201
|
Wang G, Zhu X, Hood L, Ao P. From Phage lambda to human cancer: endogenous molecular-cellular network hypothesis. QUANTITATIVE BIOLOGY 2013. [DOI: 10.1007/s40484-013-0007-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
202
|
Zhu C, Cheng KW, Ouyang N, Huang L, Sun Y, Constantinides P, Rigas B. Phosphosulindac (OXT-328) selectively targets breast cancer stem cells in vitro and in human breast cancer xenografts. Stem Cells 2013; 30:2065-75. [PMID: 22653497 DOI: 10.1002/stem.1139] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pharmacological targeting of breast cancer stem cells (CSCs) is highly promising for the treatment of breast cancer, as the small population of CSCs appears responsible for tumor initiation and progression and also for resistance to conventional treatment. Here we report that the novel phosphosulindac (OXT-328, PS) selectively and effectively eliminates breast CSCs both in vitro and in vivo. PS reduced cell proliferation and induced apoptosis in various breast CSCs. Breast CSCs are resistant to conventional cancer drugs but are sensitive to PS. Long-term treatment of mixtures of cultured breast CSCs and breast cancer cells with PS preferentially eliminated the CSCs. PS impaired the ability of CSCs to form mammospheres and markedly suppressed the expression of CSC-related genes. More importantly, PS prevented by half (p = .06) the formation of tumors initiated by CSCs in immunodeficient mice, and inhibited by 83% (p < .05) the growth of already formed breast cancer xenografts, reducing the proportion of CSCs in them. PS suppressed the Wnt/β-catenin pathway by stimulating the degradation of β-catenin and its relocalization to the cell membrane and also blocked the epithelial-mesenchymal transition and the generation of breast CSCs. These results indicate that PS has a strong inhibitory effect against breast cancer, acting, at least in part, by targeting CSCs through a signaling mechanism involving Wnt signaling.
Collapse
Affiliation(s)
- Caihua Zhu
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, New York 11794-8173, USA
| | | | | | | | | | | | | |
Collapse
|
203
|
Abdullah LN, Chow EKH. Mechanisms of chemoresistance in cancer stem cells. Clin Transl Med 2013; 2:3. [PMID: 23369605 PMCID: PMC3565873 DOI: 10.1186/2001-1326-2-3] [Citation(s) in RCA: 554] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 01/11/2013] [Indexed: 12/13/2022] Open
Abstract
Chemotherapy is one of the standard methods of treatment in many cancers. While chemotherapy is often capable of inducing cell death in tumors and reducing the tumor bulk, many cancer patients experience recurrence and ultimately death because of treatment failure. In recent years, cancer stem cells (CSCs) have gained intense interest as key tumor-initiating cells that may also play an integral role in recurrence following chemotherapy. As such, a number of mechanisms of chemoresistance have been identified in CSCs. In this review, we describe a number of these mechanisms of chemoresistance including ABC transporter expression, aldehyde dehydrogenase (ALDH) activity, B-cell lymphoma-2 (BCL2) related chemoresistance, enhanced DNA damage response and activation of key signaling pathways. Furthermore, we evaluate studies that demonstrate potential methods for overcoming chemoresistance and treating chemoresistant cancers that are driven by CSCs. By understanding how tumor-initiating cells such as CSCs escape chemotherapy, more informed approaches to treating cancer will develop and may improve clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Lissa Nurrul Abdullah
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive #12-01, Singapore, 117599, Singapore.
| | | |
Collapse
|
204
|
Abstract
Hepatocellular carcinoma (HCC) is the most common primary hepatic malignancy. Its incidence and prevalence is globally heterogeneous with the highest rates in Southeast Asia and Sub-Saharan Africa. In Western Industry nations, its incidence has significantly increased throughout the previous three decades. Its global heterogeneity is in part a reflection of the global distribution of its risk factors. Its prognosis is dismal with a 5-year survival of 11 %. The only potentially curative treatment is surgical with either resection or orthotopic liver transplantation. However, the majority of HCC patients are diagnosed at an advanced stage at which surgical therapies are not feasible. HCC is considered chemotherapy-resistant-a characteristic thought to be mediated in part through stem-like tumor initiating cells (STICs). Recent studies have provided significant insights in the hepatocarcinogenesis and the molecular signaling pathways of this malignancy resulting in the development of novel, molecular targeted therapies with modest therapeutic benefit. Our growing understanding of the biology of this malignancy will help in the development of novel, molecular-targeted therapies.
Collapse
Affiliation(s)
- Boris Blechacz
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | |
Collapse
|
205
|
Liver Stem Cells. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
206
|
Lan X, Wu YZ, Wang Y, Wu FR, Zang CB, Tang C, Cao S, Li SL. CD133 silencing inhibits stemness properties and enhances chemoradiosensitivity in CD133-positive liver cancer stem cells. Int J Mol Med 2012; 31:315-24. [PMID: 23233126 DOI: 10.3892/ijmm.2012.1208] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 09/04/2012] [Indexed: 12/27/2022] Open
Abstract
Cancer stem cells (CSCs) are considered the source of the initial tumor formation and postoperative recurrence and metastasis. CD133(+) cells in hepatocellular carcinoma (HCC) display cancer stem-like properties and are thought to be responsible for chemoradioresistance. To explore the functional role of CD133 in liver cancer stem cells (LCSCs), we isolated CD133(+) cells from the HCC cell line HepG2, which were tested and confirmed to be CSC-like cells in HCC, downregulated CD133 expression in HepG2-CD133(+) cells by lentivirus-mediated short hairpin (shRNA) and analyzed the effects of CD133 on the modulation of stemness properties and chemoradiosensitivity in LCSCs. Our results showed that the in vitro cell proliferation, tumorsphere formation, colony formation and in vivo tumor growth in NOD/SCID mouse xenografts of LCSCs were significantly repressed after CD133 silencing. We also found that suppression of CD133 enhances the sensitivity of LCSCs to chemotherapy and radiotherapy. Knockdown of CD133 reduced G0/G1 phase cells and increased cellular apoptosis via modulation of Bcl-2 and Bax. Collectively, the stem-targeted therapy via CD133 could provide a novel strategy for the treatment of HCC.
Collapse
Affiliation(s)
- Xi Lan
- Department of Radiology, Chongqing Medical University, Chongqing, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
207
|
Wu K, Ding J, Chen C, Sun W, Ning BF, Wen W, Huang L, Han T, Yang W, Wang C, Li Z, Wu MC, Feng GS, Xie WF, Wang HY. Hepatic transforming growth factor beta gives rise to tumor-initiating cells and promotes liver cancer development. Hepatology 2012; 56:2255-67. [PMID: 22898879 DOI: 10.1002/hep.26007] [Citation(s) in RCA: 165] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 07/07/2012] [Indexed: 12/16/2022]
Abstract
UNLABELLED Liver cirrhosis is a predominant risk factor for hepatocellular carcinoma (HCC). However, the mechanism underlying the progression from cirrhosis to HCC remains unclear. Herein we report the concurrent increase of liver progenitor cells (LPCs) and transforming growth factor-β (TGF-β) in diethylnitrosamine (DEN)-induced rat hepatocarcinogenesis and cirrhotic livers of HCC patients. Using several experimental approaches, including 2-acetylaminofluorene/partial hepatectomy (2-AAF/PHx) and 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-elicited murine liver regeneration, we found that activation of LPCs in the absence of TGF-β induction was insufficient to trigger hepatocarcinogenesis. Moreover, a small fraction of LPCs was detected to coexpress tumor initiating cell (T-IC) markers during rat hepatocarcinogenesis and in human HCCs, and TGF-β levels were positively correlated with T-IC marker expression, which indicates a role of TGF-β in T-IC generation. Rat pluripotent LPC-like WB-F344 cells were exposed to low doses of TGF-β for 18 weeks imitating the enhanced TGF-β expression in cirrhotic liver. Interestingly, long-term treatment of TGF-β on WB-F344 cells impaired their LPC potential but granted them T-IC properties including expression of T-IC markers, increased self-renewal capacity, stronger chemoresistance, and tumorigenicity in NOD-SCID mice. Hyperactivation of Akt but not Notch, signal transducer and activator of transcription 3 (STAT3), or mammalian target of rapamycin (mTOR) was detected in TGF-β-treated WB-F344 cells. Introduction of the dominant-negative mutant of Akt significantly attenuated T-IC properties of those transformed WB-F344 cells, indicating Akt was required in TGF-β-mediated-generation of hepatic T-ICs. We further demonstrate that TGF-β-induced Akt activation and LPC transformation was mediated by microRNA-216a-modulated phosphatase and tensin homolog deleted on chromosome 10 (PTEN) suppression. CONCLUSION Hepatoma-initiating cells may derive from hepatic progenitor cells exposed to chronic and constant TGF-β stimulation in cirrhotic liver, and pharmaceutical inhibition of microRNA-216a/PTEN/Akt signaling could be a novel strategy for HCC prevention and therapy targeting hepatic T-ICs.
Collapse
Affiliation(s)
- Kun Wu
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
208
|
Mavila N, James D, Utley S, Cu N, Coblens O, Mak K, Rountree CB, Kahn M, Wang KS. Fibroblast growth factor receptor-mediated activation of AKT-β-catenin-CBP pathway regulates survival and proliferation of murine hepatoblasts and hepatic tumor initiating stem cells. PLoS One 2012; 7:e50401. [PMID: 23308088 PMCID: PMC3540100 DOI: 10.1371/journal.pone.0050401] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 10/19/2012] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Fibroblast Growth Factor (FGF)-10 promotes the proliferation and survival of murine hepatoblasts during early stages of hepatogenesis through a Wnt-β-catenin dependent pathway. To determine the mechanism by which this occurs, we expanded primary culture of hepatoblasts enriched for progenitor markers CD133 and CD49f from embryonic day (E) 12.5 fetal liver and an established tumor initiating stem cell line from Mat1a(-/-) livers in media conditioned with recombinant (r) FGF10 or rFGF7. FGF Receptor (R) activation resulted in the downstream activation of MAPK, PI3K-AKT, and β-catenin pathways, as well as cellular proliferation. Additionally, increased levels of nuclear β-catenin phosphorylated at Serine-552 in cultured primary hepatoblasts, Mat1a(-/-) cells, and also in ex vivo embryonic liver explants indicate AKT-dependent activation of β-catenin downstream of FGFR activation; conversely, the addition of AKT inhibitor Ly294002 completely abrogated β-catenin activation. FGFR activation-induced cell proliferation and survival were also inhibited by the compound ICG-001, a small molecule inhibitor of β-catenin-CREB Binding Protein (CBP) in hepatoblasts, further indicating a CBP-dependent regulatory mechanism of β-catenin activity. CONCLUSION FGF signaling regulates the proliferation and survival of embryonic and transformed progenitor cells in part through AKT-mediated activation of β-catenin and downstream interaction with the transcriptional co-activator CBP.
Collapse
Affiliation(s)
- Nirmala Mavila
- Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, California, United States of America
| | - David James
- Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, California, United States of America
| | - Sarah Utley
- Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, California, United States of America
| | - Nguyen Cu
- Department of Biochemistry and Molecular Biology and Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Orly Coblens
- Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, California, United States of America
| | - Katrina Mak
- Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, California, United States of America
| | - C. Bart Rountree
- Pediatric Gastroenterology, Bon Secours St. Mary’s Hospital, Richmond, Virginia, United States of America
| | - Michael Kahn
- Department of Biochemistry and Molecular Biology and Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Kasper S. Wang
- Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
209
|
Liu WH, You N, Zhang N, Yan HT, Wang T, Huang Z, Liu HB, Tang LJ. Interpretation of interlocking key issues of cancer stem cells in malignant solid tumors. Cell Oncol (Dordr) 2012. [PMID: 23179790 DOI: 10.1007/s13402-012-0110-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE In this review, several interlinking issues related to cancer stem cells (CSCs) in malignant solid tumors are sequentially discussed. METHODS A literature search was performed using PubMed, Web of Science and the Cochrane library, combining the words CSCs, solid tumor, isolation, identification, origination, therapy, target and epithelial-mesenchymal transition. RESULTS Because a primary problem is the isolation of CSCs, we first analyzed the advantages and disadvantages of recently used methods, which were mostly based on the physical or immunochemical characteristics of CSCs. Once CSCs are isolated, they should be identified by their stem cell properties. Here, we suggest how to establish a standard identification strategy. We also focused on the origination hypotheses of CSCs. The supporting molecular mechanisms for each theory were thoroughly analyzed and integrated. Especially, epithelial- mesenchymal transition is an increasingly recognized mechanism to generate CSCs that are endowed with a more invasive and metastatic phenotype. Finally, we discuss putative strategies of eliminating CSCs as effective cancer therapies. CONCLUSION After several interlocking issues of CSCs are thoroughly clarified, these CSCs in solid malignant tumors may specifically be targeted, which raises a new hope for eliminating these tumors.
Collapse
Affiliation(s)
- Wei-Hui Liu
- PLA Center of General Surgery, General Hospital of Chengdu Army Region, Chengdu, Sichuan Province, China
| | | | | | | | | | | | | | | |
Collapse
|
210
|
Tsai ST, Tsou CC, Mao WY, Chang WC, Han HY, Hsu WL, Li CL, Shen CN, Chen CH. Label-free quantitative proteomics of CD133-positive liver cancer stem cells. Proteome Sci 2012; 10:69. [PMID: 23170877 PMCID: PMC3576254 DOI: 10.1186/1477-5956-10-69] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 11/02/2012] [Indexed: 01/15/2023] Open
Abstract
UNLABELLED BACKGROUND CD133-positive liver cancer stem cells, which are characterized by their resistance to conventional chemotherapy and their tumor initiation ability at limited dilutions, have been recognized as a critical target in liver cancer therapeutics. In the current work, we developed a label-free quantitative method to investigate the proteome of CD133-positive liver cancer stem cells for the purpose of identifying unique biomarkers that can be utilized for targeting liver cancer stem cells. Label-free quantitation was performed in combination with ID-based Elution time Alignment by Linear regression Quantitation (IDEAL-Q) and MaxQuant. RESULTS Initially, IDEAL-Q analysis revealed that 151 proteins were differentially expressed in the CD133-positive hepatoma cells when compared with CD133-negative cells. We then analyzed these 151 differentially expressed proteins by MaxQuant software and identified 10 significantly up-regulated proteins. The results were further validated by RT-PCR, western blot, flow cytometry or immunofluorescent staining which revealed that prominin-1, annexin A1, annexin A3, transgelin, creatine kinase B, vimentin, and EpCAM were indeed highly expressed in the CD133-positive hepatoma cells. CONCLUSIONS These findings confirmed that mass spectrometry-based label-free quantitative proteomics can be used to gain insights into liver cancer stem cells.
Collapse
Affiliation(s)
- Sheng-Ta Tsai
- Institute of Biochemistry & Molecular Biology, National Yang-Ming University, Taipei, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
211
|
Zhang W, Li Y, Yang L, Zhou B, Chen KL, Meng WJ, Liu Y, Hu JK, Sun XF, Zhou ZG. Knockdown of MMP-7 inhibits cell proliferation and enhances sensitivity to 5-Fluorouracil and X-ray irradiation in colon cancer cells. Clin Exp Med 2012; 14:99-106. [PMID: 23086188 DOI: 10.1007/s10238-012-0212-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 09/13/2012] [Indexed: 02/05/2023]
Abstract
The role of matrix metalloproteinase-7 (MMP-7) in the pathogenesis of colon cancer is not understood thoroughly. Previous studies from our group have shown that the expression levels of MMP-7 were highly elevated in colorectal cancer patient specimens and were correlated with Dukes Staging, histological differentiation grade and CEA level. The goal of this study was to investigate the cellular impact of MMP-7 in colon cancer. In this study, we used the SW480 colon cancer cell lines of MMP-7 knockdown by lentivirus-mediated RNA interference as a model system to investigate the impact of MMP-7 on cell proliferation and sensitivity to 5-Fluorouracil (5-FU) and X-ray irradiation (IR). Cell proliferation and sensitivity to 5-FU and IR were measured by MTT assay and colony formation assay. Cell cycle was evaluated by flow cytometry. We showed that the down regulation of MMP-7 inhibits colon cancer cell proliferation and sensitizes tumour cells to 5-FU and IR (P < 0.05). Decreased MMP-7 expression in SW480 cells by RNA interference triggered cell cycle arrest at G1 phase (P < 0.05). Down regulation of MMP-7 may inhibit the cell proliferation of colon cancer cells and increase tumour cells sensitivity to radiotherapy and chemotherapy. RNAi-mediated silencing of MMP-7 may represent a powerful therapeutic approach for controlling human colorectal cancer growth.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Gastrointestinal Surgery, Institute of Digestive Surgery and Organ Microcirculation, West China Hospital, Sichuan University, No. 37 on Guo-Xue, Chengdu, 610041, Sichuan Provence, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
212
|
SONG KAI, WU JUNHUA, JIANG CHUNPING. Dysregulation of signaling pathways and putative biomarkers in liver cancer stem cells (Review). Oncol Rep 2012; 29:3-12. [DOI: 10.3892/or.2012.2082] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 09/25/2012] [Indexed: 02/06/2023] Open
|
213
|
Abstract
Solid tumors are thought to contain cancer stem cells (CSCs) as a distinct population responsible for tumor relapse and metastasis due to their abilities to self-renew, differentiate, and give rise to a new tumor in local or distant organs. CSCs have been identified in many tumor types, including hepatocellular carcinoma (HCC), the fifth most common and third most deadly malignancy with observable heterogeneity. Numerous studies have shown that hepatic CSCs could be enriched via different cell surface markers, eg, CD13, CD24, CD44, CD90, CD133, EpCAM (CD326), and OV6. They also could be identified through functional assays such as isolating the side population cells by Hoechst dye staining or screening cells with a high activity of aldehyde dehydrogenase. Functional characterization of hepatic CSCs has revealed several deregulated signaling pathways, such as Wnt/β-catenin, AKT, transforming growth factor-beta (TGF-β), interleukin (IL)-6/STAT3 pathways to be critical in inducing "stemness" of HCC and in promoting self-renewal, tumorigenicity, and chemoresistance. An increased understanding of hepatic CSC biology facilitated the development of new diagnostic, prognostic, and therapeutic strategies for improving HCC clinical management. In this review, we summarize recent evidence including the identification of the hepatic CSC and its underlying biological mechanisms, and discuss the potential clinical implications in HCC.
Collapse
Affiliation(s)
- Junfang Ji
- Liver Carcinogenesis Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
214
|
TIAN YE, WAN HAN, TAN GUANG. Cell cycle-related kinase in carcinogenesis. Oncol Lett 2012; 4:601-606. [PMID: 23205069 PMCID: PMC3506610 DOI: 10.3892/ol.2012.828] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 07/24/2012] [Indexed: 12/16/2022] Open
Abstract
Cell cycle-related kinase (CCRK) is a novel protein kinase homologous to both cyclin-dependent kinase 7 (Cdk7) and Cak1p groups of CDK-activating kinase (CAK). CCRK activates Cdk2, which controls the cell-cycle progression by phosphorylating a threonine residue conserved in Cdk2. Previous studies have indicated that the CCRK protein levels were elevated by more than 1.5-fold in tumor tissue, and that the overexpression of CCRK is associated with poor prognosis of the patients. Moreover, recent studies have shown that CCRK is involved in the Wnt signaling pathway associated with the genesis and evolution of cancer. This review aims to systematically present the information currently available on CCRK obtained from in vitro and in vivo studies and highlight its significance to tumorigenesis.
Collapse
Affiliation(s)
- YE TIAN
- Department of General Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning 116011,
P.R. China
| | - HAN WAN
- Department of General Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning 116011,
P.R. China
| | - GUANG TAN
- Department of General Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning 116011,
P.R. China
| |
Collapse
|
215
|
Fan L, Xu C, Wang C, Tao J, Ho C, Jiang L, Gui B, Huang S, Evert M, Calvisi DF, Chen X. Bmi1 is required for hepatic progenitor cell expansion and liver tumor development. PLoS One 2012; 7:e46472. [PMID: 23029524 PMCID: PMC3460872 DOI: 10.1371/journal.pone.0046472] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 09/02/2012] [Indexed: 12/13/2022] Open
Abstract
Bmi1 is a polycomb group transcriptional repressor and it has been implicated in regulating self-renewal and proliferation of many types of stem or progenitor cells. In addition, Bmi1 has been shown to function as an oncogene in multiple tumor types. In this study, we investigated the functional significance of Bmi1 in regulating hepatic oval cells, the major type of bipotential progenitor cells in adult liver, as well as the role of Bmi1 during hepatocarcinogenesis using Bmi1 knockout mice. We found that loss of Bmi1 significantly restricted chemically induced oval cell expansion in the mouse liver. Concomitant deletion of Ink4a/Arf in Bmi1 deficient mice completely rescued the oval cell expansion phenotype. Furthermore, ablation of Bmi1 delayed hepatocarcinogenesis induced by AKT and Ras co-expression. This antineoplastic effect was accompanied by the loss of hepatic oval cell marker expression in the liver tumor samples. In summary, our data demonstrated that Bmi1 is required for hepatic oval cell expansion via deregulating the Ink4a/Arf locus in mice. Our study also provides the evidence, for the first time, that Bmi1 expression is required for liver cancer development in vivo, thus representing a promising target for innovative treatments against human liver cancer.
Collapse
Affiliation(s)
- Lingling Fan
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuanrui Xu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunmei Wang
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Junyan Tao
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Coral Ho
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Lijie Jiang
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Bing Gui
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Shiang Huang
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Matthias Evert
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Diego F. Calvisi
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
- Liver Center, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
216
|
Yang W, Wang C, Lin Y, Liu Q, Yu LX, Tang L, Yan HX, Fu J, Chen Y, Zhang HL, Tang L, Zheng LY, He YQ, Li YQ, Wu FQ, Zou SS, Li Z, Wu MC, Feng GS, Wang HY. OV6⁺ tumor-initiating cells contribute to tumor progression and invasion in human hepatocellular carcinoma. J Hepatol 2012; 57:613-20. [PMID: 22612999 DOI: 10.1016/j.jhep.2012.04.024] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Revised: 04/02/2012] [Accepted: 04/19/2012] [Indexed: 12/23/2022]
Abstract
BACKGROUND & AIMS Accumulating evidence suggests the involvement of tumor-initiating cells (T-ICs) in cancer genesis, but whether liver T-ICs contribute to HCC invasion and metastasis remains unclear. METHODS OV6(+) T-ICs were isolated from SMMC7721 and HuH7 cell lines by magnetic sorting. Characteristics of T-ICs were assessed by in vitro and mouse xenograft assays. Expression of OV6 was determined by immunostaining in specimens from 218 HCC patients, and Kaplan-Meier survival analysis was used to determine the correlation of OV6 expression with prognosis. RESULTS OV6(+) T-ICs isolated from HCC cell lines not only possess a higher capacity to form tumor spheroids in vitro, but also had a greater potential to form tumors when implanted in non-obese diabetic/severe combined immunodeficient mice, suggesting their elevated self-renewal capacity and tumorigenicity. Moreover, OV6(+) T-ICs exhibited more invasive and metastatic potentials both in vitro and in vivo. Patients with more OV6(+) tumor cells were associated with aggressive clinicopathologic features and poor prognosis. CXCR4 is expressed at higher levels in OV6(+) cells. Recombinant stromal cell-derived factor-1 (SDF-1) treatment expanded the OV6(+) HCC T-ICs population, by sustaining the stem cell property of OV6(+) cells. The SDF-1 effect was blocked by a specific CXCR4 inhibitor, AMD3100, or transfection of siRNA targeting CXCR4. CONCLUSIONS OV6(+) HCC cells may represent a subpopulation of T-ICs with augmented invasion and metastasis potential, which contribute to progression and metastasis of HCC. The SDF-1/CXCR4 axis also provides therapeutic targets for elimination of liver T-ICs.
Collapse
Affiliation(s)
- Wen Yang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai 200438, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
217
|
Economopoulou P, Kaklamani VG, Siziopikou K. The role of cancer stem cells in breast cancer initiation and progression: potential cancer stem cell-directed therapies. Oncologist 2012; 17:1394-401. [PMID: 22941971 DOI: 10.1634/theoncologist.2012-0163] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Recent studies have identified a small population of highly tumorigenic cells with stem cell properties in human breast and other solid tumors that are considered to be the source of tumor initiation and maintenance; these cells are referred to as cancer stem cells (CSCs). Preclinical data suggest that current breast cancer treatment strategies lead to CSC enrichment, contributing to chemotherapy and radiotherapy resistance, although a strong correlation with clinical parameters and prognosis is yet to be established. Importantly, overcoming treatment failure by effective targeting of CSCs may be an appealing approach, potentially leading to improved clinical outcomes for patients with breast cancer. Several preclinical studies provide promising results that support this hypothesis. The purpose of this review is to summarize the role of CSCs in breast cancer recurrence and resistance and to discuss current attempts of CSC targeting.
Collapse
|
218
|
Chen Y, Yu D, Zhang H, He H, Zhang C, Zhao W, Shao RG. CD133(+)EpCAM(+) phenotype possesses more characteristics of tumor initiating cells in hepatocellular carcinoma Huh7 cells. Int J Biol Sci 2012; 8:992-1004. [PMID: 22904667 PMCID: PMC3421230 DOI: 10.7150/ijbs.4454] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 07/24/2012] [Indexed: 12/26/2022] Open
Abstract
Background: EpCAM or CD133 has been used as the tumor initiating cells (TICs) marker in hepatocellular carcinoma (HCC). We investigated whether cells expressing with both EpCAM and CD133 surface marker were more representative for TICs in hepatocellular carcinoma Huh7 cells. Methods: Four different phenotypes of CD133+EpCAM+, CD133+EpCAM-, CD133-EpCAM+ and CD133-EpCAM- in Huh7 cells were sorted by flow cytometry. Then cell differentiation, self-renewal, drug-resistance, spheroid formation and the levels of stem cell-related genes were detected to compare the characteristics of TICs. The ability of tumorigenicity was measured in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice to verify TICs. Results: CD133+EpCAM+ cells have many characteristics of TICs in Huh7 cells compared with CD133+EpCAM-, CD133-EpCAM+, CD133-EpCAM- cells, including enrichment in side population cells, higher differentiation capacity, increased colony-formation ability, preferential expression of stem cell-related genes, appearance of drug-resistant to some chemotherapeutics, more spheroid formation of culture cells and stronger tumorigenicity in NOD/SCID mice. Conclusion: CD133+EpCAM+ phenotype is precisely represented TICs in Huh7 cells. It might be useful for studying biology mechanism of TICs in hepatocellular carcinoma and screening new targets for cancer therapy.
Collapse
Affiliation(s)
- Yi Chen
- Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Tiantan Xili, Beijing 100050, China
| | | | | | | | | | | | | |
Collapse
|
219
|
Chen Y, Yu D, Zhang H, He H, Zhang C, Zhao W, Shao RG. CD133(+)EpCAM(+) phenotype possesses more characteristics of tumor initiating cells in hepatocellular carcinoma Huh7 cells. Int J Biol Sci 2012. [PMID: 22904667 DOI: 10.7150/ijbs.4454ijbsv08p0992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND EpCAM or CD133 has been used as the tumor initiating cells (TICs) marker in hepatocellular carcinoma (HCC). We investigated whether cells expressing with both EpCAM and CD133 surface marker were more representative for TICs in hepatocellular carcinoma Huh7 cells. METHODS Four different phenotypes of CD133(+)EpCAM(+), CD133(+)EpCAM(-), CD133(-)EpCAM(+) and CD133(-)EpCAM(-) in Huh7 cells were sorted by flow cytometry. Then cell differentiation, self-renewal, drug-resistance, spheroid formation and the levels of stem cell-related genes were detected to compare the characteristics of TICs. The ability of tumorigenicity was measured in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice to verify TICs. RESULTS CD133(+)EpCAM(+) cells have many characteristics of TICs in Huh7 cells compared with CD133(+)EpCAM(-), CD133(-)EpCAM(+), CD133(-)EpCAM(-) cells, including enrichment in side population cells, higher differentiation capacity, increased colony-formation ability, preferential expression of stem cell-related genes, appearance of drug-resistant to some chemotherapeutics, more spheroid formation of culture cells and stronger tumorigenicity in NOD/SCID mice. CONCLUSION CD133(+)EpCAM(+) phenotype is precisely represented TICs in Huh7 cells. It might be useful for studying biology mechanism of TICs in hepatocellular carcinoma and screening new targets for cancer therapy.
Collapse
Affiliation(s)
- Yi Chen
- Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Tiantan Xili, Beijing 100050, China
| | | | | | | | | | | | | |
Collapse
|
220
|
LeCluyse EL, Witek RP, Andersen ME, Powers MJ. Organotypic liver culture models: meeting current challenges in toxicity testing. Crit Rev Toxicol 2012; 42:501-48. [PMID: 22582993 PMCID: PMC3423873 DOI: 10.3109/10408444.2012.682115] [Citation(s) in RCA: 248] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Revised: 03/26/2012] [Accepted: 03/30/2012] [Indexed: 02/07/2023]
Abstract
Prediction of chemical-induced hepatotoxicity in humans from in vitro data continues to be a significant challenge for the pharmaceutical and chemical industries. Generally, conventional in vitro hepatic model systems (i.e. 2-D static monocultures of primary or immortalized hepatocytes) are limited by their inability to maintain histotypic and phenotypic characteristics over time in culture, including stable expression of clearance and bioactivation pathways, as well as complex adaptive responses to chemical exposure. These systems are less than ideal for longer-term toxicity evaluations and elucidation of key cellular and molecular events involved in primary and secondary adaptation to chemical exposure, or for identification of important mediators of inflammation, proliferation and apoptosis. Progress in implementing a more effective strategy for in vitro-in vivo extrapolation and human risk assessment depends on significant advances in tissue culture technology and increasing their level of biological complexity. This article describes the current and ongoing need for more relevant, organotypic in vitro surrogate systems of human liver and recent efforts to recreate the multicellular architecture and hemodynamic properties of the liver using novel culture platforms. As these systems become more widely used for chemical and drug toxicity testing, there will be a corresponding need to establish standardized testing conditions, endpoint analyses and acceptance criteria. In the future, a balanced approach between sample throughput and biological relevance should provide better in vitro tools that are complementary with animal testing and assist in conducting more predictive human risk assessment.
Collapse
Affiliation(s)
- Edward L LeCluyse
- The Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, NC, USA.
| | | | | | | |
Collapse
|
221
|
Han ZG. Functional genomic studies: insights into the pathogenesis of liver cancer. Annu Rev Genomics Hum Genet 2012; 13:171-205. [PMID: 22703171 DOI: 10.1146/annurev-genom-090711-163752] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Liver cancer is the sixth-most-common cancer overall but the third-most-frequent cause of cancer death. Among primary liver cancers, hepatocellular carcinoma (HCC), the major histological subtype, is associated with multiple risk factors, including hepatitis B and C virus infection, alcohol consumption, obesity, and diet contamination. Although previous studies have revealed that certain genetic and epigenetic changes, such as TP53 and β-catenin mutations, occur in HCC cells, the pathogenesis of this cancer remains obscure. Functional genomic approaches-including genome-wide association studies, whole-genome and whole-exome sequencing, array-based comparative genomic hybridization, global DNA methylome mapping, and gene or noncoding RNA expression profiling-have recently been applied to HCC patients with different clinical features to uncover the genetic risk factors and underlying molecular mechanisms involved in this cancer's initiation and progression. The genome-wide analysis of germline and somatic genetic and epigenetic events facilitates understanding of the pathogenesis and molecular classification of liver cancer as well as the identification of novel diagnostic biomarkers and therapeutic targets for cancer.
Collapse
Affiliation(s)
- Ze-Guang Han
- National Human Genome Center of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
222
|
Qian YW, Chen Y, Yang W, Fu J, Cao J, Ren YB, Zhu JJ, Su B, Luo T, Zhao XF, Dai RY, Li JJ, Sun W, Wu MC, Feng GS, Wang HY. p28(GANK) prevents degradation of Oct4 and promotes expansion of tumor-initiating cells in hepatocarcinogenesis. Gastroenterology 2012; 142:1547-58.e14. [PMID: 22387393 DOI: 10.1053/j.gastro.2012.02.042] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 02/01/2012] [Accepted: 02/22/2012] [Indexed: 01/06/2023]
Abstract
BACKGROUND & AIMS Hepatocellular carcinoma (HCC) is believed to arise from tumor-initiating cells (T-ICs), although little is known about their stem cell-like properties. METHODS We quantified levels of p28(GANK) (Gankyrin), OV6, and Oct4 in 130 human HCC samples using immunohistochemistry. Magnetic-activated cell sorting was used to isolate OV6+ HCC cells. T-IC properties were evaluated by quantitative reverse-transcription polymerase chain reaction, flow cytometry, and spheroid formation. We used a coimmunoprecipitation assay to study interactions among p28(GANK), Oct4, and WWP2. Tumorigenicity and pulmonary metastasis were examined in nonobese diabetic and severe combined immunodeficient mice. RESULTS In HCC samples, high levels of p28(GANK) correlated with expansion of OV6+ tumor cells; the combination of high levels of p28(GANK) and OV6 was associated with progression of HCC. p28(GANK) was predominantly expressed in liver T-ICs, isolated by magnetic sorting, and undifferentiated primary HCC spheroids. Increased levels of p28(GANK) in T-ICs increased their percentages in HCC samples, expression of stem cell genes, self-renewal potential, chemoresistance in vitro, and tumorigenicity and ability to develop into pulmonary metastases in mice. Conversely, knockdown of p28(GANK) reduced their T-IC properties. p28(GANK) likely activates liver T-ICs by impeding ubiquitination and degradation of the transcription factor Oct4 by WWP2. In support of this concept, levels of p28(GANK) correlated with those of Oct4 in HCC samples. CONCLUSIONS p28(GANK) activates and maintains liver T-ICs in HCCs by preventing degradation of Oct4. Inhibitors of p28(GANK) might therefore be developed to inactivate T-ICs and slow tumor progression.
Collapse
Affiliation(s)
- You-Wen Qian
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
223
|
Favier RP, Spee B, Penning LC, Rothuizen J. Copper-induced hepatitis: the COMMD1 deficient dog as a translational animal model for human chronic hepatitis. Vet Q 2012; 31:49-60. [PMID: 22029820 DOI: 10.1080/01652176.2011.563146] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Chronic inflammatory liver disease regardless of aetiology leads to failing regeneration and fibrosis, ending in cirrhosis. Both in man and in animals this worldwide health problem has no definitive cure. Chronic liver injury causes hepatic stellate cells to proliferate and differentiate into matrix-producing cells. New therapeutic options will be developed upon detailed understanding of the molecular mechanisms driving liver fibrosis. This may lead to new anti-fibrotic therapies which need to be tested in suitable models before application in the veterinary and human clinic. On the other side, to restore the failing regenerative capacity of the diseased liver cells, adult progenitor cells are of interest, as an alternative to whole organ transplantation. In order to find the most suitable large animal model it is important to recognise that the typical histopathological reaction pattern of the liver can differ between mammalian species. It is therefore imperative that specialists in veterinary internal medicine and pathology, being familiar with the diseases and pathologies of the liver in different animal species, are teaming-up in finding the best models for veterinary and human liver diseases. Several large animal models have been mentioned, like pigs, sheep, and dogs. Based on the observations that man and dog share the same hepatopathies and have identical clinical, pathological and pathogenetic reaction patterns during the development of liver disease, the dog seems to be a properly suited species to test new therapeutic strategies for pets and their best friends.
Collapse
Affiliation(s)
- R P Favier
- Faculty of Veterinary Medicine, Department of Clinical Sciences of Companion Animals, Utrecht University, Utrecht, the Netherlands.
| | | | | | | |
Collapse
|
224
|
Lade A, Ranganathan S, Luo J, Monga SPS. Calpain induces N-terminal truncation of β-catenin in normal murine liver development: diagnostic implications in hepatoblastomas. J Biol Chem 2012; 287:22789-98. [PMID: 22613727 DOI: 10.1074/jbc.m112.378224] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hepatic competence, specification, and liver bud expansion during development depend on precise temporal modulation of the Wnt/β-catenin signaling. Also, loss- and gain-of-function studies have revealed pleiotropic roles of β-catenin in proliferation and hepatocyte and biliary epithelial cell differentiation, but precise mechanisms remain unknown. Here we utilize livers from different stages of murine development to determine β-catenin signaling and downstream targets. Although during early liver development full-length β-catenin is the predominant form, at late stages, where full-length β-catenin localizes to developing biliary epithelial cells only, a 75-kDa truncated β-catenin species is the principal form localizing at the membrane and in the nucleus of differentiating hepatocytes. The truncated species lacks 95 N-terminal amino acids and is transcriptionally active. Our evidence points to proteolytic cleavage of β-catenin by calpain as the mechanism of truncation in cell-free and cell-based assays. Intraperitoneal injection of a short term calpain inhibitor to timed pregnant female mice abrogated β-catenin truncation in the embryonic livers. RNA-seq revealed a unique set of targets transcribed in cells expressing truncated versus full-length β-catenin, consistent with different functionalities. A further investigation using N- and C-terminal-specific β-catenin antibodies on human hepatoblastomas revealed a correlation between full-length versus truncated β-catenin and differentiation status, with embryonal hepatoblastomas expressing full-length β-catenin and fetal hepatoblastomas expressing β-catenin lacking its N terminus. Thus we conclude that calpain-mediated cleavage of β-catenin plays a role in regulating hepatoblast differentiation in mouse and human liver, and the presence of the β-catenin N terminus correlates with differentiation status in hepatoblastomas.
Collapse
Affiliation(s)
- Abigale Lade
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | |
Collapse
|
225
|
Lee TKW, Cheung VCH, Ng IOL. Liver tumor-initiating cells as a therapeutic target for hepatocellular carcinoma. Cancer Lett 2012; 338:101-9. [PMID: 22579789 DOI: 10.1016/j.canlet.2012.05.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 04/27/2012] [Accepted: 05/02/2012] [Indexed: 01/03/2023]
Abstract
Hepatocellular carcinoma (HCC) is a common malignancy worldwide and has poor prognosis. Existing treatment modalities, including surgery, chemotherapy, and radiofrequency ablation, which target tumor bulk, have demonstrated limited therapeutic efficacy. In the past 10years, accumulating evidence has supported the existence of cancer stem cells (CSCs) or tumor initiating cells (T-ICs) within tumors including HCC. Identification of liver T-ICs and the signaling pathways that they are involved in may shed light on novel therapeutic strategies against this deadly disease. In this review, we will discuss recent progresses made in the research of liver T-ICs with regard to identification, functional characterization, regulation and therapeutic implications.
Collapse
Affiliation(s)
- Terence Kin Wah Lee
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong
| | | | | |
Collapse
|
226
|
Liu WH, Wang X, You N, Tao KS, Wang T, Tang LJ, Dou KF. Efficient enrichment of hepatic cancer stem-like cells from a primary rat HCC model via a density gradient centrifugation-centered method. PLoS One 2012; 7:e35720. [PMID: 22558209 PMCID: PMC3338461 DOI: 10.1371/journal.pone.0035720] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 03/20/2012] [Indexed: 12/26/2022] Open
Abstract
Background Because few definitive markers are available for hepatic cancer stem cells (HCSCs), based on physical rather than immunochemical properties, we applied a novel method to enrich HCSCs. Methodology After hepatic tumor cells (HTCs) were first isolated from diethylinitrosamine-induced F344 rat HCC model using percoll discontinuous gradient centrifugation (PDGC) and purified via differential trypsinization and differential attachment (DTDA), they were separated into four fractions using percoll continuous gradient centrifugation (PCGC) and sequentially designated as fractions I–IV (FI–IV). Morphological characteristics, mRNA and protein levels of stem cell markers, proliferative abilities, induced differentiation, in vitro migratory capacities, in vitro chemo-resistant capacities, and in vivo malignant capacities were determined for the cells of each fraction. Findings As the density of cells increased, 22.18%, 11.62%, 4.73% and 61.47% of primary cultured HTCs were segregated in FI–FIV, respectively. The cells from FIII (density between 1.041 and 1.062 g/ml) displayed a higher nuclear-cytoplasmic ratio and fewer organelles and expressed higher levels of stem cell markers (AFP, EpCAM and CD133) than cells from other fractions (P<0.01). Additionally, in vitro, the cells from FIII showed a greater capacity to self-renew, differentiate into mature HTCs, transit across membranes, close scratches, and carry resistance to chemotherapy than did cells from any other fraction; in vivo, injection of only 1×104 cells from FIII could generate tumors not only in subcutaneous tissue but also in the livers of nude mice. Conclusions Through our novel method, HCSC-like cells were successfully enriched in FIII. This study will greatly contribute to two important areas of biological interest: CSC isolation and HCC therapy.
Collapse
Affiliation(s)
- Wei-hui Liu
- PLA Center of General Surgery, General Hospital of Chengdu Army Region, Chengdu, Sichuan Province, China
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Xing Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Nan You
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Kai-shan Tao
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Tao Wang
- PLA Center of General Surgery, General Hospital of Chengdu Army Region, Chengdu, Sichuan Province, China
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Li-jun Tang
- PLA Center of General Surgery, General Hospital of Chengdu Army Region, Chengdu, Sichuan Province, China
- * E-mail: (KD); (LT)
| | - Ke-feng Dou
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
- * E-mail: (KD); (LT)
| |
Collapse
|
227
|
Li ZQ, Ding W, Sun SJ, Li J, Pan J, Zhao C, Wu WR, Si WK. Cyr61/CCN1 is regulated by Wnt/β-catenin signaling and plays an important role in the progression of hepatocellular carcinoma. PLoS One 2012; 7:e35754. [PMID: 22540002 PMCID: PMC3335098 DOI: 10.1371/journal.pone.0035754] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 03/26/2012] [Indexed: 12/20/2022] Open
Abstract
Abnormal activation of the canonical Wnt signaling pathway has been implicated in carcinogenesis. Transcription of Wnt target genes is regulated by nuclear β-catenin, whose over-expression is observed in Hepatocellular Carcinoma (HCC) tissue. Cyr61, a member of the CCN complex family of multifunctional proteins, is also found over-expressed in many types of tumor and plays dramatically different roles in tumorigenesis. In this study, we investigated the relationship between Cyr61 and β-catenin in HCC. We found that while Cyr61 protein was not expressed at a detectable level in the liver tissue of healthy individuals, its expression level was elevated in the HCC and HCC adjacent tissues and was markedly increased in cancer-adjacent hepatic cirrhosis tissue. Over-expression of Cyr61 was positively correlated with increased levels of β-catenin in human HCC samples. Activation of β-catenin signaling elevated the mRNA level of Cyr61 in HepG2 cells, while inhibition of β-catenin signaling reduced both mRNA and protein levels of Cyr61. We identified two TCF4-binding elements in the promoter region of human Cyr61 gene and demonstrated that β-catenin/TCF4 complex specifically bound to the Cyr61 promoter in vivo and directly regulated its promoter activity. Furthermore, we found that over-expression of Cyr61 in HepG2 cells promoted the progression of HCC xenografts in SCID mice. These findings indicate that Cyr61 is a direct target of β-catenin signaling in HCC and may play an important role in the progression of HCC.
Collapse
Affiliation(s)
- Zhi-Qiang Li
- Department of Clinical Hematology, Third Military Medical University, Chongqing, China
| | - Wei Ding
- Department of Clinical Hematology, Third Military Medical University, Chongqing, China
- Department of Clinical Laboratory, KunMing General Hospital of PLA, KunMing, China
| | - Shi-Jun Sun
- Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jun Li
- Department of Clinical Hematology, Third Military Medical University, Chongqing, China
| | - Jing Pan
- Department of Clinical Hematology, Third Military Medical University, Chongqing, China
| | - Chen Zhao
- Department of Clinical Hematology, Third Military Medical University, Chongqing, China
| | - Wei-Ru Wu
- Department of Clinical Hematology, Third Military Medical University, Chongqing, China
| | - Wei-Ke Si
- Department of Clinical Hematology, Third Military Medical University, Chongqing, China
- * E-mail:
| |
Collapse
|
228
|
Zheng LY, Zhou DX, Lu J, Zhang WJ, Zou DJ. Down-regulated expression of the protein-tyrosine phosphatase 1B (PTP1B) is associated with aggressive clinicopathologic features and poor prognosis in hepatocellular carcinoma. Biochem Biophys Res Commun 2012; 420:680-4. [DOI: 10.1016/j.bbrc.2012.03.066] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 03/12/2012] [Indexed: 12/27/2022]
|
229
|
Greenow K, Clarke AR. Controlling the stem cell compartment and regeneration in vivo: the role of pluripotency pathways. Physiol Rev 2012; 92:75-99. [PMID: 22298652 DOI: 10.1152/physrev.00040.2010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Since the realization that embryonic stem cells are maintained in a pluripotent state through the interplay of a number of key signal transduction pathways, it is becoming increasingly clear that stemness and pluripotency are defined by the complex molecular convergence of these pathways. Perhaps this has most clearly been demonstrated by the capacity to induce pluripotency in differentiated cell types, so termed iPS cells. We are therefore building an understanding of how cells may be maintained in a pluripotent state, and how we may manipulate cells to drive them between committed and pluripotent compartments. However, it is less clear how cells normally pass in and out of the stem cell compartment under normal and diseased physiological states in vivo, and indeed, how important these pathways are in these settings. It is also clear that there is a potential "dark side" to manipulating the stem cell compartment, as deregulation of somatic stem cells is being increasingly implicated in carcinogenesis and the generation of "cancer stem cells." This review explores these relationships, with a particular focus on the role played by key molecular regulators of stemness in tissue repair, and the possibility that a better understanding of this control may open the door to novel repair strategies in vivo. The successful development of such strategies has the potential to replace or augment intervention-based strategies (cell replacement therapies), although it is clear they must be developed with a full understanding of how such approaches might also influence tumorigenesis.
Collapse
Affiliation(s)
- Kirsty Greenow
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | | |
Collapse
|
230
|
Jeong HT, Kim MJ, Kim YE, Park YN, Choi GH, Choi JS. MRI features of hepatocellular carcinoma expressing progenitor cell markers. Liver Int 2012; 32:430-40. [PMID: 22097930 DOI: 10.1111/j.1478-3231.2011.02640.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 08/15/2011] [Indexed: 01/08/2023]
Abstract
BACKGROUND & AIMS To determine whether magnetic resonance (MR) imaging features differ between hepatocellular carcinomas (HCCs) with and without expression of progenitor cell markers, such as cytokeratin (CK) 19 and epithelial cell adhesion molecule (EpCAM). METHODS Sixty-three patients with 71 HCCs who underwent surgery after preoperative gadoxetic acid-enhanced MR imaging were evaluated. HCCs expressing progenitor cell markers were defined as showing CK19 or EpCAM expression. MR imaging features, including the fat component, arterial enhancement (global vs. peripheral), dynamic enhancement (washout vs. progressive or persistent), nodule-in-nodule appearance and MR gross morphology (expanding vs. non-expanding), were compared between HCCs with and without progenitor cell markers expression. Lesion-to-liver signal intensity ratio (SIR) and apparent diffusion coefficient values were compared using an independent samples t-test. Early recurrence rates were also compared. RESULTS HCCs expressing progenitor cell markers were more commonly of the non-expanding type (P = 0.016), more frequently had a progressive or persistent dynamic enhancement pattern (P = 0.008) and less frequently demonstrated a nodule-in-nodule appearance (P = 0.009). HCCs expressing progenitor cell markers had significantly higher SIRs on diffusion-weighted images (DWIs) (b = 50 and 800, P < 0.001; b = 400, P = 0.001) and a significantly lower SIR on hepatobiliary phase images (P = 0.024). The early recurrence rate was significantly higher in patients with prior HCCs that expressed progenitor cell markers (P = 0.045). CONCLUSIONS HCCs expressing progenitor cell markers can be characterized according to their non-expanding MR gross morphology, persistent or progressive dynamic enhancement patterns, higher SIRs on DWIs, lower SIRs on hepatobiliary phase images and less frequent nodule-in-nodule appearance.
Collapse
Affiliation(s)
- Hyeon Tae Jeong
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | |
Collapse
|
231
|
Wilkens L, Hammer C, Glombitza S, Müller DE. Hepatocellular and cholangiolar carcinoma-derived cell lines reveal distinct sets of chromosomal imbalances. Pathobiology 2012; 79:115-26. [PMID: 22261732 DOI: 10.1159/000334100] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 10/03/2011] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Hepatocellular carcinoma (HCC) and cholangiolar carcinoma (CC) cell lines are used to analyze the basic mechanisms of carcinogenesis and target therapies. However, it is not yet clear which chromosomal aberrations are to be typically expected in such cell lines. It is also not clear whether there are prerequisites for in vitro growth on the genomic and/or expression level. We therefore analyzed HCC and CC cell lines for typical genetic settings. METHODS The HCC cell lines HLE, HLF, Huh7, HepG2 and Hep3b and the CC cell lines EGI1, MzCha1 and TFK-1 were analyzed using high-density arrays for comparative genomic hybridization (aCGH; 244,000 oligonucleotides). Additional fluorescence in situ hybridization analyses were done to confirm the aCGH results and to add information regarding the aneuploidy of cell lines. RESULTS The gain of 1q, in particular q21-22, was detected in all HCC cell lines also as a partial loss of 13q. In contrast, a loss of 8p in combination with a relative gain of 8q was seen in all CC but no HCC cell lines. Interestingly, a gain of 17q was seen in all cell lines. These aberrations are also well documented for surgical tumor specimens. Besides these imbalances, the cell lines revealed imbalances for 11p, 12p, 14q, 16p, 16q, 21q and 22q, respectively, only rarely seen in surgical tumor specimens. These aberrations could be of importance for the in vitro cultivation of tumor cells. Structural aberrations were accompanied by aneuploidy in 3 of 5 HCC cell lines and 2 of 3 CC cell lines. Ploidy status was not correlated to any of the imbalances mentioned above. CONCLUSIONS HCC and CC cell lines revealed characteristic chromosomal imbalances similar to those seen in surgical tumor specimens including chromosomes 1, 8, 13 and 17, respectively. These aberrations are characteristic of the histogenetic origin of the tumor cells. However, the chromosomal imbalances that occurred probably led to the ability of tumor cells to grow in vitro.
Collapse
Affiliation(s)
- Ludwig Wilkens
- Institute of Pathology, University of Bern, Bern, Switzerland.
| | | | | | | |
Collapse
|
232
|
Xu X, Liu RF, Zhang X, Huang LY, Chen F, Fei QL, Han ZG. DLK1 as a potential target against cancer stem/progenitor cells of hepatocellular carcinoma. Mol Cancer Ther 2012; 11:629-38. [PMID: 22238367 DOI: 10.1158/1535-7163.mct-11-0531] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Delta-like 1 homolog (DLK1; Drosophila) is a hepatic stem/progenitor cell marker in fetal livers that plays a vital role in oncogenesis of hepatocellular carcinoma (HCC). The aim of this study is to investigate whether DLK1 could serve as a potential therapeutic target against cancer stem/progenitor cells of HCC. DLK1(+) and DLK1(-) cells were sorted by fluorescence-activated cell sorting and magnetic-activated cell sorting, respectively, and then were evaluated by flow cytometry. The biological behaviors of these isolated cells and those with DLK1 knockdown were assessed by growth curve, colony formation assay, spheroid colony formation, chemoresistance, and in vivo tumorigenicity. Adenovirus-mediated RNA interference was used to knockdown the endogenous DLK1. We found that DLK1(+) population was less than 10% in almost all 17 HCC cell lines examined. DLK1(+) HCC cells showed stronger ability of chemoresistance, colony formation, spheroid colony formation, and in vivo tumorigenicity compared with DLK1(-) cells. The DLK1(+) HCC cells could generate the progeny without DLK1 expression. Furthermore, DLK1 knockdown could suppress the ability of proliferation, colony formation, spheroid colony formation, and in vivo tumorigenicity of Hep3B and Huh-7 HCC cells. Our data suggested that DLK1(+) HCC cells have characteristics similar to those of cancer stem/progenitor cells. RNA interference against DLK1 can suppress the malignant behaviors of HCC cells, possibly through directly disrupting cancer stem/progenitor cells, which suggested that DLK1 could be a potential therapeutic target against the HCC stem/progenitor cells.
Collapse
Affiliation(s)
- Xiao Xu
- National Human Genome Center of Rui-Jin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
233
|
Hirner H, Günes C, Bischof J, Wolff S, Grothey A, Kühl M, Oswald F, Wegwitz F, Bösl MR, Trauzold A, Henne-Bruns D, Peifer C, Leithäuser F, Deppert W, Knippschild U. Impaired CK1 delta activity attenuates SV40-induced cellular transformation in vitro and mouse mammary carcinogenesis in vivo. PLoS One 2012; 7:e29709. [PMID: 22235331 PMCID: PMC3250488 DOI: 10.1371/journal.pone.0029709] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 12/01/2011] [Indexed: 02/05/2023] Open
Abstract
Simian virus 40 (SV40) is a powerful tool to study cellular transformation in vitro, as well as tumor development and progression in vivo. Various cellular kinases, among them members of the CK1 family, play an important role in modulating the transforming activity of SV40, including the transforming activity of T-Ag, the major transforming protein of SV40, itself. Here we characterized the effects of mutant CK1δ variants with impaired kinase activity on SV40-induced cell transformation in vitro, and on SV40-induced mammary carcinogenesis in vivo in a transgenic/bi-transgenic mouse model. CK1δ mutants exhibited a reduced kinase activity compared to wtCK1δ in in vitro kinase assays. Molecular modeling studies suggested that mutation N172D, located within the substrate binding region, is mainly responsible for impaired mutCK1δ activity. When stably over-expressed in maximal transformed SV-52 cells, CK1δ mutants induced reversion to a minimal transformed phenotype by dominant-negative interference with endogenous wtCK1δ. To characterize the effects of CK1δ on SV40-induced mammary carcinogenesis, we generated transgenic mice expressing mutant CK1δ under the control of the whey acidic protein (WAP) gene promoter, and crossed them with SV40 transgenic WAP-T-antigen (WAP-T) mice. Both WAP-T mice as well as WAP-mutCK1δ/WAP-T bi-transgenic mice developed breast cancer. However, tumor incidence was lower and life span was significantly longer in WAP-mutCK1δ/WAP-T bi-transgenic animals. The reduced CK1δ activity did not affect early lesion formation during tumorigenesis, suggesting that impaired CK1δ activity reduces the probability for outgrowth of in situ carcinomas to invasive carcinomas. The different tumorigenic potential of SV40 in WAP-T and WAP-mutCK1δ/WAP-T tumors was also reflected by a significantly different expression of various genes known to be involved in tumor progression, specifically of those involved in wnt-signaling and DNA repair. Our data show that inactivating mutations in CK1δ impair SV40-induced cellular transformation in vitro and mouse mammary carcinogenesis in vivo.
Collapse
MESH Headings
- Animals
- Antigens, Viral, Tumor/immunology
- Casein Kinase Idelta/chemistry
- Casein Kinase Idelta/genetics
- Casein Kinase Idelta/metabolism
- Cell Line
- Cell Line, Tumor
- Cell Transformation, Viral/genetics
- Disease Progression
- Female
- Gene Expression Regulation
- Male
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/pathology
- Mammary Glands, Animal/virology
- Mammary Neoplasms, Experimental/enzymology
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/virology
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Milk Proteins/genetics
- Models, Molecular
- Mutation
- Phenotype
- Phosphorylation
- Promoter Regions, Genetic/genetics
- Protein Structure, Tertiary
- Simian virus 40/immunology
- Simian virus 40/physiology
- Survival Analysis
Collapse
Affiliation(s)
- Heidrun Hirner
- Department of General-, Visceral- and Transplantation Surgery, University of Ulm, Ulm, Germany
| | - Cagatay Günes
- Institute of Molecular Medicine and Max-Planck-Research Group on Stem Cell Aging, University of Ulm, Ulm, Germany
| | - Joachim Bischof
- Department of General-, Visceral- and Transplantation Surgery, University of Ulm, Ulm, Germany
| | - Sonja Wolff
- Department of General-, Visceral- and Transplantation Surgery, University of Ulm, Ulm, Germany
| | - Arnhild Grothey
- Department of General-, Visceral- and Transplantation Surgery, University of Ulm, Ulm, Germany
| | - Marion Kühl
- Department of Tumor Virology, Heinrich-Pette-Institute, Leibniz-Center for Experimental Virology, Hamburg, Germany
| | - Franz Oswald
- Department of Internal Medicine I, University of Ulm, Ulm, Germany
| | - Florian Wegwitz
- Department of Tumor Virology, Heinrich-Pette-Institute, Leibniz-Center for Experimental Virology, Hamburg, Germany
| | - Michael R. Bösl
- Max Planck Institute of Neurobiology Transgenic Mouse Models, Max Planck Institute, Martinsried, Germany
| | - Anna Trauzold
- Division of Molecular Oncology, Institute for Experimental Cancer Research, CCCNorth, UK S-H, Kiel, Germany
| | - Doris Henne-Bruns
- Department of General-, Visceral- and Transplantation Surgery, University of Ulm, Ulm, Germany
| | | | | | - Wolfgang Deppert
- Department of Tumor Virology, Heinrich-Pette-Institute, Leibniz-Center for Experimental Virology, Hamburg, Germany
| | - Uwe Knippschild
- Department of General-, Visceral- and Transplantation Surgery, University of Ulm, Ulm, Germany
- * E-mail:
| |
Collapse
|
234
|
A Complex Interplay between Wnt/β-Catenin Signalling and the Cell Cycle in the Adult Liver. Int J Hepatol 2012; 2012:816125. [PMID: 22973520 PMCID: PMC3438741 DOI: 10.1155/2012/816125] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 08/02/2012] [Indexed: 12/19/2022] Open
Abstract
Canonical Wnt signalling, governed by its effector β-catenin, is known for a long time as playing an important role in development, tissue homeostasis, and cancer. In the liver, it was unravelled as both an oncogenic pathway involved in a subset of liver cancers and a physiological signalling identified as the "zonation-keeper" of the quiescent liver lobule. This duality has encouraged to explore the role of canonical Wnt in liver regeneration and liver-cell proliferation mainly using murine genetic models of β-catenin overactivation or inactivation. These studies definitely integrate Wnt signalling within the hepatic network driving regeneration and proliferation. We will review here the current knowledge concerning the mitogenic effect of Wnt, to switch on its specific role in the liver, which is quiescent but with a great capacity to regenerate. The duality of β-catenin signalling, associated both with liver quiescence and liver-cell proliferation, will be brought forward.
Collapse
|
235
|
Wang C, Yang W, Yan HX, Luo T, Zhang J, Tang L, Wu FQ, Zhang HL, Yu LX, Zheng LY, Li YQ, Dong W, He YQ, Liu Q, Zou SS, Lin Y, Hu L, Li Z, Wu MC, Wang HY. Hepatitis B virus X (HBx) induces tumorigenicity of hepatic progenitor cells in 3,5-diethoxycarbonyl-1,4-dihydrocollidine-treated HBx transgenic mice. Hepatology 2012; 55:108-20. [PMID: 21932402 DOI: 10.1002/hep.24675] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 08/19/2011] [Indexed: 12/12/2022]
Abstract
UNLABELLED Hepatitis B virus X (HBx) protein is implicated in hepatitis B virus (HBV)-associated liver carcinogenesis. However, it remains unclear whether HBx-expressing hepatic progenitor cells (HPCs) are attributed to liver tumor formation. In this study, by using HBx transgenic mice and a 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-induced liver injury model, the relationship between HBx expression and tumorigenicity of HPCs was analyzed. Compared with control mice, an elevated number of EpCAM(+) cells with characteristics of HPCs was observed in HBx mice after 1 month and 4 months of DDC diet feeding. All HBx transgenic mice developed liver tumors characterized by histological features of both hepatocellular carcinoma (HCC) and cholangiocarcinoma after 7 months of DDC feeding. Notably, EpCAM(+) HPCs isolated from premalignant HBx mice exposed to a DDC diet for 4 months formed subcutaneous mixed-lineage tumors (four out of six) in nonobese diabetic/severe-combined immunodeficient (NOD/SCID) mice, and none of the cells from wildtype (WT) induced tumor, indicating that HBx may induce malignant transformation of HPCs that contributes to tumorigenesis. We also found higher titers of circulating interleukin (IL)-6, activities of IL-6/STAT3, and Wnt/β-catenin signaling pathways in HBx transgenic mice, suggesting HBx may induce intrinsic changes in HPCs by way of the above signaling that enables HPCs with tumorigenicity potential. Finally, clinical evidence showed that high HBx expression in human HBV-related HCC was statistically associated with expansion of EpCAM(+) or OV6(+) tumor cells and aggressive clinicopathologic features. CONCLUSION HBx induces intrinsic cellular transformation promoting the expansion and tumorigenicity of HPCs in DDC-treated mice, which may be a possible origin for liver cancer induced by chronic hepatitis infection.
Collapse
Affiliation(s)
- Chao Wang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
236
|
Rong Y, Wang YX, Sun LX, Zhang XY, Yang ZH, Ran YL, Sun LC. CD117-positive cells in human hepatocarcinoma cell line HepG2: isolation and evaluation of their stem cell properties. Shijie Huaren Xiaohua Zazhi 2011; 19:3649-3655. [DOI: 10.11569/wcjd.v19.i36.3649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To isolate CD117+ cells from human hepatocarcinoma cell line HepG2 and to observe their biological behavior and stem cell properties.
METHODS: HepG2 cells were cultured in serum-free medium. Flow cytometry was used to detect the expression of CD117 in HepG2 cells and sphere cells. CD117+ and CD117- cells were sorted by FACS and their ability of proliferation and self-renewal was observed in vitro. Chemosensitivity to cisplatin was determined by CCK-8 assay. The inhibitory rate of cells treated with cisplatin, the half maximal inhibitory concentration (IC50) and resistance index (RI) were measured.
RESULTS: HepG2 cells could survive, proliferate and form sphere cells in serum-free medium. The sphere-forming rate was 6.21% ± 2.03%. The percentage of CD117+ cells population in sphere cells increased by 9 folds compared to HepG2 cells. The sphere-forming ability and proliferation ability of CD117+ subpopulation in serum-free medium were significantly higher than un-sorted cells and CD117- cells. The inhibitory rate of CD117+ cells treated with cisplatin of different concentrations was lower than un-sorted cells and CD117- cells. The IC50 were 12.229 μmol/L in CD117+ cells, 7.970 μmol/L in un-sorted cells, and 7.345 μmol/L in CD117- cells, and the RI of CD117+ cells and un-sorted cells were 1.165 and 1.076, respectively.
CONCLUSION: CD117+ cells are a subpopulation of HepG2 cells with stem cell properties. CD117 may be a candidate surface marker for liver cancer stem cells.
Collapse
|
237
|
Li X, Jia Y, Zhang W, Zhang Y, Li B, Huang M, Bao F, Wu J, Lou Y. [The research progress about Wnt pathway of lung cancer stem cells]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2011; 14:695-8. [PMID: 21859553 PMCID: PMC5999629 DOI: 10.3779/j.issn.1009-3419.2011.08.11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Being the most critical signaling molecule in the Wnt pathway, the Wnt/β-catenin signaling pathway plays an important role in the maintenance of the cell proliferation and clone formation of lung cancer stem cells. Since it is closely related to the WNT pathway, the proliferation of lung cancer stem cells can be restrained by blocking the WNT pathway or influencing its key protein. Such method provides a new method for the treatment of lung cancer. By summarizing the state of-the-art research of lung cancer stem cells and the Wnt pathway from 2005 to 2010, their relationship is investigated.
Collapse
Affiliation(s)
- Xiaojiang Li
- Department of Oncology, the First Teaching Hospital, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | | | | | | | | | | | | | | | | |
Collapse
|
238
|
Xu W, Lin H, Zhang Y, Chen X, Hua B, Hou W, Qi X, Pei Y, Zhu X, Zhao Z, Yang L. Compound Kushen Injection suppresses human breast cancer stem-like cells by down-regulating the canonical Wnt/β-catenin pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2011; 30:103. [PMID: 22032476 PMCID: PMC3219673 DOI: 10.1186/1756-9966-30-103] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 10/28/2011] [Indexed: 11/10/2022]
Abstract
BACKGROUND Cancer stem cells (CSCs) play an important role in cancer initiation, relapse and metastasis. To date, no specific medicine has been found to target CSCs as they are resistant to most conventional therapies and proliferate indefinitely. Compound Kushen Injection (CKI) has been widely used for cancer patients with remarkable therapeutic effects in Chinese clinical settings for many years. This study focused on whether CKI could inhibit MCF-7 SP cells in vitro and in vivo. METHODS The analysis of CKI on SP population and the main genes of Wnt signaling pathway were studied first. Then we studied the tumorigenicity of SP cells and the effects of CKI on SP cells in vivo. The mice inoculated with 10,000 SP cells were randomly divided into three groups (6 in each group) and treated with CKI, cisplatin and saline (as a control) respectively for 7 weeks. The tumor formation rates of each group were compared. The main genes and proteins of the Wnt signaling pathway were analyzed by RT-PCR and western blot. RESULTS CKI suppressed the size of SP population (approximately 90%), and down-regulated the main genes of Wnt signaling pathway. We also determined that MCF-7 SP cells were more tumorigenic than non-SP and unsorted cells. The Wnt signaling pathway was up-regulated in tumors derived from SP cells compared with that in tumors from non-SP cells. The tumor formation rate of the CKI Group was 33% (2/6, P < 0.05), and that of Cisplatin Group was 50%(3/6, P < 0.05), whereas that of the Control Group was 100% (6/6).The RT-PCR and western blot results indicated that CKI suppressed tumor growth by down-regulating the Wnt/β-catenin pathway, while cisplatin activated the Wnt/β-catenin pathway and might spare SP cells. CONCLUSIONS It suggested that CKI may serve as a novel drug targeting cancer stem-like cells, though further studies are recommended.
Collapse
Affiliation(s)
- Weiru Xu
- Oncology Department, Guang An Men Hospital, China Academy of Chinese Medical Sciences, Xicheng District, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
239
|
Sun W, Ding J, Wu K, Ning BF, Wen W, Sun HY, Han T, Huang L, Dong LW, Yang W, Deng X, Li Z, Wu MC, Feng GS, Xie WF, Wang HY. Gankyrin-mediated dedifferentiation facilitates the tumorigenicity of rat hepatocytes and hepatoma cells. Hepatology 2011; 54:1259-72. [PMID: 21735473 DOI: 10.1002/hep.24530] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
UNLABELLED Gankyrin is a critical oncoprotein overexpressed in human hepatocellular carcinoma (HCC). However, the mechanism underlying gankyrin-mediated hepatocarcinogenesis remains elusive. Herein, we provide evidence that gankyrin expression was progressively elevated in liver fibrosis, cirrhosis, and HCC. Levels of gankyrin expression were closely associated with the dedifferentiation status of hepatoma in patients. Decrease of hepatocyte characteristic markers and increase of cholangiocyte-specific markers were observed in rat primary hepatocytes with enforced gankyrin expression and diethylnitrosamine (DEN)-triggered rat hepatocarcinogenesis. Overexpression of gankyrin also attenuated the hepatic function of primary hepatocytes, which further suggests that gankyrin promotes the dedifferentiation of hepatocytes. Moreover, elevated expression of gankyrin closely correlated with the expression of HCC stem/progenitor cell markers in DEN-triggered hepatocarcinogenesis and human HCCs. Hepatoma cells derived from suspension-cultured spheroids exhibited a higher gankyrin level, and enforced gankyrin expression in hepatoma cells remarkably enhanced cluster of differentiation (CD)133, CD90, and epithelial cellular adhesion molecule expression, indicating a role of gankyrin in hepatoma cell dedifferentiation and the generation of hepatoma stem/progenitor cells. In contrast, down-regulation of gankyrin in hepatoma cells by lentivirus-mediated microRNA delivery significantly improved their differentiation status and attenuated malignancy. Interference of gankyrin expression in hepatoma cells also diminished the proportion of cancer stem/progenitor cells and their self-renewal capacity. Furthermore, gankyrin was found to bind hepatocyte nuclear factor 4α (HNF4α), which determines hepatocyte differentiation status and enhances proteasome-dependent HNF4α degradation in hepatoma cells. The inverse correlation of gankyrin and HNF4α was further confirmed in primary hepatocytes, DEN-induced hepatocarcinogenesis, and human HCCs. CONCLUSION Gankyrin-mediated dedifferentiation of hepatocytes and hepatoma cells via, at least partially, down-regulation of HNF4α facilitates HCC development, and interference of gankyrin expression could be a novel strategy for HCC prevention and differentiation therapy.
Collapse
Affiliation(s)
- Wen Sun
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
240
|
Marquardt JU, Raggi C, Andersen JB, Seo D, Avital I, Geller D, Lee YH, Kitade M, Holczbauer A, Gillen MC, Conner EA, Factor VM, Thorgeirsson SS. Human hepatic cancer stem cells are characterized by common stemness traits and diverse oncogenic pathways. Hepatology 2011; 54:1031-42. [PMID: 21618577 PMCID: PMC3179780 DOI: 10.1002/hep.24454] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 05/15/2011] [Indexed: 12/25/2022]
Abstract
UNLABELLED Epigenetic mechanisms play critical roles in stem cell biology by maintaining pluripotency of stem cells and promoting differentiation of more mature derivatives. If similar mechanisms are relevant for the cancer stem cell (CSC) model, then epigenetic modulation might enrich the CSC population, thereby facilitating CSC isolation and rigorous evaluation. To test this hypothesis, primary human cancer cells and liver cancer cell lines were treated with zebularine (ZEB), a potent DNA methyltransferase-1 inhibitor, and putative CSCs were isolated using the side population (SP) approach. The CSC properties of ZEB-treated and untreated subpopulations were tested using standard in vitro and in vivo assays. Whole transcriptome profiling of isolated CSCs was performed to generate CSC signatures. Clinical relevance of the CSC signatures was evaluated in diverse primary human cancers. Epigenetic modulation increased frequency of cells with CSC properties in the SP fraction isolated from human cancer cells as judged by self-renewal, superior tumor-initiating capacity in serial transplantations, and direct cell tracking experiments. Integrative transcriptome analysis revealed common traits enriched for stemness-associated genes, although each individual CSC gene expression signature exhibited activation of different oncogenic pathways (e.g., EGFR, SRC, and MYC). The common CSC signature was associated with malignant progression, which is enriched in poorly differentiated tumors, and was highly predictive of prognosis in liver and other cancers. CONCLUSION Epigenetic modulation may provide a tool for prospective isolation and in-depth analysis of CSC. The liver CSC gene signatures are defined by a pernicious interaction of unique oncogene-specific and common stemness traits. These data should facilitate the identifications of therapeutic tools targeting both unique and common features of CSCs.
Collapse
Affiliation(s)
- Jens U. Marquardt
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH
| | - Chiara Raggi
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH
| | - Jesper B. Andersen
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH
| | - Daekwan Seo
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH
| | - Itzhak Avital
- Surgery Branch, Center for Cancer Research, National Cancer Institute, NIH
| | - David Geller
- UPMC Liver Cancer Center, University of Pittsburgh
| | - Yun-Han Lee
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH
| | - Mitsuteru Kitade
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH
| | - Agnes Holczbauer
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH
| | - Matthew C. Gillen
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH
| | - Elizabeth A. Conner
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH
| | - Valentina M. Factor
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH
| | - Snorri S. Thorgeirsson
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH
| |
Collapse
|
241
|
Lahijani MS, Farivar S, Khodaeian M. Effects of 50 Hz electromagnetic fields on the histology, apoptosis, and expression ofc-Fosandβ-Cateninon the livers of preincubated white leghorn chicken embryos. Electromagn Biol Med 2011; 30:158-69. [DOI: 10.3109/15368378.2011.596603] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
242
|
Zhu YZ, Fu D, Liu LL, Ma YS, Shen XZ, Chen XM. A high-throughput two-dimensional screening technique for cellular recognition and localization in hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2011; 19:2455-2460. [DOI: 10.11569/wcjd.v19.i23.2455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To establish a novel high-throughput two-dimensional screening technique for cellular recognition and localization in hepatocellular carcinoma (HCC).
METHODS: HCC specimen was collected from a patient who underwent radical resection. The paraffin-embedded specimen was serially sectioned at a thickness of 1 μm. Five serial sections were used for staining: one for H&E staining and the other four for immunofluorescence staining for detecting eight reported liver cancer stem cell (LCSC) markers. Fluorescein isothiocyanate (FITC) and tetramethyl rhodamine isothiocyanate (rhodamine) were used for fluorescent imaging for double staining. The sections were counterstained with Hoechst33342 to demonstrate the nuclei for cellular localization. Fluorescence microscopy was used to detect the fluorescence intensity and localization.
RESULTS: We identified the valid number of cells in a visual field (1 × 100) of microscopy and delineated the merged cell map. The results showed that 8 LCSCs biomarkers could be detected in 2 772 valid cells. The expression levels of biomarkers were different in these cells and a single valid cell could express 0-8 biomarkers. No biomarkers could be detected in 2 453 cells (88.5%).
CONCLUSION: A high-throughput two-dimensional screening technique for cellular recognition and localization has been successfully developed and can be used to detect the expression of two or more LCSC markers in one liver cancer cell.
Collapse
|
243
|
Liu LL, Fu D, Ma Y, Shen XZ. The power and the promise of liver cancer stem cell markers. Stem Cells Dev 2011; 20:2023-30. [PMID: 21651381 DOI: 10.1089/scd.2011.0012] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Recently, there has been growing support for the cancer stem cell (CSC) hypothesis, which states that primary tumors are initiated and maintained by a small subpopulation of cancer cells that possess "stem-like" characteristics. CSCs have been identified in many tumor types, including hepatocellular carcinoma (HCC). The dye, Hoechst 33342, has been used to enrich CSCs into a side population. Alternatively, liver CSCs (LCSCs) can be identified by several cell surface antigens, including CD133, CD90, CD44, EpCAM, and CD13. In this review, we summarized the recent evidence regarding LCSC markers and discussed the origin and function of these markers. LCSC markers are essential to identify and isolate these cells, to develop future therapies targeting CSCs, and to predict prognosis and efficacy of these therapies. However, definite LCSC markers are still controversial, because none of these markers is exclusively expressed by LCSCs in HCC. By combining several positive or negative markers, it may be possible to isolate and identify CSC fractions beyond the ability of each individual assay. By grouping LCSC markers according to their cellular origin, the properties of LCSC markers may be better studied and new markers may be found. Lastly, markers could be used to estimate the number of LCSCs and therefore predict outcomes. From our point of view, selecting HCC tissue samples from patients with different prognoses and detecting expression patterns of marker combinations may be a new method to identify new and unique markers.
Collapse
Affiliation(s)
- Li-Li Liu
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | | | | | | |
Collapse
|
244
|
Abstract
Hepatocellular carcinoma (HCC) is the most commonly diagnosed malignancy of the liver and is the third most frequent cause of cancer death worldwide. Although advances in HCC detection and treatment have increased the likelihood of a cure at early stages of the disease, HCC remains largely incurable because of late presentation and tumor recurrence. Only 25% of HCC patients are deemed suitable for curative treatment, with the overall survival at just a few months for inoperable patients. Additionally, this disease is particularly difficult to treat because of the high recurrence rate, its chemotherapy-resistant nature and the premalignant nature of surrounding cirrhotic liver disease. In the past few years, compelling evidence has emerged in support of the hierarchic cancer stem cell (CSC)/tumor-initiating cell (T-IC) model for solid tumors, including HCC. Understanding the characteristics and function of CSCs in the liver has also shed light on HCC management and treatment, including the implications for prognosis, prediction and treatment resistance. In this review, a detailed summary of the recent progress in liver CSC research with regard to identification, regulation and therapeutic implications will be discussed.
Collapse
Affiliation(s)
- Carol Man Tong
- Department of Pathology, Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | | | | |
Collapse
|
245
|
Dong LW, Yang GZ, Pan YF, Chen Y, Tan YX, Dai RY, Ren YB, Fu J, Wang HY. The oncoprotein p28GANK establishes a positive feedback loop in β-catenin signaling. Cell Res 2011; 21:1248-61. [PMID: 21691299 DOI: 10.1038/cr.2011.103] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
p28(GANK) (also known as PSMD10 or gankyrin) is a novel oncoprotein that is highly expressed in hepatocellular carcinoma (HCC). Through its interaction with various proteins, p28(GANK) mediates the degradation of the tumor suppressor proteins Rb and p53. Although p53 was reported to downregulate β-catenin, whether p28(GANK) is involved in the regulation of β-catenin remains uncertain. Here we report that both growth factors and Ras upregulate p28(GANK) expression through the activation of the phosphoinositide 3-kinase-AKT pathway. Upregulation of p28(GANK) expression subsequently enhanced the transcription activity of β-catenin. This effect was observed in p53-deficient cells, suggesting a p53-independent mechanism for the p28(GANK)-mediated activation of β-catenin. p28(GANK) overexpression also reduced E-cadherin protein levels, leading to increased release of free β-catenin into the cytoplasm from the cadherin-bound pool. Interestingly, exogenous expression of p28(GANK) resulted in elevated expression of the endogenous protein. We also observed that both β-catenin and c-Myc were transcriptional activators of p28(GANK), and a correlation between p28(GANK) overexpression and c-Myc, cyclin D1 and β-catenin activation in primary human HCC. Together, these results suggest that p28(GANK) expression is regulated by a positive feedback loop involving β-catenin, which may play a critical role in tumorigenesis and the progression of HCC.
Collapse
Affiliation(s)
- Li-wei Dong
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, 225 Changhai Road, Shanghai 200438, China
| | | | | | | | | | | | | | | | | |
Collapse
|
246
|
Cao L, Zhou Y, Zhai B, Liao J, Xu W, Zhang R, Li J, Zhang Y, Chen L, Qian H, Wu M, Yin Z. Sphere-forming cell subpopulations with cancer stem cell properties in human hepatoma cell lines. BMC Gastroenterol 2011; 11:71. [PMID: 21669008 PMCID: PMC3136412 DOI: 10.1186/1471-230x-11-71] [Citation(s) in RCA: 211] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Accepted: 06/14/2011] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are regarded as the cause of tumor formation and recurrence. The isolation and identification of CSCs could help to develop novel therapeutic strategies specifically targeting CSCs. METHODS Human hepatoma cell lines were plated in stem cell conditioned culture system allowed for sphere forming. To evaluate the stemness characteristics of spheres, the self-renewal, proliferation, chemoresistance, tumorigenicity of the PLC/PRF/5 sphere-forming cells, and the expression levels of stem cell related proteins in the PLC/PRF/5 sphere-forming cells were assessed, comparing with the parental cells. The stem cell RT-PCR array was performed to further explore the biological properties of liver CSCs. RESULTS The PLC/PRF/5, MHCC97H and HepG2 cells could form clonal nonadherent 3-D spheres and be serially passaged. The PLC/PRF/5 sphere-forming cells possessed a key criteria that define CSCs: persistent self-renewal, extensive proliferation, drug resistance, overexpression of liver CSCs related proteins (Oct3/4, OV6, EpCAM, CD133 and CD44). Even 500 sphere-forming cells were able to form tumors in NOD/SCID mice, and the tumor initiating capability was not decreased when spheres were passaged. Besides, downstream proteins DTX1 and Ep300 of the CSL (CBF1 in humans, Suppressor of hairless in Drosophila and LAG1 in C. elegans) -independent Notch signaling pathway were highly expressed in the spheres, and a gamma-secretase inhibitor MRK003 could significantly inhibit the sphere formation ability. CONCLUSIONS Nonadherent tumor spheres from hepatoma cell lines cultured in stem cell conditioned medium possess liver CSC properties, and the CSL-independent Notch signaling pathway may play a role in liver CSCs.
Collapse
Affiliation(s)
- Lu Cao
- Department of Molecular Oncology, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Procházková J, Kabátková M, Bryja V, Umannová L, Bernatík O, Kozubík A, Machala M, Vondráček J. The Interplay of the Aryl Hydrocarbon Receptor and β-Catenin Alters Both AhR-Dependent Transcription and Wnt/β-Catenin Signaling in Liver Progenitors. Toxicol Sci 2011; 122:349-60. [DOI: 10.1093/toxsci/kfr129] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
248
|
Roskams T. Anatomic pathology of hepatocellular carcinoma: impact on prognosis and response to therapy. Clin Liver Dis 2011; 15:245-59, vii-x. [PMID: 21689611 DOI: 10.1016/j.cld.2011.03.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A better understanding of signaling pathways in HCC pathogenesis has led to targeted therapies against HCC. Identification of liver cancer stem cell markers and their related pathways is one of the most important goals of liver cancer research. New therapies should ideally target cancer stem cells and not normal stem/progenitor cells, because the latter are very important in regeneration and repair. Individualized HCC therapy will require better definition of patient subgroups that benefit most or should be protected from therapy failure and unwanted side effects. Tumor tissue acquisition should be mandatory, reversing the practice that was established years ago when targeted HCC therapy was but a pipe dream.
Collapse
Affiliation(s)
- Tania Roskams
- Department of Pathology, Laboratory of Morphology and Molecular Pathology, University Hospitals of Leuven, Minderbroederstraat 12, B-3000 Leuven, Belgium.
| |
Collapse
|
249
|
Xu C, Zhang X, Wang G, Chang C, Zhang L, Cheng Q, Lu A. Role of the autonomic nervous system in rat liver regeneration. Cell Mol Neurobiol 2011; 31:527-40. [PMID: 21264506 PMCID: PMC11498377 DOI: 10.1007/s10571-011-9646-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Accepted: 01/04/2011] [Indexed: 01/12/2023]
Abstract
To study the regulatory role of autonomic nervous system in rat regenerating liver, surgical operations of rat partial hepatectomy (PH) and its operation control (OC), sympathectomy combining partial hepatectomy (SPH), vagotomy combining partial hepatectomy (VPH), and total liver denervation combining partial hepatectomy (TDPH) were performed, then expression profiles of regenerating livers at 2 h after operation were detected using Rat Genome 230 2.0 array. It was shown that the expressions of 97 genes in OC, 230 genes in PH, 253 genes in SPH, 187 genes in VPH, and 177 genes in TDPH were significantly changed in biology. The relevance analysis showed that in SPH, genes involved in stimulus response, immunity response, amino acids and K(+) transport, amino acid catabolism, cell adhesion, cell proliferation mediated by JAK-STAT, Ca(+), and platelet-derived growth factor receptor, cell growth and differentiation through JAK-STAT were up-regulated, while the genes involved in chromatin assembly and disassembly, and cell apoptosis mediated by MAPK were down-regulated. In VPH, the genes associated with chromosome modification-related transcription factor, oxygen transport, and cell apoptosis mediated by MAPK pathway were up-regulated, but the genes associated with amino acid catabolism, histone acetylation-related transcription factor, and cell differentiation mediated by Wnt pathway were down-regulated. In TDPH, the genes related to immunity response, growth and development of regenerating liver, cell growth by MAPK pathway were up-regulated. Our data suggested that splanchnic and vagal nerves could regulate the expressions of liver regeneration-related genes.
Collapse
Affiliation(s)
- Cunshuan Xu
- College of Life Science, Henan Normal University, No. 46, Construction East Road, Xinxiang, 453007, Henan, China.
| | | | | | | | | | | | | |
Collapse
|
250
|
Oishi N, Wang XW. Novel therapeutic strategies for targeting liver cancer stem cells. Int J Biol Sci 2011; 7:517-35. [PMID: 21552419 PMCID: PMC3088875 DOI: 10.7150/ijbs.7.517] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 04/14/2011] [Indexed: 12/15/2022] Open
Abstract
The cancer stem cell (CSC) hypothesis was first proposed over 40 years ago. Advances in CSC isolation were first achieved in hematological malignancies, with the first CSC demonstrated in acute myeloid leukemia. However, using similar strategies and technologies, and taking advantage of available surface markers, CSCs have been more recently demonstrated in a growing range of epithelial and other solid organ malignancies, suggesting that the majority of malignancies are dependent on such a compartment. Primary liver cancer consists predominantly of hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). It is believed that hepatic progenitor cells (HPCs) could be the origin of some HCCs and ICCs. Furthermore, stem cell activators such as Wnt/β-catenin, TGF-β, Notch and Hedgehog signaling pathways also expedite tumorigenesis, and these pathways could serve as molecular targets to assist in designing cancer prevention strategies. Recent studies indicate that additional factors such as EpCAM, Lin28 or miR-181 may also contribute to HCC progression by targeting HCC CSCs. Various therapeutic drugs that directly modulate CSCs have been examined in vivo and in vitro. However, CSCs clearly have a complex pathogenesis, with a considerable crosstalk and redundancy in signaling pathways, and hence targeting single molecules or pathways may have a limited benefit for treatment. Many of the key signaling molecules are shared by both CSCs and normal stem cells, which add further challenges for designing molecularly targeted strategies specific to CSCs but sparing normal stem cells to avoid side effects. In addition to the direct control of CSCs, many other factors that are needed for the maintenance of CSCs, such as angiogenesis, vasculogenesis, invasion and migration, hypoxia, immune evasion, multiple drug resistance, and radioresistance, should be taken into consideration when designing therapeutic strategies for HCC. Here we provide a brief review of molecular signaling in liver CSCs and present insights into new therapeutic strategies for targeting liver CSCs.
Collapse
Affiliation(s)
- Naoki Oishi
- Liver Carcinogenesis Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892-4258, USA
| | | |
Collapse
|