101
|
Yang X, Xie X, Xiao YF, Xie R, Hu CJ, Tang B, Li BS, Yang SM. The emergence of long non-coding RNAs in the tumorigenesis of hepatocellular carcinoma. Cancer Lett 2015; 360:119-24. [PMID: 25721084 DOI: 10.1016/j.canlet.2015.02.035] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 02/16/2015] [Accepted: 02/16/2015] [Indexed: 12/13/2022]
Abstract
Hepatocellular carcinoma (HCC) is the third cause of cancer-related death worldwide. However, the treatments for HCC are limited, and most of them are only available to the early stage. In the later stages, traditional chemotherapy has only marginal effects and may include toxicity. Thus, the identification of new predictive markers is urgently needed. New targets for non-conventional treatments will help to accelerate research on the molecular pathogenesis of HCC. A new class of transcripts, long non-coding RNAs (lncRNAs), has recently been found to be pervasively transcribed in the human genome. Aberrant expression of several lncRNAs was found to be involved in the tumorigenesis of HCC. In this review, we describe the possible molecular mechanisms that underlie lncRNA expression changes in HCC, as well as potential future applications of lncRNA research in the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Xin Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Xia Xie
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Yu-Feng Xiao
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Rei Xie
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Chang-Jiang Hu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Bo-Sheng Li
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Shi-Ming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
102
|
Zaghloul RA, El-Shishtawy MM, El Galil KHA, Ebrahim MA, Metwaly AA, Al-Gayyar MM. Evaluation of antiglypican-3 therapy as a promising target for amelioration of hepatic tissue damage in hepatocellular carcinoma. Eur J Pharmacol 2015; 746:353-362. [PMID: 25449037 DOI: 10.1016/j.ejphar.2014.11.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 11/04/2014] [Accepted: 11/05/2014] [Indexed: 01/16/2023]
Abstract
In Egypt, hepatocellular carcinoma (HCC) was predicted to continue to rise over the next few decades causing a national problem. Meanwhile, glypican-3 (GPC3), a highly expressed glypican, has emerged as a potential target for HCC immunotherapy. Therefore, we aimed to identify the impact of blocking GPC3 on liver damage in HCC as well as a possible mechanism. Fifty four HCC patients, 20 cirrhotic patients and 10 healthy subjects were recruited. Serum levels of GPC3, sulfatase-2 (SULF-2), heparan sulfate proteoglycan (HSPG), insulin-like growth factor-II (IGF-II) were measured by ELISA. In parallel, HCC was induced in 40 male Sprague-Dawley rats in presence/absence of antiGPC-3. Liver impairment was detected by investigating liver sections stained with hematoxylin/eosin and serum α-fetoprotein (AFP). Liver homogenates of GPC3, SULF-2, and HSPG were measured by ELISA. Gene expression of caspase-3 and IGF-II were assayed by RT-PCR. HCC patients showed significant elevated serum levels of GPC3, IGF-II and SULF-2 accompanied by decreased HSPG. However, treatment of HCC rats with antiGPC-3 significantly reduced serum AFP and showed nearly normal hepatocytes. In addition, antiGPC-3 significantly reduced elevated liver homogenates protein levels of GPC3 and SULF-2 and gene expression of IGF-II and caspase-3. antiGPC-3 restored the reduced hepatic HSPG. antiGPC-3 showed anti-tumor activity as well as hepatoprotective effects. antiGPC-3-chemoprotective effect can be explained by forced reduction of IGF-II expression, restoration of HSPGs, deactivation of SULF-2 and reduction of gene expression of caspase-3. Targeting GPC3 is a promising therapeutic approach for HCC.
Collapse
Affiliation(s)
- Randa A Zaghloul
- Dept. of Biochemistry, Faculty of Pharmacy, University of Mansoura, Mansoura 35516, Egypt.
| | - Mamdouh M El-Shishtawy
- Dept. of Biochemistry, Faculty of Pharmacy, University of Mansoura, Mansoura 35516, Egypt
| | - Khaled H Abd El Galil
- Dept. of Microbiology, Faculty of Pharmacy, University of Mansoura, Mansoura 35516, Egypt
| | | | - AbdelHamid A Metwaly
- Dept. of Internal Medicine, Faculty of Medicine, University of Mansoura, Mansoura 35516, Egypt
| | - Mohammed M Al-Gayyar
- Dept. of Biochemistry, Faculty of Pharmacy, University of Mansoura, Mansoura 35516, Egypt; Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| |
Collapse
|
103
|
Beebe SJ. Hepatocellular carcinoma ablation and possible immunity in the age of nanosecond pulsed electric fields. J Hepatocell Carcinoma 2015; 2:49-55. [PMID: 27508194 PMCID: PMC4918284 DOI: 10.2147/jhc.s83941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Affiliation(s)
- Stephen J Beebe
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, USA
| |
Collapse
|
104
|
Li Y, Jiang F, Liu Q, Shen J, Wang X, Li Z, Zhang J, Lu X. Inhibition of the cancer stem cells-like properties by arsenic trioxide, involved in the attenuation of endogenous transforming growth factor beta signal. Toxicol Sci 2015; 143:156-164. [PMID: 25304214 DOI: 10.1093/toxsci/kfu218] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The elevation of cancer stem cells (CSCs)-like properties is involved in the initiation and progression of various human cancers. Current standard practices for treatment of cancers are less than satisfactory because of CSCs-mediated recurrence. For this reason, targeting the CSCs or the cancer cells with CSCs-like properties has become the new approach for the cancer treatments. In addition to treating leukemia, arsenic trioxide (As₂O₃) also suppresses other solid tumors. However, the roles of As₂O₃ in the regulation of CSCs-like properties remain largely uninvestigated. Here by using sphere formation assay, luciferase reporter assay, and some other molecular biology approaches, we found that As₂O₃ attenuated the CSCs-like properties in human hepatocellular carcinoma (HCC). Briefly, in HCC cells and mice xenograft models, As₂O₃ improved the expression of miR-491 by DNA-demethylation. MiR-491, which targeted the SMAD3-3'-UTR, decreased the expressions of SMAD3, and inhibited the CSCs-like properties in HCC cells. Knockdown of either miR-491 or SMAD3 attenuated the As₂O₃-induced inhibition of endogenous transforming growth factor beta signal and the CSCs-like properties. Further, in HCC patients, miR-491 is inversely correlated with the expressions of SMAD3, CD133, and the metastasis/recurrence outcome. By understanding a novel mechanism whereby As₂O₃ inhibits the CSCs-like properties in HCC, our study would help in the design of future strategies of developing As₂O₃ as a potential HCC chemopreventive agent when used alone or in combination with other current drugs.
Collapse
MESH Headings
- 3' Untranslated Regions
- AC133 Antigen
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antineoplastic Agents/pharmacology
- Arsenic Trioxide
- Arsenicals/pharmacology
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/mortality
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Down-Regulation
- Gene Expression Regulation, Neoplastic
- Glycoproteins/genetics
- Glycoproteins/metabolism
- Humans
- Kaplan-Meier Estimate
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/mortality
- Liver Neoplasms/pathology
- Mice, Inbred BALB C
- Mice, Nude
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Oxides/pharmacology
- Peptides/genetics
- Peptides/metabolism
- RNA Interference
- Signal Transduction/drug effects
- Smad3 Protein/genetics
- Smad3 Protein/metabolism
- Time Factors
- Transfection
- Transforming Growth Factor beta/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Yuan Li
- *The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Department of General Surgery and Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China
| | - Fei Jiang
- *The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Department of General Surgery and Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China
| | - Qinqiang Liu
- *The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Department of General Surgery and Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China
| | - Jian Shen
- *The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Department of General Surgery and Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China
| | - Xingxing Wang
- *The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Department of General Surgery and Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China
| | - Zhong Li
- *The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Department of General Surgery and Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China
| | - Jianping Zhang
- *The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Department of General Surgery and Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China
| | - Xiang Lu
- *The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Department of General Surgery and Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, China
| |
Collapse
|
105
|
Nikolaou KC, Moulos P, Chalepakis G, Hatzis P, Oda H, Reinberg D, Talianidis I. Spontaneous development of hepatocellular carcinoma with cancer stem cell properties in PR-SET7-deficient livers. EMBO J 2014; 34:430-47. [PMID: 25515659 PMCID: PMC4330999 DOI: 10.15252/embj.201489279] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
PR-SET7-mediated histone 4 lysine 20 methylation has been implicated in mitotic condensation, DNA damage response and replication licensing. Here, we show that PR-SET7 function in the liver is pivotal for maintaining genome integrity. Hepatocyte-specific deletion of PR-SET7 in mouse embryos resulted in G2 phase arrest followed by massive cell death and defect in liver organogenesis. Inactivation at postnatal stages caused cell duplication-dependent hepatocyte necrosis, accompanied by inflammation, fibrosis and compensatory growth induction of neighboring hepatocytes and resident ductal progenitor cells. Prolonged necrotic regenerative cycles coupled with oncogenic STAT3 activation led to the spontaneous development of hepatic tumors composed of cells with cancer stem cell characteristics. These include a capacity to self-renew in culture or in xenografts and the ability to differentiate to phenotypically distinct hepatic cells. Hepatocellular carcinoma in PR-SET7-deficient mice displays a cancer stem cell gene signature specified by the co-expression of ductal progenitor markers and oncofetal genes.
Collapse
Affiliation(s)
| | | | | | - Pantelis Hatzis
- Biomedical Sciences Research Center Al. Fleming, Vari, Greece
| | - Hisanobu Oda
- Medical Institute of Bioregulation Kyusyu University, Fukuoka, Japan Gastrointestinal and Oncology Division, National Kyusyu Cancer Center, Fukuoka, Japan
| | - Danny Reinberg
- HHMI, Department of Biochemistry, New York University School of Medicine, New York, NY USA
| | | |
Collapse
|
106
|
Xu LB, Liu C. Role of liver stem cells in hepatocarcinogenesis. World J Stem Cells 2014; 6:579-590. [PMID: 25426254 PMCID: PMC4178257 DOI: 10.4252/wjsc.v6.i5.579] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 08/24/2014] [Accepted: 09/01/2014] [Indexed: 02/06/2023] Open
Abstract
Liver cancer is an aggressive disease with a high mortality rate. Management of liver cancer is strongly dependent on the tumor stage and underlying liver disease. Unfortunately, most cases are discovered when the cancer is already advanced, missing the opportunity for surgical resection. Thus, an improved understanding of the mechanisms responsible for liver cancer initiation and progression will facilitate the detection of more reliable tumor markers and the development of new small molecules for targeted therapy of liver cancer. Recently, there is increasing evidence for the “cancer stem cell hypothesis”, which postulates that liver cancer originates from the malignant transformation of liver stem/progenitor cells (liver cancer stem cells). This cancer stem cell model has important significance for understanding the basic biology of liver cancer and has profound importance for the development of new strategies for cancer prevention and treatment. In this review, we highlight recent advances in the role of liver stem cells in hepatocarcinogenesis. Our review of the literature shows that identification of the cellular origin and the signaling pathways involved is challenging issues in liver cancer with pivotal implications in therapeutic perspectives. Although the dedifferentiation of mature hepatocytes/cholangiocytes in hepatocarcinogenesis cannot be excluded, neoplastic transformation of a stem cell subpopulation more easily explains hepatocarcinogenesis. Elimination of liver cancer stem cells in liver cancer could result in the degeneration of downstream cells, which makes them potential targets for liver cancer therapies. Therefore, liver stem cells could represent a new target for therapeutic approaches to liver cancer in the near future.
Collapse
|
107
|
Wang F, Yuan JH, Wang SB, Yang F, Yuan SX, Ye C, Yang N, Zhou WP, Li WL, Li W, Sun SH. Oncofetal long noncoding RNA PVT1 promotes proliferation and stem cell-like property of hepatocellular carcinoma cells by stabilizing NOP2. Hepatology 2014; 60:1278-90. [PMID: 25043274 DOI: 10.1002/hep.27239] [Citation(s) in RCA: 359] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 05/21/2014] [Indexed: 12/14/2022]
Abstract
UNLABELLED Many protein-coding oncofetal genes are highly expressed in murine and human fetal liver and silenced in adult liver. The protein products of these hepatic oncofetal genes have been used as clinical markers for the recurrence of hepatocellular carcinoma (HCC) and as therapeutic targets for HCC. Herein we examined the expression profiles of long noncoding RNAs (lncRNAs) found in fetal and adult liver in mice. Many fetal hepatic lncRNAs were identified; one of these, lncRNA-mPvt1, is an oncofetal RNA that was found to promote cell proliferation, cell cycling, and the expression of stem cell-like properties of murine cells. Interestingly, we found that human lncRNA-hPVT1 was up-regulated in HCC tissues and that patients with higher lncRNA-hPVT1 expression had a poor clinical prognosis. The protumorigenic effects of lncRNA-hPVT1 on cell proliferation, cell cycling, and stem cell-like properties of HCC cells were confirmed both in vitro and in vivo by gain-of-function and loss-of-function experiments. Moreover, mRNA expression profile data showed that lncRNA-hPVT1 up-regulated a series of cell cycle genes in SMMC-7721 cells. By RNA pulldown and mass spectrum experiments, we identified NOP2 as an RNA-binding protein that binds to lncRNA-hPVT1. We confirmed that lncRNA-hPVT1 up-regulated NOP2 by enhancing the stability of NOP2 proteins and that lncRNA-hPVT1 function depends on the presence of NOP2. CONCLUSION Our study demonstrates that the expression of many lncRNAs is up-regulated in early liver development and that the fetal liver can be used to search for new diagnostic markers for HCC. LncRNA-hPVT1 promotes cell proliferation, cell cycling, and the acquisition of stem cell-like properties in HCC cells by stabilizing NOP2 protein. Regulation of the lncRNA-hPVT1/NOP2 pathway may have beneficial effects on the treatment of HCC.
Collapse
Affiliation(s)
- Fang Wang
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China; Center of Reproductive Medicine, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
108
|
TGF-β signaling alters the pattern of liver tumorigenesis induced by Pten inactivation. Oncogene 2014; 34:3273-82. [PMID: 25132272 PMCID: PMC4333137 DOI: 10.1038/onc.2014.258] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 06/04/2014] [Accepted: 06/15/2014] [Indexed: 12/14/2022]
Abstract
Hepatocarcinogenesis results from the accumulation of genetic and epigenetic changes in liver cells. A common mechanism through which these alterations induce liver cancer is by deregulating signaling pathways. A number of signaling pathways, including the PI3K/PTEN/AKT and transforming growth factor β (TGF-β) pathways have been implicated in normal liver development as well as in cancer formation. In this study, we assessed the effect of the TGF-β signaling pathway on liver tumors induced by phosphatase and tensin homolog (Pten) loss. Inactivation of only the TGF-β receptor type II, Tgfbr2, in the mouse liver (Tgfbr2(LKO)) had no overt phenotype, while inactivation of Pten alone (Pten(LKO)), resulted in the formation of both hepatocellular carcinomas and cholangiocarcinomas (CC). Interestingly, deletion of both Pten and Tgfbr2 (Pten(LKO);Tgfbr2(LKO)) in the mouse liver resulted in a dramatic shift in tumor type to predominantly CC. Assessment of the PI3K/PTEN/AKT pathway revealed increased phosphorylation of AKT and glycogen synthase kinase 3 beta (GSK-3β) in both the Pten(LKO) and Pten(LKO);Tgfbr2(LKO) mice, suggesting that this pathway is constitutively active regardless of the status of the TGF-β signaling pathway. However, phosphorylation of p70 S6 kinase was observed in the liver of all three phenotypes (Tgfbr2(LKO), Pten(LKO), Pten(LKO);Tgfbr2(LKO)) indicating that the loss of Tgfbr2 and/or Pten leads to an increase in this signaling pathway. Analysis of markers of liver progenitor/stem cells revealed that the loss of TGF-β signaling resulted in increased expression of c-Kit and CD133. Furthermore, in addition to increased c-Kit and CD133, Scf and EpCam expression were also increased in the double knock-out mice. These results suggest that the alteration in tumor types between the Pten(LKO) mice and Pten(LKO);Tgfbr2(LKO) mice is secondary to the altered regulation of stem-cell features induced by the loss of TGF-β signaling.
Collapse
|
109
|
Yuan JH, Yang F, Wang F, Ma JZ, Guo YJ, Tao QF, Liu F, Pan W, Wang TT, Zhou CC, Wang SB, Wang YZ, Yang Y, Yang N, Zhou WP, Yang GS, Sun SH. A long noncoding RNA activated by TGF-β promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer Cell 2014; 25:666-81. [PMID: 24768205 DOI: 10.1016/j.ccr.2014.03.010] [Citation(s) in RCA: 1251] [Impact Index Per Article: 113.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 01/07/2014] [Accepted: 03/11/2014] [Indexed: 02/06/2023]
Abstract
The role of TGF-β-induced epithelial-mesenchymal transition (EMT) in cancer cell dissemination is well established, but the involvement of lncRNAs in TGF-β signaling is still unknown. In this study, we observed that the lncRNA-activated by TGF-β (lncRNA-ATB) was upregulated in hepatocellular carcinoma (HCC) metastases and associated with poor prognosis. lncRNA-ATB upregulated ZEB1 and ZEB2 by competitively binding the miR-200 family and then induced EMT and invasion. In addition, lncRNA-ATB promoted organ colonization of disseminated tumor cells by binding IL-11 mRNA, autocrine induction of IL-11, and triggering STAT3 signaling. Globally, lncRNA-ATB promotes the invasion-metastasis cascade. Thus, these findings suggest that lncRNA-ATB, a mediator of TGF-β signaling, could predispose HCC patients to metastases and may serve as a potential target for antimetastatic therapies.
Collapse
Affiliation(s)
- Ji-hang Yuan
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Fu Yang
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Fang Wang
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Jin-zhao Ma
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Ying-jun Guo
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Qi-fei Tao
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Feng Liu
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Wei Pan
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Tian-tian Wang
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Chuan-chuan Zhou
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Shao-bing Wang
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Yu-zhao Wang
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Yuan Yang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Ning Yang
- The Fifth Department of Hepatic Surgery, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Wei-ping Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Guang-shun Yang
- The Fifth Department of Hepatic Surgery, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Shu-han Sun
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
110
|
Takahashi K, Yan IK, Kogure T, Haga H, Patel T. Extracellular vesicle-mediated transfer of long non-coding RNA ROR modulates chemosensitivity in human hepatocellular cancer. FEBS Open Bio 2014; 4:458-67. [PMID: 24918061 PMCID: PMC4050189 DOI: 10.1016/j.fob.2014.04.007] [Citation(s) in RCA: 363] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 04/04/2014] [Accepted: 04/22/2014] [Indexed: 12/28/2022] Open
Abstract
Selected long non-coding RNA are aberrantly expressed in hepatocellular cancers. linc-ROR is a highly upregulated lncRNA that is expressed in response to TGFβ. linc-ROR contributes to chemoresistance of HCC cells. Intercellular transfer of linc-ROR occurs within extracellular vesicles.
Hepatocellular cancers (HCC) are highly resistant to chemotherapy. TGFβ has been associated with chemoresistance in some human cancers but the mechanisms involved are unknown. We explored how TGFβ might contribute to altered responses to therapy by assessing the involvement and mechanistic contribution of extracellular vesicle long non-coding RNA (lncRNA) in mediating TGFβ-dependent chemoresistance. TGFβ reduced the sensitivity of HCC cells to sorafenib or doxorubicin and altered the release of both extracellular vesicles and of selected lncRNA within these vesicles. Amongst these, lincRNA-ROR (linc-ROR), a stress-responsive lncRNA was highly expressed in HCC cells and enriched within extracellular vesicles derived from tumor cells. Incubation with HCC-derived extracellular vesicles increased linc-ROR expression and reduced chemotherapy-induced cell death in recipient cells. Sorafenib increased linc-ROR expression in both tumor cells and extracellular vesicles, whereas siRNA to linc-ROR increased chemotherapy-induced apoptosis and cytotoxicity. Tumor-initiating cells that express CD133 have an increased resistance to therapy. TGFβ increased expression of CD133+ cells and colony growth in limiting dilution assays, both of which were attenuated by linc-ROR knockdown. These data provide mechanistic insights into primary chemoresistance in HCC by showing that: (a) TGFβ selectively enriches linc-RoR within extracellular vesicles, which has a potential role in intercellular signaling in response to TGFβ; (b) expression and enrichment of linc-ROR during chemotherapeutic stress plays a functional role in chemoresistance; and (c) the effects of TGFβ on chemoresistance in HCC may involve linc-RoR-dependent effects on tumor-initiating cells. These findings implicate extracellular vesicle lncRNA as mediators of the chemotherapeutic response, and support targeting linc-ROR to enhance chemosensitivity in HCC.
Collapse
Affiliation(s)
- Kenji Takahashi
- Department of Transplantation, Mayo Clinic, Jacksonville, FL, United States
| | - Irene K Yan
- Department of Transplantation, Mayo Clinic, Jacksonville, FL, United States
| | - Takayuki Kogure
- Department of Transplantation, Mayo Clinic, Jacksonville, FL, United States
| | - Hiroaki Haga
- Department of Transplantation, Mayo Clinic, Jacksonville, FL, United States
| | - Tushar Patel
- Department of Transplantation, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
111
|
Chu TH, Chan HH, Kuo HM, Liu LF, Hu TH, Sun CK, Kung ML, Lin SW, Wang EM, Ma YL, Cheng KH, Lai KH, Wen ZH, Hsu PI, Tai MH. Celecoxib suppresses hepatoma stemness and progression by up-regulating PTEN. Oncotarget 2014; 5:1475-1490. [PMID: 24721996 PMCID: PMC4039225 DOI: 10.18632/oncotarget.1745] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 12/28/2013] [Indexed: 12/13/2022] Open
Abstract
Celecoxib, a COX-2 inhibitor and non-steroidal anti-inflammatory drug, can prevent several types of cancer, including hepatocellular carcinoma (HCC). Here we show that celecoxib suppressed the self-renewal and drug-pumping functions in HCC cells. Besides, celecoxib depleted CD44+/CD133+ hepatic cancer stem cells (hCSC). Prostaglandin E2 (PGE2) and CD133 overexpression did not reverse the celecoxib-induced depletion of hCSC. Also, celecoxib inhibited progression of rat Novikoff hepatoma. Moreover, a 60-day celecoxib program increased the survival rate of rats with hepatoma. Histological analysis revealed that celecoxib therapy reduced the abundance of CD44+/CD133+ hCSCs in hepatoma tissues. Besides, the hCSCs depletion was associated with elevated apoptosis and blunted proliferation and angiogenesis in hepatoma. Celecoxib therapy activated peroxisome proliferator-activated receptor γ (PPARγ) and up-regulated PTEN, thereby inhibiting Akt and disrupting hCSC expansion. PTEN gene delivery by adenovirus reduced CD44/CD133 expression in vitro and hepatoma formation in vivo. This study suggests that celecoxib suppresses cancer stemness and progression of HCC via activation of PPARγ/PTEN signaling.
Collapse
Affiliation(s)
- Tian-Huei Chu
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Hoi-Hung Chan
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- College of Pharmacy & Health Care, Tajen University, Pingtung County, Taiwan
| | - Hsiao-Mei Kuo
- Mitochondrial Research Unit, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Li-Fen Liu
- Department of Biological Science and Technology, I-Shou University, Kaohsiung, Taiwan
| | - Tsung-Hui Hu
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Chang Gung Memorial Hospital Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Cheuk-Kwan Sun
- Department of Medical Education, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Mei-Lang Kung
- Department of Chemistry, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Shih-Wei Lin
- Institute of Marine Biotechnology, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - E-Ming Wang
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Yi-Ling Ma
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Kwan-Hung Cheng
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Kwok Hung Lai
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, Asia-Pacific Ocean Research Center, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Ping-I Hsu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ming-Hong Tai
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Center for Neuroscience, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| |
Collapse
|
112
|
Parent R, Plissonnier ML, Bancel B, Liao WL, Rumin S, Asaad R, Till M, Sanlaville D, Zoulim F, Trépo C, Marion MJ. Diversity of Hepatocellular Carcinoma Clones Bearing Hematopoietic Malignancies-Related Chromosomal Translocation. J Cell Biochem 2014; 115:666-77. [DOI: 10.1002/jcb.24706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 10/23/2013] [Indexed: 01/05/2023]
Affiliation(s)
- Romain Parent
- DevWeCan Laboratories of Excellence Network (Labex); Université de Lyon, Lyon Cancer Research Center; Inserm U1052-CNRS UMR5286; Lyon France
| | - Marie-Laure Plissonnier
- DevWeCan Laboratories of Excellence Network (Labex); Université de Lyon, Lyon Cancer Research Center; Inserm U1052-CNRS UMR5286; Lyon France
| | - Brigitte Bancel
- Laboratoire d'Anatomie Pathologique; Hôpital de la Croix-Rousse; Lyon France
| | - Wan-Li Liao
- DevWeCan Laboratories of Excellence Network (Labex); Université de Lyon, Lyon Cancer Research Center; Inserm U1052-CNRS UMR5286; Lyon France
| | - Sylvie Rumin
- DevWeCan Laboratories of Excellence Network (Labex); Université de Lyon, Lyon Cancer Research Center; Inserm U1052-CNRS UMR5286; Lyon France
| | - Remal Asaad
- DevWeCan Laboratories of Excellence Network (Labex); Université de Lyon, Lyon Cancer Research Center; Inserm U1052-CNRS UMR5286; Lyon France
| | - Marianne Till
- Laboratoire de Cytogénétique; Hôpital Edouard Herriot; Lyon France
| | | | - Fabien Zoulim
- DevWeCan Laboratories of Excellence Network (Labex); Université de Lyon, Lyon Cancer Research Center; Inserm U1052-CNRS UMR5286; Lyon France
| | - Christian Trépo
- DevWeCan Laboratories of Excellence Network (Labex); Université de Lyon, Lyon Cancer Research Center; Inserm U1052-CNRS UMR5286; Lyon France
| | - Marie-Jeanne Marion
- DevWeCan Laboratories of Excellence Network (Labex); Université de Lyon, Lyon Cancer Research Center; Inserm U1052-CNRS UMR5286; Lyon France
| |
Collapse
|
113
|
Hung WL, Tsai ML, Sun PP, Tsai CY, Yang CC, Ho CT, Cheng AC, Pan MH. Protective effects of garcinol on dimethylnitrosamine-induced liver fibrosis in rats. Food Funct 2014; 5:2883-91. [DOI: 10.1039/c4fo00342j] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Garcinol markedly reduced DMN-induced liver fibrosis in rats.
Collapse
Affiliation(s)
- Wei-Lun Hung
- Institute of Food Science and Technology
- National Taiwan University
- Taipei 10617, Taiwan
| | - Mei-Ling Tsai
- Department of Seafood Science
- National Kaohsiung Marine University
- Kaohsiung 811, Taiwan
| | - Pei-Pei Sun
- Department of Seafood Science
- National Kaohsiung Marine University
- Kaohsiung 811, Taiwan
| | - Chen-Yu Tsai
- Institute of Food Science and Technology
- National Taiwan University
- Taipei 10617, Taiwan
| | - Chin-Chou Yang
- Department of Seafood Science
- National Kaohsiung Marine University
- Kaohsiung 811, Taiwan
| | - Chi-Tang Ho
- Department of Food Science
- Rutgers University
- New Brunswick, USA
| | - An-Chin Cheng
- Department of Nutrition and Health Sciences
- Chang Jung Christian University
- Tainan 71101, Taiwan
| | - Min-Hsiung Pan
- Institute of Food Science and Technology
- National Taiwan University
- Taipei 10617, Taiwan
- Department of Medical Research
- China Medical University Hospital
| |
Collapse
|
114
|
Svinka J, Mikulits W, Eferl R. STAT3 in hepatocellular carcinoma: new perspectives. Hepat Oncol 2014; 1:107-120. [PMID: 30190945 PMCID: PMC6114013 DOI: 10.2217/hep.13.7] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Chronic liver damage and inflammation are strong promoters of hepatocellular carcinoma (HCC) formation. HCC cells communicate with inflammatory and stromal cells via cytokine/chemokine signals. These heterotypic interactions inhibit immunologic anticancer activities and promote protumorigenic activities, such as angiogenesis or invasiveness. STAT3 mediates several reciprocal interactions between liver cancer cells and stromal cells and modulates preconditions of tumor formation such as chronic inflammation. Therefore, activation of STAT3 is considered as a tumor-promoting event in HCC formation. However, the oncogenic role of STAT3 in cancers has been challenged by several reports that suggest a tumor-suppressive activity. Here we discuss tumor-promoting and tumor-suppressive effects of cytokine-activated STAT3 in HCC.
Collapse
Affiliation(s)
- Jasmin Svinka
- Medical University Vienna & Comprehensive Cancer Center, Institute for Cancer Research, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Wolfgang Mikulits
- Medical University Vienna & Comprehensive Cancer Center, Institute for Cancer Research, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Robert Eferl
- Medical University Vienna & Comprehensive Cancer Center, Institute for Cancer Research, Borschkegasse 8a, A-1090 Vienna, Austria
| |
Collapse
|
115
|
Miao CG, Yang YY, He X, Huang C, Huang Y, Zhang L, Lv XW, Jin Y, Li J. Wnt signaling in liver fibrosis: progress, challenges and potential directions. Biochimie 2013; 95:2326-35. [PMID: 24036368 DOI: 10.1016/j.biochi.2013.09.003] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 09/02/2013] [Indexed: 12/25/2022]
Abstract
Liver fibrosis is a common wound-healing response to chronic liver injuries, including alcoholic or drug toxicity, persistent viral infection, and genetic factors. Myofibroblastic transdifferentiation (MTD) is the pivotal event during liver fibrogenesis, and research in the past few years has identified key mediators and molecular mechanisms responsible for MTD of hepatic stellate cells (HSCs). HSCs are undifferentiated cells which play an important role in liver regeneration. Recent evidence demonstrates that HSCs derive from mesoderm and at least in part via septum transversum and mesothelium, and HSCs express markers for different cell types which derive from multipotent mesenchymal progenitors. There is a regulatory commonality between differentiation of adipocytes and that of HSC, and the shift from adipogenic to myogenic or neuronal phenotype characterizes HSC MTD. Central of this shift is a loss of expression of the master adipogenic regulator peroxisome proliferator activated receptor γ (PPARγ). Restored expression of PPARγ and/or other adipogenic transcription genes can reverse myofibroblastic HSCs to differentiated cells. Vertebrate Wnt and Drosophila wingless are homologous genes, and their translated proteins have been shown to participate in the regulation of cell proliferation, cell polarity, cell differentiation, and other biological roles. More recently, Wnt signaling is implicated in human fibrosing diseases, such as pulmonary fibrosis, renal fibrosis, and liver fibrosis. Blocking the canonical Wnt signal pathway with the co-receptor antagonist Dickkopf-1 (DKK1) abrogates these epigenetic repressions and restores the gene PPARγ expression and HSC differentiation. The identified morphogen mediated epigenetic regulation of PPARγ and HSC differentiation also serves as novel therapeutic targets for liver fibrosis and liver regeneration. In conclusion, the Wnt signaling promotes liver fibrosis by enhancing HSC activation and survival, and we herein discuss what we currently know and what we expect will come in this field in the next future.
Collapse
Affiliation(s)
- Cheng-gui Miao
- School of Pharmacy, Institute for Liver Diseases of Anhui Medical University, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Mei Shan Road, Hefei 230032, Anhui Province, China; School of Food and Drug, Anhui Science and Technology University, Bengbu 233100, China
| | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Dzieran J, Fabian J, Feng T, Coulouarn C, Ilkavets I, Kyselova A, Breuhahn K, Dooley S, Meindl-Beinker NM. Comparative analysis of TGF-β/Smad signaling dependent cytostasis in human hepatocellular carcinoma cell lines. PLoS One 2013; 8:e72252. [PMID: 23991075 PMCID: PMC3750029 DOI: 10.1371/journal.pone.0072252] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 07/11/2013] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a major public health problem due to increased incidence, late diagnosis and limited treatment options. TGF-β is known to provide cytostatic signals during early stages of liver damage and regeneration, but exerts tumor promoting effects in onset and progression of liver cancer. To understand the mechanistic background of such a switch, we systematically correlated loss of cytostatic TGF-β effects with strength and dynamics of its downstream signaling in 10 HCC cell lines. We demonstrate that TGF-β inhibits proliferation and induces apoptosis in cell lines with low endogenous levels of TGF-β and Smad7 and strong transcriptional Smad3 activity (PLC/PRF/5, HepG2, Hep3B, HuH7), previously characterized to express early TGF-β signatures correlated with better outcome in HCC patients. TGF-β dependent cytostasis is blunted in another group of cell lines (HLE, HLF, FLC-4) expressing high amounts of TGF-β and Smad7 and showing significantly reduced Smad3 signaling. Of those, HLE and HLF exhibit late TGF-β signatures, which is associated with bad prognosis in HCC patients. RNAi with Smad3 blunted cytostatic effects in PLC/PRF/5, Hep3B and HuH7. HCC-M and HCC-T represent a third group of cell lines lacking cytostatic TGF-β signaling despite strong and prolonged Smad3 phosphorylation and low Smad7 and TGF-β expression. Inhibitory linker phosphorylation, as in HCC-T, may disrupt C-terminally phosphorylated Smad3 function. In summary, we assort 10 HCC cell lines in at least two clusters with respect to TGF-β sensitivity. Cell lines responsive to the TGF-β cytostatic program, which recapitulate early stage of liver carcinogenesis exhibit transcriptional Smad3 activity. Those with disturbed TGF-β/Smad3 signaling are insensitive to TGF-β dependent cytostasis and might represent late stage of the disease. Regulation of this switch remains complex and cell line specific. These features may be relevant to discriminate stage dependent TGF-β functions for the design of efficient TGF-β directed therapy in liver cancer.
Collapse
Affiliation(s)
- Johanna Dzieran
- Molecular Hepatology – Alcohol Associated Diseases, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jasmin Fabian
- Molecular Hepatology – Alcohol Associated Diseases, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Teng Feng
- Molecular Hepatology – Alcohol Associated Diseases, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Cédric Coulouarn
- Institut National de la Sante et de la recherche Medicale UMR991, University of Rennes, Pontchaillou University Hospital, Rennes, France
| | - Iryna Ilkavets
- Molecular Hepatology – Alcohol Associated Diseases, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Anastasia Kyselova
- Molecular Hepatology – Alcohol Associated Diseases, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kai Breuhahn
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Steven Dooley
- Molecular Hepatology – Alcohol Associated Diseases, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Nadja M. Meindl-Beinker
- Molecular Hepatology – Alcohol Associated Diseases, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
117
|
Wang L, Guo ZY, Zhang R, Xin B, Chen R, Zhao J, Wang T, Wen WH, Jia LT, Yao LB, Yang AG. Pseudogene OCT4-pg4 functions as a natural micro RNA sponge to regulate OCT4 expression by competing for miR-145 in hepatocellular carcinoma. Carcinogenesis 2013; 34:1773-1781. [PMID: 23615404 DOI: 10.1093/carcin/bgt139] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The POU transcription factor OCT4 is a pleiotropic regulator of gene expression in embryonic stem cells. Recent studies demonstrated that OCT4 is aberrantly expressed in multiple types of human cancer; however, the underlying molecular mechanism remains largely unknown. In this study, we report that OCT4-pg4, a pseudogene of OCT4, is abnormally activated in hepatocellular carcinoma (HCC). The expression level of OCT4-pg4 is positively correlated with that of OCT4, and both gene transcripts can be directly targeted by a tumor-suppressive micro RNA miR-145. We find that the non-coding RNA OCT4-pg4 is biologically active, as it can upregulate OCT4 protein level in HCC. Mechanistic analysis revealed that OCT4-pg4 functions as a natural micro RNA sponge to protect OCT4 transcript from being inhibited by miR-145. In addition, our study also showed that OCT4-pg4 can promote growth and tumorigenicity of HCC cells, thus exerting an oncogenic role in hepatocarcinogenesis. Furthermore, survival analysis suggests that high OCT4-pg4 level is significantly correlated with poor prognosis of HCC patients. Taken together, our finding adds a new layer of post-transcriptional regulation of OCT4 and sheds new light on the treatment of human HCC.
Collapse
Affiliation(s)
- Lei Wang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Yang L, Inokuchi S, Roh YS, Song J, Loomba R, Park EJ, Seki E. Transforming growth factor-β signaling in hepatocytes promotes hepatic fibrosis and carcinogenesis in mice with hepatocyte-specific deletion of TAK1. Gastroenterology 2013; 144:1042-1054.e4. [PMID: 23391818 PMCID: PMC3752402 DOI: 10.1053/j.gastro.2013.01.056] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 01/07/2013] [Accepted: 01/24/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Transforming growth factor (TGF)-β-activated kinase 1 (TAK1) is activated in different cytokine signaling pathways. Deletion of Tak1 from hepatocytes results in spontaneous development of hepatocellular carcinoma (HCC), liver inflammation, and fibrosis. TGF-β activates TAK1 and Smad signaling, which regulate cell death, proliferation, and carcinogenesis. However, it is not clear whether TGF-β signaling in hepatocytes, via TGF-β receptor-2 (Tgfbr2), promotes HCC and liver fibrosis. METHODS We generated mice with hepatocyte-specific deletion of Tak1 (Tak1ΔHep), as well as Tak1/Tgfbr2DHep and Tak1/Smad4ΔHep mice. Tak1flox/flox, Tgfbr2ΔHep, and Smad4ΔHep mice were used as controls, respectively. We assessed development of liver injury, inflammation, fibrosis, and HCC. Primary hepatocytes isolated from these mice were used to assess TGF-β-mediated signaling. RESULTS Levels of TGF-β, TGF-βR2, and phospho-Smad2/3 were increased in HCCs from Tak1ΔHep mice, which developed liver fibrosis and inflammation by 1 month and HCC by 9 months. However, Tak1/Tgfbr2ΔHep mice did not have this phenotype, and their hepatocytes did not undergo spontaneous cell death or compensatory proliferation. Hepatocytes from Tak1ΔHep mice incubated with TGF-β did not activate p38, c-Jun N-terminal kinase, or nuclear factor-κB; conversely, TGF-β-mediated cell death and phosphorylation of Smad2/3 were increased, compared with control hepatocytes. Blocking the Smad pathway inhibited TGF-β-mediated death of Tak1-/- hepatocytes. Accordingly, disruption of Smad4 reduced the spontaneous liver injury, inflammation, fibrosis, and HCC that develops in Tak1ΔHep mice. Levels of the anti-apoptotic protein Bcl-xL, β-catenin, connective tissue growth factor, and vascular endothelial growth factor were increased in HCC from Tak1ΔHep mice, but not in HCCs from Tak1/Tgfbr2ΔHep mice. Injection of N-nitrosodiethylamine induced HCC formation in wild-type mice, but less in Tgfbr2ΔHep mice. CONCLUSIONS TGF-β promotes development of HCC in Tak1ΔHep mice by inducing hepatocyte apoptosis and compensatory proliferation during early phases of tumorigenesis, and inducing expression of anti-apoptotic, pro-oncogenic, and angiogenic factors during tumor progression.
Collapse
Affiliation(s)
- Ling Yang
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | | | | | | | | | | | | |
Collapse
|
119
|
Abstract
Cancer stem cells (CSCs) drive solid tumor formation. In this issue of Cancer Cell, Zhao and colleagues indentify the calcium channel α2δ1 subunit as a new functional hepatocellular carcinoma (HCC) CSC biomarker, which is vital for CSC biology as blocking α2δ1 in combination with doxorubicin treatment hinders HCC tumor formation.
Collapse
Affiliation(s)
- Bruno Sainz
- Stem Cells and Cancer Group, Molecular Pathology Programme, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | | |
Collapse
|
120
|
Uehara T, Ainslie GR, Kutanzi K, Pogribny IP, Muskhelishvili L, Izawa T, Yamate J, Kosyk O, Shymonyak S, Bradford BU, Boorman GA, Bataller R, Rusyn I. Molecular mechanisms of fibrosis-associated promotion of liver carcinogenesis. Toxicol Sci 2013; 132:53-63. [PMID: 23288052 DOI: 10.1093/toxsci/kfs342] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) mostly develops in patients with advanced fibrosis; however, the mechanisms of interaction between a genotoxic insult and fibrogenesis are not well understood. This study tested a hypothesis that fibrosis promotes HCC via a mechanism that involves activation of liver stem cells. First, B6C3F1 mice were administered diethylnitrosamine (DEN; single ip injection of 1mg/kg at 14 days of age). Second, carbon tetrachloride (CCl(4); 0.2ml/kg, 2/week ip starting at 8 weeks of age) was administered for 9 or 14 weeks to develop advanced liver fibrosis. In animals treated with DEN as neonates, presence of liver fibrosis led to more than doubling (to 100%) of the liver tumor incidence as early as 5 months of age. This effect was associated with activation of cells with progenitor features in noncancerous liver tissue, including markers of replicative senescence (p16), oncofetal transformation (Afp, H19, and Bex1), and increased "stemness" (Prom1 and Epcam). In contrast, the dose of DEN used did not modify the extent of liver inflammation, fibrogenesis, oxidative stress, proliferation, or apoptosis induced by subchronic CCl(4) administration. This study demonstrates the potential role of liver stem-like cells in the mechanisms of chemical-induced, fibrosis-promoted HCC. We posit that the combination of genotoxic and fibrogenic insults is a sensible approach to model liver carcinogenesis in experimental animals. These results may contribute to identification of cirrhotic patients predisposed to HCC by analyzing the expression of hepatic progenitor cell markers in the noncancerous liver tissue.
Collapse
Affiliation(s)
- Takeki Uehara
- Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, NC, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
121
|
Nie J, Liu L, He F, Fu X, Han W, Zhang L. CKIP-1: a scaffold protein and potential therapeutic target integrating multiple signaling pathways and physiological functions. Ageing Res Rev 2013; 12:276-81. [PMID: 22878216 DOI: 10.1016/j.arr.2012.07.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 07/04/2012] [Accepted: 07/11/2012] [Indexed: 11/16/2022]
Abstract
The PH domain-containing casein kinase 2 interacting protein-1 (CKIP-1, also known as PLEKHO1) acts as a scaffold protein mediating interactions with multiple proteins, including CK2α, CPα, AP-1/c-Jun, Akt, ATM, IFP35/Nmi and Smurf1. CKIP-1 functions through different ways, such as plasma membrane recruitment, transcriptional activity modulation and posttranscriptional modification regulation. Moreover, the subcellular localization of CKIP-1 is determined by several key amino acids in a cell type dependent style, and the nucleus/plasma membrane shuttle of CKIP-1 is regulated by different cell stresses. As an adaptor protein, CKIP-1 is involved in various important signaling pathways, controlling cell growth, apoptosis, differentiation, cytoskeleton and bone formation. Strikingly, CKIP-1 has been recently demonstrated to be a promising target for treatment of osteoporosis in rat models. In addition, more evidences suggest that CKIP-1 might also function as a potential tumor suppressor.
Collapse
Affiliation(s)
- Jing Nie
- Department of Molecular Biology, Institute of Basic Medical Science, PLA General Hospital, Beijing, China
| | | | | | | | | | | |
Collapse
|
122
|
Dzieran J, Fabian J, Feng T, Coulouarn C, Ilkavets I, Kyselova A, Breuhahn K, Dooley S, Meindl-Beinker NM. Comparative analysis of TGF-β/Smad signaling dependent cytostasis in human hepatocellular carcinoma cell lines. PLoS One 2013. [PMID: 23991075 DOI: 10.1371/journal.pone.0072252.erratum.in:plosone.2014;9(5):e95952.pmid:23991075;pmcid:pmc3750029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a major public health problem due to increased incidence, late diagnosis and limited treatment options. TGF-β is known to provide cytostatic signals during early stages of liver damage and regeneration, but exerts tumor promoting effects in onset and progression of liver cancer. To understand the mechanistic background of such a switch, we systematically correlated loss of cytostatic TGF-β effects with strength and dynamics of its downstream signaling in 10 HCC cell lines. We demonstrate that TGF-β inhibits proliferation and induces apoptosis in cell lines with low endogenous levels of TGF-β and Smad7 and strong transcriptional Smad3 activity (PLC/PRF/5, HepG2, Hep3B, HuH7), previously characterized to express early TGF-β signatures correlated with better outcome in HCC patients. TGF-β dependent cytostasis is blunted in another group of cell lines (HLE, HLF, FLC-4) expressing high amounts of TGF-β and Smad7 and showing significantly reduced Smad3 signaling. Of those, HLE and HLF exhibit late TGF-β signatures, which is associated with bad prognosis in HCC patients. RNAi with Smad3 blunted cytostatic effects in PLC/PRF/5, Hep3B and HuH7. HCC-M and HCC-T represent a third group of cell lines lacking cytostatic TGF-β signaling despite strong and prolonged Smad3 phosphorylation and low Smad7 and TGF-β expression. Inhibitory linker phosphorylation, as in HCC-T, may disrupt C-terminally phosphorylated Smad3 function. In summary, we assort 10 HCC cell lines in at least two clusters with respect to TGF-β sensitivity. Cell lines responsive to the TGF-β cytostatic program, which recapitulate early stage of liver carcinogenesis exhibit transcriptional Smad3 activity. Those with disturbed TGF-β/Smad3 signaling are insensitive to TGF-β dependent cytostasis and might represent late stage of the disease. Regulation of this switch remains complex and cell line specific. These features may be relevant to discriminate stage dependent TGF-β functions for the design of efficient TGF-β directed therapy in liver cancer.
Collapse
Affiliation(s)
- Johanna Dzieran
- Molecular Hepatology - Alcohol Associated Diseases, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Muñoz P, Iliou MS, Esteller M. Epigenetic alterations involved in cancer stem cell reprogramming. Mol Oncol 2012; 6:620-36. [PMID: 23141800 PMCID: PMC5528346 DOI: 10.1016/j.molonc.2012.10.006] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 10/17/2012] [Indexed: 02/07/2023] Open
Abstract
Current hypotheses suggest that tumors originate from cells that carry out a process of "malignant reprogramming" driven by genetic and epigenetic alterations. Multiples studies reported the existence of stem-cell-like cells that acquire the ability to self-renew and are able to generate the bulk of more differentiated cells that form the tumor. This population of cancer cells, called cancer stem cells (CSC), is responsible for sustaining the tumor growth and, under determined conditions, can disseminate and migrate to give rise to secondary tumors or metastases to distant organs. Furthermore, CSCs have shown to be more resistant to anti-tumor treatments than the non-stem cancer cells, suggesting that surviving CSCs could be responsible for tumor relapse after therapy. These important properties have raised the interest in understanding the mechanisms that govern the generation and maintenance of this special population of cells, considered to lie behind the on/off switches of gene expression patterns. In this review, we summarize the most relevant epigenetic alterations, from DNA methylation and histone modifications to the recently discovered miRNAs that contribute to the regulation of cancer stem cell features in tumor progression, metastasis and response to chemotherapy.
Collapse
Affiliation(s)
- Purificación Muñoz
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | | | | |
Collapse
|