101
|
Epithelial-mesenchymal transition promotes SOX2 and NANOG expression in bladder cancer. J Transl Med 2017; 97:567-576. [PMID: 28240746 DOI: 10.1038/labinvest.2017.17] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 01/05/2017] [Accepted: 01/09/2017] [Indexed: 12/26/2022] Open
Abstract
Bladder cancer is the most common malignant tumor of the urothelium and is classified into non-muscle-invasive bladder cancer (NMIBC) and muscle-invasive bladder cancer (MIBC). Stemness markers such as SOX2 and NANOG are frequently overexpressed in various aggressive cancers, including MIBC; epithelial-mesenchymal transition (EMT) has been proposed as a potential trigger of stemness in cancers. To determine whether cancer stemness is acquired via EMT in bladder cancer, we studied the effect of EMT on the expression of SOX2 and NANOG in bladder cancer cell lines. We also analyzed their expression in clinical tissue samples. Our results revealed that a potent EMT inducer (transforming growth factor β1) reduced the expression of the epithelial marker E-cadherin and increased expression of both SOX2 and NANOG in epithelial-type bladder cancer cells. As for clinical bladder cancer samples, in NMIBC, E-cadherin expression was slightly diminished, and the expression of both SOX2 and NANOG was negligible. In contrast, in MIBC, E-cadherin expression was highly and heterogeneously diminished, while the expression of both SOX2 and NANOG was increased. We also noticed that either E-cadherin or SOX2 (or NANOG) was expressed (ie, in a manner exclusive of each other). In addition, the concentration of E-cadherin showed a significant negative correlation with tumor grade and stage, while expression of SOX2 and NANOG positively correlated with those clinicopathological parameters. These findings suggest that EMT promotes stemness of bladder cancer cells, contributing to tumor aggressiveness. This EMT-cancer stemness axis may also play an important role in the pathogenesis of NMIBC and MIBC.Laboratory Investigation advance online publication, 27 February 2017; doi:10.1038/labinvest.2017.17.
Collapse
|
102
|
Liu B, Gong S, Li Q, Chen X, Moore J, Suraneni MV, Badeaux MD, Jeter CR, Shen J, Mehmood R, Fan Q, Tang DG. Transgenic overexpression of NanogP8 in the mouse prostate is insufficient to initiate tumorigenesis but weakly promotes tumor development in the Hi-Myc mouse model. Oncotarget 2017; 8:52746-52760. [PMID: 28881767 PMCID: PMC5581066 DOI: 10.18632/oncotarget.17186] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 03/21/2017] [Indexed: 11/25/2022] Open
Abstract
This project was undertaken to address a critical cancer biology question: Is overexpression of the pluripotency molecule Nanog sufficient to initiate tumor development in a somatic tissue? Nanog1 is critical for the self-renewal and pluripotency of ES cells, and its retrotransposed homolog, NanogP8 is preferentially expressed in somatic cancer cells. Our work has shown that shRNA-mediated knockdown of NanogP8 in prostate, breast, and colon cancer cells inhibits tumor regeneration whereas inducible overexpression of NanogP8 promotes cancer stem cell phenotypes and properties. To address the key unanswered question whether tissue-specific overexpression of NanogP8 is sufficient to promote tumor development in vivo, we generated a NanogP8 transgenic mouse model, in which the ARR2PB promoter was used to drive NanogP8 cDNA. Surprisingly, the ARR2PB-NanogP8 transgenic mice were viable, developed normally, and did not form spontaneous tumors in >2 years. Also, both wild type and ARR2PB-NanogP8 transgenic mice responded similarly to castration and regeneration and castrated ARR2PB-NanogP8 transgenic mice also did not develop tumors. By crossing the ARR2PB-NanogP8 transgenic mice with ARR2PB-Myc (i.e., Hi-Myc) mice, we found that the double transgenic (i.e., ARR2PB-NanogP8; Hi-Myc) mice showed similar tumor incidence and histology to the Hi-Myc mice. Interestingly, however, we observed white dots in the ventral lobes of the double transgenic prostates, which were characterized as overgrown ductules/buds featured by crowded atypical Nanog-expressing luminal cells. Taken together, our present work demonstrates that transgenic overexpression of NanogP8 in the mouse prostate is insufficient to initiate tumorigenesis but weakly promotes tumor development in the Hi-Myc mouse model.
Collapse
Affiliation(s)
- Bigang Liu
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, TX 78957, USA
| | - Shuai Gong
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, TX 78957, USA.,Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Qiuhui Li
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, TX 78957, USA.,Department of Pharmacology & Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Xin Chen
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, TX 78957, USA.,Department of Pharmacology & Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - John Moore
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, TX 78957, USA
| | - Mahipal V Suraneni
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, TX 78957, USA
| | - Mark D Badeaux
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, TX 78957, USA
| | - Collene R Jeter
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, TX 78957, USA
| | - Jianjun Shen
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, TX 78957, USA
| | - Rashid Mehmood
- Department of Pharmacology & Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Qingxia Fan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Dean G Tang
- Department of Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, Science Park, Smithville, TX 78957, USA.,Department of Pharmacology & Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.,Cancer Stem Cell Institute, Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| |
Collapse
|
103
|
Freitag D, McLean AL, Simon M, Koch A, Grube S, Walter J, Kalff R, Ewald C. NANOG overexpression and its correlation with stem cell and differentiation markers in meningiomas of different WHO grades. Mol Carcinog 2017; 56:1953-1964. [PMID: 28345785 DOI: 10.1002/mc.22653] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 03/10/2017] [Accepted: 03/23/2017] [Indexed: 01/15/2023]
Abstract
NANOG, as a key regulator of pluripotency and acting synergistically with other factors, has been described as a crucial transcription factor in various types of cancer. In meningiomas the expression of this marker has not yet been described. With our study, we aimed to identify and localize NANOG and other possible markers of pluripotency, stem cell properties and differentiation in meningioma tissue, to elucidate a possible effect on tumorigenesis. The gene expression levels of NANOG (NANOG1 and NANOGP8), SOX2, OCT4, KLF4, ABCG2, CMYC, MSI1, CD44, NOTCH1, NES, SALL4B, TP53, and EPAS1 were quantitatively examined using RT-qPCR in 33 surgical specimens of low- (WHO grade I) as well as in high-grade (WHO grade II/III) meningiomas with dural tissue as reference. Immunofluorescence co-localization analysis following confocal fluorescence microscopy for NANOG, OCT4, SOX2, Nestin, KI-67, and CD44 was also performed. There was a significant overexpression of NANOG, MSI1, and EPAS1 and a downregulation of NES in all examined tumors. Subgroup analysis (WHO grade I versus grade II/III) revealed differences in the expression of NANOG, CD44, and MSI1. We found 1% NANOG-positive (NANOG+) cells in low-grade and 2% in grade II/III meningiomas co-expressing the other mentioned markers in various compositions. In particular, NANOG+ cells expressing SOX2 and OCT4 were successfully identified (26% low-grade versus 20% high-grade). Our data reveal an overexpression of NANOG and other markers of pluripotency and stemness in meningiomas. Such potentially pluripotent "stem cell-like" cells may have an impact on tumorigenesis and progression in human meningiomas.
Collapse
Affiliation(s)
- Diana Freitag
- Department of Neurosurgery, Section of Experimental Neurooncology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Aaron Lawson McLean
- Department of Neurosurgery, Section of Experimental Neurooncology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Michèle Simon
- Department of Neurosurgery, Section of Experimental Neurooncology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany.,Department of Neurosurgery, Vivantes Klinikum im Friedrichshain, Berlin, Germany
| | - Arend Koch
- Department of Neuropathology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Susanne Grube
- Department of Neurosurgery, Section of Experimental Neurooncology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Jan Walter
- Department of Neurosurgery, Section of Experimental Neurooncology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Rolf Kalff
- Department of Neurosurgery, Section of Experimental Neurooncology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Christian Ewald
- Department of Neurosurgery, Section of Experimental Neurooncology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany.,Department of Neurosurgery, Städtisches Klinikum Brandenburg, Brandenburg an der Havel, Germany
| |
Collapse
|
104
|
Guo T, Kong J, Liu Y, Li Z, Xia J, Zhang Y, Zhao S, Li F, Li J, Gu C. Transcriptional activation of NANOG by YBX1 promotes lung cancer stem-like properties and metastasis. Biochem Biophys Res Commun 2017; 487:153-159. [PMID: 28400280 DOI: 10.1016/j.bbrc.2017.04.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 04/07/2017] [Indexed: 01/11/2023]
Abstract
Aberrant overexpression of the transcription/translation factor Y-box-binding protein-1 (YBX1) is associated with non-small cell lung cancer (NSCLC) aggressiveness. Cancer stem cells (CSCs) contribute to the tumorigenesis and metastasis of NSCLC. Hitherto, the mechanism by which YBX1 regulates CSCs and metastasis in NSCLC remains unclear. Here, we demonstrated that YBX1 levels were elevated in NSCLC tissues and cell lines. Enforced expression of YBX1 promoted NSCLC cells invasion, sphere forming ability and ALDH1+ population. Conversely, reduced YBX1 impaired CSC properties of NSCLC cells in vitro and tumor-initiating frequencies, as well as metastasis in vivo. Importantly, we described a mechanism whereby YBX1 directly promoted NANOG, a transcription factor, transcriptional activation. Depletion of NANOG abolished the enhanced ability of invasion and sphere formation in YBX1 elevated-A549 cells. Collectively, these findings demonstrate a novel role of YBX1 in maintaining the stemness of CSCs and metastasis, unveiling YBX1 as promising therapeutic target for NSCLC treatments.
Collapse
Affiliation(s)
- Tao Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China; Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Jing Kong
- College of Stomatology, Dalian Medical University, Dalian, 116044, China
| | - Yang Liu
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Zhuoshi Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China; Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Jianglong Xia
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Yan Zhang
- Department of Radiation Oncology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, 250000, China
| | - Shilei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China; Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Fengzhou Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China; Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Jinxiu Li
- Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| | - Chundong Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China; Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
105
|
Adamowicz J, Pakravan K, Bakhshinejad B, Drewa T, Babashah S. Prostate cancer stem cells: from theory to practice. Scand J Urol 2017. [DOI: 10.1080/21681805.2017.1283360] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Jan Adamowicz
- Chair of Urology, Department of Regenerative Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Katayoon Pakravan
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Babak Bakhshinejad
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Tomasz Drewa
- Chair of Urology, Department of Regenerative Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
106
|
Stress granule-associated protein G3BP2 regulates breast tumor initiation. Proc Natl Acad Sci U S A 2017; 114:1033-1038. [PMID: 28096337 DOI: 10.1073/pnas.1525387114] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Breast tumors contain tumorigenic cancer cells, termed "tumor-initiating cells" (TICs), which are capable of both replenishing themselves and giving rise to populations of nontumorigenic breast cancer cells (non-TICs). However, the molecular mechanisms responsible for breast tumor initiation remain poorly understood. Here we describe a chemical screening strategy to identify small molecules that enhance the effect of chemotherapeutic agents on TIC-enriched breast cancer cells. We identified proteins that interact with the lead compound C108, including the stress granule-associated protein, GTPase-activating protein (SH3 domain)-binding protein 2, G3BP2. G3BP2 regulates breast tumor initiation through the stabilization of Squamous cell carcinoma antigen recognized by T cells 3 (SART3) mRNA, which leads to increased expression of the pluripotency transcription factors Octamer-binding protein 4 (Oct-4) and Nanog Homeobox (Nanog). Our findings suggest that G3BP2 is important for the process of breast cancer initiation. Furthermore, these data suggest a possible connection between stress granule formation and tumor initiation in breast cancer cells.
Collapse
|
107
|
Xu DD, Zhou PJ, Wang Y, Zhang Y, Zhang R, Zhang L, Chen SH, Fu WY, Ruan BB, Xu HP, Hu CZ, Tian L, Qin JH, Wang S, Wang X, Liu QY, Ren Z, Gu XK, Li YH, Liu Z, Wang YF. miR-150 Suppresses the Proliferation and Tumorigenicity of Leukemia Stem Cells by Targeting the Nanog Signaling Pathway. Front Pharmacol 2016; 7:439. [PMID: 27917123 PMCID: PMC5114241 DOI: 10.3389/fphar.2016.00439] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 11/03/2016] [Indexed: 12/18/2022] Open
Abstract
Proliferation, a key feature of cancer cells, accounts for the majority of cancer-related diseases resulting in mortality. MicroRNAs (miRNAs) plays important post-transcriptional modulation roles by acting on multiple signaling pathways, but the underlying mechanism in proliferation and tumorigenicity is unclear. Here, we identified the role of miR-150 in proliferation and tumorigenicity in leukemia stem cells (LSCs; CD34+CD38- cells). miR-150 expression was significantly down-regulated in LSCs from leukemia cell lines and clinical samples. Functional assays demonstrated that increased miR-150 expression inhibited proliferation and clonal and clonogenic growth, enhanced chemosensitivity, and attenuated tumorigenic activity of LSCs in vitro. Transplantation animal studies revealed that miR-150 overexpression progressively abrogates tumor growth. Immunohistochemistry assays demonstrated that miR-150 overexpression enhanced caspase-3 level and reduced Ki-67 level. Moreover, luciferase reporter assays indicated Nanog is a direct and functional target of miR-150. Nanog silencing using small interfering RNA recapitulated anti-proliferation and tumorigenicity inhibition effects. Furthermore, miR-150 directly down-regulated the expression of other cancer stem cell factors including Notch2 and CTNNB1. These results provide insights into the specific biological behavior of miR-150 in regulating LSC proliferation and tumorigenicity. Targeting this miR-150/Nanog axis would be a helpful therapeutic strategy to treat acute myeloid leukemia.
Collapse
Affiliation(s)
- Dan-Dan Xu
- College of Life Science and Technology, Jinan UniversityGuangzhou, China; College of Biology Technolgy, Guangdong Food and Drug Vocational CollegeGuangzhou, China
| | - Peng-Jun Zhou
- College of Life Science and Technology, Jinan University Guangzhou, China
| | - Ying Wang
- College of Life Science and Technology, Jinan UniversityGuangzhou, China; Faculty of Environmental and Biological Engineering, Guangdong University of Petrochemical TechnologyMaoming, China
| | - Yi Zhang
- Section of Otolaryngology, Department of Surgery, Yale School of Medicine, New Haven CT, USA
| | - Rong Zhang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center Guangzhou, China
| | - Li Zhang
- College of Life Science and Technology, Jinan University Guangzhou, China
| | - Su-Hong Chen
- College of Biology Technolgy, Guangdong Food and Drug Vocational College Guangzhou, China
| | - Wu-Yu Fu
- Faculty of Environmental and Biological Engineering, Guangdong University of Petrochemical Technology Maoming, China
| | - Bi-Bo Ruan
- Faculty of Environmental and Biological Engineering, Guangdong University of Petrochemical Technology Maoming, China
| | - Hai-Peng Xu
- College of Life Science and Technology, Jinan University Guangzhou, China
| | - Chao-Zhi Hu
- College of Life Science and Technology, Jinan University Guangzhou, China
| | - Lu Tian
- College of Life Science and Technology, Jinan University Guangzhou, China
| | - Jin-Hong Qin
- College of Life Science and Technology, Jinan University Guangzhou, China
| | - Sheng Wang
- College of Life Science and Technology, Jinan University Guangzhou, China
| | - Xiao Wang
- College of Life Science and Technology, Jinan University Guangzhou, China
| | - Qiu-Ying Liu
- College of Life Science and Technology, Jinan University Guangzhou, China
| | - Zhe Ren
- College of Life Science and Technology, Jinan University Guangzhou, China
| | - Xue-Kui Gu
- The First Affiliated Hospital, Guangzhou Hospital of Traditional Chinese Medicine Guangzhou, China
| | - Yao-He Li
- The First Affiliated Hospital, Guangzhou Hospital of Traditional Chinese Medicine Guangzhou, China
| | - Zhong Liu
- College of Life Science and Technology, Jinan University Guangzhou, China
| | - Yi-Fei Wang
- College of Life Science and Technology, Jinan University Guangzhou, China
| |
Collapse
|
108
|
Jeter CR, Liu B, Lu Y, Chao HP, Zhang D, Liu X, Chen X, Li Q, Rycaj K, Calhoun-Davis T, Yan L, Hu Q, Wang J, Shen J, Liu S, Tang DG. NANOG reprograms prostate cancer cells to castration resistance via dynamically repressing and engaging the AR/FOXA1 signaling axis. Cell Discov 2016; 2:16041. [PMID: 27867534 PMCID: PMC5109294 DOI: 10.1038/celldisc.2016.41] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/18/2016] [Indexed: 12/24/2022] Open
Abstract
The pluripotency transcription factor NANOG has been implicated in tumor development, and NANOG-expressing cancer cells manifest stem cell properties that sustain tumor homeostasis, mediate therapy resistance and fuel tumor progression. However, how NANOG converges on somatic circuitry to trigger oncogenic reprogramming remains obscure. We previously reported that inducible NANOG expression propels the emergence of aggressive castration-resistant prostate cancer phenotypes. Here we first show that endogenous NANOG is required for the growth of castration-resistant prostate cancer xenografts. Genome-wide chromatin immunoprecipitation sequencing coupled with biochemical assays unexpectedly reveals that NANOG co-occupies a distinctive proportion of androgen receptor/Forkhead box A1 genomic loci and physically interacts with androgen receptor and Forkhead box A1. Integrative analysis of chromatin immunoprecipitation sequencing and time-resolved RNA sequencing demonstrates that NANOG dynamically alters androgen receptor/Forkhead box A1 signaling leading to both repression of androgen receptor-regulated pro-differentiation genes and induction of genes associated with cell cycle, stem cells, cell motility and castration resistance. Our studies reveal global molecular mechanisms whereby NANOG reprograms prostate cancer cells to a clinically relevant castration-resistant stem cell-like state driven by distinct NANOG-regulated gene clusters that correlate with patient survival. Thus, reprogramming factors such as NANOG may converge on and alter lineage-specific master transcription factors broadly in somatic cancers, thereby facilitating malignant disease progression and providing a novel route for therapeutic resistance.
Collapse
Affiliation(s)
- Collene R Jeter
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center , Smithville, TX, USA
| | - Bigang Liu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center , Smithville, TX, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center , Smithville, TX, USA
| | - Hsueh-Ping Chao
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center , Smithville, TX, USA
| | - Dingxiao Zhang
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX, USA; Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Xin Liu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center , Smithville, TX, USA
| | - Xin Chen
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX, USA; Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Qiuhui Li
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX, USA; Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Kiera Rycaj
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX, USA; Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Tammy Calhoun-Davis
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center , Smithville, TX, USA
| | - Li Yan
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute , Buffalo, NY, USA
| | - Qiang Hu
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute , Buffalo, NY, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute , Buffalo, NY, USA
| | - Jianjun Shen
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center , Smithville, TX, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute , Buffalo, NY, USA
| | - Dean G Tang
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX, USA; Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA; Cancer Stem Cell Institute, Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China; Centers for Cancer Epigenetics, Stem Cell and Developmental Biology, RNA Interference and Non-coding RNAs and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
109
|
Gawlik-Rzemieniewska N, Bednarek I. The role of NANOG transcriptional factor in the development of malignant phenotype of cancer cells. Cancer Biol Ther 2016; 17:1-10. [PMID: 26618281 DOI: 10.1080/15384047.2015.1121348] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
NANOG is a transcription factor that is involved in the self-renewal of embryonic stem cells (ES) and is a critical factor for the maintenance of the undifferentiated state of pluripotent cells. Extensive data in the literature show that the NANOG gene is aberrantly expressed during the development of malignancy in cancer cells. ES and cancer stem cells (CSCs), a subpopulation of cancer cells within the tumor, are thought to share common phenotypic properties. This review describes the role of NANOG in cancer cell proliferation, epithelial-mesenchymal transition (EMT), apoptosis and metastasis. In addition, this paper illustrates a correlation between NANOG and signal transducer and activator of transcription 3 (STAT3) in the maintenance of cancer stem cell properties and multidrug resistance. Together, the available data demonstrate that NANOG is strictly involved in the process of carcinogenesis and is a potential prognostic marker of malignant tumors.
Collapse
Affiliation(s)
- Natalia Gawlik-Rzemieniewska
- a School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Biotechnology and Genetic Engineering, Medical University of Silesia , Katowice , Poland
| | - Ilona Bednarek
- a School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Biotechnology and Genetic Engineering, Medical University of Silesia , Katowice , Poland
| |
Collapse
|
110
|
Park YS, Nemeño JGE, Choi NY, Lee JI, Ko K, Choi SC, Kim WS, Han DW, Tapia N, Ko K. Ectopic overexpression of Nanog induces tumorigenesis in non-tumorous fibroblasts. Biol Chem 2016; 397:249-55. [PMID: 26733157 DOI: 10.1515/hsz-2015-0255] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/18/2015] [Indexed: 01/06/2023]
Abstract
Key regulatory genes in pluripotent stem cells are of interest not only as reprogramming factors but also as regulators driving tumorigenesis. Nanog is a transcription factor involved in the maintenance of embryonic stem cells and is one of the reprogramming factors along with Oct4, Sox2, and Lin28. Nanog expression has been detected in different types of tumors, and its expression is a poor prognosis for cancer patients. However, there is no clear evidence that Nanog is functionally involved in tumorigenesis. In this study, we induced overexpression of Nanog in mouse embryonic fibroblast cells and subsequently assessed their morphological changes, proliferation rate, and tumor formation ability. We found that Nanog overexpression induced immortalization of mouse embryonic fibroblast cells (MEFs) and increased their proliferation rate in vitro. We also found that formation of tumors after subcutaneous injection of retroviral-Nanog infected MEFs (N-MEFs) into athymic mouse. Cancer-related genes such as Bmi1 were expressed at high levels in N-MEFs. Hence, our results demonstrate that Nanog is able to transform normal somatic cells into tumor cells.
Collapse
|
111
|
CRISPR/Cas9-mediated gene knockout of NANOG and NANOGP8 decreases the malignant potential of prostate cancer cells. Oncotarget 2016; 6:22361-74. [PMID: 26087476 PMCID: PMC4673169 DOI: 10.18632/oncotarget.4293] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/03/2015] [Indexed: 01/03/2023] Open
Abstract
NANOG expression in prostate cancer is highly correlated with cancer stem cell characteristics and resistance to androgen deprivation. However, it is not clear whether NANOG or its pseudogenes contribute to the malignant potential of cancer. We established NANOG- and NANOGP8-knockout DU145 prostate cancer cell lines using the CRISPR/Cas9 system. Knockouts of NANOG and NANOGP8 significantly attenuated malignant potential, including sphere formation, anchorage-independent growth, migration capability, and drug resistance, compared to parental DU145 cells. NANOG and NANOGP8 knockout did not inhibit in vitro cell proliferation, but in vivo tumorigenic potential decreased significantly. These phenotypes were recovered in NANOG- and NANOGP8-rescued cell lines. These results indicate that NANOG and NANOGP8 proteins are expressed in prostate cancer cell lines, and NANOG and NANOGP8 equally contribute to the high malignant potential of prostate cancer.
Collapse
|
112
|
Toraih EA, Fawzy MS, El-Falouji AI, Hamed EO, Nemr NA, Hussein MH, Abd El Fadeal NM. Stemness-related transcriptional factors and homing gene expression profiles in hepatic differentiation and cancer. Mol Med 2016; 22:653-663. [PMID: 27623812 DOI: 10.2119/molmed.2016.00096] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 08/26/2016] [Indexed: 12/12/2022] Open
Abstract
Stem cell transcriptional signature activation is an essential event in the development of cancer. This study aimed to investigate the differential expression profile of three pluripotency-associated genes (OCT4, NANOG, and SOX2), G-protein-coupled chemokine receptor 4 (CXCR4) and the ligand (CXCL2), and alpha feto-protein (AFP) in hepatogenic differentiated stem cells and in sera of hepatitis C virus (HCV) and HCV-induced hepatocellular carcinoma (HCC) patients. Mesenchymal stem cells derived from umbilical cord blood were differentiated using hepatogenic differentiation media. Serum specimens were collected from 96 patients (32 cirrhotic HCV, 32 early HCC, and 32 late HCC) and 96 controls. Real-time quantitative reverse transcription polymerase chain reaction was performed for relative quantification of the 6 target genes using LIVAC method. In silico network analysis was also executed to explore the pluripotency and tumorigenic regulatory circuits in liver cancer. The expression levels of all genes declined gradually during the stages of stem cell differentiation. On univariate and multivariate analyses, NANOG, CXCR4 and AFP were significantly up-regulated in HCC patients with late clinical stage. In contrast, SOX2 and CXCL2 were markedly over-expressed in cirrhotic patients and could be used for clear demarcation between cirrhotic and HCC patients in our cases. In conclusion, our data highlight the potential role of SOX2 stem cell marker and CXCL2 chemokine in liver cell degeneration and fibrogenesis in HCV-induced hepatic cirrhosis in our sample of the Egyptian population. In addition, the significant association of NANOG and CXCR4 high-expression with late HCC, could contribute to the acquisition of stem cell-like properties in hepatic cancer and dissemination in late stages, respectively. Taken together, our results could have a potential application in HCC prognosis and treatment.
Collapse
Affiliation(s)
- Eman A Toraih
- Department of Histology and Cell Biology, Genetics Unit, Faculty of Medicine, Suez Canal University, Ismailia, Egypt, P.O. 41522
| | - Manal S Fawzy
- Department of Medical Biochemistry, Faculty of Medicine, Suez Canal University, Ismailia, Egypt, P.O. 41522
| | | | - Elham O Hamed
- Clinical Pathology Department, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Nader A Nemr
- Endemic and Infectious diseases Department, Suez Canal University, Ismailia, Egypt
| | | | - Noha M Abd El Fadeal
- Department of Medical Biochemistry, Faculty of Medicine, Suez Canal University, Ismailia, Egypt, P.O. 41522
| |
Collapse
|
113
|
Hattermann K, Flüh C, Engel D, Mehdorn HM, Synowitz M, Mentlein R, Held-Feindt J. Stem cell markers in glioma progression and recurrence. Int J Oncol 2016; 49:1899-1910. [PMID: 27600094 DOI: 10.3892/ijo.2016.3682] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 06/13/2016] [Indexed: 11/05/2022] Open
Abstract
Aggressive cancer cells show histological similarities to embryonic stem cells. As differentiated cells can re-acquire pluripotency and self-renewal by transfection with the transcription factors OCT4, SOX2, KLF4 and MYC, with Nanog as readout for success, we comprehensively investigated their occurrence and frequency in human astrocytomas of different malignancy grades, primary and matched recurrent glioblastomas, short- and long-term glioblastoma cultures and glioma cell lines. Among astrocytomas, mRNA expression of OCT4, MYC and (less robust) KLF4 increased with malignancy, while in recurrent glioblastomas MYC expression slightly decreased. Correlation analysis revealed distinct positive correlation between distinct stem cell markers, and this effect was most prominent in the recurrent glioblastoma cohort. In situ, embryonic stem cell factors were found also in more differentiated tumor regions. Respective cells were rarely actively proliferating and showed single or combined expression signatures, which, at least in parts, corresponded to observed positive correlations of mRNA expression. However, a 'master-marker' defining the complete glioma stem cell subset could not be confirmed. In glioma cell lines, long- and short-term cultures, embryonic markers were detected at comparable levels. Upon exposure to temozolomide, increased expression of KLF4 (and lesser Nanog and OCT4) was observed. Experimental intrinsic overexpression of SOX2, KLF4 or OCT4 did not affect the other stem cell factors. The embryonic stem cell factors comprehensively investigated in this project can control self-renewal and pluripotency, and therefore tumorigenicity. They should be considered for the development of future diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
| | - Charlotte Flüh
- Department of Neurosurgery, University Medical Center Schleswig-Holstein UKSH, Campus Kiel, D-24105 Kiel, Germany
| | - Dorothee Engel
- Department of Anatomy, University of Kiel, D-24098 Kiel, Germany
| | - H Maximilian Mehdorn
- Department of Neurosurgery, University Medical Center Schleswig-Holstein UKSH, Campus Kiel, D-24105 Kiel, Germany
| | - Michael Synowitz
- Department of Neurosurgery, University Medical Center Schleswig-Holstein UKSH, Campus Kiel, D-24105 Kiel, Germany
| | - Rolf Mentlein
- Department of Anatomy, University of Kiel, D-24098 Kiel, Germany
| | - Janka Held-Feindt
- Department of Neurosurgery, University Medical Center Schleswig-Holstein UKSH, Campus Kiel, D-24105 Kiel, Germany
| |
Collapse
|
114
|
Zhang S, Shu R, Yue M, Zhang S. Effect of Over-Expression of Zinc-Finger Protein (ZFX) on Self-Renewal and Drug-Resistance of Hepatocellular Carcinoma. Med Sci Monit 2016; 22:3025-34. [PMID: 27566731 PMCID: PMC5012459 DOI: 10.12659/msm.897699] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background X-chromosome-coupled zinc finger protein (ZFX) in the Zfy protein family is abundantly expressed in both embryonic and hematopoietic stem cells (HSCs). ZFX exist in various tumor cells and is correlated with proliferation and survival of tumor cells. As a malignant tumor with high invasiveness, hepatocellular carcinoma (HCC) may present resistance against chemotherapy and features of stem cells. This study aimed to explore the expression of ZFX in HCC cells, in an attempt to illustrate the role of ZFX in tumorigenesis. Material/Methods The expression of ZFX in tumor tissues was quantified by RT-PCR. The ZFX expression was then silenced to evaluate the stem cell-like features of HCC cells, including self-renewal, colony formation, and cell cycle, along with the sensitivity to cisplatin. Xenograft of ZFX-overexpressed HCC on nude mice was performed to evaluate the in vivo effect of ZFX on tumor growth. Results Quantitative RT-PCR showed over-expression of ZFX in 51.8% of HCC tumors. The silencing of ZFX gene inhibited the self-renewal, colony formation, and proliferation ability of HCC cells (p<0.05 in all cases) via the cell cycle arrest at G0/G1 phase, in addition to the elevated sensitivity of tumor cells to cisplatin (p<0.001). Further studies showed that binding between ZFX and promoter regions of Nanog or SOX-2 regulatory factor initiate their expression in HCC cells. The xenograft experiment indicated the potentiation of tumor growth by ZFX over-expression. Conclusions ZFX is over-expressed in HCC cells, and correlates with stem cell-like features and pleiotropic characteristics.
Collapse
Affiliation(s)
- Shuhong Zhang
- Department of Gastroenterology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Ronghua Shu
- Department of Gastroenterology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Meng Yue
- Department of Gastroenterology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Shuhong Zhang
- Department of Gastroenterology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
115
|
Jiang Z, Liu Y, Wang C. Oncogenic NanogP8 expression regulates cell proliferation and migration through the Akt/mTOR signaling pathway in human gastric cancer - SGC-7901cell line. Onco Targets Ther 2016; 9:4859-66. [PMID: 27563247 PMCID: PMC4984828 DOI: 10.2147/ott.s97861] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Although elevated expression of NanogP8 has been detected in many human tumor tissues, its role in gastric tumorigenesis remains unclear. Therefore, this study aimed to investigate the function and regulatory mechanism of NanogP8 in gastric cancer. METHODS In this study, NanogP8 cDNA was amplified by real time polymerase chain reaction from the human gastric cancer cell line SGC-7901. The shRNA for RNA interference was established. The NanogP8, pAkt, Akt, pERK, ERK, p-mTOR, and mTOR proteins were detected by using the Western blot assay. Cell viability was evaluated by using the CCK-8 assay. Cell migration and invasion were also examined by using the transwell assay. RESULTS The results indicated that the NanogP8 overexpression promoted proliferation and migration of SGC-7901 cell line, whereas its ablation exerted opposite effects. Interestingly, NanogP8 activated Akt, a key mediator of survival signals, and without affecting total Akt protein level. The NanogP8-increased gastric cell proliferation was downregulated by Akt inhibition. Our results further showed that increasing NanogP8 expression in human gastric cancer cells promoted cell proliferation by activating the AKT/mTOR pathway and further maintained gastric cell survival. CONCLUSION Our findings extend the knowledge regarding the oncogenic functions and proved that the NanogP8 regulates cell proliferation and migration by Akt/mTOR signaling pathway in human gastric cancer SGC-7901cell line.
Collapse
Affiliation(s)
- Zheng Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, People's Republic of China
| | - Yao Liu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, People's Republic of China
| | - Chuan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, People's Republic of China
| |
Collapse
|
116
|
Gawlik-Rzemieniewska N, Galilejczyk A, Krawczyk M, Bednarek I. Silencing expression of the NANOG gene and changes in migration and metastasis of urinary bladder cancer cells. Arch Med Sci 2016; 12:889-97. [PMID: 27478472 PMCID: PMC4947613 DOI: 10.5114/aoms.2015.55368] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 01/01/2015] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION It has been proved that expression of the NANOG gene is observed not only in embryonic-derived malignancies, but also in breast cancer, ovarian cancer, cervix cancer and bladder cancer. NANOG overexpression is correlated with high activity of MMP-2 and MMP-9. The aim of the study was to evaluate the changes in the malignant phenotype of T24 bladder cancer cells with modulated expression of the NANOG gene. MATERIAL AND METHODS Human urinary bladder cancer cells T24 (HTB-4) were cultivated under standard conditions. Transfection of the cells with silencing constructions was performed with the application of Lipofectamine 2000 (Invitrogen) reagent. Evaluation of changes in the expression level of individual genes was performed using qRTPCR. Changes in the protein level were evaluated using the Human ELISA Kit (Abcam). The invasion capability of transfected cells was tested using Matrigel Invasion Chambers (BD Biosciences). The changes in cell migration were assessed with a wound-healing assay. RESULTS The qRTPCR evaluation showed that silencing the NANOG gene in T24 cells led to the decrease of mRNA for the MMP-2 gene to the level of 62.4% and the MMP-9 gene to the level of 76%. The cells with modulated expression of the NANOG gene migrated slower in the Matrigel invasion assay and in the wound-healing assay. The immunoenzymatic test showed a decrease in the protein level of MMP-9. CONCLUSIONS The transcriptional activity of the NANOG gene might be connected with some aspects of bladder cancer cell metastasis in vitro and has an influence on MMP-2 and MMP-9 expression levels.
Collapse
Affiliation(s)
- Natalia Gawlik-Rzemieniewska
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Biotechnology and Genetic Engineering, Medical University of Silesia, Katowice, Poland
| | - Anna Galilejczyk
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Biotechnology and Genetic Engineering, Medical University of Silesia, Katowice, Poland
| | - Michał Krawczyk
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Biotechnology and Genetic Engineering, Medical University of Silesia, Katowice, Poland
| | - Ilona Bednarek
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Department of Biotechnology and Genetic Engineering, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
117
|
Kozlov AP. Expression of evolutionarily novel genes in tumors. Infect Agent Cancer 2016; 11:34. [PMID: 27437030 PMCID: PMC4949931 DOI: 10.1186/s13027-016-0077-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/18/2016] [Indexed: 01/29/2023] Open
Abstract
The evolutionarily novel genes originated through different molecular mechanisms are expressed in tumors. Sometimes the expression of evolutionarily novel genes in tumors is highly specific. Moreover positive selection of many human tumor-related genes in primate lineage suggests their involvement in the origin of new functions beneficial to organisms. It is suggested to consider the expression of evolutionarily young or novel genes in tumors as a new biological phenomenon, a phenomenon of TSEEN (tumor specifically expressed, evolutionarily novel) genes.
Collapse
Affiliation(s)
- A. P. Kozlov
- The Biomedical Center and Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| |
Collapse
|
118
|
Wang D, Guo Q, Ba H, Li C. Cloning and Characterization of a Nanog Pseudogene in Sika Deer (Cervus nippon). DNA Cell Biol 2016; 35:576-584. [PMID: 27351458 DOI: 10.1089/dna.2016.3303] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Nanog plays a crucial role in the maintenance of stem cell pluripotency. Annual full regeneration of deer antlers has been shown to be a stem cell-based process, and antler stem cells (ASCs) reportedly express Nanog. In the present study, we found that Nanog RNA expressed by ASCs was a pseudogene (Nanog-ps). The coding sequence of Nanog-ps was 93.1% homologous to that of bovine Nanog, but with two missing nucleotides after position 391. Deletion of the two nucleotides in Nanog-ps resulted in a frame-shift mutation, suggesting that Nanog-ps would not encode a normal Nanog protein. Overexpression of Nanog-ps failed to affect downstream genes of Nanog or to enhance cell proliferation in the ASCs. However, this pseudogene was transcribed in the ASCs and encoded a nuclear protein; the expression levels of Nanog-ps were also related to the degree of stemness in antler cells. Here, we reported this pseudogene, because it could serve as a useful marker for identifying ASCs and evaluating the degree of their stemness.
Collapse
Affiliation(s)
- Datao Wang
- 1 Chinese Academy of Agricultural Sciences, Institute of Special Wild Economic Animal and Plant Science , Jilin, China .,2 State Key Laboratory for Molecular Biology of Special Economical Animals , Chinese Academy of Agricultural Sciences, Jilin, China
| | - Qianqian Guo
- 1 Chinese Academy of Agricultural Sciences, Institute of Special Wild Economic Animal and Plant Science , Jilin, China
| | - Hengxing Ba
- 1 Chinese Academy of Agricultural Sciences, Institute of Special Wild Economic Animal and Plant Science , Jilin, China
| | - Chunyi Li
- 1 Chinese Academy of Agricultural Sciences, Institute of Special Wild Economic Animal and Plant Science , Jilin, China .,2 State Key Laboratory for Molecular Biology of Special Economical Animals , Chinese Academy of Agricultural Sciences, Jilin, China
| |
Collapse
|
119
|
Nagata T, Shimada Y, Sekine S, Moriyama M, Hashimoto I, Matsui K, Okumura T, Hori T, Imura J, Tsukada K. KLF4 and NANOG are prognostic biomarkers for triple-negative breast cancer. Breast Cancer 2016; 24:326-335. [PMID: 27300169 DOI: 10.1007/s12282-016-0708-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 05/23/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Prognosis of breast cancer patients has been reported to depend on the expression of induced pluripotent stem (iPS) cell-inducing factors: KLF4 and NANOG. However, the relationship between KLF4 or NANOG expression in each breast cancer subtype and the life prognosis has not been elucidated. METHOD KLF4 and NANOG expression levels were evaluated in 208 patients using a newly developed tissue microarray (TMA). In vitro, siRNA against klf4 (siKLF4) was transfected in TNBC cell line MDA-MB-231, and the expression of KLF4 was inhibited. RESULTS Triple-negative breast cancer (TNBC) patients in KLF4 high-expression (upper) group had more favorable overall survival (OS) and disease-free survival (DFS) rates than KLF4 lower group (p = 0.0453 and p = 0.0427). In contrast, patients in the NANOG upper group had significantly poorer prognosis than lower group in TNBC breast cancer subtypes (p < 0.0001). Multivariate analysis showed that KLF4 (p = 0.0313), NANOG (p = 0.0002), and TNM stage (p = 0.0001) are mutually independent prognostic factors. It was also shown that the proliferation and invasion ability of siKLF4-induced TNBC cells were up-regulated significantly. CONCLUSION Our findings suggested that KLF4 and NANOG expression levels were favorable prognostic factors for TNBC patients. KLF4 also had an ability to inhibit the proliferation and invasion of TNBC.
Collapse
Affiliation(s)
- Takuya Nagata
- Department of Surgery and Science, Graduate school of Medicine and Pharmaceutical Sciences for Research, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| | - Yutaka Shimada
- Department of Nanobio Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Shinichi Sekine
- Department of Surgery and Science, Graduate school of Medicine and Pharmaceutical Sciences for Research, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Makoto Moriyama
- Department of Surgery and Science, Graduate school of Medicine and Pharmaceutical Sciences for Research, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Isaya Hashimoto
- Department of Surgery and Science, Graduate school of Medicine and Pharmaceutical Sciences for Research, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Koshi Matsui
- Department of Surgery and Science, Graduate school of Medicine and Pharmaceutical Sciences for Research, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Tomoyuki Okumura
- Department of Surgery and Science, Graduate school of Medicine and Pharmaceutical Sciences for Research, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Takashi Hori
- Department of Pathology, Graduate School of Research Into Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Johji Imura
- Department of Pathology, Graduate School of Research Into Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Kazuhiro Tsukada
- Department of Surgery and Science, Graduate school of Medicine and Pharmaceutical Sciences for Research, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| |
Collapse
|
120
|
Evidence that high-migration drug-surviving MOLT4 leukemia cells exhibit cancer stem cell-like properties. Int J Oncol 2016; 49:343-51. [PMID: 27210806 DOI: 10.3892/ijo.2016.3526] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 04/28/2016] [Indexed: 11/05/2022] Open
Abstract
Leukemia represents a spectrum of hematological malignancies threatening human health. Resistance to treatments and metastasis of leukemia are the main causes of death in patients. Leukemia stem cells (LSCs) are the initiating cells of leukemia as well as the main source of drug resistance, invasion and metastasis. Consequently, eliminating LSCs is a prerequisite to eradicate leukemia. Preliminary studies in our laboratory have shown that chemokines and their related receptors play an important role in the drug resistance and metastasis of leukemic cells. In this study, we obtained high migration drug-surviving (short term) MOLT4 cells (hMDSCs-MOLT4) with treatment of doxorubicin (DOX) after Transwell assay. Then we detected stem cell-associated molecular markers on hMDSCs-MOLT4 cells and the parental MOLT4 cells by FCM, QPCR, western blotting, H&E staining and immunohisto-chemistry experimental techniques in vitro and in vivo. Moreover, we explored its impact on drug resistance and tumor formation. Then we found that compared with the parental MOLT4 cells, the mRNA expression levels of stem cell-related factors Sox2, Oct4, C-myc, Klf4, Nanog, Bmi-1, CXCR4 are increased in hMDSCs-MOLT4 cells, together with the protein expression levels of Sox2, Oct4, Klf4, Nanog, CXCR4 and CD34. Our results indicated that hMDSCs-MOLT4 cells exhibited strong drug resistance and certain cancer stem cell-like characteristics. It is the first indication that the targeting stemness factors such as Sox2, Oct4, Klf4, Nanog and CXCR4 may represent plausible options for eliminating T-ALL stem-like cells. The present findings shed light on the relationship between drug-tolerant leukemic cells and cancer stem cells.
Collapse
|
121
|
Zhang D, Park D, Lu Y, Shen J, Iyer VR, Tang DG. Deep RNA-Seq analysis reveals unexpected features of human prostate basal epithelial cells. GENOMICS DATA 2016. [DOI: 10.1016/j.gdata.2015.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
122
|
Yao C, Su L, Shan J, Zhu C, Liu L, Liu C, Xu Y, Yang Z, Bian X, Shao J, Li J, Lai M, Shen J, Qian C. IGF/STAT3/NANOG/Slug Signaling Axis Simultaneously Controls Epithelial-Mesenchymal Transition and Stemness Maintenance in Colorectal Cancer. Stem Cells 2016; 34:820-31. [PMID: 26840943 DOI: 10.1002/stem.2320] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 10/29/2015] [Indexed: 12/19/2022]
Abstract
Discovery of epithelial-mesenchymal transition (EMT) and cancer stem cells (CSCs) are two milestones in people exploring the nature of malignant tumor in recent decades. Although some studies have presented the potential connections between them, the link details, underneath their superficial correlation, are largely unknown. In this study, we identified a small subpopulation of NANOG-positive colorectal cancer (CRC) cells, and demonstrated that they exhibited characteristics of CSCs and EMT traits simultaneously. Furthermore, we found that NANOG was a core factor in regulating both of EMT and stemness in CRC cells, NANOG modulate EMT and metastasis by binding to Slug promoter and transcriptionally regulate Slug expression. For the first time, we demonstrated that NANOG was regulated by extracellular IGF signaling pathway via STAT3 phosphorylation in CRC. This coincides with that IGF receptor IGF-1R is often increasing expressed in malignant metastasis colon cancer. Taken together, our data define the crucial functions of IGF/STAT3/NANOG/Slug signaling axis in the progression of CRC by operating EMT and CSCs properties, which make them served as potential therapeutic targets for treatment of CRC.
Collapse
Affiliation(s)
- Chao Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Li Su
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Juanjuan Shan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Chuanlin Zhu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Limei Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Chungang Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yanmin Xu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Zhi Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jimin Shao
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianming Li
- Department of Pathology, Soochow University School of Medicine, Suzhou, China
| | - Maode Lai
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Junjie Shen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Cheng Qian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
123
|
Chen CC, Hsieh TF, Huang CP, Yu AL, Chang WL, Shyr CR. Androgen receptor expands the population of cancer stem cells in upper urinary tract urothelial cell carcinoma cells. Am J Cancer Res 2016; 6:238-248. [PMID: 27186399 PMCID: PMC4859656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 11/27/2015] [Indexed: 06/05/2023] Open
Abstract
Androgen receptor (AR) affects the development and progression of upper urinary tract urothelial cell carcinoma (UUTUC). However, the regulatory mechanism exerted by AR to affect UUTUC cells remains unclear. Here we investigated whether AR promotes UUTUC development and progression, possibly by expanding the population of cancer stem cells (CSCs), which are a particular population of cells within cancer cells responsible for tumor initiation, drug resistance and metastasis. We compared UUTUC cells with or without the addition of AR on their CSC population with flow cytometry, colony formation and sphere formation assay to determine the effect of AR on CSC activity, and real-time PCR was used to detect the expression stemness genes and miRNAs. In vivo tumor formation was evaluated with the implantation of cancer cells in nude mice. We found that the addition of AR in UUTUC cells, significantly increased the population of CSC, clonogenicity, sphere formation and the expression of stemness genes (Oct4, Bmi1 and Nanog), altered CSC-related miRNA profile, as well as promoted epithelial mesenchymal transition (EMT). And AR inhibitor, enzalutamide was shown to suppress AR's effect on tumorsphere formation. Furthermore, in an immune-deficient mouse model, the addition of AR in UUTUC cells also increased the tumor formation capacity. This study will help us better understand the extent to which AR contributes to UUTUC progression by expanding their CSC population and capacity. Our findings could explain high incidence of UUTUC observed in males. And targeting AR may lead to novel therapeutic approaches for genetically diversified urothelial carcinomas in precision medicine era.
Collapse
Affiliation(s)
- Chi-Cheng Chen
- Sex Hormone Research Center, Departments of Urology/Surgery and Medical Laboratory Science and Biotechnology, Graduate Institute of Clinical Medical Science, China Medical University/HospitalTaichung 404, Taiwan
- Department of Urology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical FoundationTaichung 404, Taiwan
| | - Teng-Fu Hsieh
- Sex Hormone Research Center, Departments of Urology/Surgery and Medical Laboratory Science and Biotechnology, Graduate Institute of Clinical Medical Science, China Medical University/HospitalTaichung 404, Taiwan
- Department of Urology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical FoundationTaichung 404, Taiwan
| | - Chi-Ping Huang
- Sex Hormone Research Center, Departments of Urology/Surgery and Medical Laboratory Science and Biotechnology, Graduate Institute of Clinical Medical Science, China Medical University/HospitalTaichung 404, Taiwan
| | - Ai-Lin Yu
- Sex Hormone Research Center, Departments of Urology/Surgery and Medical Laboratory Science and Biotechnology, Graduate Institute of Clinical Medical Science, China Medical University/HospitalTaichung 404, Taiwan
| | - Wen-Lin Chang
- Sex Hormone Research Center, Departments of Urology/Surgery and Medical Laboratory Science and Biotechnology, Graduate Institute of Clinical Medical Science, China Medical University/HospitalTaichung 404, Taiwan
| | - Chih-Rong Shyr
- Sex Hormone Research Center, Departments of Urology/Surgery and Medical Laboratory Science and Biotechnology, Graduate Institute of Clinical Medical Science, China Medical University/HospitalTaichung 404, Taiwan
| |
Collapse
|
124
|
The Androgen Receptor Bridges Stem Cell-Associated Signaling Nodes in Prostate Stem Cells. Stem Cells Int 2016; 2016:4829602. [PMID: 26880966 PMCID: PMC4737002 DOI: 10.1155/2016/4829602] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/04/2015] [Accepted: 12/13/2015] [Indexed: 11/18/2022] Open
Abstract
The therapeutic potential of stem cells relies on dissecting the complex signaling networks that are thought to regulate their pluripotency and self-renewal. Until recently, attention has focused almost exclusively on a small set of "core" transcription factors for maintaining the stem cell state. It is now clear that stem cell regulatory networks are far more complex. In this review, we examine the role of the androgen receptor (AR) in coordinating interactions between signaling nodes that govern the balance of cell fate decisions in prostate stem cells.
Collapse
|
125
|
Huang CK, Luo J, Lee SO, Chang C. Concise review: androgen receptor differential roles in stem/progenitor cells including prostate, embryonic, stromal, and hematopoietic lineages. Stem Cells 2015; 32:2299-308. [PMID: 24740898 DOI: 10.1002/stem.1722] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 03/07/2014] [Accepted: 03/12/2014] [Indexed: 01/07/2023]
Abstract
Stem/progenitor (S/P) cells are special types of cells that have the ability to generate tissues throughout their entire lifetime and play key roles in the developmental process. Androgen and the androgen receptor (AR) signals are the critical determinants in male gender development, suggesting that androgen and AR signals might modulate the behavior of S/P cells. In this review, we summarize the AR effects on the behavior of S/P cells, including self-renewal, proliferation, apoptosis, and differentiation in normal S/P cells, as well as proliferation, invasion, and self-renewal in prostate cancer S/P cells. AR plays a protective role in the oxidative stress-induced apoptosis in embryonic stem cells. AR inhibits the self-renewal of embryonic stem cells, bone marrow stromal cells, and prostate S/P cells, but promotes their differentiation except for adipogenesis. However, AR promotes the proliferation of hematopoietic S/P cells and stimulates hematopoietic lineage differentiation. In prostate cancer S/P cells, AR suppresses their self-renewal, metastasis, and invasion. Together, AR differentially influences the characteristics of normal S/P cells and prostate cancer S/P cells, and targeting AR might improve S/P cell transplantation therapy, especially in embryonic stem cells and bone marrow stromal cells.
Collapse
Affiliation(s)
- Chiung-Kuei Huang
- Departments of Pathology, Urology, Radiation Oncology, the George Whipple Lab for Cancer Research, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA
| | | | | | | |
Collapse
|
126
|
Wong OGW, Cheung ANY. Stem cell transcription factor NANOG in cancers--is eternal youth a curse? Expert Opin Ther Targets 2015; 20:407-17. [PMID: 26634876 DOI: 10.1517/14728222.2016.1112791] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Targeting cancer stem cells can be a more effective approach to treat cancer. NANOG is one of the key factors for maintaining the self-renewal ability and pluripotency of stem cells, including cancer stem cells. Overexpression of NANOG has been observed in various human malignancies. Several reports have suggested that NANOG contributes to carcinogenesis by initiating and preserving cancer stem cells. It is obvious that NANOG is also involved in establishing other hallmarks of cancer such as uncontrolled cell growth, chemoresistance, metastasis, and immune evasion. AREAS COVERED This review will discuss the molecular properties and oncogenic roles of NANOG. The idea of using agents that inhibit the transcription factor to treat cancer is presented. Interfering with NANOG-mediated transcriptions using small interfering RNA, transcription factor decoy, genome editing, and small-molecule inhibitors may provide novel strategies to target cancer stem cells. EXPERT OPINION As a pivotal controller in cancer stem cell maintenance and a positive regulator of various oncogenic pathways, NANOG may be an important target for cancer therapy. However, as a transcription factor, it is inherently difficult to target by pharmacological means. Novel approaches need to be explored before the inhibition of NANOG can be applied in a clinical setting.
Collapse
Affiliation(s)
- Oscar G W Wong
- a Department of Pathology , The University of Hong Kong , Hong Kong Special Administrative Region , People's Republic of China
| | - Annie N Y Cheung
- a Department of Pathology , The University of Hong Kong , Hong Kong Special Administrative Region , People's Republic of China
| |
Collapse
|
127
|
Deng Q, Tang DG. Androgen receptor and prostate cancer stem cells: biological mechanisms and clinical implications. Endocr Relat Cancer 2015; 22:T209-20. [PMID: 26285606 PMCID: PMC4646167 DOI: 10.1530/erc-15-0217] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/18/2015] [Indexed: 12/13/2022]
Abstract
Prostate cancer (PCa) contains phenotypically and functionally distinct cells, and this cellular heterogeneity poses clinical challenges as the distinct cell types likely respond differently to various therapies. Clonal evolution, driven by genetic instability, and intraclonal cancer cell diversification, driven by cancer stem cells (CSCs), together create tumor cell heterogeneity. In this review, we first discuss PCa stem cells (PCSCs) and heterogeneity of androgen receptor (AR) expression in primary, metastatic, and treatment-failed PCa. Based on literature reports and our own studies, we hypothesize that, whereas PCSCs in primary and untreated tumors and models are mainly AR(-), PCSCs in CRPCs could be either AR(+) or AR(-/lo). We illustrate the potential mechanisms AR(+) and AR(-) PCSCs may employ to propagate PCa at the population level, mediate therapy resistance, and metastasize. As a result, targeting AR alone may not achieve long-lasting therapeutic efficacy. Elucidating the roles of AR and PCSCs should provide important clues to designing novel personalized combinatorial therapeutic protocols targeting both AR(+) and AR(-) PCa cells.
Collapse
Affiliation(s)
- Qu Deng
- Department of Epigenetics and Molecular CarcinogenesisUniversity of Texas MD Anderson Cancer Center, Science Park, Park Road 1C, Smithville, Texas 78957, USAProgram in Molecular CarcinogenesisUniversity of Texas Graduate School of Biomedical Sciences, Houston, Texas, USACancer Stem Cell InstituteResearch Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China Department of Epigenetics and Molecular CarcinogenesisUniversity of Texas MD Anderson Cancer Center, Science Park, Park Road 1C, Smithville, Texas 78957, USAProgram in Molecular CarcinogenesisUniversity of Texas Graduate School of Biomedical Sciences, Houston, Texas, USACancer Stem Cell InstituteResearch Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Dean G Tang
- Department of Epigenetics and Molecular CarcinogenesisUniversity of Texas MD Anderson Cancer Center, Science Park, Park Road 1C, Smithville, Texas 78957, USAProgram in Molecular CarcinogenesisUniversity of Texas Graduate School of Biomedical Sciences, Houston, Texas, USACancer Stem Cell InstituteResearch Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China Department of Epigenetics and Molecular CarcinogenesisUniversity of Texas MD Anderson Cancer Center, Science Park, Park Road 1C, Smithville, Texas 78957, USAProgram in Molecular CarcinogenesisUniversity of Texas Graduate School of Biomedical Sciences, Houston, Texas, USACancer Stem Cell InstituteResearch Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China Department of Epigenetics and Molecular CarcinogenesisUniversity of Texas MD Anderson Cancer Center, Science Park, Park Road 1C, Smithville, Texas 78957, USAProgram in Molecular CarcinogenesisUniversity of Texas Graduate School of Biomedical Sciences, Houston, Texas, USACancer Stem Cell InstituteResearch Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| |
Collapse
|
128
|
Sławek S, Szmyt K, Fularz M, Dziudzia J, Boruczkowski M, Sikora J, Kaczmarek M. Pluripotency transcription factors in lung cancer-a review. Tumour Biol 2015; 37:4241-9. [PMID: 26581906 DOI: 10.1007/s13277-015-4407-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/09/2015] [Indexed: 12/28/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide. Diagnosis of lung cancer in an early stage is still a challenge due to the asymptomatic course of early stages of the disease and the lack of a standard screening program for the population. Nowadays, learning about the mechanisms that lead to cancerogenesis in the lung is crucial for the development of new diagnostic and therapeutic strategies. Recently, many studies have proved that cancer stem cells (CSCs) are responsible for the initiation, progression, metastasis, recurrence, and even resistance of chemo- and radiotherapeutic treatment in patients with lung cancer. The expression of pluripotency transcription factors is responsible for stemness properties. In this review, we summarize the current knowledge on the role of CSCs and pluripotency transcription factors in lung carcinogenesis.
Collapse
Affiliation(s)
- Sylwia Sławek
- Department of Immunology, Chair of Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland.
| | - Krzysztof Szmyt
- Department of Immunology, Chair of Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Fularz
- Department of Immunology, Chair of Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Joanna Dziudzia
- Department of Immunology, Chair of Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Boruczkowski
- Department of Immunology, Chair of Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Jan Sikora
- Department of Immunology, Chair of Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Mariusz Kaczmarek
- Department of Immunology, Chair of Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
129
|
Ota I, Masui T, Kurihara M, Yook JI, Mikami S, Kimura T, Shimada K, Konishi N, Yane K, Yamanaka T, Kitahara T. Snail-induced EMT promotes cancer stem cell-like properties in head and neck cancer cells. Oncol Rep 2015; 35:261-6. [PMID: 26498709 DOI: 10.3892/or.2015.4348] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/22/2015] [Indexed: 11/05/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a key process involved in the invasion and metastasis of cancer cells. Furthermore, EMT can induce a cancer stem cell (CSC)-like phenotype in a number of tumor types. We demonstrated that Snail is one of the master regulators that promotes EMT and mediates cancer cell migration and invasion in many types of malignancies including head and neck squamous cell carcinoma (HNSCC). In the present study, we investigated the role of Snail in inducing and maintaining CSC-like properties through EMT in HNSCC. We established HNSCC cell lines transfected with Snail. Stem cell markers were evaluated with real-time RT-PCR and western blot analysis. CSC properties were assessed using sphere formation and WST-8 assays as well as chemosensitivity and chick chorioallantoic membrane in vivo invasion assays. Introduction of Snail induced EMT properties in HNSCC cells. Moreover, Snail-induced EMT maintained the CSC-like phenotype, and enhanced sphere formation capability, chemoresistance and invasive ability. These data suggest that Snail could be one of the critical molecular targets for the development of therapeutic strategies for HNSCC.
Collapse
Affiliation(s)
- Ichiro Ota
- Department of Otolaryngology-Head and Neck Surgery, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Takashi Masui
- Department of Otolaryngology-Head and Neck Surgery, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Miyako Kurihara
- Department of Molecular Pathology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Jong-In Yook
- Department of Oral Pathology, Oral Cancer Research Institute, College of Dentistry, Yonsei University, Seoul 120-752, Republic of Korea
| | - Shinji Mikami
- Department of Otolaryngology-Head and Neck Surgery, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Takahiro Kimura
- Department of Otolaryngology-Head and Neck Surgery, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Keiji Shimada
- Department of Pathology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Noboru Konishi
- Department of Pathology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Katsunari Yane
- Department of Otolaryngology, Kinki University School of Medicine, Nara Hospital, Ikoma, Nara 630-0293, Japan
| | - Toshiaki Yamanaka
- Department of Otolaryngology-Head and Neck Surgery, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Tadashi Kitahara
- Department of Otolaryngology-Head and Neck Surgery, Nara Medical University, Kashihara, Nara 634-8522, Japan
| |
Collapse
|
130
|
Talebi A, Kianersi K, Beiraghdar M. Comparison of gene expression of SOX2 and OCT4 in normal tissue, polyps, and colon adenocarcinoma using immunohistochemical staining. Adv Biomed Res 2015; 4:234. [PMID: 26645019 PMCID: PMC4647122 DOI: 10.4103/2277-9175.167958] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 07/29/2015] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Cancer stem cells have been isolated and characterized in all common cancers. SOX2 and OCT4 are important genes to enhance the self-renewal ability as activate stem cells and inhibit the genes that start differentiation and thus maintain the self-renewal ability of stem cells. Also, the aim of this study is "Comparison of gene expression of SOX2 and OCT4 in normal tissue, polyps, and colon adenocarcinoma using immunohistochemical staining." MATERIALS AND METHODS This cross-sectional study conducted on 20 patients so that for each patient, a sample of healthy tissue, dysplastic polyp tissue, and colon adenocarcinoma were provided as microscopic sections and staining on each tissue was performed through immunohistochemistry method by markers OCT4 and SOX2. The collected data were interred into SPSS version 18.0, (SPSS Inc., Chicago, IL, USA) software and the level of significance were considered as <0.05. RESULTS The study sample consisted of 20 patients including 11 men (55%) and 9 women (45%) with a mean age of 55.6 ± 9.88 years. There was no association between Oct4 and colorectal cancer (CRC) patients (P > 0.05), but there was a significant correlation between Sox2 expression and CRC (P < 0.05). Patients in many aspects such as race, type of polyp, presence of lymph node, grade and intensity of Sox2 in different types of patients' tissues (P < 0.05). CONCLUSION Regarding our findings, the expression of Sox2 would be a liable marker for evaluating of cancer progression and could be a treatment target of CRC cells.
Collapse
Affiliation(s)
- Ardeshir Talebi
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Kianoosh Kianersi
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mozhdeh Beiraghdar
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
131
|
Habibi L, Pedram M, AmirPhirozy A, Bonyadi K. Mobile DNA Elements: The Seeds of Organic Complexity on Earth. DNA Cell Biol 2015. [DOI: 10.1089/dna.2015.2938] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Laleh Habibi
- Department of Pharmaceutics, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Nutrition Department, School of Nutritional Science and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Pedram
- Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Akbar AmirPhirozy
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Khadijeh Bonyadi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
132
|
Affiliation(s)
- Mireia Mato Prado
- Division of Cancer, Department of Surgery & Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College, Hammersmith Hospital Campus, London, UK
| | - Adam E Frampton
- Division of Cancer, Department of Surgery & Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College, Hammersmith Hospital Campus, London, UK HPB Surgical Unit, Department of Surgery & Cancer, Imperial College, Hammersmith Hospital Campus, London, UK
| | - Justin Stebbing
- Division of Cancer, Department of Surgery & Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College, Hammersmith Hospital Campus, London, UK
| | - Jonathan Krell
- Division of Cancer, Department of Surgery & Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College, Hammersmith Hospital Campus, London, UK
| |
Collapse
|
133
|
An H, Kim JY, Lee N, Cho Y, Oh E, Seo JH. Salinomycin possesses anti-tumor activity and inhibits breast cancer stem-like cells via an apoptosis-independent pathway. Biochem Biophys Res Commun 2015; 466:696-703. [PMID: 26407842 DOI: 10.1016/j.bbrc.2015.09.108] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 09/19/2015] [Indexed: 01/16/2023]
Abstract
Cancer stem cells (CSCs) play important roles in the formation, growth and recurrence of tumors, particularly following therapeutic intervention. Salinomycin has received recent attention for its ability to target breast cancer stem cells (BCSCs), but the mechanisms of action involved are not fully understood. In the present study, we sought to investigate the mechanisms responsible for salinomycin's selective targeting of BCSCs and its anti-tumor activity. Salinomycin suppressed cell viability, concomitant with the downregulation of cyclin D1 and increased p27(kip1) nuclear accumulation. Mammosphere formation assays revealed that salinomycin suppresses self-renewal of ALDH1-positive BCSCs and downregulates the transcription factors Nanog, Oct4 and Sox2. TUNEL analysis of MDA-MB-231-derived xenografts revealed that salinomycin administration elicited a significant reduction in tumor growth with a marked downregulation of ALDH1 and CD44 levels, but seemingly without the induction of apoptosis. Our findings shed further light on the mechanisms responsible for salinomycin's effects on BCSCs.
Collapse
Affiliation(s)
- Hyunsook An
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 152-703, Republic of Korea; Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul 152-703, Republic of Korea
| | - Ji Young Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 152-703, Republic of Korea; Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul 152-703, Republic of Korea
| | - Nahyun Lee
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 152-703, Republic of Korea; Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul 152-703, Republic of Korea
| | - Youngkwan Cho
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 152-703, Republic of Korea; Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul 152-703, Republic of Korea
| | - Eunhye Oh
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 152-703, Republic of Korea; Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul 152-703, Republic of Korea
| | - Jae Hong Seo
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul 152-703, Republic of Korea; Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul 152-703, Republic of Korea.
| |
Collapse
|
134
|
Voutsadakis IA. The network of pluripotency, epithelial-mesenchymal transition, and prognosis of breast cancer. BREAST CANCER-TARGETS AND THERAPY 2015; 7:303-19. [PMID: 26379447 PMCID: PMC4567227 DOI: 10.2147/bctt.s71163] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Breast cancer is the leading female cancer in terms of prevalence. Progress in molecular biology has brought forward a better understanding of its pathogenesis that has led to better prognostication and treatment. Subtypes of breast cancer have been identified at the genomic level and guide therapeutic decisions based on their biology and the expected benefit from various interventions. Despite this progress, a significant percentage of patients die from their disease and further improvements are needed. The cancer stem cell theory and the epithelial-mesenchymal transition are two comparatively novel concepts that have been introduced in the area of cancer research and are actively investigated. Both processes have their physiologic roots in normal development and common mediators have begun to surface. This review discusses the associations of these networks as a prognostic framework in breast cancer.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Division of Medical Oncology, Department of Internal Medicine, Sault Area Hospital, Sault Ste Marie, ON, Canada ; Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada
| |
Collapse
|
135
|
Stoyanova E, Mourdjeva M, Kyurkchiev S. Early selection of human fibroblast-derived induced pluripotent stem cells. BIOTECHNOL BIOTEC EQ 2015. [DOI: 10.1080/13102818.2015.1052015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
136
|
Xu C, Xia C, Zhang SJ, Qin H, Li DM, Zhao Q. Effect of salinomycin on growth and migration of pancreatic cancer cells in vitro. Shijie Huaren Xiaohua Zazhi 2015; 23:3831-3837. [DOI: 10.11569/wcjd.v23.i24.3831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of salinomycin on proliferation and migration of pancreatic cancer cell lines Capan-2 and PANC-1 and the possible mechanisms of inhibition of cell migration.
METHODS: Canpan-2 and PANC-1 cells were treated with different concentrations of salinomycin for 24 h, while the cells treated with dimethyl sulphoxide (DMSO) were used as controls. Cell proliferation and the half maximal inhibitory concentration (IC50) of salinomycin were measured by CCK8 assay. Transwell assay was used to measure the migration of these cells which were pretreated with salinomycin at a concentration of IC50. The protein and mRNA expression of biomarkers for epithelial-mesenchymal transition (EMT) was detected by Western blot and Real-time PCR, respectively.
RESULTS: Salinomycin inhibited the proliferation of Capan-2 and PANC-1 cells in a dose-dependent manner. The IC50 values for Capan-2 and PANC-1 cells were 20 μmol/L and 10 μmol/L, respectively. Salinomycin significantly decreased the migration ability of pancreatic cancer cells as revealed by Transwell migration assay (P < 0.05). The protein expression of E-cadherin was increased while that of Vimentin was decreased after salinomycin treatment. The expression of E-cadherin mRNA was increased in both Capan-2 and PANC-1 cells (P < 0.05), while the expression of Vimentin mRNA was decreased in both Capan-2 and PANC-1 cells (P < 0.05).
CONCLUSION: Salinomycin can inhibit the proliferation and migration of Capan-2 and PANC-1 cells possibly by inhibiting the process of EMT.
Collapse
|
137
|
Zhang W, Meng Y, Liu N, Wen XF, Yang T. Insights into Chemoresistance of Prostate Cancer. Int J Biol Sci 2015; 11:1160-70. [PMID: 26327810 PMCID: PMC4551752 DOI: 10.7150/ijbs.11439] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 06/29/2015] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer (PCa) remains the most prevalent malignancy among males in the western world. Though hormonal therapies through chemical or surgical castration have been proposed many years ago, heretofore, such mainstay for the treatment on advanced PCa has not fundamentally changed. These therapeutic responses are temporary and most cases will eventually undergo PCa recurrence and metastasis, or even progress to castration-resistant prostate cancer (CRPC) due to persistent development of drug resistance. Prostate cancer stem cells (PCSCs) are a small population of cells, which possess unlimited self-renewal capacities, and can regenerate tumorigenic progenies, and play an essential role in PCa therapy resistance, metastasis and recurrence. Nowadays advanced progresses have been made in understanding of PCSC properties, roles of androgen receptor signaling and ATP-binding cassette sub-family G member 2 (ABCG2), as well as roles of genomic non-coding microRNAs and key signaling pathways, which have led to the development of novel therapies which are active against chemoresistant PCa and CRPC. Based on these progresses, this review is dedicated to address mechanisms underlying PCa chemoresistance, unveil crosstalks among pivotal signaling pathways, explore novel biotherapeutic agents, and elaborate functional properties and specific roles of chemoresistant PCSCs, which may act as a promising target for novel therapies against chemoresistant PCa.
Collapse
Affiliation(s)
- Wei Zhang
- 1. Department of Pharmacology, School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yan Meng
- 2. Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Na Liu
- 3. Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xiao-Fei Wen
- 4. Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Tao Yang
- 2. Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| |
Collapse
|
138
|
Li J, Lam M. Registered report: the microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. eLife 2015; 4:e06434. [PMID: 26231042 PMCID: PMC4521141 DOI: 10.7554/elife.06434] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 07/10/2015] [Indexed: 01/11/2023] Open
Abstract
The Reproducibility Project: Cancer Biology seeks to address growing concerns about reproducibility in scientific research by conducting replications of selected experiments from a number of high-profile papers in the field of cancer biology. The papers, which were published between 2010 and 2012, were selected on the basis of citations and Altimetric scores (Errington et al., 2014). This Registered report describes the proposed replication plan of key experiments from ‘The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44’ by Liu and colleagues published in Nature Medicine in 2011 (Liu et al., 2011). Liu and colleagues first demonstrated that miR-34a levels were reduced in CD44+ prostate cancer cells (Figure 1B). They then showed that xenograft tumors from cells expressing exogenous miR-34a were smaller in size than control tumors (Supplemental Figure 5C). Tumors with exogenous miR-34a showed reduced levels of CD44 expression (Figure 4A), and mutation of two putative miR-34a binding sites in the CD33 3′ UTR partially abrogated signal repression in a luciferase assay (Figure 4D). The Reproducibility Project: Cancer Biology is a collaboration between the Center for Open Science and Science Exchange, and the results of the replications will be published by eLife. DOI:http://dx.doi.org/10.7554/eLife.06434.001
Collapse
Affiliation(s)
- Jia Li
- Crown Bioscience, Santa Clara, California, United States
| | - Matthew Lam
- Breakthrough Breast Cancer, London, United Kingdom
| | | |
Collapse
|
139
|
Ding Y, Yu AQ, Li CL, Fang J, Zeng Y, Li DS. TALEN-mediated Nanog disruption results in less invasiveness, more chemosensitivity and reversal of EMT in Hela cells. Oncotarget 2015; 5:8393-401. [PMID: 25245189 PMCID: PMC4226691 DOI: 10.18632/oncotarget.2298] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Emerging evidence suggests that Nanog is involved in cervical tumorigenesis. However, the regulating role of Nanog in tumorigenesis and chemosensitivity are still poorly understood. In this study, Nanog was disrupted by transcription activatorlike effector nucleases (TALEN) in Hela cells and its expression was significantly decreased in a single-cell derived sub-clone with biallelic mutations. The disruption of Nanog not only induced down regulation of some other core transcription factor genes for cell self-renewal, such as Oct4, Sox2 and FoxD3, but also led to the down regulation of some mesenchymal representative genes, vimentin and N-adherin, and up regulation of the epithelial gene, E-cadherin. In addition, the invasiveness and clonogenicity of the Hela cells were obviously affected, and surprisingly their sensitivities to anti-cancer drugs were also significantly increased in vitro. After Xenograft into nude mice, the growth volumes of the neoplasms from the Nanog disrupted Hela cells were significantly smaller compared with those from wild type ones. In conclusion, these results suggest that disruption of Nanog may reverse the status of epithelial-mesenchymal transition, which is critical in tumorigenesis, and alleviate chemoresistance, as well as their invasiveness, in cervical cancer cells.
Collapse
Affiliation(s)
- Yan Ding
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China. College of Life Science and Bioengineering, Beijing University of Industry, Beijing, China
| | - Ai Qing Yu
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Cheng Lin Li
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Juan Fang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yi Zeng
- College of Life Science and Bioengineering, Beijing University of Industry, Beijing, China
| | - Dong Sheng Li
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
140
|
|
141
|
Zhang M, Cui F, Lu S, Lu H, Xue Y, Wang J, Chen J, Zhao S, Ma S, Zhang Y, Yu Y, Peng Z, Tang H. Developmental pluripotency-associated 4: a novel predictor for prognosis and a potential therapeutic target for colon cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:60. [PMID: 26063247 PMCID: PMC4466839 DOI: 10.1186/s13046-015-0176-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 05/27/2015] [Indexed: 12/12/2022]
Abstract
Backgrounds Developmental pluripotency-associated 4 (Dppa4) gene plays an important role in self-renewal and pluripotency sustainability in embryonic stem cells. It is re-expressed in several malignant tumors and is identified as a new pluripotency-related oncogene. The present study investigates the expression and clinical significance of Dppa4 in colon cancer. Methods Real-time polymerase chain reaction and Western blotting were used to evaluate Dppa4 mRNA and protein expression in 39 pairs of fresh-frozzen colon cancer samples, which were compared with adjacent normal mucosa. The Dppa4 protein was evaluated by immunohistochemical techniques using colon tissue microarrays (TMA). The sample included 185 cancer specimens and corresponding normal colorectal mucosa. The effect of Dppa4 knockdown on colorectal cancer cell proliferation was investigated using Cell Counting Kit-8 (CCK8) assays and colony-formation assays. Results Both the mRNA and protein level expression of Dppa4 gene was found to be upregulated in colon cancer tissues. Furthermore, the upregulated expression of Dppa4 was significantly correlated with the results of American Joint Committee on Cancer (AJCC) stage (P = 0.01), invasion depth (P = 0.028), nodal involvement (P = 0.012), distant metastasis (P = 0.003), and differentiation (P = 0.002). Dppa4 was also shown to be an independent prognostic indicator of disease-free survival (HR 6.118, 95 % CI 3.004–12.462) and overall survival (HR 6.348, 95 % CI 2.875–14.014) for patients with colon cancer. Knockdown of Dppa4 expression inhibited the proliferation of colorectal cancer cell lines through G1/S transition regulation. Conclusion The results indicate that Dppa4 might play an important role in colon cancer progression and function as a novel prognostic indicator and a potential therapeutic target.
Collapse
Affiliation(s)
- Meng Zhang
- Departments of Pathology, Shanghai Jiaotong University Affiiated First People's Hospital, Shanghai, 200080, People's Republic of China.
| | - Feifei Cui
- Departments of General Surgery, Shanghai Jiaotong University Affiiated First People's Hospital, Shanghai, 200080, People's Republic of China.
| | - Su Lu
- Departments of Pathology, Shanghai Jiaotong University Affiiated First People's Hospital, Shanghai, 200080, People's Republic of China.
| | - Huijun Lu
- Departments of Pathology, Shanghai Jiaotong University Affiiated First People's Hospital, Shanghai, 200080, People's Republic of China.
| | - Yingming Xue
- Departments of General Surgery, Shanghai Jiaotong University Affiiated First People's Hospital, Shanghai, 200080, People's Republic of China.
| | - Jingtao Wang
- Departments of General Surgery, Shanghai Jiaotong University Affiiated First People's Hospital, Shanghai, 200080, People's Republic of China.
| | - Jian Chen
- Departments of General Surgery, Shanghai Jiaotong University Affiiated First People's Hospital, Shanghai, 200080, People's Republic of China.
| | - Senlin Zhao
- Departments of General Surgery, Shanghai Jiaotong University Affiiated First People's Hospital, Shanghai, 200080, People's Republic of China.
| | - Shaofei Ma
- Departments of Pathology, Shanghai Jiaotong University Affiiated First People's Hospital, Shanghai, 200080, People's Republic of China.
| | - Yu Zhang
- Departments of Pathology, Shanghai Jiaotong University Affiiated First People's Hospital, Shanghai, 200080, People's Republic of China.
| | - Yang Yu
- Departments of General Surgery, Shanghai Jiaotong University Affiiated First People's Hospital, Shanghai, 200080, People's Republic of China.
| | - Zhihai Peng
- Departments of General Surgery, Shanghai Jiaotong University Affiiated First People's Hospital, Shanghai, 200080, People's Republic of China.
| | - Huamei Tang
- Departments of Pathology, Shanghai Jiaotong University Affiiated First People's Hospital, Shanghai, 200080, People's Republic of China.
| |
Collapse
|
142
|
Arif K, Hussain I, Rea C, El-Sheemy M. The role of Nanog expression in tamoxifen-resistant breast cancer cells. Onco Targets Ther 2015; 8:1327-34. [PMID: 26082649 PMCID: PMC4461083 DOI: 10.2147/ott.s67835] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
There is an accumulation of evidence that shows a significant role of cancer stem cells in tumor initiation, proliferation, relapse, and metastasis. Nanog is the most important core transcription marker of stem cells, known by its role in maintaining pluripotency, proliferation, and differentiation. Therefore, this study aimed to examine the role of Nanog in breast cancer cell tamoxifen resistance and its implications in breast cancer treatment. In this study, the expression of the three core transcription markers Nanog, Oct3/4, and Sox2 were quantitatively evaluated using flow cytometry. Then, small interfering RNA (siRNA) against human Nanog was transfected into tamoxifen-resistant breast cancer cells via Lipofectamine 2000. Nanog gene expression in the cells was detected using reverse transcription polymerase chain reaction (RT-PCR). The change in cell proliferation was evaluated using the tetrazolium bromide method. An enzyme-linked immunosorbent assay was used to detect apoptosis of the transfected cells alone and in combination with 4-hydroxytamoxifen. The results showed a high level expression of Nanog, Oct3/4, and Sox2 in MDA-MB-231 and MCF7/tamoxifen resistant cells compared with MCF7/wild-type. siRNA-mediated Nanog gene silencing can efficiently inhibit cell proliferation and induce apoptosis of tamoxifen-resistant breast cancer cells. This study provides a basis for further study of the role of Nanog in developing resistance to tamoxifen, its implication in breast cancer management, and as a new strategy to enhance response to endocrine therapy.
Collapse
Affiliation(s)
- Khalid Arif
- School of Life Sciences, University of Lincoln, Brayford Pool, UK
| | - Issam Hussain
- School of Life Sciences, University of Lincoln, Brayford Pool, UK
| | - Carol Rea
- School of Life Sciences, University of Lincoln, Brayford Pool, UK
| | | |
Collapse
|
143
|
Singovski G, Bernal C, Kuciak M, Siegl-Cachedenier I, Conod A, Ruiz i Altaba A. In vivo epigenetic reprogramming of primary human colon cancer cells enhances metastases. J Mol Cell Biol 2015; 8:157-73. [PMID: 26031752 PMCID: PMC4816146 DOI: 10.1093/jmcb/mjv034] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 04/27/2015] [Indexed: 01/06/2023] Open
Abstract
How metastases develop is not well understood and no genetic mutations have been reported as specific metastatic drivers. Here we have addressed the idea that epigenetic reprogramming by GLI-regulated pluripotent stemness factors promotes metastases. Using primary human colon cancer cells engrafted in mice, we find that transient expression of OCT4, SOX2, KLF4 +/− cMYC establishes an enhanced pro-metastatic state in the primary tumor that is stable through sequential engraftments and is transmitted through clonogenic cancer stem cells. Metastatic reprogramming alters NANOG methylation and stably boosts NANOG and NANOGP8 expression. Metastases and reprogrammed EMT-like phenotypes require endogenous NANOG, but enhanced NANOG is not sufficient to induce these phenotypes. Finally, reprogrammed tumors enhance GLI2, and we show that GLI2high and AXIN2low, which are markers of the metastatic transition of colon cancers, are prognostic of poor disease outcome in patients. We propose that metastases arise through epigenetic reprogramming of cancer stem cells within primary tumors.
Collapse
Affiliation(s)
- Grigori Singovski
- Department of Genetic Medicine and Development, University of Geneva Medical School, CH-1211 Geneva, Switzerland
| | - Carolina Bernal
- Department of Genetic Medicine and Development, University of Geneva Medical School, CH-1211 Geneva, Switzerland
| | - Monika Kuciak
- Department of Genetic Medicine and Development, University of Geneva Medical School, CH-1211 Geneva, Switzerland
| | - Irene Siegl-Cachedenier
- Department of Genetic Medicine and Development, University of Geneva Medical School, CH-1211 Geneva, Switzerland
| | - Arwen Conod
- Department of Genetic Medicine and Development, University of Geneva Medical School, CH-1211 Geneva, Switzerland
| | - Ariel Ruiz i Altaba
- Department of Genetic Medicine and Development, University of Geneva Medical School, CH-1211 Geneva, Switzerland
| |
Collapse
|
144
|
Gong S, Li Q, Jeter CR, Fan Q, Tang DG, Liu B. Regulation of NANOG in cancer cells. Mol Carcinog 2015; 54:679-87. [PMID: 26013997 DOI: 10.1002/mc.22340] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/19/2015] [Accepted: 05/01/2015] [Indexed: 12/14/2022]
Abstract
As one of the key pluripotency transcription factors, NANOG plays a critical role in maintaining the self-renewal and pluripotency in normal embryonic stem cells. Recent data indicate that NANOG is expressed in a variety of cancers and its expression correlates with poor survival in cancer patients. Of interest, many studies suggest that NANOG enhances the defined characteristics of cancer stem cells and may thus function as an oncogene to promote carcinogenesis. Therefore, NANOG expression determines the cell fate not only in pluripotent cells but also in cancer cells. Although the regulation of NANOG in normal embryonic stem cells is reasonably well understood, the regulation of NANOG in cancer cells has only emerged recently. The current review provides a most updated summary on how NANOG expression is regulated during tumor development and progression.
Collapse
Affiliation(s)
- Shuai Gong
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, city, Smithville, Texas.,The First Affiliated Hospital of Zhengzhou University, city, Henan, China
| | - Qiuhui Li
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, city, Smithville, Texas
| | - Collene R Jeter
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, city, Smithville, Texas
| | - Qingxia Fan
- The First Affiliated Hospital of Zhengzhou University, city, Henan, China
| | - Dean G Tang
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, city, Smithville, Texas.,Cancer Stem Cell Institute, Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bigang Liu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D Anderson Cancer Center, city, Smithville, Texas
| |
Collapse
|
145
|
Thiagarajan PS, Hitomi M, Hale JS, Alvarado AG, Otvos B, Sinyuk M, Stoltz K, Wiechert A, Mulkearns-Hubert E, Jarrar A, Zheng Q, Thomas D, Egelhoff T, Rich JN, Liu H, Lathia JD, Reizes O. Development of a Fluorescent Reporter System to Delineate Cancer Stem Cells in Triple-Negative Breast Cancer. Stem Cells 2015; 33:2114-2125. [PMID: 25827713 DOI: 10.1002/stem.2021] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 02/17/2015] [Accepted: 02/28/2015] [Indexed: 02/06/2023]
Abstract
Advanced cancers display cellular heterogeneity driven by self-renewing, tumorigenic cancer stem cells (CSCs). The use of cell lines to model CSCs is challenging due to the difficulty of identifying and isolating cell populations that possess differences in self-renewal and tumor initiation. To overcome these barriers in triple-negative breast cancer (TNBC), we developed a CSC system using a green fluorescent protein (GFP) reporter for the promoter of the well-established pluripotency gene NANOG. NANOG-GFP+ cells gave rise to both GFP+ and GFP(-) cells, and GFP+ cells possessed increased levels of the embryonic stem cell transcription factors NANOG, SOX2, and OCT4 and elevated self-renewal and tumor initiation capacities. GFP+ cells also expressed mesenchymal markers and demonstrated increased invasion. Compared with the well-established CSC markers CD24(-) /CD44(+) , CD49f, and aldehyde dehydrogenase (ALDH) activity, our NANOG-GFP reporter system demonstrated increased enrichment for CSCs. To explore the utility of this system as a screening platform, we performed a flow cytometry screen that confirmed increased CSC marker expression in the GFP+ population and identified new cell surface markers elevated in TNBC CSCs, including junctional adhesion molecule-A (JAM-A). JAM-A was highly expressed in GFP+ cells and patient-derived xenograft ALDH+ CSCs compared with the GFP(-) and ALDH(-) cells, respectively. Depletion of JAM-A compromised self-renewal, whereas JAM-A overexpression induced self-renewal in GFP(-) cells. Our data indicate that we have defined and developed a robust system to monitor differences between CSCs and non-CSCs in TNBC that can be used to identify CSC-specific targets for the development of future therapeutic strategies.
Collapse
Affiliation(s)
- Praveena S Thiagarajan
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44915, USA
| | - Masahiro Hitomi
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44915, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| | - James S Hale
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44915, USA
| | - Alvaro G Alvarado
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44915, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| | - Balint Otvos
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44915, USA
| | - Maksim Sinyuk
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44915, USA
| | - Kevin Stoltz
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44915, USA
| | - Andrew Wiechert
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44915, USA
| | - Erin Mulkearns-Hubert
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44915, USA
| | - Awad Jarrar
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44915, USA
| | - Qiao Zheng
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44915, USA
| | - Dustin Thomas
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44915, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| | - Thomas Egelhoff
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44915, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA.,Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Jeremy N Rich
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA.,Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, OH 44195, USA.,Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Huiping Liu
- Case Comprehensive Cancer Center, Cleveland, OH 44106, USA.,Department of Pathology and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, United States
| | - Justin D Lathia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44915, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA.,Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Ofer Reizes
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44915, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA.,Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| |
Collapse
|
146
|
Jeter CR, Yang T, Wang J, Chao HP, Tang DG. Concise Review: NANOG in Cancer Stem Cells and Tumor Development: An Update and Outstanding Questions. Stem Cells 2015; 33:2381-90. [PMID: 25821200 DOI: 10.1002/stem.2007] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/08/2015] [Indexed: 12/22/2022]
Abstract
The homeobox domain transcription factor NANOG, a key regulator of embryonic development and cellular reprogramming, has been reported to be broadly expressed in human cancers. Functional studies have provided strong evidence that NANOG possesses protumorigenic attributes. In addition to promoting self-renewal and long-term proliferative potential of stem-like cancer cells, NANOG-mediated oncogenic reprogramming may underlie clinical manifestations of malignant disease. In this review, we examine the molecular origin, expression, biological activities, and mechanisms of action of NANOG in various malignancies. We also consider clinical implications such as correlations between NANOG expression and cancer prognosis and/or response to therapy. We surmise that NANOG potentiates the molecular circuitry of tumorigenesis, and thus may represent a novel therapeutic target or biomarker for the diagnosis, prognosis, and treatment outcome of cancer. Finally, we present critical pending questions relating NANOG to cancer stem cells and tumor development.
Collapse
Affiliation(s)
- Collene R Jeter
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, Texas, USA
| | - Tao Yang
- Cancer Stem Cell Institute, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Junchen Wang
- Cancer Stem Cell Institute, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Hsueh-Ping Chao
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, Texas, USA
| | - Dean G Tang
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, Texas, USA.,Cancer Stem Cell Institute, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
147
|
Cancer stem cells and tumor-associated macrophages: a roadmap for multitargeting strategies. Oncogene 2015; 35:671-82. [PMID: 25961921 DOI: 10.1038/onc.2015.132] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 03/16/2015] [Accepted: 03/20/2015] [Indexed: 12/12/2022]
Abstract
The idea that tumor initiation and progression are driven by a subset of cells endowed with stem-like properties was first described by Rudolf Virchow in 1855. 'Cancer stem cells', as they were termed more than a century later, represent a subset of tumor cells that are able to generate all tumorigenic and nontumorigenic cell types within the malignancy. Although their existence was hypothesized >150 years ago, it was only recently that stem-like cells started to be isolated from different neoplastic malignancies. Interestingly, Virchow, in suggesting a correlation between cancer and the inflammatory microenvironment, also paved the way for the 'Seed and Soil' theory proposed by Paget a few years later. Despite the time that has passed since these two important concepts were suggested, the relationships between Virchow's 'stem-like cells' and Paget's 'soil' are far from being fully understood. One emerging topic is the importance of a stem-like niche in modulating the biological properties of stem-like cancer cells and thus in affecting the response of the tumor to drugs. This review aims to summarize the recent molecular data concerning the multilayered relationship between cancer stem cells and tumor-associated macrophages that form a key component of the tumor microenvironment. We also discuss the therapeutic implications of targeting this synergistic interplay.
Collapse
|
148
|
Yang T, Rycaj K. Targeted therapy against cancer stem cells. Oncol Lett 2015; 10:27-33. [PMID: 26170972 DOI: 10.3892/ol.2015.3172] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 04/21/2015] [Indexed: 01/05/2023] Open
Abstract
Research into cancer stem cells (CSCs), which have the ability to self-renew and give rise to more mature (differentiated) cancer cells, and which may be the cells responsible for the overall organization of a tumor, has progressed rapidly and concomitantly with recent advances in studies of normal tissue stem cells. CSCs have been reported in a wide spectrum of human tumors. Like normal tissue stem cells, CSCs similarly exhibit significant phenotypic and functional heterogeneity. The ability of CSCs to self-renew results in the immortality of malignant cells at the population level, whereas the ability of CSCs to differentiate, either fully or partially, generates the cellular hierarchy and heterogeneity commonly observed in solid tumors. CSCs also appear to have maximized their pro-survival mechanisms leading to their relative resistance to anti-cancer therapies and subsequent relapse. Studies in animal models of human cancers have also provided insight into the heterogeneity and characteristics of CSCs, helping to establish a platform for the development of novel targeted therapies against specific CSCs. In the present study, we briefly review the most recent progress in dissecting CSC heterogeneity and targeting CSCs in various human tumor systems. We also highlight a few examples of CSC-targeted drug development and clinical trials, with the ultimate aim of developing more effective therapeutic regimens that are capable of preventing tumor recurrence and metastasis.
Collapse
Affiliation(s)
- Tao Yang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Kiera Rycaj
- Department of Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA
| |
Collapse
|
149
|
Yang F, Zhang J, Liu Y, Cheng D, Wang H. Structure and functional evaluation of porcine NANOG that is a single-exon gene and has two pseudogenes. Int J Biochem Cell Biol 2015; 59:142-52. [DOI: 10.1016/j.biocel.2014.12.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 12/11/2014] [Accepted: 12/15/2014] [Indexed: 12/25/2022]
|
150
|
Deng XB, Xiao L, Wu Y, Jin F, Mossman B, Testa JR, Xiao GH. Inhibition of mesothelioma cancer stem-like cells with adenovirus-mediated NK4 gene therapy. Int J Cancer 2014; 137:481-90. [PMID: 25501304 DOI: 10.1002/ijc.29391] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 11/24/2014] [Indexed: 02/05/2023]
Abstract
Malignant mesothelioma (MM) is a highly invasive and chemoresistant malignancy induced by asbestos fibers. NK4, a hepatocyte growth factor antagonist and angiogenesis inhibitor, consists of the N-terminal hairpin domain and four kringle domains of the α-chain of hepatocyte growth factor. The therapeutic potential of NK4 has been demonstrated in a variety of tumor types. However, the mechanisms by which NK4 inhibits tumor growth have not been well delineated. In this study, it is shown that the NK4 adenovirus (Ad-NK4) potently inhibits cell viability, invasiveness and tumorigenicity of human MM cells. Significantly, this study demonstrates for the first time that Ad-NK4 inhibits cancer stem-like cell (CSC) properties as assessed by spheroid formation assay, side population analysis and flow cytometric sorting of CD24 cells. In addition to inhibiting phosphorylation of Met and AKT, Ad-NK4 markedly suppressed the active form of β-catenin, a key mediator of both Wnt and AKT pathways. It is further demonstrated that expression of NK4 suppresses β-catenin nuclear localization and transcriptional activity. Intriguingly, the expression levels of Oct4 and Myc, two critical stem cell factors and downstream targets of β-catenin, were also diminished by Ad-NK4. Furthermore, the strong antitumor effect of NK4 was found to be linked to its ability to inhibit CSCs as revealed by immunohistochemical examination of tumor specimens from a mouse xenograft model of human MM. These findings suggest that NK4 acts as a CSC inhibitor by impeding Met/AKT/β-catenin signaling and holds promise for achieving durable therapeutic responses in MM by constraining the CSC component of these aggressive tumors.
Collapse
Affiliation(s)
- Xu-Bin Deng
- Cancer Institute, Southern Medical University, Guangzhou, China
| | - Li Xiao
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Yue Wu
- Cancer Institute, Southern Medical University, Guangzhou, China
| | - Fang Jin
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Brooke Mossman
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT
| | - Joseph R Testa
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Guang-Hui Xiao
- Cancer Institute, Southern Medical University, Guangzhou, China.,Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA
| |
Collapse
|