) test on 230 serum samples including 47 HCC, 60 normal controls (NC), 68 breast cancer (BC) and 55 lung cancer (LC) by mixing Au@AgNRs with serum directly. Based on the observed SERS spectra, discriminative metabolites including tryptophan, phenylalanine, and etc. were found in HCC, when compared with BC, LC, and NC (P<0.05 in all). Common metabolites-proline, valine, adenine and thymine were found in HCC, BC and LC with compared to NC group (P<0.05). Importantly, Raman spectra of HCC serum biomarker AFP were firstly detected to analyze the HCC prominent peak. Orthogonal partial least squares discriminant analysis was adopted to assess the diagnostic accuracy; area under curve value of HCC is 0.991. This study provides new insights into the HCC metabolites detection through Raman spectroscopy.
Collapse
Affiliation(s)
- Rui Xiao
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Xuhui Zhang
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, Beijing, China
| | - Zhen Rong
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Bingshui Xiu
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, Beijing, China
| | - Xiqin Yang
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, Beijing, China
| | - Chongwen Wang
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Wende Hao
- Affiliated 307 Hospital, Beijing, China
| | - Qi Zhang
- National Center of Biomedical Analysis, Beijing, China
| | - Zhiqiang Liu
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, Beijing, China
| | - Cuimi Duan
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, Beijing, China
| | - Kai Zhao
- Department of Radiotherapy and Chemotherapy, Tangshan City People's Hospital, Tangshan, China
| | - Xu Guo
- Clinical Laboratory, 252 Hospital of PLA, Baoding, China
| | - Yawen Fan
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, Beijing, China
| | - Yanfeng Zhao
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, Beijing, China
| | | | - Yan Huang
- Affiliated 307 Hospital, Beijing, China
| | - Xiaoyan Feng
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, Beijing, China.
| | | | - Heqiu Zhang
- Department of Bio-diagnosis, Institute of Basic Medical Sciences, Beijing, China.
| | - Shengqi Wang
- Beijing Institute of Radiation Medicine, Beijing, China.
| |
Collapse
|
26
|
Ma J, Duan W, Han S, Lei J, Xu Q, Chen X, Jiang Z, Nan L, Li J, Chen K, Han L, Wang Z, Li X, Wu E, Huo X. Ginkgolic acid suppresses the development of pancreatic cancer by inhibiting pathways driving lipogenesis. Oncotarget 2016; 6:20993-1003. [PMID: 25895130 PMCID: PMC4673245 DOI: 10.18632/oncotarget.3663] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 02/28/2015] [Indexed: 12/16/2022] Open
Abstract
Ginkgolic acid (GA) is a botanical drug extracted from the seed coat of Ginkgo biloba L. with a wide range of bioactive properties, including anti-tumor effect. However, whether GA has antitumor effect on pancreatic cancer cells and the underlying mechanisms have yet to be investigated. In this study, we show that GA suppressed the viability of cancer cells but has little toxicity on normal cells, e.g, HUVEC cells. Furthermore, treatment of GA resulted in impaired colony formation, migration, and invasion ability and increased apoptosis of cancer cells. In addition, GA inhibited the de novo lipogenesis of cancer cells through inducing activation of AMP-activated protein kinase (AMPK) signaling and downregulated the expression of key enzymes (e.g. acetyl-CoA carboxylase [ACC], fatty acid synthase [FASN]) involved in lipogenesis. Moreover, the in vivo experiment showed that GA reduced the expression of the key enzymes involved in lipogenesis and restrained the tumor growth. Taken together, our results suggest that GA may serve as a new candidate against tumor growth of pancreatic cancer partially through targeting pathway driving lipogenesis.
Collapse
Affiliation(s)
- Jiguang Ma
- Department of Oncology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Wanxing Duan
- Department of Oncology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Suxia Han
- Department of Oncology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jianjun Lei
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qinhong Xu
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xin Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Zhengdong Jiang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Ligang Nan
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jiahui Li
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Ke Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Liang Han
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xuqi Li
- Department of General Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Erxi Wu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota, USA
| | - Xiongwei Huo
- Department of General Surgery, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
27
|
Turmeric enhancing anti-tumor effect of Rhizoma paridis saponins by influencing their metabolic profiling in tumors of H22 hepatocarcinoma mice. Pathol Res Pract 2015; 211:948-54. [PMID: 26471217 DOI: 10.1016/j.prp.2015.09.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 08/15/2015] [Accepted: 09/09/2015] [Indexed: 11/21/2022]
Abstract
Rhizoma Paridis saponins combined with turmeric (RT) showed well anti-hepatocarcinoma activities in our previous research. The aim of this study was to investigate the progression of the biochemical response to RT and capture metabolic variations during intragastric administration of their compatibility. In the experiment, histopathological examination and (1)H NMR method were developed and validated for the metabolic profiling of RT intervention in H22 tumor growth. Data were analyzed with principal components analysis (PCA) and partial least-squares discrimination analysis (PLS-DA). As a result, Rhizoma paridis saponins (RPS) or RT induced inflammatory cell infiltration in tumors. RT also mediated the tumor microenvironment to promote anti-tumor immunity of mice. RT significantly inhibited tumor growth rate through suppressing levels of amino acids containing alanine, asparagine, glutamine, putrescine, and sarcosine, lipid compounds, and carbohydrates like myo-inositol and arabinose in the tumor tissues. In conclusion, these results uncovered unexpectedly poor nutritional conditions in the RT-treated tumor tissues whose effect was stronger than RPS's. Therefore, RT could be a novel anticancer agent that targets on cancer metabolism through starving tumors reducing viability of cancer cells.
Collapse
|
28
|
Territo PR, Maluccio M, Riley AA, McCarthy BP, Fletcher J, Tann M, Saxena R, Skill NJ. Evaluation of 11C-acetate and 18F-FDG PET/CT in mouse multidrug resistance gene-2 deficient mouse model of hepatocellular carcinoma. BMC Med Imaging 2015; 15:15. [PMID: 25981587 PMCID: PMC4493966 DOI: 10.1186/s12880-015-0058-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 05/08/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) remains a global health problem with unique diagnostic and therapeutic challenges, including difficulties in identifying the highest risk patients. Previous work from our lab has established the murine multidrug resistance-2 mouse (MDR2) model of HCC as a reasonable preclinical model that parallels the changes seen in human inflammatory associated HCC. The purpose of this study is to evaluate modalities of PET/CT in MDR2(-/-) mice in order to facilitate therapeutic translational studies from bench to bedside. METHODS 18F-FDG and 11C-acetate PET/CT was performed on 12 m MDR2(-/-) mice (n = 3/tracer) with HCC and 12 m MDR2(-/+) control mice (n = 3/tracer) without HCC. To compare PET/CT to biological markers of HCC and cellular function, serum alpha-fetoprotein (AFP), lysophosphatidic acid (LPA), cAMP and hepatic tumor necrosis factor α (TNFα) were quantified in 3-12 m MDR2(-/-) (n = 10) mice using commercially available ELISA analysis. To translate results in mice to patients 11C-acetate PET/CT was also performed in 8 patents suspected of HCC recurrence following treatment and currently on the liver transplant wait list. RESULTS Hepatic18F-FDG metabolism was not significantly increased in MDR2(-/-) mice. In contrast, hepatic 11C-acetate metabolism was significantly elevated in MDR2(-/-) mice when compared to MDR2(-/+) controls. Serum AFP and LPA levels increased in MDR2(-/-) mice contemporaneous with the emergence of HCC. This was accompanied by a significant decrease in serum cAMP levels and an increase in hepatic TNFα. In patients suspected of HCC recurrence there were 5 true positives, 2 true negatives and 1 suspected false 11C-acetate negative. CONCLUSIONS Hepatic 11C-acetate PET/CT tracks well with HCC in MDR2(-/-) mice and patients with underlying liver disease. Consequently 11C-acetate PET/CT is well suited to study (1) HCC emergence/progression in patients and (2) reduce animal numbers required to study new chemotherapeutics in murine models of HCC.
Collapse
Affiliation(s)
- Paul R Territo
- Department of Surgery, Radiology and Imaging Sciences, Indianapolis, IN, 46202, USA.
| | - Mary Maluccio
- Department of Surgery, Indiana University School of Medicine, C519 Walthur Cancer Research Building (R3), 980 W Walnut Street, Indianapolis, IN, 46077, USA.
| | - Amanda A Riley
- Department of Surgery, Radiology and Imaging Sciences, Indianapolis, IN, 46202, USA.
| | - Brian P McCarthy
- Department of Surgery, Radiology and Imaging Sciences, Indianapolis, IN, 46202, USA.
| | - James Fletcher
- Department of Surgery, Radiology and Imaging Sciences, Indianapolis, IN, 46202, USA.
| | - Mark Tann
- Department of Surgery, Radiology and Imaging Sciences, Indianapolis, IN, 46202, USA.
| | - Romil Saxena
- Department of Surgery, Radiology and Imaging Sciences, Indianapolis, IN, 46202, USA.
| | - Nicholas J Skill
- Department of Surgery, Indiana University School of Medicine, C519 Walthur Cancer Research Building (R3), 980 W Walnut Street, Indianapolis, IN, 46077, USA.
| |
Collapse
|
29
|
Simo KA, Niemeyer DJ, Hanna EM, Swet JH, Thompson KJ, Sindram D, Iannitti DA, Eheim AL, Sokolov E, Zuckerman V, McKillop IH. Altered lysophosphatidic acid (LPA) receptor expression during hepatic regeneration in a mouse model of partial hepatectomy. HPB (Oxford) 2014; 16:534-42. [PMID: 24750398 PMCID: PMC4048075 DOI: 10.1111/hpb.12176] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 07/28/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND Hepatic regeneration requires coordinated signal transduction for efficient restoration of functional liver mass. This study sought to determine changes in lysophosphatidic acid (LPA) and LPA receptor (LPAR) 1-6 expression in regenerating liver following two-thirds partial hepatectomy (PHx). METHODS Liver tissue and blood were collected from male C57BL/6 mice following PHx. Circulating LPA was measured by enzyme-linked immunosorbent assay (ELISA) and hepatic LPAR mRNA and protein expression were determined. RESULTS Circulating LPA increased 72 h after PHx and remained significantly elevated for up to 7 days post-PHx. Analysis of LPAR expression after PHx demonstrated significant increases in LPAR1, LPAR3 and LPAR6 mRNA and protein in a time-dependent manner for up to 7 days post-PHx. Conversely, LPAR2, LPAR4 and LPAR5 mRNA were barely detected in normal liver and did not significantly change after PHx. Changes in LPAR1 expression were confined to non-parenchymal cells following PHx. CONCLUSIONS Liver regeneration following PHx is associated with significant changes in circulating LPA and hepatic LPAR1, LPAR3 and LPAR6 expression in a time- and cell-dependent manner. Furthermore, changes in LPA-LPAR post-PHx occur after the first round of hepatocyte division is complete.
Collapse
Affiliation(s)
- Kerri A Simo
- Department of Surgery, Carolinas Medical Center, Charlotte, NC, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Beyoğlu D, Imbeaud S, Maurhofer O, Bioulac-Sage P, Zucman-Rossi J, Dufour JF, Idle JR. Tissue metabolomics of hepatocellular carcinoma: tumor energy metabolism and the role of transcriptomic classification. Hepatology 2013; 58:229-38. [PMID: 23463346 PMCID: PMC3695036 DOI: 10.1002/hep.26350] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 02/19/2013] [Indexed: 12/12/2022]
Abstract
UNLABELLED Hepatocellular carcinoma (HCC) is one of the commonest causes of death from cancer. A plethora of metabolomic investigations of HCC have yielded molecules in biofluids that are both up- and down-regulated but no real consensus has emerged regarding exploitable biomarkers for early detection of HCC. We report here a different approach, a combined transcriptomics and metabolomics study of energy metabolism in HCC. A panel of 31 pairs of HCC tumors and corresponding nontumor liver tissues from the same patients was investigated by gas chromatography-mass spectrometry (GCMS)-based metabolomics. HCC was characterized by ∼2-fold depletion of glucose, glycerol 3- and 2-phosphate, malate, alanine, myo-inositol, and linoleic acid. Data are consistent with a metabolic remodeling involving a 4-fold increase in glycolysis over mitochondrial oxidative phosphorylation. A second panel of 59 HCC that had been typed by transcriptomics and classified in G1 to G6 subgroups was also subjected to GCMS tissue metabolomics. No differences in glucose, lactate, alanine, glycerol 3-phosphate, malate, myo-inositol, or stearic acid tissue concentrations were found, suggesting that the Wnt/β-catenin pathway activated by CTNNB1 mutation in subgroups G5 and G6 did not exhibit specific metabolic remodeling. However, subgroup G1 had markedly reduced tissue concentrations of 1-stearoylglycerol, 1-palmitoylglycerol, and palmitic acid, suggesting that the high serum α-fetoprotein phenotype of G1, associated with the known overexpression of lipid catabolic enzymes, could be detected through metabolomics as increased lipid catabolism. CONCLUSION Tissue metabolomics yielded precise biochemical information regarding HCC tumor metabolic remodeling from mitochondrial oxidation to aerobic glycolysis and the impact of molecular subtypes on this process.
Collapse
Affiliation(s)
- Diren Beyoğlu
- Hepatology Research Group, Department of Clinical Research, University of Bern, Switzerland
| | - Sandrine Imbeaud
- Inserm, UMR-674, Génomiquefonctionnelle des tumeurssolides, IUH, Paris, F-75010 France,Université Paris Descartes, LabexImmuno-oncology, Sorbonne Paris Cité, Faculté de Médecine, Assistance Publique-Hôpitaux de Paris, France
| | - Olivier Maurhofer
- Hepatology Research Group, Department of Clinical Research, University of Bern, Switzerland
| | - Paulette Bioulac-Sage
- Inserm, UMR-1053; Université Victor Segalen Bordeaux 2, Bordeaux, F-33076, France,CHU de Bordeaux, Pellegrin Hospital, Department of Pathology, Bordeaux, F-33076, France
| | - Jessica Zucman-Rossi
- Inserm, UMR-674, Génomiquefonctionnelle des tumeurssolides, IUH, Paris, F-75010 France,Université Paris Descartes, LabexImmuno-oncology, Sorbonne Paris Cité, Faculté de Médecine, Assistance Publique-Hôpitaux de Paris, France
| | - Jean-François Dufour
- Hepatology Research Group, Department of Clinical Research, University of Bern, Switzerland
| | - Jeffrey R. Idle
- Hepatology Research Group, Department of Clinical Research, University of Bern, Switzerland
| |
Collapse
|
31
|
Skill NJ, Jianmin W, Yan X, Zhao Z, Tector AJ, Maluccio MA. Lysophospholipid variants in hepatocellular carcinoma. J Surg Res 2013; 182:241-9. [DOI: 10.1016/j.jss.2012.10.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 09/26/2012] [Accepted: 10/18/2012] [Indexed: 12/17/2022]
|
32
|
Skill N, Wu J, Xu Y, Zhao Z, Maluccio M. Lysophosphatidic acid aberrancies and hepatocellular carcinoma: studies in the MDR2 gene knockout mouse. Cancer Invest 2013; 31:145-55. [PMID: 23362952 DOI: 10.3109/07357907.2012.762779] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Studies show that lysophosphatidic acid (LPA) reprogramming is associated with the development of hepatocellular carcinoma (HCC). This manuscript evaluates the MDR2(-/-) model of HCC as a tool to examine the role of LPA reprogramming in the initiation/progression of HCC and identify novel treatment targets. Hepatic tumors developed in MDR2(-/-) mice between 9-12 m and serum LPA levels were greater in MDR2(-/-) when compared to controls. Blocking LPA biosynthesis/signaling significantly reduced tumor burden. LPA biosynthesis/signaling plays an important role in murine MDR2(-/-) model and is potentially linked to regulation of TNFα or other cytokines that are relevant to high-risk patients.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/deficiency
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- Animals
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Drug Resistance, Multiple
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms, Experimental/blood
- Liver Neoplasms, Experimental/genetics
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/pathology
- Lysophospholipids/blood
- Lysophospholipids/genetics
- Lysophospholipids/metabolism
- Mice
- Mice, Knockout
- Phosphodiesterase Inhibitors/pharmacology
- Phospholipids/metabolism
- Phosphoric Diester Hydrolases/metabolism
- Receptors, Lysosphingolipid/antagonists & inhibitors
- Receptors, Lysosphingolipid/metabolism
- Signal Transduction/drug effects
- ATP-Binding Cassette Sub-Family B Member 4
Collapse
Affiliation(s)
- Nicholas Skill
- Department of Surgery, Indiana University, Indianapolis, IN, USA.
| | | | | | | | | |
Collapse
|
33
|
Sokolov E, Eheim AL, Ahrens WA, Walling TL, Swet JH, McMillan MT, Simo KA, Thompson KJ, Sindram D, McKillop IH. Lysophosphatidic acid receptor expression and function in human hepatocellular carcinoma. J Surg Res 2012. [PMID: 23182454 DOI: 10.1016/j.jss.2012.10.054] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Lysophosphatidic acid (LPA) is a ubiquitously expressed phospholipid that regulates diverse cellular functions. Previously identified LPA receptor subtypes (LPAR1-5) are weakly expressed or absent in the liver. This study sought to determine LPAR expression, including the newly identified LPAR6, in normal human liver (NL), hepatocellular carcinoma (HCC), and non-tumor liver tissue (NTL), and LPAR expression and function in human hepatoma cells in vitro. METHODS We determined LPAR1-6 expression by quantitative reverse transcriptase polymerase chain reaction, Western blot, or immunohistochemistry in NL, NTL, and HCC, and HuH7, and HepG2 cells. Hepatoma cells were treated with LPA in the absence or presence of LPAR1-3 (Ki16425) or pan-LPAR (α-bromomethylene phosphonate) antagonists and proliferation and motility were measured. RESULTS We report HCC-associated changes in LPAR1, 3, and 6 mRNA and protein expression, with significantly increased LPAR6 in HCC versus NL and NTL. Analysis of human hepatoma cells demonstrated significantly higher LPAR1, 3, and 6 mRNA and protein expression in HuH7 versus HepG2 cells. Treatment with LPA (0.05-10 μg/mL) led to dose-dependent HuH7 growth and increased motility. In HepG2 cells, LPA led to moderate, although significant, increases in proliferation but not motility. Pretreatment with α-bromomethylene phosphonate inhibited LPA-dependent proliferation and motility to a greater degree than Ki16425. CONCLUSIONS Multiple LPAR forms are expressed in human HCC, including the recently described LPAR6. Inhibition of LPA-LPAR signaling inhibits HCC cell proliferation and motility, the extent of which depends on LPAR subtype expression.
Collapse
Affiliation(s)
- Eugene Sokolov
- Department of Surgery, Carolinas Medical Center, Charlotte, NC 28203, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Functional imaging techniques in hepatocellular carcinoma. Eur J Nucl Med Mol Imaging 2012; 39:1070-9. [PMID: 22434049 DOI: 10.1007/s00259-012-2096-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Accepted: 02/20/2012] [Indexed: 02/07/2023]
Abstract
Novel biological therapies, including tyrosine kinase inhibitors such as sorafenib, improve the survival of patients with unresectable hepatocellular carcinoma. However, assessment of therapeutic efficacy remains challenging with conventional imaging techniques such as ultrasonography, CT or MRI that predominantly rely on size change to detect a treatment response. A beneficial tumour effect may go unrecognized in some patients who do not show tumour shrinkage and conversely, some patients may be maintained on treatment that is not active. This paper explores the use of functional imaging methods that are showing promise in the assessment of hepatocellular carcinoma.
Collapse
|
35
|
Korkusuz H, Knau L, Kromen W, Huebner F, Hammerstingl R, Lindemayr S, Bihrer V, Piiper A, Vogl TJ. Gadoxetate acid-enhanced MRI of hepatocellular carcinoma in a c-myc/TGFα transgenic mouse model including signal intensity and fat content: initial experience. Cancer Imaging 2012; 12:72-8. [PMID: 22418445 PMCID: PMC3335333 DOI: 10.1102/1470-7330.2012.0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Genetically engineered mouse models, such as double transgenic c-myc/TGFα mice, with specific pathway abnormalities might be more successful at predicting the clinical response of hepatocellular carcinoma (HCC) treatment. But a major drawback of the tumour models is the difficulty of visualizing endogenously formed tumours. The optimal imaging procedure should be brief and minimally invasive. Magnetic resonance imaging (MRI) satisfies these criteria and gadoxetate acid-enhanced MRI improves the detection of HCC. Fat content is stated to be an additional tool to help assess tumour responses, for example, in cases of radiofrequency ablation. Therefore the aim of this study was to investigate if gadoxetate acid-enhanced MRI could be used to detect HCC in c-myc/TGFα transgenic mice by determining the relation between the signal intensity of HCC and normal liver parenchyma and the corresponding fat content as a diagnostic marker of HCC. In our study, 20 HCC in c-myc/TGFα transgenic male mice aged 20–34 weeks were analyzed. On gadoxetate acid-enhanced MRI, the signal intensity was 752.4 for liver parenchyma and 924.5 for HCC. The contrast to noise ratio was 20.4, the percentage enhancement was 267.1% for normal liver parenchyma and 353.9% for HCC. The fat content was 11.2% for liver parenchyma and 16.2% for HCC. There was a correlation between fat content and signal intensity with r = 0.7791. All parameters were statistically significant with P < 0.05. Our data indicate that gadoxetate acid contrast enhancement allows sensitive detection of HCC in c-myc/TGFα transgenic mice and determination of the fat content seems to be an additional useful parameter for HCC.
Collapse
Affiliation(s)
- Huedayi Korkusuz
- Department of Nuclear Medicine, Johann Wolfgang Goethe University Hospital, Frankfurt, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Mazzocca A, Dituri F, Lupo L, Quaranta M, Antonaci S, Giannelli G. Tumor-secreted lysophostatidic acid accelerates hepatocellular carcinoma progression by promoting differentiation of peritumoral fibroblasts in myofibroblasts. Hepatology 2011; 54:920-30. [PMID: 21674557 DOI: 10.1002/hep.24485] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 05/26/2011] [Indexed: 12/11/2022]
Abstract
UNLABELLED Hepatocellular carcinoma (HCC) occurs in fibrotic liver as a consequence of underlying cirrhosis. The goal of this study was to investigate how the interaction between HCC cells and stromal fibroblasts affects tumor progression. We isolated and characterized carcinoma-associated fibroblasts (CAFs) and paired peritumoral tissue fibroblasts (PTFs) from 10 different patients with HCC and performed coculture experiments. We demonstrated a paracrine mechanism whereby HCC cells secrete lysophostatidic acid (LPA), which promotes transdifferentiation of PTFs to a CAF-like myofibroblastic phenotype. This effect is mediated by up-regulation of specific genes related to a myo/contractile phenotype. After transdifferentiation, PTFs expressed α-smooth muscle actin (α-SMA) and enhanced proliferation, migration, and invasion of HCC cells occur. A pan-LPA inhibitor (α-bromomethylene phosphonate [BrP]-LPA), or autotaxin gene silencing, inhibited this PTF transdifferentiation and the consequent enhanced proliferation, migration, and invasion of HCC cells. In vivo, PTFs coinjected with HCC cells underwent transdifferentiation and promoted tumor progression. Treatment with BrP-LPA blocked transdifferentiation of PTFs, down-regulated myofibroblast-related genes, and slowed HCC growth and progression. Patients with larger and metastatic HCC and shorter survival displayed higher serum levels of LPA. Analysis of microdissected tissues indicated that stroma is the main target of the LPA paracrine loop in HCC. As a consequence, α-SMA-positive cells were more widespread in tumoral compared with paired peritumoral stroma. CONCLUSION Our data indicate that LPA accelerates HCC progression by recruiting PTFs and promoting their transdifferentiation into myofibroblasts. Inhibition of LPA could prove effective in blocking transdifferentiation of myofibroblasts and tumor progression.
Collapse
Affiliation(s)
- Antonio Mazzocca
- Department of Internal Medicine, Immunology, and Infectious Diseases, Section of Internal Medicine, University of Bari Medical School, Bari, Italy
| | | | | | | | | | | |
Collapse
|