401
|
Lee TKW, Cheung VCH, Lu P, Lau EYT, Ma S, Tang KH, Tong M, Lo J, Ng IOL. Blockade of CD47-mediated cathepsin S/protease-activated receptor 2 signaling provides a therapeutic target for hepatocellular carcinoma. Hepatology 2014; 60:179-91. [PMID: 24523067 DOI: 10.1002/hep.27070] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 01/30/2014] [Accepted: 02/07/2014] [Indexed: 12/12/2022]
Abstract
UNLABELLED Identification of therapeutic targets against tumor-initiating cells (TICs) is a priority in the development of new therapeutic paradigms against cancer. We enriched a TIC population capable of tumor initiation and self-renewal by serial passages of hepatospheres with chemotherapeutic agents. In chemoresistant hepatospheres, CD47 was found to be up-regulated, when compared with differentiated progenies. CD47 is preferentially expressed in liver TICs, which contributed to tumor initiation, self-renewal, and metastasis and significantly affected patients' clinical outcome. Knockdown of CD47 suppressed stem/progenitor cell characteristics. CD47(+) hepatocellular carcinoma (HCC) cells preferentially secreted cathepsin S (CTSS), which regulates liver TICs through the CTSS/protease-activated receptor 2 (PAR2) loop. Suppression of CD47 by morpholino approach suppressed growth of HCC in vivo and exerted a chemosensitization effect through blockade of CTSS/PAR2 signaling. CONCLUSION These data suggest that CD47 may be an attractive therapeutic target for HCC therapy.
Collapse
Affiliation(s)
- Terence Kin-Wah Lee
- State Key Laboratory for Liver Research, The University of Hong Kong, Pokfulam, Hong Kong; Departments of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
| | | | | | | | | | | | | | | | | |
Collapse
|
402
|
Lu W, Feng F, Xu J, Lu X, Wang S, Wang L, Lu H, Wei M, Yang G, Wang L, Lu Z, Liu Y, Lei X. QKI impairs self-renewal and tumorigenicity of oral cancer cells via repression of SOX2. Cancer Biol Ther 2014; 15:1174-84. [PMID: 24918581 DOI: 10.4161/cbt.29502] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Cancer stem cells (CSCs) may contribute to tumor initiation, distant metastasis and chemo-resistance. One of RNA-binding proteins, Quaking (QKI), was reported to be a tumor suppressor. Here we showed that reduced QKI levels were observed in many human oral cancer samples. Moreover further reduction of QKI expression in CSCs was detected compared with non-CSCs in oral cancer cell lines. Overexpressing QKI in oral cancer cells significantly reduced CSC sphere formation and stem cell-associated genes. In tumor implanting nude mice model, QKI significantly impeded tumor initiation rates, tumor sizes and lung metastasis rates. As a contrast, knocking down QKI enhanced the above effects. Among the putative CSC target genes, SOX2 expression was negatively affected by QKI, mechanism study revealed that QKI may directly regulate SOX2 expression via specific binding with its 3'UTR in a cis element-dependent way. Loss of SOX2 even completely reversed the sphere forming ability in QKI knockdown cell line. Taken together, these data demonstrated that SOX2 is an important CSC regulator in oral cancer. QKI is a novel CSC inhibitor and impaired multiple oral CSC properties via partial repression of SOX2. Therefore, reduced expression of QKI may provide a novel diagnostic marker for oral cancer.
Collapse
Affiliation(s)
- Wei Lu
- Department of Oral and Maxillofacial Surgery; School of Stomatology; the Fourth Military Medical University; Xi'an, PR China; Department of Stomatology; 101 Hospital of PLA; Wuxi, PR China
| | - Feixue Feng
- The State Key Laboratory of Cancer Biology; Department of Pharmacogenomics; the Fourth Military Medical University; Xi'an, PR China
| | - Jinke Xu
- Department of Oral and Maxillofacial Surgery; School of Stomatology; the Fourth Military Medical University; Xi'an, PR China
| | - Xiaozhao Lu
- The State Key Laboratory of Cancer Biology; Department of Pharmacogenomics; the Fourth Military Medical University; Xi'an, PR China
| | - Shan Wang
- The State Key Laboratory of Cancer Biology; Department of Pharmacogenomics; the Fourth Military Medical University; Xi'an, PR China
| | - Lifeng Wang
- The State Key Laboratory of Cancer Biology; Department of Biochemistry and Molecular Biology; the Fourth Military Medical University; Xi'an, PR China
| | - Huanyu Lu
- The State Key Laboratory of Cancer Biology; Department of Pharmacogenomics; the Fourth Military Medical University; Xi'an, PR China
| | - Mengying Wei
- The State Key Laboratory of Cancer Biology; Department of Biochemistry and Molecular Biology; the Fourth Military Medical University; Xi'an, PR China
| | - Guodong Yang
- The State Key Laboratory of Cancer Biology; Department of Biochemistry and Molecular Biology; the Fourth Military Medical University; Xi'an, PR China
| | - Li Wang
- The State Key Laboratory of Cancer Biology; Department of Pharmacogenomics; the Fourth Military Medical University; Xi'an, PR China
| | - Zifan Lu
- The State Key Laboratory of Cancer Biology; Department of Pharmacogenomics; the Fourth Military Medical University; Xi'an, PR China
| | - Yanpu Liu
- Department of Oral and Maxillofacial Surgery; School of Stomatology; the Fourth Military Medical University; Xi'an, PR China
| | - Xiaoying Lei
- The State Key Laboratory of Cancer Biology; Department of Pharmacogenomics; the Fourth Military Medical University; Xi'an, PR China
| |
Collapse
|
403
|
Liu M, Jiang L, Guan XY. The genetic and epigenetic alterations in human hepatocellular carcinoma: a recent update. Protein Cell 2014. [PMID: 24916440 DOI: 10.1007/s13238- 014-0065-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most frequent human malignancies worldwide with very poor prognosis. It is generally accepted that the progression of HCC is a long-term process with accumulation of multiple genetic and epigenetic alterations, which further lead to the activation of critical oncogenes or inactivation of tumor suppressor genes. HCC is characterized with multiple cancer hallmarks including their ability to proliferate, anti-apoptosis, invade, metastasis, as well as the emerging features such as stem cell properties and energy metabolic switch. The irreversible alterations at genetic level could be detected as early as in the pre-neoplastic stages and accumulate during cancer progression. Thus, they might account for the cancer initiating steps and further malignant transformation. In addition to genetic alterations, epigenetic alterations can affect the cancer transcriptome more extensively. Alterations in DNA methylation, histone modification, miRNAs, RNA editing, and lncRNAs might result in disrupted gene regulation networks and substantially contribute to HCC progression. In this review, the genetic and epigenetic alterations which significantly contribute to the malignant capabilities of HCC will be updated and summarized in detail. Further characterization of those critical molecular events might better elucidate the pathogenesis of HCC and provide novel therapeutic targets for treatment of this deadly disease.
Collapse
Affiliation(s)
- Ming Liu
- Department of Clinical Oncology, University of Hong Kong, Hong Kong, China
| | | | | |
Collapse
|
404
|
Yan S, Xu D, Jiang T, Wang P, Yin Y, Wang X, Hua C, Zhang B, Li Z, Lu L, Liu X, Wang B, Zhang D, Zhang R, Sun B, Wang X. CD24 single nucleotide polymorphisms and cancer risk. Tumour Biol 2014; 35:8927-32. [PMID: 24894672 DOI: 10.1007/s13277-014-2127-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 05/20/2014] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED Cluster of differentiation 24 (CD24) has been implicated in the development of cancer. Several single nucleotide polymorphisms (SNPs) in CD24 gene are reported to exert diverse effect on cancer risk. However, the association between CD24 SNPs and cancer risk remains unclear due to contradictory published findings. We performed a meta-analysis by pooling all available published studies on the susceptibility of CD24 rs52812045 and rs3838646 polymorphisms to cancer. The pooled odds ratios (ORs) with 95 % confidence intervals (95 % CIs) were calculated. There were five independent case-control studies with 5,539 cases and 10,241 controls included into the present study. The pooled results showed that no appreciable relationship was identified between any of the SNPs of CD24 and cancer risk. Interestingly, a protective role of the CD24 rs3838646 polymorphism was found in the risk of breast cancer, but lack of statistical significance (del allele vs. TG allele: OR = 0.89; 95 % CI, 0.79-1.01; P OR = 0.063; del/del vs. TG/TG OR = 0.70; 95 % CI, 0.44-1.12; P OR = 0.135; del/TG vs. TG/TG OR = 0.91; 95 % CI, 0.80-1.04, P OR = 0.180; del/del + del/TG vs. TG/TG OR = 0.90; 95 % CI, 0.79-1.03; P OR = 0.123; del/del vs. TG/TG + del/TG: OR = 0.69; 95 % CI, 0.44-1.08, P OR = 0.105). Our study firstly provides the evidence that SNPs (rs52812045 and rs3838646) of CD24 may not modify the risk of cancer. Nonetheless, more individual studies with high quality are needed for further elucidation.
Collapse
Affiliation(s)
- Shushan Yan
- Department of Surgical Oncology, The Eighty-First Hospital of People's Liberation Army, Nanjing, Jiangsu Province, 210002, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
405
|
Raggi C, Factor VM, Seo D, Holczbauer A, Gillen MC, Marquardt JU, Andersen JB, Durkin M, Thorgeirsson SS. Epigenetic reprogramming modulates malignant properties of human liver cancer. Hepatology 2014; 59:2251-62. [PMID: 24449497 PMCID: PMC4043911 DOI: 10.1002/hep.27026] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 01/06/2014] [Accepted: 01/15/2014] [Indexed: 01/27/2023]
Abstract
UNLABELLED Reversal of DNA hypermethylation and associated gene silencing is an emerging cancer therapy approach. Here we addressed the impact of epigenetic alterations and cellular context on functional and transcriptional reprogramming of hepatocellular carcinoma (HCC) cells. Our strategy employed a 3-day treatment of established and primary human HCC-derived cell lines grown as a monolayer at various cell densities with the DNMT1 inhibitor zebularine (ZEB) followed by a 3D culture to identify cells endowed with self-renewal potential. Differences in self-renewal, gene expression, tumorigenicity, and metastatic potential of spheres at generations G1-G5 were examined. Transient ZEB exposure produced differential cell density-dependent responses. In cells grown at low density, ZEB caused a remarkable increase in self-renewal and tumorigenicity associated with long-lasting gene expression changes characterized by a stable overexpression of cancer stem cell-related and key epithelial-mesenchymal transition genes. These effects persisted after restoration of DNMT1 expression. In contrast, at high cell density, ZEB caused a gradual decrease in self-renewal and tumorigenicty, and up-regulation of apoptosis- and differentiation-related genes. A permanent reduction of DNMT1 protein using short hairpin RNA (shRNA)-mediated DNMT1 silencing rendered HCC cells insensitive both to cell density and ZEB effects. Similarly, WRL68 and HepG2 hepatoblastoma cells expressing low DNMT1 basal levels also possessed a high self-renewal, irrespective of cell density or ZEB exposure. Spheres formed by low-density cells treated with ZEB or shDNMT1 displayed a high molecular similarity which was sustained through consecutive generations, confirming the essential role of DNMT1 depletion in the enhancement of cancer stem cell properties. CONCLUSION These results identify DNA methylation as a key epigenetic regulatory mechanism determining the pool of cancer stem cells in liver cancer and possibly other solid tumors.
Collapse
Affiliation(s)
- Chiara Raggi
- 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
| | - Daekwan Seo
- 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
| | - Jens U. Marquardt
- 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
| | - Marian Durkin
- 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,Corresponding author: Snorri S. Thorgeirsson
| |
Collapse
|
406
|
Huang CY, Tai WT, Hsieh CY, Hsu WM, Lai YJ, Chen LJ, Shiau CW, Chen KF. A sorafenib derivative and novel SHP-1 agonist, SC-59, acts synergistically with radiotherapy in hepatocellular carcinoma cells through inhibition of STAT3. Cancer Lett 2014; 349:136-43. [PMID: 24735751 DOI: 10.1016/j.canlet.2014.04.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 03/31/2014] [Accepted: 04/07/2014] [Indexed: 12/17/2022]
Abstract
Radiotherapy shows limited benefit as treatment for hepatocellular carcinoma (HCC). In this study, we aimed to overcome the radioresistance of HCC by using a novel sorafenib derivative, SC-59 that targets SHP-1-related signaling. HCC cell lines (SK-Hep1, Hep3B, and Huh7) were treated with sorafenib, SC-59, radiation, sorafenib plus radiation, or SC-59 plus radiation, and then apoptosis, colony formation, signal transduction and the phosphatase activity were analyzed. The synergistic effect of radiotherapy and SC-59 was analyzed using a combination index (CI) approach. In vivo efficacy was determined in a Huh7-bearing subcutaneous model. Mice were treated with radiation (5 Gy, one fraction per day) for 4 days, SC-59 (10mg/kg/day) for 24 days, or a combination. Tumor samples were further analyzed for p-STAT3 and SHP-1 activity. SC-59 displayed a better synergistic effect when used in combination with radiotherapy than sorafenib in HCC cell lines. SC-59 downregulated p-STAT3 and its downstream targets and increased SHP-1 phosphatase activity. Both ectopic STAT3 and inhibition of SHP-1 abolished SC-59-induced radiosensitization. Moreover, SC-59 significantly synergized radiotherapy in a Huh7 xenograft model by targeting SHP-1/STAT3 signaling. The novel sorafenib derivative, SC-59, acting as a SHP-1 agonist, displays a better synergistic effect when used in combination with radiotherapy than sorafenib for the treatment of HCC. Further clinical investigation is warranted.
Collapse
Affiliation(s)
- Chao-Yuan Huang
- Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; Department of Radiological Technology, Yuanpei University, Hsinchu, Taiwan
| | - Wei-Tien Tai
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan; National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Chi-Ying Hsieh
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan; National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Wan-Mai Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan; National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Ying-Jiun Lai
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan; National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Li-Ju Chen
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan; National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Chung-Wai Shiau
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan.
| | - Kuen-Feng Chen
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan; National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
407
|
Yang T, Rycaj K, Liu ZM, Tang DG. Cancer stem cells: constantly evolving and functionally heterogeneous therapeutic targets. Cancer Res 2014; 74:2922-7. [PMID: 24713433 DOI: 10.1158/0008-5472.can-14-0266] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Elucidating the origin of and dynamic interrelationship between intratumoral cell subpopulations has clear clinical significance in helping to understand the cellular basis of treatment response, therapeutic resistance, and tumor relapse. Cancer stem cells (CSC), together with clonal evolution driven by genetic alterations, generate cancer cell heterogeneity commonly observed in clinical samples. The 2013 Shanghai International Symposium on Cancer Stem Cells brought together leaders in the field to highlight the most recent progress in phenotyping, characterizing, and targeting CSCs and in elucidating the relationship between the cell-of-origin of cancer and CSCs. Discussions from the symposium emphasize the urgent need in developing novel therapeutics to target the constantly evolving CSCs.
Collapse
Affiliation(s)
- Tao Yang
- Authors' Affiliations: Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China; and Department of Molecular Carcinogenesis, the University of Texas MD Anderson Cancer Center, Science Park, Smithville, Texas
| | - Kiera Rycaj
- Authors' Affiliations: Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China; and Department of Molecular Carcinogenesis, the University of Texas MD Anderson Cancer Center, Science Park, Smithville, Texas
| | - Zhong-Min Liu
- Authors' Affiliations: Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China; and Department of Molecular Carcinogenesis, the University of Texas MD Anderson Cancer Center, Science Park, Smithville, Texas
| | - Dean G Tang
- Authors' Affiliations: Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China; and Department of Molecular Carcinogenesis, the University of Texas MD Anderson Cancer Center, Science Park, Smithville, TexasAuthors' Affiliations: Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China; and Department of Molecular Carcinogenesis, the University of Texas MD Anderson Cancer Center, Science Park, Smithville, Texas
| |
Collapse
|
408
|
Zhang J, Pan YF, Ding ZW, Yang GZ, Tan YX, Yang C, Jiang TY, Liu LJ, Zhang B, Han T, Cao D, Yang T, Yang N, Wu MC, Dong LW, Wang HY. RMP promotes venous metastases of hepatocellular carcinoma through promoting IL-6 transcription. Oncogene 2014; 34:1575-83. [DOI: 10.1038/onc.2014.84] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 01/31/2014] [Accepted: 02/25/2014] [Indexed: 02/07/2023]
|
409
|
Tang J, Tao ZH, Wen D, Wan JL, Liu DL, Zhang S, Cui JF, Sun HC, Wang L, Zhou J, Fan J, Wu WZ. MiR-612 suppresses the stemness of liver cancer via Wnt/β-catenin signaling. Biochem Biophys Res Commun 2014; 447:210-5. [PMID: 24704424 DOI: 10.1016/j.bbrc.2014.03.135] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 03/26/2014] [Indexed: 12/12/2022]
Abstract
Previous research showed that microRNA-612 (miR-612) has inhibitory effects on cell proliferation, migration, invasion, and metastasis of hepatocellular carcinoma (HCC). AKT2 was confirmed to be a direct target of miR-612, through which the epithelial-mesenchymal transition (EMT) and metastasis of HCC were inhibited. Our present findings reveal that miR-612 is able to suppress the stemness of HCC by reducing the number and size of tumorspheres as well as clone formation in soft agar, and to relieve drug resistance to cisplatin and 5-fluorouracil. In addition, miR-612 hampered the capacity of tumorigenesis in NOD/SCID mice and redistributed the tumor invasive frontier of miR-612-modulating cells. Finally, our findings suggest that Wnt/β-catenin signaling is required in the regulation of EMT-associated stem cell-like traits by miR-612.
Collapse
Affiliation(s)
- Jun Tang
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Zhong-Hua Tao
- Department of Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai 200032, China
| | - Duo Wen
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Jin-Liang Wan
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Dong-Li Liu
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Shu Zhang
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Jie-Feng Cui
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Hui-Chuan Sun
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Lu Wang
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Jian Zhou
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China; Institute of Biomedical Sciences of Fudan University, Shanghai 200032, China
| | - Jia Fan
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China; Institute of Biomedical Sciences of Fudan University, Shanghai 200032, China
| | - Wei-Zhong Wu
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China.
| |
Collapse
|
410
|
Guo S, Singh KK, Lillard JW, Yang L. Leptin Signaling in the Regulation of Stem and Cancer Stem Cells. CANCER STEM CELLS 2014:347-360. [DOI: 10.1002/9781118356203.ch26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
411
|
Drosch M, Schmidt N, Markowski DN, Zollner TM, Koch M, Bullerdiek J. The CD24hi smooth muscle subpopulation is the predominant fraction in uterine fibroids. Mol Hum Reprod 2014; 20:664-76. [PMID: 24657878 DOI: 10.1093/molehr/gau022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Uterine fibroids are the most common gynecological tumors affecting women in their reproductive age. Despite this high incidence the pathogenesis of fibroids is widely unsolved. Whereas formerly only imbalances in hormonal levels were considered to account for tumor development, the identification of genetic changes likely to affect myometrial stem cell reservoirs provided a novel approach to fibroid genesis. Here, we identified a certain subset of cells by the surface marker CD24 with increased abundance in fibroids compared with myometrial tissue. Fibroid cells expressing CD24 shared certain features of immature or progenitor-like cells such as quiescence, reduced expression of smooth muscle differentiation markers and elevated expression of genes involved in the wingless-type (WNT)-pathway such as beta-catenin. In addition, a positive correlation between CD24 and wingless-type family member 4 (WNT4) expression was observed in uterine fibroids with mediator subcomplex 12 gene (MED12) mutations. Our findings suggest that cells highly expressing CD24 represent a type of immature smooth muscle progenitor cells. Their accumulation might be driven by disturbed differentiation processes caused by genetic changes possibly involving MED12 mutations or high mobility group AT-hook (HMGA)2 rearrangements.
Collapse
Affiliation(s)
- Michael Drosch
- Center of Human Genetics, University of Bremen, Leobener Strasse ZHG, Bremen 28359, Germany Global Drug Discovery-TRG Oncology/Gynecological Therapies, Bayer Healthcare Pharmaceuticals, Muellerstraße 178, Berlin 13342, Germany
| | - Nicole Schmidt
- Global Drug Discovery-TRG Oncology/Gynecological Therapies, Bayer Healthcare Pharmaceuticals, Muellerstraße 178, Berlin 13342, Germany
| | - Dominique Nadine Markowski
- Global Drug Discovery-TRG Oncology/Gynecological Therapies, Bayer Healthcare Pharmaceuticals, Muellerstraße 178, Berlin 13342, Germany
| | - Thomas Matthias Zollner
- Global Drug Discovery-TRG Oncology/Gynecological Therapies, Bayer Healthcare Pharmaceuticals, Muellerstraße 178, Berlin 13342, Germany
| | - Markus Koch
- Global Drug Discovery-TRG Oncology/Gynecological Therapies, Bayer Healthcare Pharmaceuticals, Muellerstraße 178, Berlin 13342, Germany
| | - Jörn Bullerdiek
- Center of Human Genetics, University of Bremen, Leobener Strasse ZHG, Bremen 28359, Germany
| |
Collapse
|
412
|
Zhou JJ, Deng XG, He XY, Zhou Y, Yu M, Gao WC, Zeng B, Zhou QB, Li ZH, Chen RF. Knockdown of NANOG enhances chemosensitivity of liver cancer cells to doxorubicin by reducing MDR1 expression. Int J Oncol 2014; 44:2034-40. [PMID: 24647772 DOI: 10.3892/ijo.2014.2347] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 03/04/2014] [Indexed: 11/05/2022] Open
Abstract
Multidrug resistance (MDR) is one of the major reasons for the failure of liver cancer chemotherapy, and its suppression may increase the efficacy of chemotherapy. NANOG plays a key role in the regulation of embryonic stem cell self-renewal and pluripotency. Recent studies reported that NANOG was abnormally expressed in several types of tumors, indicating that NANOG is related to tumor development. However, the correlation between NANOG and liver cancer chemoresistance remains uncertain. In this study, RNA interfere technology was employed to knock down NANOG expression in HepG2 human liver cancer cells. We found that the knockdown of NANOG expression in NANOG siRNA-transfected HepG2 cells resulted in decreased colony formation rate and cell migration compared to control HepG2 cells. In addition, HepG2 cells were treated with doxorubicin to evaluate the chemosensitivity to doxorubicin. We found that the doxorubicin sensitivity of HepG2 cells was increased with downregulation of NANOG expression. The expression of MDR1 at both mRNA and protein levels was decreased in HepG2 cells when NANOG was knocked down. These findings suggest that the knockdown of NANOG in HepG2 human cells resulted in decreased MDR1 expression and increased doxorubicin sensitivity, and NANOG could be used as a novel potential therapeutic target to reverse multidrug resistance of liver cancer.
Collapse
Affiliation(s)
- Jia-Jia Zhou
- Department of Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Xiao-Geng Deng
- Department of Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Xiao-Yu He
- Laboratory of Biomechanics and Physiology, Guangdong Provincial Institute of Sports Science, Guangzhou, Guangdong, P.R. China
| | - Yu Zhou
- Department of Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Min Yu
- Department of Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Wen-Chao Gao
- Department of Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Bing Zeng
- Department of Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Quan-Bo Zhou
- Department of Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Zhi-Hua Li
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Ru-Fu Chen
- Department of Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
413
|
Bu Y, Jia QA, Ren ZG, Zhang JB, Jiang XM, Liang L, Xue TC, Zhang QB, Wang YH, Zhang L, Xie XY, Tang ZY. Maintenance of stemness in oxaliplatin-resistant hepatocellular carcinoma is associated with increased autocrine of IGF1. PLoS One 2014; 9:e89686. [PMID: 24632571 PMCID: PMC3954560 DOI: 10.1371/journal.pone.0089686] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 01/23/2014] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Evidence suggests that many types of cancers are composed of different cell types, including cancer stem cells (CSCs). We have previously shown that the chemotherapeutic agent oxaliplatin induced epithelial-mesenchymal transition, which is thought to be an important mechanism for generating CSCs. In the present study, we investigate whether oxaliplatin-treated cancer tissues possess characteristics of CSCs, and explore oxaliplatin resistance in these tissues. METHODS Hepatocellular carcinoma cells (MHCC97H cells) were subcutaneously injected into mice to form tumors, and the mice were intravenously treated with either oxaliplatin or glucose. Five weeks later, the tumors were orthotopically xenografted into livers of other mice, and these mice were treated with either oxaliplatin or glucose. Metastatic potential, sensitivity to oxaliplatin, and expression of CSC-related markers in the xenografted tumor tissues were evaluated. DNA microarrays were used to measure changes in gene expression as a result of oxaliplatin treatment. Additionally, an oxaliplatin-resistant cell line (MHCC97H-OXA) was established to assess insulin-like growth factor 1 secretion, cell invasion, cell colony formation, oxaliplatin sensitivity, and expression of CSC-related markers. The effects of an insulin-like growth factor 1 receptor inhibitor were also assessed. RESULTS Oxaliplatin treatment inhibited subcutaneous tumor growth. Tumors from oxaliplatin-treated mice that were subsequently xenografted into livers of other mice exhibited that decreasing sensitivity to oxaliplatin and increasing pulmonary metastatic potential. Among the expression of CSC-related proteins, the gene for insulin-like growth factor 1, was up-regulated expecially in these tumor tissues. Additionally, MHCC97H-OXA cells demonstrated that increasing cell invasion, colony formation, and expression of insulin-like growth factor 1 and CSC-related markers, whereas treatment with an inhibitor of the insulin-like growth factor 1 receptor suppressed these effects. CONCLUSION Maintenance of stemness in oxaliplatin-resistant hepatocellular carcinoma cells is associated with increased autocrine of IGF1.
Collapse
Affiliation(s)
- Yang Bu
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Qing-An Jia
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
- Hepatobiliary Surgery, Shanxi Provincial People's Hospital, Xi'an, China
| | - Zheng-Gang Ren
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Ju-Bo Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Xue-Mei Jiang
- Department of Gastroenterology, Haikou People's Hospital, Haikou, China
| | - Lei Liang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Tong-Chun Xue
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Quan-Bao Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Yan-Hong Wang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Lan Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Xiao-Ying Xie
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Zhao-You Tang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
- * E-mail:
| |
Collapse
|
414
|
Hofner T, Macher-Goeppinger S, Klein C, Schillert A, Eisen C, Wagner S, Rigo-Watermeier T, Baccelli I, Vogel V, Trumpp A, Sprick MR. Expression and prognostic significance of cancer stem cell markers CD24 and CD44 in urothelial bladder cancer xenografts and patients undergoing radical cystectomy. Urol Oncol 2014; 32:678-86. [PMID: 24631171 DOI: 10.1016/j.urolonc.2014.01.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Revised: 12/15/2013] [Accepted: 01/02/2014] [Indexed: 01/19/2023]
Abstract
OBJECTIVES To evaluate CD24/CD44/CD47 cancer stem cell marker expressions in bladder cancer (BCa) and provide data on their prognostic significance for clinical outcome in patients undergoing radical cystectomy (RC). MATERIAL AND METHODS Primary BCa tissue was used for xenograft studies. A tissue microarray was prepared using specimens from a cohort of 132 patients. All patients underwent RC for urothelial BCa between 2001 and 2010. Expression of CD24, CD44, and CD47 was examined in primary samples and xenografts by fluorescence-activated cell sorting. Populations of CD24(low)- and CD24(high)-expressing cells were sorted and evaluated for tumorigenicity in vivo. Tissue microarray was analyzed for CD24/CD44 staining intensity and tumor-specific vs. stromal cell staining. Associations with BCa survival, BCa stage, and lymph node status were evaluated by univariate and multivariate analyses. RESULTS CD24 and CD44/CD47 expressions mark distinct cell populations within the normal urothelium as well as in BCa. CD24(high/low) expression was not sufficient to characterize CD24 as a BCa-initiating marker in in vivo primary xenotransplants. CD24 and CD44 expressions correlated with lower cancer-specific survival in patients. However, multivariate analyses of CD24 or CD44 did not demonstrate significantly increased hazards for cancer-specific death if analyzed together with stage, grade, and nodal status of patients. CONCLUSIONS Cancer stem cell markers CD24/CD44/CD47 are differentially expressed in cells of urothelial BCa in patients undergoing RC and influence cancer-specific survival of patients. Further evaluation of CD24/CD44/CD47 protein expression could be of high therapeutic value in BCa. However, both CD24 and CD44 expressions cannot be regarded as independent prognostic parameters for patients undergoing RC.
Collapse
Affiliation(s)
- Thomas Hofner
- Division of Stem Cells and Cancer, HI-STEM gGmbH, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Urology, University Hospital Heidelberg, Heidelberg, Germany.
| | - Stephan Macher-Goeppinger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Molecular Tumor Pathology, German Cancer Research Center, Heidelberg, Germany
| | - Corinna Klein
- Division of Stem Cells and Cancer, HI-STEM gGmbH, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anja Schillert
- Division of Stem Cells and Cancer, HI-STEM gGmbH, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian Eisen
- Division of Stem Cells and Cancer, HI-STEM gGmbH, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Steve Wagner
- Division of Stem Cells and Cancer, HI-STEM gGmbH, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Teresa Rigo-Watermeier
- Division of Stem Cells and Cancer, HI-STEM gGmbH, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Irène Baccelli
- Division of Stem Cells and Cancer, HI-STEM gGmbH, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Vanessa Vogel
- Division of Stem Cells and Cancer, HI-STEM gGmbH, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, HI-STEM gGmbH, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin R Sprick
- Division of Stem Cells and Cancer, HI-STEM gGmbH, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
415
|
Liu B, Badeaux MD, Choy G, Chandra D, Shen I, Jeter CR, Rycaj K, Lee CF, Person MD, Liu C, Chen Y, Shen J, Jung SY, Qin J, Tang DG. Nanog1 in NTERA-2 and recombinant NanogP8 from somatic cancer cells adopt multiple protein conformations and migrate at multiple M.W species. PLoS One 2014; 9:e90615. [PMID: 24598770 PMCID: PMC3944193 DOI: 10.1371/journal.pone.0090615] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 01/29/2014] [Indexed: 12/12/2022] Open
Abstract
Human Nanog1 is a 305-amino acid (aa) homeodomain-containing transcription factor critical for the pluripotency of embryonic stem (ES) and embryonal carcinoma (EC) cells. Somatic cancer cells predominantly express a retrogene homolog of Nanog1 called NanogP8, which is ∼99% similar to Nanog at the aa level. Although the predicted M.W of Nanog1/NanogP8 is ∼35 kD, both have been reported to migrate, on Western blotting (WB), at apparent molecular masses of 29–80 kD. Whether all these reported protein bands represent authentic Nanog proteins is unclear. Furthermore, detailed biochemical studies on Nanog1/NanogpP8 have been lacking. By combining WB using 8 anti-Nanog1 antibodies, immunoprecipitation, mass spectrometry, and studies using recombinant proteins, here we provide direct evidence that the Nanog1 protein in NTERA-2 EC cells exists as multiple M.W species from ∼22 kD to 100 kD with a major 42 kD band detectable on WB. We then demonstrate that recombinant NanogP8 (rNanogP8) proteins made in bacteria using cDNAs from multiple cancer cells also migrate, on denaturing SDS-PAGE, at ∼28 kD to 180 kD. Interestingly, different anti-Nanog1 antibodies exhibit differential reactivity towards rNanogP8 proteins, which can spontaneously form high M.W protein species. Finally, we show that most long-term cultured cancer cell lines seem to express very low levels of or different endogenous NanogP8 protein that cannot be readily detected by immunoprecipitation. Altogether, the current study reveals unique biochemical properties of Nanog1 in EC cells and NanogP8 in somatic cancer cells.
Collapse
Affiliation(s)
- Bigang Liu
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, Texas, United States of America
| | - Mark D. Badeaux
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, Texas, United States of America
| | - Grace Choy
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, Texas, United States of America
| | - Dhyan Chandra
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, Texas, United States of America
| | - Irvin Shen
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, Texas, United States of America
| | - Collene R. Jeter
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, Texas, United States of America
| | - Kiera Rycaj
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, Texas, United States of America
| | - Chia-Fang Lee
- College of Pharmacy, University of Texas, Austin, Texas, United States of America
| | - Maria D. Person
- College of Pharmacy, University of Texas, Austin, Texas, United States of America
| | - Can Liu
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, Texas, United States of America
| | - Yueping Chen
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, Texas, United States of America
| | - Jianjun Shen
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, Texas, United States of America
| | - Sung Yun Jung
- Department of Biochemistry, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jun Qin
- Department of Biochemistry, Baylor College of Medicine, Houston, Texas, United States of America
| | - Dean G. Tang
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, Texas, United States of America
- Cancer Stem Cell Institute, Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China
- * E-mail:
| |
Collapse
|
416
|
Schreiber L, Raanan C, Amsterdam A. CD24 and Nanog identify stem cells signature of ovarian epithelium and cysts that may develop to ovarian cancer. Acta Histochem 2014; 116:399-406. [PMID: 24103524 DOI: 10.1016/j.acthis.2013.09.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 09/09/2013] [Accepted: 09/10/2013] [Indexed: 11/25/2022]
Abstract
Ovarian cancer is the most lethal gynecological cancer. There is a general debate whether ovarian cancer is an intrinsic or an imported disease. We investigated whether in normal morphological appearance and in early stages of ovarian tumorgenesis typical cancer cell markers such as CD24 and Nanog are expressed. In 25% of normal appearing ovaries of post-menopausal women there was co-localization of CD24 and Nanog in the walls of the ovarian cysts, leaving the epithelial cells on the surface of these ovaries free of Nanog or CD24 expression. In benign ovarian tumors 37% of specimens were positive to CD24 and Nanog labeling while 26% of them were localized in the cyst walls. In contrast, in serous borderline tumors 79% specimens were labeled with CD24, 42% of them were localized in cysts and in 32% of them showed co-localization with CD24 and Nanog was evident: the rest were labeled in the ovarian epithelial cells. In serous ovarian carcinomas 81% specimens were labeled with CD24 antibodies. In 45% of them co-localization with Nanog was evident in the bulk of the cancerous tissue. In mucinous carcinomas no labeling with CD24 or Nanog was evident. In view of the synergistic effect of CD24 and Nanog expressed in malignant cancer development in other systems, it is suggested that such an analysis can be valuable for early detection of ovarian cancer. Moreover, the abundance of these markers in cysts in the development of ovarian cancer may suggest that they present an intrinsic source of the development of the highly malignant disease. Finally, since CD24 is exposed on the surface of the cancer cells, it may be highly beneficial to target these cells with antibodies to CD24 conjugated to cytotoxic drugs for more efficient treatment of this malignant disease.
Collapse
|
417
|
Lau AN, Curtis SJ, Fillmore CM, Rowbotham SP, Mohseni M, Wagner DE, Beede AM, Montoro DT, Sinkevicius KW, Walton ZE, Barrios J, Weiss DJ, Camargo FD, Wong KK, Kim CF. Tumor-propagating cells and Yap/Taz activity contribute to lung tumor progression and metastasis. EMBO J 2014; 33:468-81. [PMID: 24497554 DOI: 10.1002/embj.201386082] [Citation(s) in RCA: 172] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Metastasis is the leading cause of morbidity for lung cancer patients. Here we demonstrate that murine tumor propagating cells (TPCs) with the markers Sca1 and CD24 are enriched for metastatic potential in orthotopic transplantation assays. CD24 knockdown decreased the metastatic potential of lung cancer cell lines resembling TPCs. In lung cancer patient data sets, metastatic spread and patient survival could be stratified with a murine lung TPC gene signature. The TPC signature was enriched for genes in the Hippo signaling pathway. Knockdown of the Hippo mediators Yap1 or Taz decreased in vitro cellular migration and transplantation of metastatic disease. Furthermore, constitutively active Yap was sufficient to drive lung tumor progression in vivo. These results demonstrate functional roles for two different pathways, CD24-dependent and Yap/Taz-dependent pathways, in lung tumor propagation and metastasis. This study demonstrates the utility of TPCs for identifying molecules contributing to metastatic lung cancer, potentially enabling the therapeutic targeting of this devastating disease.
Collapse
Affiliation(s)
- Allison N Lau
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
418
|
Yang Y, Hou J, Lin Z, Zhuo H, Chen D, Zhang X, Chen Y, Sun B. Attenuated Listeria monocytogenes as a cancer vaccine vector for the delivery of CD24, a biomarker for hepatic cancer stem cells. Cell Mol Immunol 2014; 11:184-96. [PMID: 24488178 DOI: 10.1038/cmi.2013.64] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 12/01/2013] [Accepted: 12/05/2013] [Indexed: 01/14/2023] Open
Abstract
Attenuated Listeria monocytogenes (LM) is a promising candidate vector for the delivery of cancer vaccines. After phagocytosis by antigen-presenting cells, this bacterium stimulates the major histocompatibility complex (MHC)-I and MHC-II pathways and induces the proliferation of antigen-specific T lymphocytes. A new strategy involving genetic modification of the replication-deficient LM strain ΔdalΔdat (Lmdd) to express and secrete human CD24 protein has been developed. CD24 is a hepatic cancer stem cell biomarker that is closely associated with apoptosis, metastasis and recurrence of hepatocellular carcinoma (HCC). After intravenous administration in mice, Lmdd-CD24 was distributed primarily in the spleen and liver and did not cause severe organ injury. Lmdd-CD24 effectively increased the number of interferon (IFN)-γ-producing CD8(+) T cells and IFN-γ secretion. Lmdd-CD24 also enhanced the number of IL-4- and IL-10-producing T helper 2 cells. The efficacy of the Lmdd-CD24 vaccine was further investigated against Hepa1-6-CD24 tumors, which were inguinally inoculated into mice. Lmdd-CD24 significantly reduced the tumor size in mice and increased their survival. Notably, a reduction of T regulatory cell (Treg) numbers and an enhancement of specific CD8(+) T-cell activity were observed in the tumor-infiltrating lymphocytes (TILs). These results suggest a potential application of the Lmdd-CD24 vaccine against HCC.
Collapse
Affiliation(s)
- Yu Yang
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiajie Hou
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhe Lin
- Department of Microbiology and Immunology, Nanjing Medical University, Nanjing, China
| | - Han Zhuo
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dianyu Chen
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xudong Zhang
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yun Chen
- Department of Microbiology and Immunology, Nanjing Medical University, Nanjing, China
| | - Beicheng Sun
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
419
|
Stapelberg M, Zobalova R, Nguyen MN, Walker T, Stantic M, Goodwin J, Pasdar EA, Thai T, Prokopova K, Yan B, Hall S, de Pennington N, Thomas SR, Grant G, Stursa J, Bajzikova M, Meedeniya ACB, Truksa J, Ralph SJ, Ansorge O, Dong LF, Neuzil J. Indoleamine-2,3-dioxygenase elevated in tumor-initiating cells is suppressed by mitocans. Free Radic Biol Med 2014; 67:41-50. [PMID: 24145120 DOI: 10.1016/j.freeradbiomed.2013.10.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 10/02/2013] [Accepted: 10/02/2013] [Indexed: 01/07/2023]
Abstract
Tumor-initiating cells (TICs) often survive therapy and give rise to second-line tumors. We tested the plausibility of sphere cultures as models of TICs. Microarray data and microRNA data analysis confirmed the validity of spheres as models of TICs for breast and prostate cancer as well as mesothelioma cell lines. Microarray data analysis revealed the Trp pathway as the only pathway upregulated significantly in all types of studied TICs, with increased levels of indoleamine-2,3-dioxygenase-1 (IDO1), the rate-limiting enzyme of Trp metabolism along the kynurenine pathway. All types of TICs also expressed higher levels of the Trp uptake system consisting of CD98 and LAT1 with functional consequences. IDO1 expression was regulated via both transcriptional and posttranscriptional mechanisms, depending on the cancer type. Serial transplantation of TICs in mice resulted in gradually increased IDO1. Mitocans, represented by α-tocopheryl succinate and mitochondrially targeted vitamin E succinate (MitoVES), suppressed IDO1 in TICs. MitoVES suppressed IDO1 in TICs with functional mitochondrial complex II, involving transcriptional and posttranscriptional mechanisms. IDO1 increase and its suppression by VE analogues were replicated in TICs from primary human glioblastomas. Our work indicates that IDO1 is increased in TICs and that mitocans suppress the protein.
Collapse
Affiliation(s)
- Michael Stapelberg
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia.
| | - Renata Zobalova
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia; Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic
| | - Maria Nga Nguyen
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Tom Walker
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia; Department of Neurosurgery, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Marina Stantic
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Jacob Goodwin
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Elham Alizadeh Pasdar
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Thuan Thai
- Centre for Vascular Research, School of Medical Sciences, University of New South Wales, Sydney, 2052 NSW, Australia
| | - Katerina Prokopova
- Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic; Faculty of Science, Charles University, 11000 Prague 1, Czech Republic
| | - Bing Yan
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Susan Hall
- School of Pharmacy, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | | | - Shane R Thomas
- Centre for Vascular Research, School of Medical Sciences, University of New South Wales, Sydney, 2052 NSW, Australia
| | - Gary Grant
- School of Pharmacy, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Jan Stursa
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia; Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague 160 00, Czech Republic
| | - Martina Bajzikova
- Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic
| | - Adrian C B Meedeniya
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Jaroslav Truksa
- Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic
| | - Stephen J Ralph
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Olaf Ansorge
- Department of Neurosurgery, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Lan-Feng Dong
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia
| | - Jiri Neuzil
- School of Medical Science, Griffith Health Institute, Griffith University, Southport, 4222 QLD, Australia; Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic.
| |
Collapse
|
420
|
Michishita M, Ezaki S, Ogihara K, Naya Y, Azakami D, Nakagawa T, Sasaki N, Arai T, Shida T, Takahashi K. Identification of tumor-initiating cells in a canine hepatocellular carcinoma cell line. Res Vet Sci 2014; 96:315-22. [PMID: 24534130 DOI: 10.1016/j.rvsc.2014.01.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 12/13/2013] [Accepted: 01/26/2014] [Indexed: 12/27/2022]
Abstract
Tumor-initiating cells (TICs) or cancer stem cells (CSCs), a small subset of tumor cells, are involved in tumor initiation, progression, recurrence and metastasis. In human hepatocellular carcinoma (HCC), TICs are enriched with cell surface markers and have the ability to self-renew and differentiate tumors at a high frequency. We established a canine HCC cell line, HCC930599, and analyzed it for stem and progenitor cell marker expression using flow cytometry. HCC930599 showed high CD44 and CD29, moderate CD90, and low CD133, CD34, CD24, CD117, and CD13 expression. CD90(+)CD44(+) and CD90(-)CD44(+) cells were characterized using the in vitro sphere assay and an in vivo transplant model. CD90(+)CD44(+) cells acquired enhanced self-renewal capacity, proliferative activity and tumourigenicity compared with CD90(-)CD44(+) cells, suggesting that TICs exist in the HCC930599 cell line and that CD90 is a marker for enriched TICs. Understanding TIC characteristics may help elucidate hepatic carcinogenesis and HCC therapy development.
Collapse
Affiliation(s)
- Masaki Michishita
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, 1-7-1 Kyounan-cho, Musashino, Tokyo 180-8602, Japan.
| | - Shiori Ezaki
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, 1-7-1 Kyounan-cho, Musashino, Tokyo 180-8602, Japan
| | - Kikumi Ogihara
- Department of Pathology, School of Life and Environmental Science, Azabu University, 1-17-7 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa 252-5201, Japan
| | - Yuko Naya
- Department of Pathology, School of Life and Environmental Science, Azabu University, 1-17-7 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa 252-5201, Japan
| | - Daigo Azakami
- Department of Veterinary Nursing, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, 1-7-1 Kyounan-cho, Musashino, Tokyo 180-8602, Japan
| | - Takayuki Nakagawa
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo, Tokyo 113-8657, Japan
| | - Nobuo Sasaki
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1 Yayoi, Bunkyo, Tokyo 113-8657, Japan
| | - Toshiro Arai
- Department of Veterinary Biochemistry, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, 1-7-1 Kyounan-cho, Musashino, Tokyo 180-8602, Japan
| | - Takuo Shida
- Laboratory of Veterinary Radiology, School of Veterinary Medicine, Azabu University, 1-17-7 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa 252-5201, Japan
| | - Kimimasa Takahashi
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, 1-7-1 Kyounan-cho, Musashino, Tokyo 180-8602, Japan
| |
Collapse
|
421
|
Abstract
Many cancer patients suffer from metastatic relapse several years after they have undergone radical surgery. Early cancer cell dissemination followed by a protracted period of dormancy potentially explains this prevalent clinical behavior. Increasing evidence suggests that the metastasis-initiating cells are cancer stem cells or revert to this functional state upon infiltrating a target organ. Their entry into dormancy and subsequent reactivation are governed by intrinsic programs and by contextual cues, which resemble those regulating the self-renewal capability of adult stem cells. In addition, metastatic cells undergoing reactivation are nursed by specialized extracellular matrix niches, which support positive signals, such as Wnt and Notch, and attenuate negative signals, such as BMP. In spite of significant remaining uncertainties, these findings provide a framework to understand the logic of metastatic dormancy and reactivation and open new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Filippo G Giancotti
- Cell Biology Program, Sloan-Kettering Institute for Cancer Research and Metastasis Research Center, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
422
|
Yin S, Wu H, Lv J, Wu X, Zhang Y, Du J, Zhang Y. SHP-1 arrests mouse early embryo development through downregulation of Nanog by dephosphorylation of STAT3. PLoS One 2014; 9:e86330. [PMID: 24466030 PMCID: PMC3897670 DOI: 10.1371/journal.pone.0086330] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 12/07/2013] [Indexed: 01/14/2023] Open
Abstract
Src-homology protein tyrosine phosphatase-1 (SHP-1) is a protein tyrosine phosphatase that is implicated in the regulation of growth, differentiation, survival, apoptosis and proliferation of hematopoietic cells and other cell types. Here, we found that SHP-1 is involved in regulation of early embryonic development. Embryos overexpressing SHP-1 were mainly arrested at the 8-cell stage, and Nanog mRNA expression was first observed in the morulae that showed down-regulation of SHP-1. These results suggested an antagonistic relationship between SHP-1 and Nanog during early embryonic development. Next, the specific mechanism was examined in mouse F9 embryonal carcinoma cells. We confirmed that signal transducer and activator of transcription 3 (STAT3) was a substrate for SHP-1 by co-immunoprecipitation. Using overexpression and knockdown strategies, we found that SHP-1 participated in regulation of Nanog expression. Furthermore, site mutation of STAT3 was performed to confirm that SHP-1 was responsible for rapid STAT3 dephosphorylation and a decrease of Nanog expression in F9 cells. These findings suggest that SHP-1 plays a crucial role during early embryonic development. Thus, SHP-1 may function as a key regulator for Nanog that specifically demarcates the nascent epiblast, coincident with the domain of X chromosome reprogramming.
Collapse
Affiliation(s)
- Songna Yin
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
| | - Haibo Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
| | - Jiaxing Lv
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
| | - Xinying Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
| | - Yan Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
| | - Juan Du
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Yong Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, China
- * E-mail:
| |
Collapse
|
423
|
Gao YJ, Li B, Wu XY, Cui J, Han JK. Thyroid tumor-initiating cells: increasing evidence and opportunities for anticancer therapy (review). Oncol Rep 2014; 31:1035-42. [PMID: 24424445 PMCID: PMC3926673 DOI: 10.3892/or.2014.2978] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 12/30/2013] [Indexed: 12/18/2022] Open
Abstract
Accumulating evidence supports the notion that thyroid cancer is initiated by tumor-initiating cells (TICs) (commonly known as cancer stem cells), which are thought to play a crucial role in malignant progression, therapeutic resistance and recurrence. Thyroid TICs have been isolated and identified using specific biomarkers (such as CD133), the side population, sphere formation and aldehyde dehydrogenase activity assays. Although their characteristics remain largely unknown, TICs provide an attractive cellular mechanism to explain therapeutic refractoriness. Efforts are currently being directed toward the identification of therapeutic strategies that could target these cells. The present review discusses the cellular origins of TICs and the main approaches used to isolate and identify thyroid TICs, with a focus on the remaining challenges and opportunities for anticancer therapy.
Collapse
Affiliation(s)
- Yong-Ju Gao
- Department of Nuclear Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Bo Li
- Department of Nuclear Medicine, Henan Provincial People's Hospital and the People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Xin-Yu Wu
- Department of Nuclear Medicine, Henan Provincial People's Hospital and the People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Jing Cui
- Department of Nuclear Medicine, Henan Provincial People's Hospital and the People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Jian-Kui Han
- Department of Nuclear Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
424
|
Abstract
Spontaneous tumors often contain heterogeneous populations of tumor cells with different tumor-initiating potentials or cancer cell "stemness." Clonal heterogeneity can be traced to specific locations inside a tumor where clones with different metastatic capabilities are identified, suggesting that the tumor microenvironment can exert a significant effect on the evolution of different clonal populations. Hypoxia is a common feature of tumor microenvironments and has the potential to facilitate malignant progression. This chapter provides a synopsis of hypoxia-regulated pathways implicated in the maintenance of cancer stem cells.
Collapse
Affiliation(s)
- Zhong Yun
- Department of Therapeutic Radiology, Yale School of Medicine, 208040, New Haven, 06520-8040, CT, USA,
| | | |
Collapse
|
425
|
Thakolwiboon S, Zhu J, Liang Q, Welling TH, Zhang M, Lubman DM. Heterogeneity of The CD90 + Population in Different Stages of Hepatocarcinogenesis. ACTA ACUST UNITED AC 2014; 7:296-302. [PMID: 25580065 DOI: 10.4172/jpb.1000332] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We have characterized herein the heterogeneity of the CD90+ population at each stage of hepatocarcinogenesis using a computer-assisted immunohistochemical staining evaluation method for quantitative analysis on tissue microarrays. We found that CD90 in Hepatocellular carcinoma (HCC) tissues, which has been shown to be a marker for cancer stem cells, is expressed on tumor cells, in the stroma or on endothelial cells. Sub-classification of the CD90+ population was based on morphology and co-expression with known markers including CD45 and CD31. Multiple linear regression suggested that the percentage of CD90+ cancer cells/hepatocyte (p<0.0001), level of overall CD90 expression (p<0.0014), and level of CD90 expression in tumor islands (p<0.0001) increased significantly in each stage of liver disease progression, while the level of stromal CD90 expression (p=0.1129) did not change significantly. Additionally, only the CD90+ cancer cells were positive for other cancer stem cell (CSC) markers including CD24, CD44 and CD133 whereas the other CD90+ cells were negative for these markers. CD90 expression in cirrhosis was observed in hepatocytes, the portal tract area and fibrous septa while CD90 expression in normal liver was limited only to the portal tract area. This study demonstrates the heterogeneity of the CD90+ population in HCC where a small population of the CD90+ cells that expressed other CSC markers are CSCs and are associated with advanced stages of hepatocarcinogenesis. This heterogeneity should be emphasized in further studies where other methods may not be able to discriminate these distinct types of CD90+ cells.
Collapse
Affiliation(s)
- Smathorn Thakolwiboon
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan 48109, United States ; Department of Medicine, Faculty of Medicine Siriraj hospital, Mahidol University, Bangkok10700, Thailand
| | - Jianhui Zhu
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan 48109, United States
| | - Qixing Liang
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States
| | - Theodore H Welling
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan 48109, United States
| | - Min Zhang
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States
| | - David M Lubman
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
426
|
Zhang L, Yang Z, Ma A, Qu Y, Xia S, Xu D, Ge C, Qiu B, Xia Q, Li J, Liu Y. Growth arrest and DNA damage 45G down-regulation contributes to Janus kinase/signal transducer and activator of transcription 3 activation and cellular senescence evasion in hepatocellular carcinoma. Hepatology 2014; 59:178-89. [PMID: 23897841 DOI: 10.1002/hep.26628] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 07/07/2013] [Indexed: 12/19/2022]
Abstract
UNLABELLED Growth arrest and DNA damage 45G (GADD45G), a stress sensor with multiple implications in various biological processes, is down-regulated in a broad spectrum of cancers. However, little is known about the biological effects of GADD45G on hepatocellular carcinoma (HCC) cells and the related mechanisms. In the present study, we found that GADD45G was commonly down-regulated in oncogene-transformed mouse liver cells and in human and mouse HCC. Ectopic expression of GADD45G robustly elicited senescence in HCC cells and suppressed tumor growth in vivo. Furthermore, GADD45G-induced senescence occurred in HCC cells independently of p53, p16(INK4a) (p16), and retinoblastoma (Rb). Instead, the prompt inhibition of Janus kinase 2 (Jak2), tyrosine kinase 2 (Tyk2), and signal transducer and activator of transcription 3 (Stat3) activation was observed in cells undergoing senescence. Impairment of Jak-Stat3 activation caused by GADD45G expression was associated with activation of SH2 domain-containing protein tyrosine phosphatase-2 (Shp2). Expression of constitutively activated Stat3 or human telomerase reverse transcriptase (hTERT), as well as knockdown of Shp2f, efficiently counteracted GADD45G-induced senescence. More important, in clinical HCC specimens, we found that GADD45G expression was inversely correlated with phosphorylated Stat3 expression in tumor cells and disease progression. CONCLUSION GADD45G functions as a negative regulator of the Jak-Stat3 pathway and inhibits HCC by inducing cellular senescence. The decrease or absence of GADD45G expression may be a key event for tumor cells or premalignant liver cells to bypass cellular senescence.
Collapse
Affiliation(s)
- Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
427
|
Ding J, Wang H. Multiple interactive factors in hepatocarcinogenesis. Cancer Lett 2013; 346:17-23. [PMID: 24374016 DOI: 10.1016/j.canlet.2013.12.024] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 12/08/2013] [Accepted: 12/10/2013] [Indexed: 12/25/2022]
Abstract
Hepatocellular carcinoma (HCC) is the fifth most prevalent cancer and the third most frequent cause of cancer mortality globally. Each year there are approximately 630,000 new cases of HCC in the world and more than half of the new cases occur in China. Major risk factors of HCC include HBV or HCV infection, alcoholic liver disease, and nonalcoholic fatty liver disease. Most of these risk factors lead to chronic hepatitis and cirrhosis, which is present in 80-90% of HCC patients. Hepatocarcinogenesis has been regarded as a multi-stage process involving multiple genetic or environmental factors. Interaction and cross-regulation of distinct factors synergistically contributes to HCC occurrence. A comprehensive knowledge on the multiple factors and their interaction in hepatocarcinogenesis is necessary to improve the effectiveness of HCC intervention. In this review, we will focus on the recent progress made in understanding the mechanisms of hepatocarcinogenesis and discuss some potential issues or challenges in this area.
Collapse
Affiliation(s)
- Jin Ding
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital/Institute, Second Military Medical University, Shanghai 200433, China; National Center for Liver Cancer, Shanghai 200433, China.
| | - Hongyang Wang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital/Institute, Second Military Medical University, Shanghai 200433, China; National Center for Liver Cancer, Shanghai 200433, China.
| |
Collapse
|
428
|
Okabe H, Ishimoto T, Mima K, Nakagawa S, Hayashi H, Kuroki H, Imai K, Nitta H, Saito S, Hashimoto D, Chikamoto A, Ishiko T, Watanabe M, Nagano O, Beppu T, Saya H, Baba H. CD44s signals the acquisition of the mesenchymal phenotype required for anchorage-independent cell survival in hepatocellular carcinoma. Br J Cancer 2013; 110:958-66. [PMID: 24300972 PMCID: PMC3929866 DOI: 10.1038/bjc.2013.759] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 09/24/2013] [Accepted: 11/11/2013] [Indexed: 12/11/2022] Open
Abstract
Background: Circulating tumour cells (CTCs) have an important role in metastatic processes, but details of their basic characteristics remain elusive. We hypothesised that CD44-expressing CTCs show a mesenchymal phenotype and high potential for survival in hepatocellular carcinoma (HCC). Methods: Circulating CD44+CD90+ cells, previously shown to be tumour-initiating cells, were sorted from human blood and their genetic characteristics were compared with those of tumour cells from primary tissues. The mechanism underlying the high survival potential of CD44-expressing cells in the circulatory system was investigated in vitro. Results: CD44+CD90+ cells in the blood acquired epithelial–mesenchymal transition, and CD44 expression remarkably increased from the tissue to the blood. In Li7 and HLE cells, the CD44high population showed higher anoikis resistance and sphere-forming ability than did the CD44low population. This difference was found to be attributed to the upregulation of Twist1 and Akt signal in the CD44high population. Twist1 knockdown showed remarkable reduction in anoikis resistance, sphere formation, and Akt signal in HLE cells. In addition, mesenchymal markers and CD44s expression were downregulated in the Twist1 knockdown. Conclusions: CD44s symbolises the acquisition of a mesenchymal phenotype regulating anchorage-independent capacity. CD44s-expressing tumour cells in peripheral blood are clinically important therapeutic targets in HCC.
Collapse
Affiliation(s)
- H Okabe
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - T Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - K Mima
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - S Nakagawa
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - H Hayashi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - H Kuroki
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - K Imai
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - H Nitta
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - S Saito
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - D Hashimoto
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - A Chikamoto
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - T Ishiko
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - M Watanabe
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - O Nagano
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Minato, Japan
| | - T Beppu
- 1] Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan [2] Department of Multidisciplinary Treatment for Gastroenterological Cancer, Kumamoto University Hospital, Kumamoto 860-8556, Japan
| | - H Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Minato, Japan
| | - H Baba
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| |
Collapse
|
429
|
Zhang Y, Guan DX, Shi J, Gao H, Li JJ, Zhao JS, Qiu L, Liu J, Li N, Guo WX, Xue J, Zhou FG, Wu MC, Wang HY, Xie D, Cheng SQ. All-trans retinoic acid potentiates the chemotherapeutic effect of cisplatin by inducing differentiation of tumor initiating cells in liver cancer. J Hepatol 2013; 59:1255-63. [PMID: 23867314 DOI: 10.1016/j.jhep.2013.07.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 06/30/2013] [Accepted: 07/05/2013] [Indexed: 01/15/2023]
Abstract
BACKGROUND & AIMS Systemic chemotherapy serves as an adjuvant treatment for post-operation patients with hepatocellular carcinoma (HCC), and provides curative option for the patients with unresectable HCC. However, its efficiency is largely limited because of the high incidence of chemo-resistance. Increasing evidence has shown that tumor initiating cells (TICs) not only have the ability to self-renew and drive the initiation and progression of cancer, but also exhibit greater resistance to conventional chemo- and radio-therapies than non-TICs. It was the aim of this study to investigate the effects of ATRA with and without cisplatin on TIC differentiation and apoptosis in human HCC. METHODS In the present study, we evaluated the TICs of HCC cell differentiation induced by all-trans retinoic acid (ATRA), and developed a novel chemotherapeutic approach to HCC, by characterizing the function of combinatorial treatment with cis-diammineplatinum(II) (cisplatin) and ATRA in vitro and in vivo. RESULTS ATRA effectively induced differentiation of TICs, which potentiated the cytotoxic effects of cisplatin. The combinatorial treatment of ATRA acid and cisplatin reduced protein kinase B (AKT) (Thr308) phosphorylation, and promoted apoptosis of HCC cells more significantly than treatment with cisplatin alone. In addition, the combined treatment with the two drugs exerted stronger inhibition on either HCC cell migration in vitro or metastasis in vivo, when compared to the treatment with either drug alone. CONCLUSIONS These results indicated that ATRA could significantly improve the effect of cisplatin, which is at least partially attributed to ATRA-induced differentiation of HCC TICs, and the subsequent decrease in this chemo-resistant subpopulation.
Collapse
Affiliation(s)
- Yang Zhang
- Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai 200438, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
430
|
Liu AY, Cai Y, Mao Y, Lin Y, Zheng H, Wu T, Huang Y, Fang X, Lin S, Feng Q, Huang Z, Yang T, Luo Q, Ouyang G. Twist2 promotes self-renewal of liver cancer stem-like cells by regulating CD24. Carcinogenesis 2013; 35:537-45. [PMID: 24193512 DOI: 10.1093/carcin/bgt364] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Twist2 is a highly conserved basic helix-loop-helix transcription factor that plays a critical role in embryogenesis. Recent evidence has revealed that aberrant Twist2 expression contributes to tumor progression; however, the role of Twist2 in human hepatocellular carcinoma (HCC) and its underlying mechanisms remain undefined. In this report, we demonstrate that Twist2 is overexpressed in human HCC tumors. We show that ectopic expression of Twist2 induces epithelial-mesenchymal transition phenotypes, augments cell migration and invasion and colony-forming abilities in human HCC cells in vitro, and promotes tumor growth in vivo. Moreover, we found a higher percentage of CD24(+) liver cancer stem-like cells in Twist2-transduced HCC cells. Twist2-expressing cells exhibited an increased expression of stem cell markers Bmi-1, Sox2, CD24 and Nanog and an increased capacity for self-renewal. Knockdown of CD24 in HepG2/Twist2 cells decreased the levels of Sox2, pSTAT3 and Nanog, and reversed the cancer stem-like cell phenotypes induced by ectopic expression of Twist2. Furthermore, Twist2 regulated the CD24 expression by directly binding to the E-box region in CD24 promoter. Therefore, our data demonstrated that Twist2 augments liver cancer stem-like cell self-renewal in a CD24-dependent manner. Twist2-CD24-STAT3-Nanog pathway may play a critical role in regulating liver cancer stem-like cell self-renewal. The identification of the Twist2-CD24 signaling pathway provides a potential therapeutic approach to target cancer stem cells in HCCs.
Collapse
Affiliation(s)
- Allan Yi Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
431
|
Tomimaru Y, Xu CQ, Nambotin SB, Yan T, Wands JR, Kim M. Loss of exon 4 in a human T-cell factor-4 isoform promotes hepatic tumourigenicity. Liver Int 2013; 33:1536-48. [PMID: 23648141 PMCID: PMC3775864 DOI: 10.1111/liv.12189] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 04/01/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND T-cell factor (TCF) proteins represent key transcription factors that activate Wnt/β-catenin signalling. We have reported that a pair of TCF-4 isoforms (TCF-4C and TCF-4D) exhibit differential TCF transcriptional activity in hepatocellular carcinoma (HCC) cells, although their structure differs by only the presence (TCF-4D) or absence (TCF-4C) of exon 4. AIM To demonstrate a regulatory role of exon 4 in HCC development. METHODS TCF-4C and TCF-4D expression profiles were examined in 27 pairs of human HCC and adjacent liver tissues. The functional role of the TCF-4 isoforms was evaluated in OUMS-29 (an immortalized hepatocyte-derived) and HAK-1A (a well-differentiated HCC) cell lines using stable clones overexpressing the TCF-4 isoforms. RESULTS TCF-4C was significantly upregulated in HCC tissues compared with corresponding peritumour and normal liver tissues; in contrast, there was no difference in TCF-4D expression. TCF-4C clones derived from both cell lines exhibited increased TCF activity, Wnt-responsive target genes, cell proliferation, cell cycle progression and resistance to chemotherapeutic drugs compared with TCF-4D clones. Capability of cell migration and colony formation was significantly higher in TCF-4C than TCF-4D clones. In a nude mice xenograft model, the HAK-1A-derived TCF-4C clone rapidly developed tumours compared with the TCF-4D clone. TCF-4C clone-derived tumours exhibited upregulation of Wnt-responsive target genes compared with the slow developing and small TCF-4D-derived tumours. CONCLUSION These results demonstrate that the TCF-4C isoform lacking exon 4 is associated with a malignant phenotype compared with the exon 4-harbouring TCF-4D isoform, indicating that exon 4 of TCF-4 plays a prominent role in HCC development.
Collapse
Affiliation(s)
- Yoshito Tomimaru
- Liver Research Center, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | | | | | | | | | | |
Collapse
|
432
|
Ghanekar A, Ahmed S, Chen K, Adeyi O. Endothelial cells do not arise from tumor-initiating cells in human hepatocellular carcinoma. BMC Cancer 2013; 13:485. [PMID: 24138671 PMCID: PMC3856592 DOI: 10.1186/1471-2407-13-485] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 10/15/2013] [Indexed: 12/15/2022] Open
Abstract
Background Conventional models of carcinogenesis suggest that tumors recruit blood vessel formation from normal host tissues. This concept has recently been challenged by prominent studies of glioblastoma, which suggest that intratumoral endothelial cells (ECs) may arise from cancer stem cells/tumor-initiating cells (TICs). Hepatocellular carcinoma (HCC) is a common, highly vascularized tumor with few effective therapies, against which anti-angiogenic strategies are being actively explored. TICs are felt to play a role in HCC pathobiology, but their contributions to tumor vasculature have not been studied. Methods We examined human HCCs in settings that selected for tumor formation from functionally defined TICs, and in which the origin of intratumoral ECs from TICs as opposed to host tissues could be clearly distinguished. We generated HCC nodules in the livers of immunodeficient mice by intrasplenic injection of HCC cells from cell lines and patient specimens and studied the tumor ECs by immunohistochemistry for mouse and human markers. We then used immunohistochemistry for EC markers in combination with fluorescence in situ hybridization (FISH) for X and Y chromosomes to study the endothelium of recurrent HCC specimens resected from sex-mismatched liver allografts of patients who had undergone liver transplantation for HCC. Results We observed that all ECs in intrahepatic human HCC xenografts expressed mouse rather than human CD31. FISH analysis of recurrent HCCs resected from patients with sex-mismatched liver allografts revealed that all CD31+ and CD34+ intratumoral ECs originated from the donor allograft rather than the tumor. Conclusions These observations suggest that the vasculature of human HCC arises from normal host tissues rather than from TICs, supporting ongoing efforts to target angiogenesis in HCC as it is currently understood, and suggesting that the contribution of TICs to the vasculature of other cancers is disease-specific.
Collapse
Affiliation(s)
- Anand Ghanekar
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada.
| | | | | | | |
Collapse
|
433
|
Hou H, Sun H, Lu P, Ge C, Zhang L, Li H, Zhao F, Tian H, Zhang L, Chen T, Yao M, Li J. Tunicamycin Potentiates Cisplatin Anticancer Efficacy through the DPAGT1/Akt/ABCG2 Pathway in Mouse Xenograft Models of Human Hepatocellular Carcinoma. Mol Cancer Ther 2013; 12:2874-84. [DOI: 10.1158/1535-7163.mct-13-0201] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
434
|
Vidal SJ, Rodriguez-Bravo V, Galsky M, Cordon-Cardo C, Domingo-Domenech J. Targeting cancer stem cells to suppress acquired chemotherapy resistance. Oncogene 2013; 33:4451-63. [PMID: 24096485 DOI: 10.1038/onc.2013.411] [Citation(s) in RCA: 191] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 08/12/2013] [Accepted: 08/12/2013] [Indexed: 12/18/2022]
Abstract
Acquired resistance has curtailed cancer survival since the dawn of the chemotherapy age more than half a century ago. Although the application of stem cell (SC) concepts to cancer captured the imagination of scientists for many years, only the last decade has yielded substantial evidence that cancer SCs (CSCs) contribute to chemotherapy resistance. Recent studies suggest that the functional and molecular properties of CSCs constitute therapeutic opportunities to improve the efficacy of chemotherapy. Here we review how these properties have stimulated combination strategies that suppress acquired resistance across a spectrum of malignancies. The clinical implementation of these strategies promises to rejuvenate the effort against an enduring challenge.
Collapse
Affiliation(s)
- S J Vidal
- 1] Department of Pathology, Mount Sinai Icahn School of Medicine, New York, NY, USA [2] Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - V Rodriguez-Bravo
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - M Galsky
- Oncology Department, Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, NY, USA
| | - C Cordon-Cardo
- Department of Pathology, Mount Sinai Icahn School of Medicine, New York, NY, USA
| | - J Domingo-Domenech
- Department of Pathology, Mount Sinai Icahn School of Medicine, New York, NY, USA
| |
Collapse
|
435
|
Petkova N, Hennenlotter J, Sobiesiak M, Todenhöfer T, Scharpf M, Stenzl A, Bühring HJ, Schwentner C. Surface CD24 distinguishes between low differentiated and transit-amplifying cells in the basal layer of human prostate. Prostate 2013; 73:1576-90. [PMID: 23836489 DOI: 10.1002/pros.22708] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 06/15/2013] [Indexed: 11/11/2022]
Abstract
BACKGROUND Benign prostatic hyperplasia (BPH) and prostate cancer (PCa) are common abnormalities in elderly men. It is considered that epithelial stem cells are involved in the etiology and development of both diseases. To distinguish aberrant from normal cells, the knowledge about primary epithelial stem/progenitor cells (ES/P) is essential. The aim of this study was to examine the role of surface markers to distinguish between different subsets of prostate basal epithelium. METHODS The expression pattern of prostate tissue single cell suspensions was analyzed by flow cytometry using different markers. Sorted cell populations were examined for their clonogenic capacity and the resulted colonies were analyzed with flow cytometry, Western blot, and qPCR for stem cell, basal, and luminal epithelium markers. Additionally, the histological localization of the examined markers was determined using immunofluorescence. RESULTS Using the combination of CD49f, Trop-2, and surface CD24, basal cell subsets with distinct differentiation capacities were dissected (CD49f(+) Trop-2(+) CD24(-) and CD49f(+) Trop-2(+) CD24(+) ). Although cells from the two subsets gave rise to similar basal colonies, qPCR of primary tissue revealed that higher levels of basal marker expression were detected in the CD49f(+) Trop-2(+) CD24(-) subset. Immunofluorescence analysis showed a prominent expression of CD24 by luminal and basal cells. CONCLUSIONS Subsets with distinct differentiation capacities within the basal epithelium (CD49f(+) Trop-2(+) CD24(-) and CD49f(+) Trop-2(+) CD24(+) ) can be distinguished in human prostate. CD24 is a marker expressed on the basal transit-amplifying cells (transition cells) and may play a role in the differentiation and migration of ES/P cells to the luminal layer. The knowledge of this mechanism is of relevance for treatment of both diseases.
Collapse
Affiliation(s)
- Neli Petkova
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
436
|
Azzam DJ, Zhao D, Sun J, Minn AJ, Ranganathan P, Drews-Elger K, Han X, Picon-Ruiz M, Gilbert CA, Wander SA, Capobianco AJ, El-Ashry D, Slingerland JM. Triple negative breast cancer initiating cell subsets differ in functional and molecular characteristics and in γ-secretase inhibitor drug responses. EMBO Mol Med 2013; 5:1502-1522. [PMID: 23982961 PMCID: PMC3799576 DOI: 10.1002/emmm.201302558] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 07/15/2013] [Accepted: 07/16/2013] [Indexed: 02/06/2023] Open
Abstract
Increasing evidence suggests that stem-like cells mediate cancer therapy resistance and metastasis. Breast tumour-initiating stem cells (T-ISC) are known to be enriched in CD44(+) CD24(neg/low) cells. Here, we identify two T-ISC subsets within this population in triple negative breast cancer (TNBC) lines and dissociated primary breast cancer cultures: CD44(+) CD24(low+) subpopulation generates CD44(+) CD24(neg) progeny with reduced sphere formation and tumourigenicity. CD44(+) CD24(low+) populations contain subsets of ALDH1(+) and ESA(+) cells, yield more frequent spheres and/or T-ISC in limiting dilution assays, preferentially express metastatic gene signatures and show greater motility, invasion and, in the MDA-MB-231 model, metastatic potential. CD44(+) CD24(low+) but not CD44(+) CD24(neg) express activated Notch1 intracellular domain (N1-ICD) and Notch target genes. We show N1-ICD transactivates SOX2 to increase sphere formation, ALDH1+ and CD44(+) CD24(low+) cells. Gamma secretase inhibitors (GSI) reduced sphere formation and xenograft growth from CD44(+) CD24(low+) cells, but CD44(+) CD24(neg) were resistant. While GSI hold promise for targeting T-ISC, stem cell heterogeneity as observed herein, could limit GSI efficacy. These data suggest a breast T-ISC hierarchy in which distinct pathways drive developmentally related subpopulations with different anti-cancer drug responsiveness.
Collapse
Affiliation(s)
- Diana J Azzam
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of MedicineMiami, FL, USA
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of MedicineMiami, FL, USA
| | - Dekuang Zhao
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of MedicineMiami, FL, USA
- Sheila and David Fuente Cancer Biology ProgramMiami, FL, USA
| | - Jun Sun
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of MedicineMiami, FL, USA
| | - Andy J Minn
- Department of Radiation Oncology, Abramson Family Cancer Research Institute, University of PennsylvaniaPhiladelphia, PA, USA
| | - Prathibha Ranganathan
- Molecular Oncology, Department of Surgery, Sylvester Comprehensive Cancer CenterMiami, FL, USA
| | - Katherine Drews-Elger
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of MedicineMiami, FL, USA
| | - Xiaoqing Han
- Sheila and David Fuente Cancer Biology ProgramMiami, FL, USA
- Molecular Oncology, Department of Surgery, Sylvester Comprehensive Cancer CenterMiami, FL, USA
| | - Manuel Picon-Ruiz
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of MedicineMiami, FL, USA
| | - Candace A Gilbert
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of MedicineMiami, FL, USA
| | - Seth A Wander
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of MedicineMiami, FL, USA
- Sheila and David Fuente Cancer Biology ProgramMiami, FL, USA
| | - Anthony J Capobianco
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of MedicineMiami, FL, USA
- Molecular Oncology, Department of Surgery, Sylvester Comprehensive Cancer CenterMiami, FL, USA
| | - Dorraya El-Ashry
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of MedicineMiami, FL, USA
- Department of Medicine, University of Miami Miller School of MedicineMiami, FL, USA
| | - Joyce M Slingerland
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of MedicineMiami, FL, USA
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of MedicineMiami, FL, USA
- Sheila and David Fuente Cancer Biology ProgramMiami, FL, USA
- Department of Medicine, University of Miami Miller School of MedicineMiami, FL, USA
| |
Collapse
|
437
|
Ng L, Poon RTP, Pang R. Biomarkers for predicting future metastasis of human gastrointestinal tumors. Cell Mol Life Sci 2013; 70:3631-56. [PMID: 23370778 PMCID: PMC11113832 DOI: 10.1007/s00018-013-1266-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 01/02/2013] [Accepted: 01/10/2013] [Indexed: 12/19/2022]
Abstract
The recent advances in surgery and radiation therapy have significantly improved the prognosis of patients with primary cancer, and the major challenge of cancer treatment now is metastatic disease development. The 5-year survival rate of cancer patients who have distant metastasis at diagnosis is extremely low, suggesting that prediction and early detection of metastasis would definitely improve their prognosis because suitable patient therapeutic management and treatment strategy can be provided. Cancer cells from a primary site give rise to a metastatic tumor via a number of steps which require the involvement and altered expression of many regulators. These regulators may serve as biomarkers for predicting metastasis. Over the past few years, numerous regulators have been found correlating with metastasis. In this review, we summarize the findings of a number of potential biomarkers that are involved in cadherin-catenin interaction, integrin signaling, PI3K/Akt/mTOR signaling and cancer stem cell identification in gastrointestinal cancers. We will also discuss how certain biomarkers are associated with the tumor microenvironment that favors cancer metastasis.
Collapse
Affiliation(s)
- Lui Ng
- Department of Surgery, The University of Hong Kong, 102 Pokfulam Road, Hong Kong SAR, China,
| | | | | |
Collapse
|
438
|
Wang ML, Chiou SH, Wu CW. Targeting cancer stem cells: emerging role of Nanog transcription factor. Onco Targets Ther 2013; 6:1207-20. [PMID: 24043946 PMCID: PMC3772775 DOI: 10.2147/ott.s38114] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The involvement of stemness factors in cancer initiation and progression has drawn much attention recently, especially after the finding that introducing four stemness factors in somatic cells is able to reprogram the cells back to an embryonic stem cell-like state. Following accumulating data revealing abnormal elevated expression levels of key stemness factors, like Nanog, Oct4, and Sox2, in several types of cancer stem cells; the importance and therapeutic potential of targeting these stemness regulators in cancers has turned to research focus. Nanog determines cell fate in both embryonic and cancer stem cells; activating Nanog at an inappropriate time would result in cancer stem cells rather than normal pluripotent stem cells or differentiated somatic cells. Upregulated Nanog is correlated with poor survival outcome of patients with various types of cancer. The discoveries of downstream regulatory pathways directly or indirectly mediated by Nanog indicate that Nanog regulates several aspects of cancer development such as tumor cell proliferation, self-renewal, motility, epithelial-mesenchymal transition, immune evasion, and drug-resistance, which are all defined features for cancer stem cells. The current review paper illustrates the central role of Nanog in the regulatory networks of cancer malignant development and stemness acquirement, as well as in the communication between cancer cells and the surrounding stroma. Though a more defined model is needed to test the therapeutic efficacy of targeting Nanog as a cancer treatment method, current animal experiments using siNanog or shNanog have shown the promising therapeutic potential of Nanog targeting in several types of cancer.
Collapse
Affiliation(s)
- Mong-Lien Wang
- Institute of Biochemistry and Molecular Biology, National Yang Ming University, Taipei, Taiwan
| | | | | |
Collapse
|
439
|
Barashi N, Weiss ID, Wald O, Wald H, Beider K, Abraham M, Klein S, Goldenberg D, Axelrod J, Pikarsky E, Abramovitch R, Zeira E, Galun E, Peled A. Inflammation-induced hepatocellular carcinoma is dependent on CCR5 in mice. Hepatology 2013; 58:1021-30. [PMID: 23526353 DOI: 10.1002/hep.26403] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2012] [Accepted: 03/15/2013] [Indexed: 12/12/2022]
Abstract
UNLABELLED Human hepatocellular carcinoma (HCC) is an inflammation-induced cancer, which is the third-leading cause of cancer mortality worldwide. We investigated the role of the chemokine receptors, CCR5 and CCR1, in regulating inflammation and tumorigenesis in an inflammation-induced HCC model in mice. Multidrug resistance 2 gene (Mdr2)-knockout (Mdr2-KO) mice spontaneously develop chronic cholestatic hepatitis and fibrosis that is eventually followed by HCC. We generated two new strains from the Mdr2-KO mouse, the Mdr2:CCR5 and the Mdr2:CCR1 double knockouts (DKOs), and set out to compare inflammation and tumorigenesis among these strains. We found that in Mdr2-KO mice lacking the chemokine receptor, CCR5 (Mdr2:CCR5 DKO mice), but not CCR1 (Mdr2:CCR1 DKO), macrophage recruitment and trafficking to the liver was significantly reduced. Furthermore, in the absence of CCR5, reduced inflammation was also associated with reduced periductal accumulation of CD24(+) oval cells and abrogation of fibrosis. DKO mice for Mdr2 and CCR5 exhibited a significant decrease in tumor incidence and size. CONCLUSIONS Our results indicate that CCR5 has a critical role in both the development and progression of liver cancer. Therefore, we propose that a CCR5 antagonist can serve for HCC cancer prevention and treatment.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/deficiency
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/physiology
- Animals
- Carcinoma, Hepatocellular/epidemiology
- Carcinoma, Hepatocellular/etiology
- Carcinoma, Hepatocellular/physiopathology
- Chemokine CCL5/physiology
- Disease Models, Animal
- Disease Progression
- Hepatitis, Chronic/complications
- Hepatitis, Chronic/genetics
- Incidence
- Liver/pathology
- Liver/physiopathology
- Liver Cirrhosis/complications
- Liver Cirrhosis/genetics
- Liver Neoplasms/epidemiology
- Liver Neoplasms/etiology
- Liver Neoplasms/physiopathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, CCR1/deficiency
- Receptors, CCR1/genetics
- Receptors, CCR1/physiology
- Receptors, CCR5/deficiency
- Receptors, CCR5/genetics
- Receptors, CCR5/physiology
- ATP-Binding Cassette Sub-Family B Member 4
Collapse
Affiliation(s)
- Neta Barashi
- Goldyne Savad Institute of Gene Therapy, Human Biology Research Center, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
440
|
Liu L, Yang Z, Xu Y, Li J, Xu D, Zhang L, Sun J, Xia S, Zou F, Liu Y. Inhibition of oxidative stress-elicited AKT activation facilitates PPARγ agonist-mediated inhibition of stem cell character and tumor growth of liver cancer cells. PLoS One 2013; 8:e73038. [PMID: 24023668 PMCID: PMC3758331 DOI: 10.1371/journal.pone.0073038] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Accepted: 07/16/2013] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence suggests that tumor-initiating cells (TICs) are the most malignant cell subpopulation in tumors because of their resistance to chemotherapy or radiation treatment. Targeting TICs may be a key innovation for cancer treatment. In this study, we found that PPARγ agonists inhibited the cancer stem cell-like phenotype and attenuated tumor growth of human hepatocellular carcinoma (HCC) cells. Reactive oxygen species (ROS) initiated by NOX2 upregulation were partially responsible for the inhibitory effects mediated by PPARγ agonists. However, PPARγ agonist-mediated ROS production significantly activated AKT, which in turn promoted TIC survival by limiting ROS generation. Inhibition of AKT, by either pharmacological inhibitors or AKT siRNA, significantly enhanced PPARγ agonist-mediated inhibition of cell proliferation and stem cell-like properties in HCC cells. Importantly, in nude mice inoculated with HCC Huh7 cells, we demonstrated a synergistic inhibitory effect of the PPARγ agonist rosiglitazone and the AKT inhibitor triciribine on tumor growth. In conclusion, we observed a negative feedback loop between oxidative stress and AKT hyperactivation in PPARγ agonist-mediated suppressive effects on HCCs. Combinatory application of an AKT inhibitor and a PPARγ agonist may provide a new strategy for inhibition of stem cell-like properties in HCCs and treatment of liver cancer.
Collapse
Affiliation(s)
- Lanlan Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Developmental and Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zhaojuan Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingqian Xu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingyi Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongxu Xu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiabin Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Suhua Xia
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feiyan Zou
- Department of Developmental and Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yongzhong Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail:
| |
Collapse
|
441
|
Nishimoto A, Kugimiya N, Hosoyama T, Enoki T, Li TS, Hamano K. JAB1 regulates unphosphorylated STAT3 DNA-binding activity through protein–protein interaction in human colon cancer cells. Biochem Biophys Res Commun 2013; 438:513-518. [DOI: 10.1016/j.bbrc.2013.07.105] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 07/25/2013] [Indexed: 10/26/2022]
|
442
|
Abstract
Metastasis is an inefficient process and most cancer cells fail to colonize secondary sites. There are several possible reasons for this. First, the nature of the infiltrating cells is important as a small population of cancer stem cells has been shown to have exclusive metastasis-initiating potential. Secondly, supportive niches are required to promote the outgrowth of disseminated tumour cells. Such niches are either produced prior to the arrival of cancer cells in the target organ or are induced ad hoc upon cell infiltration. Components of the extracellular matrix (ECM) have been found to play a role in establishing these niches. This has highlighted the importance of the ECM for metastatic progression, and suggests that such components may provide alternative targets for treatment of metastatic disease.
Collapse
Affiliation(s)
- A Santamaria-Martínez
- École Polytechnique Fédérale de Lausanne (EPFL), ISREC (Swiss Institute for Experimental Cancer Research), National Center of Competence in Research (NCCR) 'Molecular Oncology', Lausanne, Switzerland
| | | |
Collapse
|
443
|
Liu C, Liu L, Shan J, Shen J, Xu Y, Zhang Q, Yang Z, Wu L, Xia F, Bie P, Cui Y, Zhang X, Bian X, Qian C. Histone deacetylase 3 participates in self-renewal of liver cancer stem cells through histone modification. Cancer Lett 2013; 339:60-9. [PMID: 23879963 DOI: 10.1016/j.canlet.2013.07.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 07/11/2013] [Accepted: 07/15/2013] [Indexed: 01/06/2023]
Abstract
Understanding molecular mechanisms in self-renewal of cancer stem cells (CSCs) is important for finding novel target in therapy of cancer. In this study, we explored potential effects of histone deacetylase (HDAC) on liver CSCs. Our data showed that HDAC inhibitors suppressed self-renewal and induced differentiation of liver CSCs. Furthermore, we demonstrated that HDAC3 was selectively expressed in liver CSCs and participated in self-renewal of liver CSCs via regulating expression of pluripotency factors. Overexpression of HDAC3 was associated with poor outcome of liver cancer. HDAC inhibitors could render liver CSCs sensitive to sorafenib. Taken together, our data suggest that HDAC3 plays a critical role in regulating self-renewal of liver CSCs.
Collapse
Affiliation(s)
- Chungang Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
444
|
Zheng Y, de la Cruz CC, Sayles LC, Alleyne-Chin C, Vaka D, Knaak TD, Bigos M, Xu Y, Hoang CD, Shrager J, Fehling HJ, French D, Forrest W, Jiang Z, Jackson EL, Sweet-Cordero EA. A rare population of CD24(+)ITGB4(+)Notch(hi) cells drives tumor propagation in NSCLC and requires Notch3 for self-renewal. Cancer Cell 2013; 24:59-74. [PMID: 23845442 PMCID: PMC3923526 DOI: 10.1016/j.ccr.2013.05.021] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 04/05/2013] [Accepted: 05/30/2013] [Indexed: 01/15/2023]
Abstract
Sustained tumor progression has been attributed to a distinct population of tumor-propagating cells (TPCs). To identify TPCs relevant to lung cancer pathogenesis, we investigated functional heterogeneity in tumor cells isolated from Kras-driven mouse models of non-small-cell lung cancer (NSCLC). CD24(+)ITGB4(+)Notch(hi) cells are capable of propagating tumor growth in both a clonogenic and an orthotopic serial transplantation assay. While all four Notch receptors mark TPCs, Notch3 plays a nonredundant role in tumor cell propagation in two mouse models and in human NSCLC. The TPC population is enriched after chemotherapy, and the gene signature of mouse TPCs correlates with poor prognosis in human NSCLC. The role of Notch3 in tumor propagation may provide a therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Yanyan Zheng
- Cancer Biology Program, Department of Pediatrics, Stanford University School of Medicine, Stanford, California, 94305. U.S.A
| | | | - Leanne C. Sayles
- Cancer Biology Program, Department of Pediatrics, Stanford University School of Medicine, Stanford, California, 94305. U.S.A
| | - Chris Alleyne-Chin
- Cancer Biology Program, Department of Pediatrics, Stanford University School of Medicine, Stanford, California, 94305. U.S.A
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco CA 94132
| | - Dedeepya Vaka
- Cancer Biology Program, Department of Pediatrics, Stanford University School of Medicine, Stanford, California, 94305. U.S.A
| | - Tim D. Knaak
- Stanford Shared FACS Facility, Center for Molecular and Genetic Medicine, Stanford University, Stanford, California, 94305
| | - Marty Bigos
- Stanford Shared FACS Facility, Center for Molecular and Genetic Medicine, Stanford University, Stanford, California, 94305
| | - Yue Xu
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California, 94305. U.S.A
| | - Chuong D. Hoang
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California, 94305. U.S.A
| | - Joseph Shrager
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California, 94305. U.S.A
| | - Hans Joerg Fehling
- Institute of Immunology, University Clinics Ulm, Albert-Einstein-Allee 11, D-89081 Ulm
| | - Dorothy French
- Genentech, Inc. 1 DNA Way, South San Francisco, 94080-4990
| | | | - Zhaoshi Jiang
- Genentech, Inc. 1 DNA Way, South San Francisco, 94080-4990
| | - Erica L. Jackson
- Genentech, Inc. 1 DNA Way, South San Francisco, 94080-4990
- Correspondence: or
| | - E. Alejandro Sweet-Cordero
- Cancer Biology Program, Department of Pediatrics, Stanford University School of Medicine, Stanford, California, 94305. U.S.A
- Correspondence: or
| |
Collapse
|
445
|
Chen CL, Tsukamoto H, Liu JC, Kashiwabara C, Feldman D, Sher L, Dooley S, French SW, Mishra L, Petrovic L, Jeong JH, Machida K. Reciprocal regulation by TLR4 and TGF-β in tumor-initiating stem-like cells. J Clin Invest 2013; 123:2832-49. [PMID: 23921128 PMCID: PMC3696549 DOI: 10.1172/jci65859] [Citation(s) in RCA: 133] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 04/08/2013] [Indexed: 12/17/2022] Open
Abstract
Tumor-initiating stem-like cells (TICs) are resistant to chemotherapy and associated with hepatocellular carcinoma (HCC) caused by HCV and/or alcohol-related chronic liver injury. Using HCV Tg mouse models and patients with HCC, we isolated CD133(+) TICs and identified the pluripotency marker NANOG as a direct target of TLR4, which drives the tumor-initiating activity of TICs. These TLR4/NANOG-dependent TICs were defective in the TGF-β tumor suppressor pathway. Functional oncogene screening of a TIC cDNA library identified Yap1 and Igf2bp3 as NANOG-dependent genes that inactivate TGF-β signaling. Mechanistically, we determined that YAP1 mediates cytoplasmic retention of phosphorylated SMAD3 and suppresses SMAD3 phosphorylation/activation by the IGF2BP3/AKT/mTOR pathway. Silencing of both YAP1 and IGF2BP3 restored TGF-β signaling, inhibited pluripotency genes and tumorigenesis, and abrogated chemoresistance of TICs. Mice with defective TGF-β signaling (Spnb2(+/-) mice) exhibited enhanced liver TLR4 expression and developed HCC in a TLR4-dependent manner. Taken together, these results suggest that the activated TLR4/NANOG oncogenic pathway is linked to suppression of cytostatic TGF-β signaling and could potentially serve as a therapeutic target for HCV-related HCC.
Collapse
MESH Headings
- AC133 Antigen
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Antigens, CD/metabolism
- Antineoplastic Agents/pharmacology
- Base Sequence
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Separation
- Drug Resistance, Neoplasm
- Flow Cytometry
- Gene Expression Regulation, Neoplastic
- Gene Knockdown Techniques
- Glycoproteins/metabolism
- Homeodomain Proteins/metabolism
- Humans
- Inhibitory Concentration 50
- Liver Neoplasms/drug therapy
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Mice, Transgenic
- Nanog Homeobox Protein
- Neoplastic Stem Cells/metabolism
- Niacinamide/analogs & derivatives
- Niacinamide/pharmacology
- Oncogenes
- Peptides/metabolism
- Phenylurea Compounds/pharmacology
- Phosphoproteins/genetics
- Phosphoproteins/metabolism
- RNA, Small Interfering/genetics
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Signal Transduction
- Sirolimus/pharmacology
- Smad Proteins/metabolism
- Sorafenib
- Spheroids, Cellular/metabolism
- Toll-Like Receptor 4/genetics
- Toll-Like Receptor 4/metabolism
- Transcription Factors
- Transcriptional Activation
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
- Tumor Burden
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
- YAP-Signaling Proteins
Collapse
Affiliation(s)
- Chia-Lin Chen
- Department of Molecular Microbiology and Immunology and
Department of Pathology, University of Southern California, Los Angeles, California, USA.
Southern California Research Center for ALPD and Cirrhosis, Los Angeles, California, USA.
Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, USA.
Department of Surgery, University of Southern California, Los Angeles, California, USA.
Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
Department of Pathology, Harbor-UCLA Medical Center, Los Angeles, California, USA.
Department of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hidekazu Tsukamoto
- Department of Molecular Microbiology and Immunology and
Department of Pathology, University of Southern California, Los Angeles, California, USA.
Southern California Research Center for ALPD and Cirrhosis, Los Angeles, California, USA.
Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, USA.
Department of Surgery, University of Southern California, Los Angeles, California, USA.
Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
Department of Pathology, Harbor-UCLA Medical Center, Los Angeles, California, USA.
Department of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jian-Chang Liu
- Department of Molecular Microbiology and Immunology and
Department of Pathology, University of Southern California, Los Angeles, California, USA.
Southern California Research Center for ALPD and Cirrhosis, Los Angeles, California, USA.
Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, USA.
Department of Surgery, University of Southern California, Los Angeles, California, USA.
Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
Department of Pathology, Harbor-UCLA Medical Center, Los Angeles, California, USA.
Department of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Claudine Kashiwabara
- Department of Molecular Microbiology and Immunology and
Department of Pathology, University of Southern California, Los Angeles, California, USA.
Southern California Research Center for ALPD and Cirrhosis, Los Angeles, California, USA.
Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, USA.
Department of Surgery, University of Southern California, Los Angeles, California, USA.
Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
Department of Pathology, Harbor-UCLA Medical Center, Los Angeles, California, USA.
Department of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Douglas Feldman
- Department of Molecular Microbiology and Immunology and
Department of Pathology, University of Southern California, Los Angeles, California, USA.
Southern California Research Center for ALPD and Cirrhosis, Los Angeles, California, USA.
Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, USA.
Department of Surgery, University of Southern California, Los Angeles, California, USA.
Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
Department of Pathology, Harbor-UCLA Medical Center, Los Angeles, California, USA.
Department of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Linda Sher
- Department of Molecular Microbiology and Immunology and
Department of Pathology, University of Southern California, Los Angeles, California, USA.
Southern California Research Center for ALPD and Cirrhosis, Los Angeles, California, USA.
Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, USA.
Department of Surgery, University of Southern California, Los Angeles, California, USA.
Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
Department of Pathology, Harbor-UCLA Medical Center, Los Angeles, California, USA.
Department of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Steven Dooley
- Department of Molecular Microbiology and Immunology and
Department of Pathology, University of Southern California, Los Angeles, California, USA.
Southern California Research Center for ALPD and Cirrhosis, Los Angeles, California, USA.
Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, USA.
Department of Surgery, University of Southern California, Los Angeles, California, USA.
Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
Department of Pathology, Harbor-UCLA Medical Center, Los Angeles, California, USA.
Department of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Samuel W. French
- Department of Molecular Microbiology and Immunology and
Department of Pathology, University of Southern California, Los Angeles, California, USA.
Southern California Research Center for ALPD and Cirrhosis, Los Angeles, California, USA.
Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, USA.
Department of Surgery, University of Southern California, Los Angeles, California, USA.
Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
Department of Pathology, Harbor-UCLA Medical Center, Los Angeles, California, USA.
Department of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lopa Mishra
- Department of Molecular Microbiology and Immunology and
Department of Pathology, University of Southern California, Los Angeles, California, USA.
Southern California Research Center for ALPD and Cirrhosis, Los Angeles, California, USA.
Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, USA.
Department of Surgery, University of Southern California, Los Angeles, California, USA.
Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
Department of Pathology, Harbor-UCLA Medical Center, Los Angeles, California, USA.
Department of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lydia Petrovic
- Department of Molecular Microbiology and Immunology and
Department of Pathology, University of Southern California, Los Angeles, California, USA.
Southern California Research Center for ALPD and Cirrhosis, Los Angeles, California, USA.
Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, USA.
Department of Surgery, University of Southern California, Los Angeles, California, USA.
Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
Department of Pathology, Harbor-UCLA Medical Center, Los Angeles, California, USA.
Department of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Joseph H. Jeong
- Department of Molecular Microbiology and Immunology and
Department of Pathology, University of Southern California, Los Angeles, California, USA.
Southern California Research Center for ALPD and Cirrhosis, Los Angeles, California, USA.
Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, USA.
Department of Surgery, University of Southern California, Los Angeles, California, USA.
Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
Department of Pathology, Harbor-UCLA Medical Center, Los Angeles, California, USA.
Department of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Keigo Machida
- Department of Molecular Microbiology and Immunology and
Department of Pathology, University of Southern California, Los Angeles, California, USA.
Southern California Research Center for ALPD and Cirrhosis, Los Angeles, California, USA.
Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, USA.
Department of Surgery, University of Southern California, Los Angeles, California, USA.
Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
Department of Pathology, Harbor-UCLA Medical Center, Los Angeles, California, USA.
Department of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
446
|
Wen W, Han T, Chen C, Huang L, Sun W, Wang X, Chen SZ, Xiang DM, Tang L, Cao D, Feng GS, Wu MC, Ding J, Wang HY. Cyclin G1 expands liver tumor-initiating cells by Sox2 induction via Akt/mTOR signaling. Mol Cancer Ther 2013; 12:1796-804. [PMID: 23804702 DOI: 10.1158/1535-7163.mct-13-0099] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recurrence and chemoresistance of liver cancer has been attributed to the existence of liver tumor-initiating cells (T-ICs). It is important to decipher the molecular mechanism for acquisition of drug resistance and to design combinatorial therapeutic strategies. Cyclin G1 has been shown to play a pivotal role in initiation and metastasis of hepatocellular carcinoma. In this study, we found that enhanced cyclin G1 expression was associated with drug resistance of hepatoma cells and higher recurrence rate in hepatocellular carcinoma patients. Expression of cyclin G1 was elevated in liver T-ICs and closely correlated with the expression of liver T-IC markers. Forced cyclin G1 expression remarkably enhanced self-renewal and tumorigenicity of hepatoma cells. Cyclin G1 overexpression dramatically upregulated the expression of Sox2 both in vitro and in vivo, which was impaired by chemical inhibitors of Akt/mTOR signaling. Furthermore, blockade of Akt/mTOR signaling or interference of Sox2 expression suppressed cyclin G1-enhanced self-renewal, chemoresistance, and tumorigenicity of hepatoma cells, indicating that cyclin G1 expands liver T-ICs through Sox2 induction via Akt/mTOR signaling pathway. These results suggest that cyclin G1-induced liver T-IC expansion contributes to the recurrence and chemoresistance of hepatoma, and cyclin G1 may be a promising biomarker for individualized therapy of hepatocellular carcinoma patients.
Collapse
Affiliation(s)
- Wen Wen
- Corresponding Authors: Hong-Yang Wang, International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital/Institute, Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
447
|
Badeaux MA, Jeter CR, Gong S, Liu B, Suraneni MV, Rundhaug J, Fischer SM, Yang T, Kusewitt D, Tang DG. In vivo functional studies of tumor-specific retrogene NanogP8 in transgenic animals. Cell Cycle 2013; 12:2395-408. [PMID: 23839044 PMCID: PMC3841319 DOI: 10.4161/cc.25402] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The current study was undertaken to investigate potential oncogenic functions of NanogP8, a tumor-specific retrogene homolog of Nanog (expressed in pluripotent cells), in transgenic animal models. To this end, human primary prostate tumor-derived NanogP8 was targeted to the cytokeratin 14 (K14) cellular compartment, and two lines of K14-NanogP8 mice were derived. The line 1 animals, expressing high levels of NanogP8, experienced perinatal lethality and developmental abnormalities in multiple organs, including the skin, tongue, eye, and thymus in surviving animals. On postnatal day 5 transgenic skin, for example, there was increased c-Myc expression and Ki-67(+) cells accompanied by profound abnormalities in skin development such as thickened interfollicular epidermis and dermis and lack of hypodermis and sebaceous glands. The line 3 mice, expressing low levels of NanogP8, were grossly normal except cataract development by 4-6 mo of age. Surprisingly, both lines of mice do not develop spontaneous tumors related to transgene expression. Even more unexpectedly, high levels of NanogP8 expression in L1 mice actually inhibited tumor development in a two-stage chemical carcinogenesis model. Mechanistic studies revealed that constitutive NanogP8 overexpression in adult L1 mice reduced CD34(+)α6(+) and Lrig-1(+) bulge stem cells, impaired keratinocyte migration, and repressed the expression of many stem cell-associated genes, including Bmp5, Fgfr2, Jmjd1a, and Jun. Our study, for the first time, indicates that transgenically expressed human NanogP8 is biologically functional, but suggests that high levels of NanogP8 may disrupt normal developmental programs and inhibit tumor development by depleting stem cells.
Collapse
Affiliation(s)
- Mark A Badeaux
- Department of Molecular Carcinogenesis; University of Texas MD Anderson Cancer Center; Smithville, TX USA; Program in Molecular Carcinogenesis; University of Texas Graduate School of Biomedical Sciences (GSBS); Houston, TX USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
448
|
Gedaly R, Galuppo R, Musgrave Y, Angulo P, Hundley J, Shah M, Daily MF, Chen C, Cohen DA, Spear BT, Evers BM. PKI-587 and sorafenib alone and in combination on inhibition of liver cancer stem cell proliferation. J Surg Res 2013; 185:225-30. [PMID: 23769634 DOI: 10.1016/j.jss.2013.05.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Revised: 05/02/2013] [Accepted: 05/03/2013] [Indexed: 12/17/2022]
Abstract
BACKGROUND Deregulated Ras/Raf/mitogen-activated protein kinase and PI3 K/AKT/mTOR signaling pathways are significant in hepatocellular carcinoma proliferation (HCC). In this study we evaluated differences in the antiproliferative effect of dual PI3 K/Akt/mTOR and Ras/Raf/mitogen-activated protein kinase inhibition of non liver cancer stem cell lines (PLC and HuH7) and liver cancer stem cell (LCSC) lines (CD133, CD44, CD24, and aldehyde dehydrogenase 1-positive cells). MATERIALS AND METHODS Flow cytometry was performed on the resulting tumors to identify the LCSC markers CD133, CD44, CD24, and aldehyde dehydrogenase 1. Methylthiazol tetrazolium assay was used to assess cellular proliferation. Finally, a Western blot assay was used to evaluate for inhibition of specific enzymes in these two signaling pathways. RESULTS Using flow cytometry, we found that LCSC contain 64.4% CD133 + cells, 83.2% CD44 + cells, and 96.4% CD24 + cells. PKI-587 and sorafenib caused inhibiton of LCSC and HCC cell proliferation. PLC cells were more sensitive to PKI-587 than LCSC or Huh7 (P < 0.001). Interestingly, HuH7 cells were more sensitive to sorafenib than LCSC or PLC cells. Additionally, combination therapy with PKI-587 and sorafenib caused significantly more inhibition than monotherapy in HuH7, PLC, and LCSC. Using the methylthiazol tetrazolium assay, we found that the LCSC proliferation was inhibited with sorafenib monotherapy 39% at 5 μM (P < 0.001; n = 12) and 67% by PKI-587 at 0.1 μM (P = 0.002, n = 12) compared with control. The combination of PKI-587 and sorafenib, however, synergistically inhibited LCSC proliferation by 86% (P = 0.002; n = 12). CONCLUSIONS LCSC (CD133+, CD44+, CD24+) were able to develop very aggressive tumors with low cell concentrations at 4 to 6 wk. Cells CD133+, CD44+, CD24+, which demonstrated at least moderate resistance to therapy in vitro. The combination of PKI-587 and sorafenib was better than either drug alone at inhibiting of LCSC and on HCC cell proliferation.
Collapse
Affiliation(s)
- Roberto Gedaly
- Department of Surgery, University of Kentucky, College of Medicine, Lexington, Kentucky.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
449
|
Chai S, Ma S. Clinical implications of microRNAs in liver cancer stem cells. CHINESE JOURNAL OF CANCER 2013; 32:419-26. [PMID: 23668930 PMCID: PMC3845583 DOI: 10.5732/cjc.013.10038] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The prognosis of patients diagnosed with hepatocellular carcinoma (HCC) is often dismal, mainly due to late presentation, high recurrence rate, and frequent resistance to chemotherapy and radiotherapy. Accumulating evidence on the differential microRNA (miRNA) expression patterns between non-tumor and HCC tissues or between liver cancer stem cells (CSCs) and non-CSC subsets and the significant clinical implications of these differences suggest that miRNAs are a promising, non-invasive marker for the prognosis and diagnosis of the disease. This perspective article summarizes the current knowledge of miRNAs in liver CSCs and highlights the need for further investigations of the role of miRNAs in regulating liver CSC subsets for possible future clinical applications.
Collapse
Affiliation(s)
- Stella Chai
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, P. R. China.
| | | |
Collapse
|
450
|
Cao J, Li L, Chen C, Lv C, Meng F, Zeng L, Li Z, Wu Q, Zhao K, Pan B, Cheng H, Chen W, Xu K. RNA interference-mediated silencing of NANOG leads to reduced proliferation and self-renewal, cell cycle arrest and apoptosis in T-cell acute lymphoblastic leukemia cells via the p53 signaling pathway. Leuk Res 2013; 37:1170-7. [PMID: 23683786 DOI: 10.1016/j.leukres.2013.04.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 04/13/2013] [Accepted: 04/18/2013] [Indexed: 01/23/2023]
Abstract
NANOG is critical for maintaining the self-renewal and proliferative properties of embryonic stem cells. Here we found that cultured T-cell acute lymphoblastic leukemia (T-ALL) cells, as well as human primary T-ALL cells, express a functional variant of NANOG. NANOG mRNA is derived predominantly from a retrogene locus termed NANOGP8. Furthermore, we showed that RNA interference-mediated NANOG knockdown inhibited cell proliferation, reduced self-renewal, promoted apoptosis and arrested the cell cycle through a p53-mediated pathway in leukemic cells. These findings demonstrate the oncogenic potential of this pluripotent gene in human T-ALL cells.
Collapse
Affiliation(s)
- Jiang Cao
- Department of Hematology, Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|