2,485 results on '"Tumor initiating cells"'
Search Results
2. Promotion of gastric tumor initiating cells in a 3D collagen gel culture model via YBX1/SPP1/NF-κB signaling
- Author
-
Deng, Shuangya, Li, Lun, Xu, Shu, Wang, Xiaobo, and Han, Tong
- Published
- 2021
- Full Text
- View/download PDF
3. Antagonists of the serotonin receptor 5A target human breast tumor initiating cells
- Author
-
Gwynne, William D., Shakeel, Mirza S., Girgis-Gabardo, Adele, Kim, Kwang H., Ford, Emily, Dvorkin-Gheva, Anna, Aarts, Craig, Isaac, Methvin, Al-awar, Rima, and Hassell, John A.
- Published
- 2020
- Full Text
- View/download PDF
4. Retraction Note: LncGPR107 drives the self-renewal of liver tumor initiating cells and liver tumorigenesis through GPR107-dependent manner
- Author
-
Huang, Guanqun, Jiang, Hui, Lin, Ye, Xia, Wuzheng, Luo, Yuanwei, Wu, Yanpeng, Cai, Weilong, Zhou, Xinke, and Jiang, Xianhan
- Published
- 2022
- Full Text
- View/download PDF
5. Characterization of EpCAM in thyroid cancer biology by three-dimensional spheroids in vitro model
- Author
-
Ghiandai, Viola, Grassi, Elisa Stellaria, Gazzano, Giacomo, Fugazzola, Laura, and Persani, Luca
- Published
- 2024
- Full Text
- View/download PDF
6. Human bronchial carcinoid tumor initiating cells are targeted by the combination of acetazolamide and sulforaphane
- Author
-
Bayat Mokhtari, Reza, Baluch, Narges, Morgatskaya, Evgeniya, Kumar, Sushil, Sparaneo, Angelo, Muscarella, Lucia Anna, Zhao, Sheyun, Cheng, Hai-Ling, Das, Bikul, and Yeger, Herman
- Published
- 2019
- Full Text
- View/download PDF
7. Functional consequences of enhanced expression of STIM1 and Orai1 in Huh-7 hepatocellular carcinoma tumor-initiating cells
- Author
-
Karacicek, B., Erac, Y., and Tosun, M.
- Published
- 2019
- Full Text
- View/download PDF
8. RETRACTED ARTICLE: LncGPR107 drives the self-renewal of liver tumor initiating cells and liver tumorigenesis through GPR107-dependent manner
- Author
-
Huang, Guanqun, Jiang, Hui, Lin, Ye, Xia, Wuzheng, Luo, Yuanwei, Wu, Yanpeng, Cai, Weilong, Zhou, Xinke, and Jiang, Xianhan
- Published
- 2018
- Full Text
- View/download PDF
9. Tumor-Initiating Cells: a criTICal review of isolation approaches and new challenges in targeting strategies
- Author
-
Komal Qureshi-Baig, Elisabeth Letellier, Pit Ullmann, Serge Haan, Fondation Cancer [sponsor], and Fondation du Pélican de Mie [sponsor]
- Subjects
0301 basic medicine ,cancer stem cells ,Cancer Research ,Colorectal cancer ,medicine.medical_treatment ,Cell Culture Techniques ,Review ,Biochemistry, biophysics & molecular biology [F05] [Life sciences] ,Tumor Initiating Cells ,Targeted therapy ,Tumor Cells, Cultured ,Molecular Targeted Therapy ,Biochimie, biophysique & biologie moléculaire [F05] [Sciences du vivant] ,Cancer stem cells ,Tumor-initiating cells ,targeted therapy ,Oncology ,Isolation (psychology) ,Neoplastic Stem Cells ,Molecular Medicine ,Metabolic identity ,Colorectal Neoplasms ,Signal Transduction ,congenital, hereditary, and neonatal diseases and abnormalities ,Tics ,colorectal cancer ,Biology ,tumor-initiating cells ,03 medical and health sciences ,Spheroid Culture System ,Genetic Heterogeneity ,Cancer stem cell ,mental disorders ,medicine ,Biomarkers, Tumor ,metabolic identity ,Humans ,Culturing conditions ,surface markers ,inter- and intratumor heterogeneity ,medicine.disease ,nervous system diseases ,body regions ,Spheroid Culture Systems ,030104 developmental biology ,Immunology ,Surface markers ,Inter- and intra-tumor heterogeneity ,Cancer research ,human activities - Abstract
Most cancers contain a subpopulation of highly tumorigenic cells, known as cancer stem cells (CSCs) or tumor-initiating cells (TICs). Targeting TICs may be essential to achieve cure, because of their self-renewal and tumorigenic properties as well as their resistance to conventional therapies. Despite significant advances in TIC biology, their isolation and identification remain largely disputed and incompletely established. In this review, we discuss the latest developments in isolation and culturing approaches of TICs, with focus on colorectal cancer (CRC). We feature recent findings on TIC-relevant signaling pathways and the metabolic identity of TICs, as well as their current clinical implications. Lastly, we highlight the influence of inter- and intra-tumoral heterogeneity on TIC function and targeting approaches.
- Published
- 2017
10. Spheres derived from the human SN12C renal cell carcinoma cell line are enriched in tumor initiating cells
- Author
-
Zhiyong Liu, Yanhui Zhang, Xin Yao, Xiulan Zhao, Baocun Sun, Xueyi Dong, Wei Cui, and Huizhi Sun
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Epithelial-Mesenchymal Transition ,Angiogenesis ,Cell Culture Techniques ,Tumor initiation ,Tumor initiating cells ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Spheroids, Cellular ,Cell Self Renewal ,Medicine ,Animals ,Humans ,Epithelial–mesenchymal transition ,Carcinoma, Renal Cell ,Cell Proliferation ,business.industry ,Cell growth ,Research ,Spheroid ,Aldehyde Dehydrogenase ,Renal cell carcinoma ,Kidney Neoplasms ,030104 developmental biology ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,embryonic structures ,Cancer research ,Neoplastic Stem Cells ,Stem cell ,business ,Neoplasm Transplantation - Abstract
Background Recently, tumor initiating cells (TICs), which possess self-renewal and other stem cell properties, are regarded as the cause of tumor initiation, recurrence and metastasis. The isolation and identification of TICs could help to develop novel therapeutic strategies. Methods In this study, we isolated spheroid cells from human renal cell carcinoma (RCC) cell line SN12C in stem cell-conditioned medium. The stemness characteristics of spheroid cells, including tumorigenicity, self-renewal, proliferation and aldehyde dehydrogenase (ALDH) activity were evaluated; the expression levels of stemness genes and related proteins were assessed. Furthermore, study examined the differentiation of TICs into endothelial cells and the relationship between TICs and EMT. Results Our data demonstrated that spheroid cells cultured in defined serum-free medium possessed TIC properties, such as high tumorigenic capacity, upregulation of TIC-related genes and proteins, persistent self-renewal and extensive proliferation. Furthermore, spheroid cells were more aggressive in growth, invasion, scratch recovery, clonogenic survival and high aldehyde dehydrogenase (ALDH) activity. Interestingly, a marked increase in tumor vascularity compared to adherent tumors in vivo, and spheroid cells can differentiate into functional endothelial-like cells in vitro suggesting a role of tumor initiating cells in tumor angiogenesis. The spheroid cells also demonstrated down-regulated E-cadherin and up-regulated Vimentin expression, which is the typical phenotype of EMT. Conclusions These results suggest that spheroid cells with tumor initiating cells-like characteristics contributed to tumor generation, progression, high tumorigenicity, pro-angiogenic capability and relationship with EMT. Further experiments using more refined selection criteria such as a combination of two or multiple markers would be useful to specifically identify and purify TICs.
- Published
- 2016
11. Generation of tumor-initiating cells by exogenous delivery of OCT4transcription factor
- Author
-
Beltran, Adriana S, Rivenbark, Ashley G, Richardson, Bryan T, Yuan, Xinni, Quian, Haili, Hunt, John P, Zimmerman, Eric, Graves, Lee M, and Blancafort, Pilar
- Published
- 2011
- Full Text
- View/download PDF
12. The inhibition of FGF receptor 1 activity mediates sorafenib antiproliferative effects in human malignant pleural mesothelioma tumor-initiating cells.
- Author
-
Pattarozzi, Alessandra, Carra, Elisa, Favoni, Roberto E., Würth, Roberto, Marubbi, Daniela, Filiberti, Rosa Angela, Mutti, Luciano, Florio, Tullio, Barbieri, Federica, and Daga, Antonio
- Subjects
- *
FIBROBLAST growth factors , *SORAFENIB , *MESOTHELIOMA , *ANTINEOPLASTIC agents , *CELL survival - Abstract
Background: Malignant pleural mesothelioma is an aggressive cancer, characterized by rapid progression and high mortality. Persistence of tumor-initiating cells (TICs, or cancer stem cells) after cytotoxic drug treatment is responsible for tumor relapse, and represents one of the main reasons for the poor prognosis of mesothelioma. In fact, identification of the molecules affecting TIC viability is still a significant challenge. Methods: TIC-enriched cultures were obtained from 10 human malignant pleural mesotheliomas and cultured in vitro. Three fully characterized tumorigenic cultures, named MM1, MM3, and MM4, were selected and used to assess antiproliferative effects of the multi-kinase inhibitor sorafenib. Cell viability was investigated by MTT assay, and cell cycle analysis as well as induction of apoptosis were determined by flow cytometry. Western blotting was performed to reveal the modulation of protein expression and the phosphorylation status of pathways associated with sorafenib treatment. Results: We analyzed the molecular mechanisms of the antiproliferative effects of sorafenib in mesothelioma TIC cultures. Sorafenib inhibited cell cycle progression in all cultures, but only in MM3 and MM4 cells was this effect associated with Mcl-1-dependent apoptosis. To investigate the mechanisms of sorafenib-mediated antiproliferative activity, TICs were treated with epidermal growth factor (EGF) or basic fibroblast growth factor (bFGF) causing, in MM3 and MM4 cells, MEK, ERK1/2, Akt, and STAT3 phosphorylation. These effects were abolished by sorafenib only in bFGF-treated cells, while a modest inhibition occurred after EGF stimulation, suggesting that sorafenib effects are mainly due to FGF receptor (FGFR) inhibition. Indeed, FGFR1 phosphorylation was inhibited by sorafenib. Moreover, in MM1 cells, which release high levels of bFGF and showed autocrine activation of FGFR1 and constitutive phosphorylation/activation of MEK-ERK1/2, sorafenib induced a more effective antiproliferative response, confirming that the main target of the drug is the inhibition of FGFR1 activity. Conclusions: These results suggest that, in malignant pleural mesothelioma TICs, bFGF signaling is the main target of the antiproliferative response of sorafenib, acting directly on the FGFR1 activation. Patients with constitutive FGFR1 activation via an autocrine loop may be more sensitive to sorafenib treatment and the analysis of this possibility warrants further clinical investigation. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
13. Tumor-Initiating Cells: a criTICal review of isolation approaches and new challenges in targeting strategies.
- Author
-
Qureshi-Baig, Komal, Ullmann, Pit, Haan, Serge, and Letellier, Elisabeth
- Subjects
- *
CANCER cells , *CANCER stem cells , *COLON cancer , *JAK-STAT pathway , *TUMORS - Abstract
Most cancers contain a subpopulation of highly tumorigenic cells, known as cancer stem cells (CSCs) or tumor-initiating cells (TICs). Targeting TICs may be essential to achieve cure, because of their self-renewal and tumorigenic properties as well as their resistance to conventional therapies. Despite significant advances in TIC biology, their isolation and identification remain largely disputed and incompletely established. In this review, we discuss the latest developments in isolation and culturing approaches of TICs, with focus on colorectal cancer (CRC). We feature recent findings on TIC-relevant signaling pathways and the metabolic identity of TICs, as well as their current clinical implications. Lastly, we highlight the influence of inter- and intra-tumoral heterogeneity on TIC function and targeting approaches. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
14. Spheres derived from the human SN12C renal cell carcinoma cell line are enriched in tumor initiating cells.
- Author
-
Yanhui Zhang, Baocun Sun, Xiulan Zhao, Huizhi Sun, Wei Cui, Zhiyong Liu, Xin Yao, and Xueyi Dong
- Subjects
- *
RENAL cell carcinoma , *METASTASIS , *TUMOR risk factors , *ALDEHYDE dehydrogenase , *THERAPEUTICS - Abstract
Background: Recently, tumor initiating cells (TICs), which possess self-renewal and other stem cell properties, are regarded as the cause of tumor initiation, recurrence and metastasis. The isolation and identification of TICs could help to develop novel therapeutic strategies. Methods: In this study, we isolated spheroid cells from human renal cell carcinoma (RCC) cell line SN12C in stem cell-conditioned medium. The stemness characteristics of spheroid cells, including tumorigenicity, self-renewal, proliferation and aldehyde dehydrogenase (ALDH) activity were evaluated; the expression levels of stemness genes and related proteins were assessed. Furthermore, study examined the differentiation of TICs into endothelial cells and the relationship between TICs and EMT. Results: Our data demonstrated that spheroid cells cultured in defined serum-free medium possessed TIC properties, such as high tumorigenic capacity, upregulation of TIC-related genes and proteins, persistent self-renewal and extensive proliferation. Furthermore, spheroid cells were more aggressive in growth, invasion, scratch recovery, clonogenic survival and high aldehyde dehydrogenase (ALDH) activity. Interestingly, a marked increase in tumor vascularity compared to adherent tumors in vivo, and spheroid cells can differentiate into functional endothelial-like cells in vitro suggesting a role of tumor initiating cells in tumor angiogenesis. The spheroid cells also demonstrated down-regulated E-cadherin and up-regulated Vimentin expression, which is the typical phenotype of EMT. Conclusions: These results suggest that spheroid cells with tumor initiating cells-like characteristics contributed to tumor generation, progression, high tumorigenicity, pro-angiogenic capability and relationship with EMT. Further experiments using more refined selection criteria such as a combination of two or multiple markers would be useful to specifically identify and purify TICs. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
15. The inhibition of FGF receptor 1 activity mediates sorafenib-induced antiproliferative effects in human mesothelioma tumor-initiating cells
- Author
-
Pattarozzi, A, Carra, E, Favoni, RE, Würth, R, Marubbi, D, Filiberti, R, Mutti, L, Florio, T, Barbieri, F, and Daga, A
- Subjects
neoplasms ,digestive system diseases - Abstract
Tumor-initiating cells (TICs), the subset of cells within tumors endowed with stem-like features, being highly resistant to conventional cytotoxic drugs, are the major cause of tumor relapse. The identification of molecules able to target TICs remains a significant challenge in cancer therapy. Using TIC-enriched cultures (MM1, MM3 and MM4), from 3 human malignant pleural mesotheliomas (MPM), we tested the effects of sorafenib on cell survival and the intracellular mechanisms involved. Sorafenib inhibited cell-cycle progression in all the TIC cultures, but only in MM3 and MM4 cells this effect was associated with induction of apoptosis via the down-regulation of Mcl-1. Although sorafenib inhibits the activity of several tyrosine kinases, its effects are mainly ascribed to Raf inhibition. To investigate the mechanisms of sorafenib-mediated antiproliferative activity, TICs were treated with EGF or bFGF causing, in MM3 and MM4 cells, MEK, ERK1/2, Akt and STAT3 phosphorylation. These effects were significantly reduced by sorafenib in bFGF-treated cells, while a slight inhibition occurred after EGF stimulation, suggesting that sorafenib effects are mainly due to FGFR inhibition. Indeed, FGFR1 phosphorylation was inhibited by sorafenib. \ud A different picture was observed in MM1 cells, which, releasing high levels of bFGF, showed an autocrine activation of FGFR1 and a constitutive phosphorylation/activation of MEK-ERK1/2. A powerful inhibitory response to sorafenib was observed in these cells, indirectly confirming the central role of sorafenib as FGFR inhibitor.\ud These results suggest that bFGF signaling may impact antiproliferative response to sorafenib of MPM TICs, which is mainly mediated by a direct FGFR targeting.
- Published
- 2017
16. Suppression of RAF/MEK or PI3K synergizes cytotoxicity of receptor tyrosine kinase inhibitors in glioma tumor-initiating cells.
- Author
-
Shingu T, Holmes L, Henry V, Wang Q, Latha K, Gururaj AE, Gibson LA, Doucette T, Lang FF, Rao G, Yuan L, Sulman EP, Farrell NP, Priebe W, Hess KR, Wang YA, Hu J, and Bögler O
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols pharmacology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Apoptosis drug effects, Autophagy drug effects, Biomarkers, Tumor metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Drug Synergism, Humans, Inhibitory Concentration 50, Male, Mice, Nude, Mitogen-Activated Protein Kinase Kinases metabolism, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells metabolism, Phosphatidylinositol 3-Kinases metabolism, Protein Kinase Inhibitors pharmacology, Signal Transduction drug effects, raf Kinases metabolism, Glioma drug therapy, Glioma pathology, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Neoplastic Stem Cells pathology, Phosphoinositide-3 Kinase Inhibitors, Protein Kinase Inhibitors therapeutic use, raf Kinases antagonists & inhibitors
- Abstract
Background: The majority of glioblastomas have aberrant receptor tyrosine kinase (RTK)/RAS/phosphoinositide 3 kinase (PI3K) signaling pathways and malignant glioma cells are thought to be addicted to these signaling pathways for their survival and proliferation. However, recent studies suggest that monotherapies or inappropriate combination therapies using the molecular targeted drugs have limited efficacy possibly because of tumor heterogeneities, signaling redundancy and crosstalk in intracellular signaling network, indicating necessity of rationale and methods for efficient personalized combination treatments. Here, we evaluated the growth of colonies obtained from glioma tumor-initiating cells (GICs) derived from glioma sphere culture (GSC) in agarose and examined the effects of combination treatments on GICs using targeted drugs that affect the signaling pathways to which most glioma cells are addicted., Methods: Human GICs were cultured in agarose and treated with inhibitors of RTKs, non-receptor kinases or transcription factors. The colony number and volume were analyzed using a colony counter, and Chou-Talalay combination indices were evaluated. Autophagy and apoptosis were also analyzed. Phosphorylation of proteins was evaluated by reverse phase protein array and immunoblotting., Results: Increases of colony number and volume in agarose correlated with the Gompertz function. GICs showed diverse drug sensitivity, but inhibitions of RTK and RAF/MEK or PI3K by combinations such as EGFR inhibitor and MEK inhibitor, sorafenib and U0126, erlotinib and BKM120, and EGFR inhibitor and sorafenib showed synergy in different subtypes of GICs. Combination of erlotinib and sorafenib, synergistic in GSC11, induced apoptosis and autophagic cell death associated with suppressed Akt and ERK signaling pathways and decreased nuclear PKM2 and β-catenin in vitro, and tended to improve survival of nude mice bearing GSC11 brain tumor. Reverse phase protein array analysis of the synergistic treatment indicated involvement of not only MEK and PI3K signaling pathways but also others associated with glucose metabolism, fatty acid metabolism, gene transcription, histone methylation, iron transport, stress response, cell cycle, and apoptosis., Conclusion: Inhibiting RTK and RAF/MEK or PI3K could induce synergistic cytotoxicity but personalization is necessary. Examining colonies in agarose initiated by GICs from each patient may be useful for drug sensitivity testing in personalized cancer therapy.
- Published
- 2016
- Full Text
- View/download PDF
17. Fusion with stem cell makes the hepatocellular carcinoma cells similar to liver tumor-initiating cells.
- Author
-
Wang R, Chen S, Li C, Ng KT, Kong CW, Cheng J, Cheng SH, Li RA, Lo CM, Man K, and Sun D
- Subjects
- Cell Line, Tumor, Flow Cytometry, Gene Expression Regulation, Neoplastic, Human Embryonic Stem Cells metabolism, Human Embryonic Stem Cells pathology, Humans, Hyaluronan Receptors biosynthesis, Lasers, Liver metabolism, Liver pathology, Neoplasm Proteins biosynthesis, Neoplastic Stem Cells metabolism, Carcinoma, Hepatocellular pathology, Cell Fusion, Liver Neoplasms pathology, Neoplastic Stem Cells pathology
- Abstract
Background: Cell fusion is a fast and highly efficient technique for cells to acquire new properties. The fusion of somatic cells with stem cells can reprogram somatic cells to a pluripotent state. Our research on the fusion of stem cells and cancer cells demonstrates that the fused cells can exhibit stemness and cancer cell-like characteristics. Thus, tumor-initiating cell-like cells are generated., Methods: We employed laser-induced single-cell fusion technique to fuse the hepatocellular carcinoma cells and human embryonic stem cells (hESC). Real-time RT-PCR, flow cytometry and in vivo tumorigenicity assay were adopted to identify the gene expression difference., Results: We successfully produced a fused cell line that coalesces the gene expression information of hepatocellular carcinoma cells and stem cells. Experimental results showed that the fused cells expressed cancer and stemness markers as well as exhibited increased resistance to drug treatment and enhanced tumorigenesis., Conclusions: Fusion with stem cells transforms liver cancer cells into tumor initiating-like cells. Results indicate that fusion between cancer cell and stem cell may generate tumor initiating-like cells.
- Published
- 2016
- Full Text
- View/download PDF
18. LAMC2 marks a tumor-initiating cell population with an aggressive signature in pancreatic cancer
- Author
-
Cave, Donatella Delle, Buonaiuto, Silvia, Sainz, Jr, Bruno, Fantuz, Marco, Mangini, Maria, Carrer, Alessandro, Di Domenico, Annalisa, Iavazzo, Tea Teresa, Andolfi, Gennaro, Cortina, Carme, Sevillano, Marta, Heeschen, Christopher, Colonna, Vincenza, Corona, Marco, Cucciardi, Antonio, Di Guida, Martina, Batlle, Eduard, De Luca, Annachiara, and Lonardo, Enza
- Published
- 2022
- Full Text
- View/download PDF
19. Fusion with stem cell makes the hepatocellular carcinoma cells similar to liver tumor-initiating cells
- Author
-
Shuxun Chen, Ronald A. Li, Kevin Tak-Pan Ng, Chang-Xian Li, Jinping Cheng, Ran Wang, Shuk Han Cheng, Chung Mau Lo, Chi-Wing Kong, Dong Sun, and Kwan Man
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,Hepatocellular carcinoma ,Cellular differentiation ,Human Embryonic Stem Cells ,Biology ,Tumor-initiating cell ,Cell Fusion ,03 medical and health sciences ,0302 clinical medicine ,Cancer stem cell ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Induced stem cells ,Stem cell ,Lasers ,Liver Neoplasms ,Flow Cytometry ,Embryonic stem cell ,Neoplasm Proteins ,Endothelial stem cell ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,P19 cell ,Hyaluronan Receptors ,Oncology ,Liver ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Neoplastic Stem Cells ,Research Article - Abstract
Background Cell fusion is a fast and highly efficient technique for cells to acquire new properties. The fusion of somatic cells with stem cells can reprogram somatic cells to a pluripotent state. Our research on the fusion of stem cells and cancer cells demonstrates that the fused cells can exhibit stemness and cancer cell-like characteristics. Thus, tumor-initiating cell-like cells are generated. Methods We employed laser-induced single-cell fusion technique to fuse the hepatocellular carcinoma cells and human embryonic stem cells (hESC). Real-time RT-PCR, flow cytometry and in vivo tumorigenicity assay were adopted to identify the gene expression difference. Results We successfully produced a fused cell line that coalesces the gene expression information of hepatocellular carcinoma cells and stem cells. Experimental results showed that the fused cells expressed cancer and stemness markers as well as exhibited increased resistance to drug treatment and enhanced tumorigenesis. Conclusions Fusion with stem cells transforms liver cancer cells into tumor initiating-like cells. Results indicate that fusion between cancer cell and stem cell may generate tumor initiating-like cells.
- Published
- 2016
20. Small nucleolar RNA signatures of lung tumor-initiating cells.
- Author
-
Mannoor, Kaiissar, Jun Shen, Jipei Liao, Zhenqiu Liu, and Feng Jiang
- Subjects
- *
LUNG cancer , *CELL proliferation , *CELL culture , *CANCER invasiveness , *CELL lines - Abstract
Background Non-small cell lung cancer (NSCLC) is the number one cancer killer. Tumor-initiating cells (TICs) are responsible for tumor progression and recurrence. Emerging evidences suggest that small nucleolar RNAs (snoRNAs) play malfunctioning roles in lung tumorigenesis. This study aims to determine if snoRNAs have important function in lung TICs by: 1) profiling and comparing snoRNA expression patterns in lung ALDH1+/- cells of 28 primary NSCLC tissues to identify new signatures of TICs; 2) determining prognostic significance of the snoRNA signatures by analyzing the expression in 82 NSCLC tissues with different stages and histological types using quantitative PCR; 3) functionally investigating if the snoRNAs contribute to stemness of lung TICs using in vitro and in vivo assays. Results Twenty-two snoRNAs were identified whose changes were specific to the TICs. The expression of two snoRNAs (snoRA3 and snoRA42) was inversely associated with survival of NSCLC patients (P = 0.002, p = 0.001, respectively). Functional analysis indicated that snoRA42 was upregulated in CD133+ cells isolated from NSCLC cell lines compared with the CD133- counterparts. snoRA42 knockdown reduced the proliferation and self-renewal of TICs in vitro. However, ectopic expression of snoRA42 in non-TICs enhanced the potentials of cell proliferation and self-renewal. snoRA42 expression was associated with expression of stem cell-core transcription factors in lung TICs. Blocking snoRA42 expression in TIC xenografts decreased tumorigenesis in mice. Conclusions The snoRNA signatures of lung TICs provide potential biomarkers for predicting outcome of NSCLC. snoRA42 is one of the important snoRNAs in regulating features of lung TICs, and thus contributes to lung tumorigenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
21. Protein markers of cancer-associated fibroblasts and tumor-initiating cells reveal subpopulations in freshly isolated ovarian cancer ascites.
- Author
-
Wintzell, My, Hjerpe, Elisabet, Lundqvist, Elisabeth Åvall, and Shoshan, Maria
- Subjects
- *
OVARIAN cancer , *TUMORS , *FIBROBLASTS , *VIMENTIN , *INTEGRINS , *STEM cells - Abstract
Background: In ovarian cancer, massive intraperitoneal dissemination is due to exfoliated tumor cells in ascites. Tumor-initiating cells (TICs or cancer stem cells) and cells showing epithelial-mesenchymal-transition (EMT) are particularly implicated. Spontaneous spherical cell aggregates are sometimes observed, but although similar to those formed by TICs in vitro, their significance is unclear. Methods: Cells freshly isolated from malignant ascites were separated into sphere samples (S-type samples, n=9) and monolayer-forming single-cell suspensions (M-type, n=18). Using western blot, these were then compared for expression of protein markers of EMT, TIC, and of cancer-associated fibroblasts (CAFs). Results: S-type cells differed significantly from M-type by expressing high levels of E-cadherin and no or little vimentin, integrin-β3 or stem cell transcription factor Oct-4A. By contrast, M-type samples were enriched for CD44, Oct-4A and for CAF markers. Independently of M- and S-type, there was a strong correlation between TIC markers Nanog and EpCAM. The CAF marker α-SMA correlated with clinical stage IV. This is the first report on CAF markers in malignant ascites and on SUMOylation of Oct-4A in ovarian cancer. Conclusions: In addition to demonstrating potentially high levels of TICs in ascites, the results suggest that the S-type population is the less tumorigenic one. Nanoghigh/EpCAMhigh samples represent a TIC subset which may be either M- or S-type, and which is separate from the CD44high/Oct-4Ahigh subset observed only in M-type samples. This demonstrates a heterogeneity in TIC populations in vivo which has practical implications for TIC isolation based on cell sorting. The biological heterogeneity will need to be addressed in future therapeutical strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
22. A mouse model for triple-negative breast cancer tumor-initiating cells (TNBC-TICs) exhibits similar aggressive phenotype to the human disease.
- Subjects
- *
TRIPLE-negative breast cancer , *CANCER cells , *LABORATORY mice , *PHENOTYPES , *CELL lines , *GENE expression - Abstract
The article discusses a study which examined a mouse model for triple-negative breast cancer tumor-initiating cells (TNBC-TICs) to find whether it exhibits similar aggressive phenotype to the human disease. To understand the mechanistic basis for the aggressiveness of TNBC, the study produced a stable TNBC cell line by sorting for 4T1 cells that do not express the estrogen receptor (ER), progesterone receptor (PgR) or the gene for human epidermal growth factor receptor 2 (HER2).
- Published
- 2012
- Full Text
- View/download PDF
23. "A novel in vivo model for the study of human breast cancer metastasis using primary breast tumor-initiating cells from patient biopsies"
- Subjects
- *
BREAST cancer , *CANCER research , *METASTASIS , *BIOPSY , *LABORATORY mice - Abstract
The article focuses on a study which aims developing a novel in vivo and reproducible breast cancer model using breast tumor-initiating cells (bTICs). The model was developed by isolating bTICs as tumorspheres from biopsies for the investigation of breast cancer metastatic process. It explains the methodology of the study, in which culture conditions were transplanted into the mammary fat pad of mice. It concludes the efficiency of this model for investigating breast cancer metastasis.
- Published
- 2012
- Full Text
- View/download PDF
24. An inhibitor of K+ channels modulates human endometrial tumor-initiating cells.
- Subjects
- *
CANCER cells , *MENSTRUAL cycle , *POTASSIUM channels , *CANCER treatment , *ONCOGENES - Abstract
The article examines the role of tetraethylammonium (TEA),an inhibitor of K+ channels, in modulating human endometrial tumor-initiating cells. It is found that TEA suppress colony formation in endometrial cancer cells through inhibition of putative. It is also found that the activity of potassium channels significantly contributes to the progression of endometrial tumors.
- Published
- 2011
- Full Text
- View/download PDF
25. Endothelial cells do not arise from tumor-initiating cells in human hepatocellular carcinoma.
- Author
-
Ghanekar A, Ahmed S, Chen K, and Adeyi O
- Subjects
- Animals, Carcinoma, Hepatocellular diagnosis, Carcinoma, Hepatocellular metabolism, Carcinoma, Hepatocellular therapy, Cell Line, Tumor, Disease Models, Animal, Endothelial Cells metabolism, Female, Heterografts, Humans, Liver metabolism, Liver pathology, Liver Neoplasms diagnosis, Liver Neoplasms metabolism, Liver Neoplasms therapy, Liver Transplantation, Magnetic Resonance Imaging, Mice, Neoplasm Recurrence, Local, Neoplastic Stem Cells metabolism, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Transplantation, Homologous, Carcinoma, Hepatocellular pathology, Endothelial Cells pathology, Liver Neoplasms pathology, Neoplastic Stem Cells pathology
- Abstract
Background: Conventional models of carcinogenesis suggest that tumors recruit blood vessel formation from normal host tissues. This concept has recently been challenged by prominent studies of glioblastoma, which suggest that intratumoral endothelial cells (ECs) may arise from cancer stem cells/tumor-initiating cells (TICs). Hepatocellular carcinoma (HCC) is a common, highly vascularized tumor with few effective therapies, against which anti-angiogenic strategies are being actively explored. TICs are felt to play a role in HCC pathobiology, but their contributions to tumor vasculature have not been studied., Methods: We examined human HCCs in settings that selected for tumor formation from functionally defined TICs, and in which the origin of intratumoral ECs from TICs as opposed to host tissues could be clearly distinguished. We generated HCC nodules in the livers of immunodeficient mice by intrasplenic injection of HCC cells from cell lines and patient specimens and studied the tumor ECs by immunohistochemistry for mouse and human markers. We then used immunohistochemistry for EC markers in combination with fluorescence in situ hybridization (FISH) for X and Y chromosomes to study the endothelium of recurrent HCC specimens resected from sex-mismatched liver allografts of patients who had undergone liver transplantation for HCC., Results: We observed that all ECs in intrahepatic human HCC xenografts expressed mouse rather than human CD31. FISH analysis of recurrent HCCs resected from patients with sex-mismatched liver allografts revealed that all CD31+ and CD34+ intratumoral ECs originated from the donor allograft rather than the tumor., Conclusions: These observations suggest that the vasculature of human HCC arises from normal host tissues rather than from TICs, supporting ongoing efforts to target angiogenesis in HCC as it is currently understood, and suggesting that the contribution of TICs to the vasculature of other cancers is disease-specific.
- Published
- 2013
- Full Text
- View/download PDF
26. PC3 prostate tumor-initiating cells with molecular profile FAM65Bhigh/MFI2low/LEF1low increase tumor angiogenesis.
- Author
-
Zhang K and Waxman DJ
- Subjects
- Animals, Cell Adhesion Molecules, Cell Line, Tumor, Flow Cytometry, Humans, Immunohistochemistry, Lymphoid Enhancer-Binding Factor 1 genetics, Male, Mice, Mice, SCID, Neoplasm Proteins genetics, Neovascularization, Pathologic genetics, Oligonucleotide Array Sequence Analysis, Platelet Endothelial Cell Adhesion Molecule-1 genetics, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Polymerase Chain Reaction, Proteins genetics, Lymphoid Enhancer-Binding Factor 1 metabolism, Neoplasm Proteins metabolism, Neovascularization, Pathologic metabolism, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Proteins metabolism
- Abstract
Background: Cancer stem-like cells are proposed to sustain solid tumors by virtue of their capacity for self-renewal and differentiation to cells that comprise the bulk of the tumor, and have been identified for a variety of cancers based on characteristic clonal morphologies and patterns of marker gene expression., Methods: Single cell cloning and spheroid culture studies were used to identify a population of cancer stem-like cells in the androgen-independent human prostate cancer cell line PC3., Results: We demonstrate that, under standard culture conditions, ~10% of PC3 cells form holoclones with cancer stem cell characteristics. These holoclones display high self-renewal capability in spheroid formation assays under low attachment and serum-free culture conditions, retain their holoclone morphology when passaged at high cell density, exhibit moderate drug resistance, and show high tumorigenicity in scid immunodeficient mice. PC3 holoclones readily form spheres, and PC3-derived spheres yield a high percentage of holoclones, further supporting their cancer stem cell-like nature. We identified one gene, FAM65B, whose expression is consistently up regulated in PC3 holoclones compared to paraclones, the major cell morphology in the parental PC3 cell population, and two genes, MFI2 and LEF1, that are consistently down regulated. This molecular profile, FAM65Bhigh/MFI2low/LEF1low, also characterizes spheres generated from parental PC3 cells. The PC3 holoclones did not show significant enriched expression of the putative prostate cancer stem cell markers CD44 and integrin α2β1. PC3 tumors seeded with holoclones showed dramatic down regulation of FAM65B and dramatic up regulation of MFI2 and LEF1, and unexpectedly, a marked increase in tumor vascularity compared to parental PC3 tumors, suggesting a role of cancer stem cells in tumor angiogenesis., Conclusions: These findings support the proposal that PC3 tumors are sustained by a small number of tumor-initiating cells with stem-like characteristics, including strong self-renewal and pro-angiogenic capability and marked by the expression pattern FAM65Bhigh/MFI2low/LEF1low. These markers may serve as targets for therapies designed to eliminate cancer stem cell populations associated with aggressive, androgen-independent prostate tumors such as PC3.
- Published
- 2010
- Full Text
- View/download PDF
27. New-generation taxoid SB-T-1214 inhibits stem cell-related gene expression in 3D cancer spheroids induced by purified colon tumor-initiating cells.
- Author
-
Botchkina GI, Zuniga ES, Das M, Wang Y, Wang H, Zhu S, Savitt AG, Rowehl RA, Leyfman Y, Ju J, Shroyer K, and Ojima I
- Subjects
- Antigens, CD immunology, Cell Line, Tumor, Colonic Neoplasms immunology, Colonic Neoplasms pathology, Humans, Polymerase Chain Reaction, Antineoplastic Agents pharmacology, Colonic Neoplasms genetics, Gene Expression drug effects, Stem Cells drug effects, Taxoids pharmacology
- Abstract
Background: Growing evidence suggests that the majority of tumors are organized hierarchically, comprising a population of tumor-initiating, or cancer stem cells (CSCs) responsible for tumor development, maintenance and resistance to drugs. Previously we have shown that the CD133high/CD44high fraction of colon cancer cells is different from their bulk counterparts at the functional, morphological and genomic levels. In contrast to the majority of colon cancer cells expressing moderate levels of CD133, CD44 and CD166, cells with a high combined expression of CD133 and CD44 possessed several characteristic stem cell features, including profound self-renewal capacity in vivo and in vitro, and the ability to give rise to different cell phenotypes. The present study was undertaken for two aims: a) to determine stem cell-related genomic characteristics of floating 3D multicellular spheroids induced by CD133high/CD44high colon cancer cells; and b) to evaluate CSC-specific alterations induced by new-generation taxoid SB-T-1214., Results: Selected CSC phenotype was isolated from three independent invasive colon cancer cell lines, HCT116, HT29 and DLD-1. A stem cell-specific PCR array assay (SABiosciences) revealed that colonospheres induced by purified CD133high/CD44high expressing cells display profound up-regulation of stem cell-related genes in comparison with their bulk counterparts. The FACS analysis has shown that the 3D colonospheres contained some minority cell populations with high levels of expression of Oct4, Sox2, Nanog and c-Myc, which are essential for stem cell pluripotency and self-renewal. Single administration of the SB-T-1214 at concentration 100 nM-1 microM for 48 hr not only induced growth inhibition and apoptotic cell death in these three types of colon cancer spheroids in 3D culture, but also mediated massive inhibition of the stem cell-related genes and significant down-regulation of the pluripotency gene expression. PCR array and FACS data were confirmed with western blotting. Importantly, viable cells that survived this treatment regimen were no longer able to induce secondary floating spheroids and exhibited significant morphological abnormalities., Conclusions: We report here that a new-generation taxoid SB-T-1214 possesses significant activity against colon cancer spheroids induced by and enriched with drug resistant tumorigenic CD133high/CD44high cells and efficiently inhibited expression of the majority of stem cell-related genes. Our data indicates that the previously observed long-term efficacy of SB-T-1214 against drug resistant colon tumors in vivo may be explained by the down-regulation of multiple stem cell-related genes in the tumorigenic cell population, in addition to its known efficacy as a mitotic poison against proliferating cancer cells.
- Published
- 2010
- Full Text
- View/download PDF
28. Small interfering RNA library screen identified polo-like kinase-1 (PLK1) as a potential therapeutic target for breast cancer that uniquely eliminates tumor-initiating cells
- Author
-
Jennifer Law, Sandra E. Dunn, Abbas Fotovati, and Kaiji Hu
- Subjects
Gene Expression ,Apoptosis ,Cell Cycle Proteins ,Polo-like kinase ,chemistry.chemical_compound ,0302 clinical medicine ,Molecular Targeted Therapy ,RNA, Small Interfering ,skin and connective tissue diseases ,Medicine(all) ,0303 health sciences ,education.field_of_study ,biology ,Kinase ,Pteridines ,3. Good health ,Hyaluronan Receptors ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,Neoplastic Stem Cells ,Female ,RNA Interference ,Fluorouracil ,Growth inhibition ,Research Article ,Paclitaxel ,Population ,Antineoplastic Agents ,Breast Neoplasms ,Protein Serine-Threonine Kinases ,03 medical and health sciences ,Breast cancer ,Cancer stem cell ,Cell Line, Tumor ,Proto-Oncogene Proteins ,medicine ,Humans ,Cyclin B1 ,education ,030304 developmental biology ,Cell Proliferation ,Gene Library ,Cell growth ,CD44 ,medicine.disease ,Molecular biology ,chemistry ,Doxorubicin ,biology.protein ,Cancer research - Abstract
Introduction Triple-negative breast cancer (TNBC) high rate of relapse is thought to be due to the presence of tumor-initiating cells (TICs), molecularly defined as being CD44high/CD24-/low. TICs are resilient to chemotherapy and radiation. However, no currently accepted molecular target exists against TNBC and, moreover, TICs. Therefore, we sought the identification of kinase targets that inhibit TNBC growth and eliminate TICs. Methods A genome-wide human kinase small interfering RNA (siRNA) library (691 kinases) was screened against the TNBC cell line SUM149 for growth inhibition. Selected siRNAs were then tested on four different breast cancer cell lines to confirm the spectrum of activity. Their effect on the CD44high subpopulation and sorted CD44high/CD24-/low cells of SUM149 also was studied. Further studies were focused on polo-like kinase 1 (PLK1), including its expression in breast cancer cell lines, effect on the CD44high/CD24-/low TIC subpopulation, growth inhibition, mammosphere formation, and apoptosis, as well as the activity of the PLK1 inhibitor, BI 2536. Results Of the 85 kinases identified in the screen, 28 of them were further silenced by siRNAs on MDA-MB-231 (TNBC), BT474-M1 (ER+/HER2+, a metastatic variant), and HR5 (ER+/HER2+, a trastuzumab-resistant model) cells and showed a broad spectrum of growth inhibition. Importantly, 12 of 28 kinases also reduced the CD44high subpopulation compared with control in SUM149. Further tests of these 12 kinases directly on a sorted CD44high/CD24-/low TIC subpopulation of SUM149 cells confirmed their effect. Blocking PLK1 had the greatest growth inhibition on breast cancer cells and TICs by about 80% to 90% after 72 hours. PLK1 was universally expressed in breast cancer cell lines, representing all of the breast cancer subtypes, and was positively correlated to CD44. The PLK1 inhibitor BI 2536 showed similar effects on growth, mammosphere formation, and apoptosis as did PLK1 siRNAs. Finally, whereas paclitaxel, doxorubicin, and 5-fluorouracil enriched the CD44high/CD24-/low population compared with control in SUM149, subsequent treatment with BI 2536 killed the emergent population, suggesting that it could potentially be used to prevent relapse. Conclusion Inhibiting PLK1 with siRNA or BI 2536 blocked growth of TNBCs including the CD44high/CD24-/low TIC subpopulation and mammosphere formation. Thus, PLK1 could be a potential therapeutic target for the treatment of TNBC as well as other subtypes of breast cancer.
- Published
- 2012
29. Suppression of RAF/MEK or PI3K synergizes cytotoxicity of receptor tyrosine kinase inhibitors in glioma tumor-initiating cells.
- Author
-
Takashi Shingu, Holmes, Lindsay, Henry, Verlene, Qianghu Wang, Latha, Khatri, Gururaj, Anupama E., Gibson, Laura A., Doucette, Tiffany, Lang, Frederick F., Ganesh Rao, Liang Yuan, Sulman, Erik P., Farrell, Nicholas P., Priebe, Waldemar, Hess, Kenneth R., Wang, Yaoqi A., Jian Hu, Bögler, Oliver, Shingu, Takashi, and Wang, Qianghu
- Subjects
GLIOMAS ,CANCER cell culture ,TARGETED drug delivery ,PROTEIN-tyrosine kinase inhibitors ,AUTOPHAGY ,APOPTOSIS ,PHOSPHORYLATION ,CELLULAR signal transduction ,ANIMAL experimentation ,ANTINEOPLASTIC agents ,CELL lines ,CELL physiology ,DRUG synergism ,MICE ,PHOSPHOTRANSFERASES ,RESEARCH funding ,STEM cells ,TRANSFERASES ,CHEMICAL inhibitors ,PROTEIN kinase inhibitors ,PHARMACODYNAMICS ,THERAPEUTICS - Abstract
Background: The majority of glioblastomas have aberrant receptor tyrosine kinase (RTK)/RAS/phosphoinositide 3 kinase (PI3K) signaling pathways and malignant glioma cells are thought to be addicted to these signaling pathways for their survival and proliferation. However, recent studies suggest that monotherapies or inappropriate combination therapies using the molecular targeted drugs have limited efficacy possibly because of tumor heterogeneities, signaling redundancy and crosstalk in intracellular signaling network, indicating necessity of rationale and methods for efficient personalized combination treatments. Here, we evaluated the growth of colonies obtained from glioma tumor-initiating cells (GICs) derived from glioma sphere culture (GSC) in agarose and examined the effects of combination treatments on GICs using targeted drugs that affect the signaling pathways to which most glioma cells are addicted.Methods: Human GICs were cultured in agarose and treated with inhibitors of RTKs, non-receptor kinases or transcription factors. The colony number and volume were analyzed using a colony counter, and Chou-Talalay combination indices were evaluated. Autophagy and apoptosis were also analyzed. Phosphorylation of proteins was evaluated by reverse phase protein array and immunoblotting.Results: Increases of colony number and volume in agarose correlated with the Gompertz function. GICs showed diverse drug sensitivity, but inhibitions of RTK and RAF/MEK or PI3K by combinations such as EGFR inhibitor and MEK inhibitor, sorafenib and U0126, erlotinib and BKM120, and EGFR inhibitor and sorafenib showed synergy in different subtypes of GICs. Combination of erlotinib and sorafenib, synergistic in GSC11, induced apoptosis and autophagic cell death associated with suppressed Akt and ERK signaling pathways and decreased nuclear PKM2 and β-catenin in vitro, and tended to improve survival of nude mice bearing GSC11 brain tumor. Reverse phase protein array analysis of the synergistic treatment indicated involvement of not only MEK and PI3K signaling pathways but also others associated with glucose metabolism, fatty acid metabolism, gene transcription, histone methylation, iron transport, stress response, cell cycle, and apoptosis.Conclusion: Inhibiting RTK and RAF/MEK or PI3K could induce synergistic cytotoxicity but personalization is necessary. Examining colonies in agarose initiated by GICs from each patient may be useful for drug sensitivity testing in personalized cancer therapy. [ABSTRACT FROM AUTHOR]- Published
- 2016
- Full Text
- View/download PDF
30. Hydrodynamic shear stress promotes epithelial-mesenchymal transition by downregulating ERK and GSK3β activities
- Author
-
Choi, Hye Yeon, Yang, Gwang-Mo, Dayem, Ahmed Abdal, Saha, Subbroto Kumar, Kim, Kyeongseok, Yoo, Youngbum, Hong, Kwonho, Kim, Jin-Hoi, Yee, Cassian, Lee, Kyung-Mi, and Cho, Ssang-Goo
- Published
- 2019
- Full Text
- View/download PDF
31. Monoamine oxidase-A activity is required for clonal tumorsphere formation by human breast tumor cells
- Author
-
Gwynne, William D., Shakeel, Mirza S., Wu, Jianhan, Hallett, Robin M., Girgis-Gabardo, Adele, Dvorkin-Gheva, Anna, and Hassell, John A.
- Published
- 2019
- Full Text
- View/download PDF
32. LncHOXA10 drives liver TICs self-renewal and tumorigenesis via HOXA10 transcription activation
- Author
-
Shao, Ming, Yang, Qiankun, Zhu, Weitao, Jin, Huifang, Wang, Jing, Song, Jie, Kong, Yongkui, and Lv, Xianping
- Published
- 2018
- Full Text
- View/download PDF
33. Enrichment of superoxide dismutase 2 in glioblastoma confers to acquisition of temozolomide resistance that is associated with tumor-initiating cell subsets
- Author
-
Chien, Chia-Hung, Chuang, Jian-Ying, Yang, Shun-Tai, Yang, Wen-Bin, Chen, Pin-Yuan, Hsu, Tsung-I, Huang, Chih-Yuan, Lo, Wei-Lun, Yang, Ka-Yen, Liu, Ming-Sheng, Chu, Jui-Mei, Chung, Pei-Hsuan, Liu, Jr-Jiun, Chou, Shao-Wen, Chen, Shang-Hung, and Chang, Kwang-Yu
- Published
- 2019
- Full Text
- View/download PDF
34. Generation of tumor-initiating cells by exogenous delivery of OCT4 transcription factor.
- Subjects
TUMORS ,CANCER cells ,CELL lines ,ECTOPIC tissue ,BREAST cancer - Abstract
The article focuses on the research related to determine the formation of tumor-initiating cells with the help of exogenous delivery of octamer-binding transcription factor 4 (OCT4) transcription factor. It offers information on tumor-initiating cells (TICs) or cancer stem cells, tumor cells with the ability of self renewing and generate tumors. It mentions that formation of TIC-like cell lines with ectopic expression of the OCT4 transcription factor (TF) in used for breast cell preparations.
- Published
- 2011
- Full Text
- View/download PDF
35. Enrichment of cancer stem cells via β-catenin contributing to the tumorigenesis of hepatocellular carcinoma
- Author
-
Pandit, Harshul, Li, Yan, Li, Xuanyi, Zhang, Weizhong, Li, Suping, and Martin, Robert C. G.
- Published
- 2018
- Full Text
- View/download PDF
36. Inhibition of CREB binding protein-beta-catenin signaling down regulates CD133 expression and activates PP2A-PTEN signaling in tumor initiating liver cancer cells
- Author
-
Tang, Yuanyuan, Berlind, Joshua, and Mavila, Nirmala
- Published
- 2018
- Full Text
- View/download PDF
37. Fusion with stem cell makes the hepatocellular carcinoma cells similar to liver tumor-initiating cells.
- Author
-
Ran Wang, Shuxun Chen, Changxian Li, Kevin Tak Pan Ng, Chi-wing Kong, Jinping Cheng, Shuk Han Cheng, Li, Ronald A., Chung Mau Lo, Kwan Man, and Dong Sun
- Subjects
LIVER cancer ,CANCER cells ,SOMATIC hybrids ,SOMATIC cells ,PLURIPOTENT stem cells ,EMBRYONIC stem cells ,GENE expression ,NEOPLASTIC cell transformation - Abstract
Background: Cell fusion is a fast and highly efficient technique for cells to acquire new properties. The fusion of somatic cells with stem cells can reprogram somatic cells to a pluripotent state. Our research on the fusion of stem cells and cancer cells demonstrates that the fused cells can exhibit stemness and cancer cell-like characteristics. Thus, tumor-initiating cell-like cells are generated. Methods: We employed laser-induced single-cell fusion technique to fuse the hepatocellular carcinoma cells and human embryonic stem cells (hESC). Real-time RT-PCR, flow cytometry and in vivo tumorigenicity assay were adopted to identify the gene expression difference. Results: We successfully produced a fused cell line that coalesces the gene expression information of hepatocellular carcinoma cells and stem cells. Experimental results showed that the fused cells expressed cancer and stemness markers as well as exhibited increased resistance to drug treatment and enhanced tumorigenesis. Conclusions: Fusion with stem cells transforms liver cancer cells into tumor initiating-like cells. Results indicate that fusion between cancer cell and stem cell may generate tumor initiating-like cells. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
38. Small interfering RNA library screen identified polo-like kinase-1 (PLK1) as a potential therapeutic target for breast cancer that uniquely eliminates tumor-initiating cells
- Author
-
Hu, Kaiji, Law, Jennifer H, Fotovati, Abbas, and Dunn, Sandra E
- Subjects
skin and connective tissue diseases ,3. Good health - Abstract
Introduction: Triple-negative breast cancer (TNBC) high rate of relapse is thought to be due to the presence of tumor-initiating cells (TICs), molecularly defined as being CD44high/CD24-/low. TICs are resilient to chemotherapy and radiation. However, no currently accepted molecular target exists against TNBC and, moreover, TICs. Therefore, we sought the identification of kinase targets that inhibit TNBC growth and eliminate TICs. Methods: A genome-wide human kinase small interfering RNA (siRNA) library (691 kinases) was screened against the TNBC cell line SUM149 for growth inhibition. Selected siRNAs were then tested on four different breast cancer cell lines to confirm the spectrum of activity. Their effect on the CD44high subpopulation and sorted CD44high/CD24-/low cells of SUM149 also was studied. Further studies were focused on polo-like kinase 1 (PLK1), including its expression in breast cancer cell lines, effect on the CD44high/CD24-/low TIC subpopulation, growth inhibition, mammosphere formation, and apoptosis, as well as the activity of the PLK1 inhibitor, BI 2536. Results: Of the 85 kinases identified in the screen, 28 of them were further silenced by siRNAs on MDA-MB-231 (TNBC), BT474-M1 (ER+/HER2+, a metastatic variant), and HR5 (ER+/HER2+, a trastuzumab-resistant model) cells and showed a broad spectrum of growth inhibition. Importantly, 12 of 28 kinases also reduced the CD44high subpopulation compared with control in SUM149. Further tests of these 12 kinases directly on a sorted CD44high/CD24-/low TIC subpopulation of SUM149 cells confirmed their effect. Blocking PLK1 had the greatest growth inhibition on breast cancer cells and TICs by about 80% to 90% after 72 hours. PLK1 was universally expressed in breast cancer cell lines, representing all of the breast cancer subtypes, and was positively correlated to CD44. The PLK1 inhibitor BI 2536 showed similar effects on growth, mammosphere formation, and apoptosis as did PLK1 siRNAs. Finally, whereas paclitaxel, doxorubicin, and 5-fluorouracil enriched the CD44high/CD24-/low population compared with control in SUM149, subsequent treatment with BI 2536 killed the emergent population, suggesting that it could potentially be used to prevent relapse. Conclusion: Inhibiting PLK1 with siRNA or BI 2536 blocked growth of TNBCs including the CD44high/CD24-/low TIC subpopulation and mammosphere formation. Thus, PLK1 could be a potential therapeutic target for the treatment of TNBC as well as other subtypes of breast cancer.
39. PC3 prostate tumor-initiating cells with molecular profile FAM65Bhigh/MFI2low/LEF1low increase tumor angiogenesis.
- Author
-
Kexiong Zhang and Waxman, David J.
- Subjects
CELL proliferation ,NEOVASCULARIZATION ,ONCOLOGY ,GENETICS ,BLOOD-vessel development - Abstract
Background: Cancer stem-like cells are proposed to sustain solid tumors by virtue of their capacity for self-renewal and differentiation to cells that comprise the bulk of the tumor, and have been identified for a variety of cancers based on characteristic clonal morphologies and patterns of marker gene expression. Methods: Single cell cloning and spheroid culture studies were used to identify a population of cancer stem-like cells in the androgen-independent human prostate cancer cell line PC3. Results: We demonstrate that, under standard culture conditions, ∼10% of PC3 cells form holoclones with cancer stem cell characteristics. These holoclones display high self-renewal capability in spheroid formation assays under low attachment and serum-free culture conditions, retain their holoclone morphology when passaged at high cell density, exhibit moderate drug resistance, and show high tumorigenicity in scid immunodeficient mice. PC3 holoclones readily form spheres, and PC3-derived spheres yield a high percentage of holoclones, further supporting their cancer stem cell-like nature. We identified one gene, FAM65B, whose expression is consistently up regulated in PC3 holoclones compared to paraclones, the major cell morphology in the parental PC3 cell population, and two genes, MFI2 and LEF1, that are consistently down regulated. This molecular profile, FAM65B
high /MFI2low /LEF1low , also characterizes spheres generated from parental PC3 cells. The PC3 holoclones did not show significant enriched expression of the putative prostate cancer stem cell markers CD44 and integrin a2b1. PC3 tumors seeded with holoclones showed dramatic down regulation of FAM65B and dramatic up regulation of MFI2 and LEF1, and unexpectedly, a marked increase in tumor vascularity compared to parental PC3 tumors, suggesting a role of cancer stem cells in tumor angiogenesis. Conclusions: These findings support the proposal that PC3 tumors are sustained by a small number of tumorinitiating cells with stem-like characteristics, including strong self-renewal and pro-angiogenic capability and marked by the expression pattern FAM65Bhigh /MFI2low /LEF1low . These markers may serve as targets for therapies designed to eliminate cancer stem cell populations associated with aggressive, androgen-independent prostate tumors such as PC3. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
40. LncGPR107 drives the self-renewal of liver tumor initiating cells and liver tumorigenesis through GPR107-dependent manner.
- Author
-
Huang, Guanqun, Jiang, Hui, Lin, Ye, Xia, Wuzheng, Luo, Yuanwei, Wu, Yanpeng, Cai, Weilong, Zhou, Xinke, and Jiang, Xianhan
- Subjects
LIVER cancer ,LIVER diseases ,LIVER transplantation ,GENE expression ,CANCER cells - Abstract
Background: With self-renewal and differentiation properties, liver tumor initiating cells (TICs) are the reasons for tumor initiation, metastasis and drug resistance. G protein coupled receptors (GPCR) are critical modulators in many physiological and pathological processes. While, their roles in liver TICs are unknown. Methods: An unbiased screening was performed using online-available data dataset. Liver TICs were sorted by FACS with surface marker CD133, or enriched by oncosphere formation. TIC self-renewal was examined by oncosphere formation and tumor initiation assay. Loss of function and gain of function assays were performed to examine the role of lncRNA. RNA pulldown, RNA immunoprecipitation, ChIP, western blot and double FISH were used explore the molecular mechanism of lncRNA. Results: We performed an unbiased screening for GPCR expression in liver cancers, and found GPR107 was the most highly expressed GPCR in liver cancer and liver TICs. GPR107 was essential for the self-renewal of liver TICs. The expression of GPR107 was regulated by a long noncoding RNA lncGPR107. LncGPR107 was also highly expressed in liver cancers and liver TICs. LncGPR107 drove the self-renewal of liver TICs through GPR107. Moreover, lncGPR107 recruited SRCAP complex to
GPR107 promoter to drive its transcriptional activation. LncGPR107 depletion inhibited the binding of SRCAP complex andGPR107 promoter and subsequent GPR107 expression. Moreover, LncGPR107-SRCAP-GPR107 can be targeted for liver TIC elimination. Conclusion: GPR107 was the most highly expressed GPCR in liver cancer and liver TICs. LncGPR107 participated in the transcriptional regulation of GPR107 in cis, through recruiting SRCAP remodeling complex toGPR107 promoter. This work revealed the important role of GPCR signaling in liver TIC self-renewal and added a new layer for liver TIC and GPCR regulation. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
41. scMuffin: an R package to disentangle solid tumor heterogeneity by single-cell gene expression analysis.
- Author
-
Nale, Valentina, Chiodi, Alice, Di Nanni, Noemi, Cifola, Ingrid, Moscatelli, Marco, Cocola, Cinzia, Gnocchi, Matteo, Piscitelli, Eleonora, Sula, Ada, Zucchi, Ileana, Reinbold, Rolland, Milanesi, Luciano, Mezzelani, Alessandra, Pelucchi, Paride, and Mosca, Ettore
- Subjects
GENE expression ,HETEROGENEITY ,USER interfaces ,TUMORS ,CANCER invasiveness - Abstract
Introduction: Single-cell (SC) gene expression analysis is crucial to dissect the complex cellular heterogeneity of solid tumors, which is one of the main obstacles for the development of effective cancer treatments. Such tumors typically contain a mixture of cells with aberrant genomic and transcriptomic profiles affecting specific sub-populations that might have a pivotal role in cancer progression, whose identification eludes bulk RNA-sequencing approaches. We present scMuffin, an R package that enables the characterization of cell identity in solid tumors on the basis of a various and complementary analyses on SC gene expression data. Results: scMuffin provides a series of functions to calculate qualitative and quantitative scores, such as: expression of marker sets for normal and tumor conditions, pathway activity, cell state trajectories, Copy Number Variations, transcriptional complexity and proliferation state. Thus, scMuffin facilitates the combination of various evidences that can be used to distinguish normal and tumoral cells, define cell identities, cluster cells in different ways, link genomic aberrations to phenotypes and identify subtle differences between cell subtypes or cell states. We analysed public SC expression datasets of human high-grade gliomas as a proof-of-concept to show the value of scMuffin and illustrate its user interface. Nevertheless, these analyses lead to interesting findings, which suggest that some chromosomal amplifications might underlie the invasive tumor phenotype and the presence of cells that possess tumor initiating cells characteristics. Conclusions: The analyses offered by scMuffin and the results achieved in the case study show that our tool helps addressing the main challenges in the bioinformatics analysis of SC expression data from solid tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
42. The role of the aryl hydrocarbon receptor in the development of cells with the molecular and functional characteristics of cancer stem-like cells
- Author
-
George J. Murphy, Elizabeth A. Stanford, Esther Landesman-Bollag, Stefano Monti, Brenden W. Smith, Olga Novikov, David H. Sherr, David C. Seldin, Zhongyan Wang, and Francesca Mulas
- Subjects
0301 basic medicine ,Physiology ,Cell ,Sox2 ,Breast Neoplasms ,Plant Science ,Tumor initiation ,Environment ,Tumor initiating cells ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,SOX2 ,Structural Biology ,Cancer stem cell ,Cell Movement ,Cell Line, Tumor ,medicine ,Humans ,Breast ,Ecology, Evolution, Behavior and Systematics ,Aryl hydrocarbon receptor ,Agricultural and Biological Sciences(all) ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,Cancer ,Cell Biology ,respiratory system ,medicine.disease ,respiratory tract diseases ,3. Good health ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Receptors, Aryl Hydrocarbon ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,biology.protein ,Neoplastic Stem Cells ,Female ,Stem cell ,Signal transduction ,General Agricultural and Biological Sciences ,Developmental Biology ,Biotechnology ,Research Article - Abstract
Background Self-renewing, chemoresistant breast cancer stem cells are believed to contribute significantly to cancer invasion, migration and patient relapse. Therefore, the identification of signaling pathways that regulate the acquisition of stem-like qualities is an important step towards understanding why patients relapse and towards development of novel therapeutics that specifically target cancer stem cell vulnerabilities. Recent studies identified a role for the aryl hydrocarbon receptor (AHR), an environmental carcinogen receptor implicated in cancer initiation, in normal tissue-specific stem cell self-renewal. These studies inspired the hypothesis that the AHR plays a role in the acquisition of cancer stem cell-like qualities. Results To test this hypothesis, AHR activity in Hs578T triple negative and SUM149 inflammatory breast cancer cells were modulated with AHR ligands, shRNA or AHR-specific inhibitors, and phenotypic, genomic and functional stem cell-associated characteristics were evaluated. The data demonstrate that (1) ALDHhigh cells express elevated levels of Ahr and Cyp1b1 and Cyp1a1, AHR-driven genes, (2) AHR knockdown reduces ALDH activity by 80 %, (3) AHR hyper-activation with several ligands, including environmental ligands, significantly increases ALDH1 activity, expression of stem cell- and invasion/migration-associated genes, and accelerates cell migration, (4) a significant correlation between Ahr or Cyp1b1 expression (as a surrogate marker for AHR activity) and expression of stem cell- and invasion/migration-associated gene sets is seen with genomic data obtained from 79 human breast cancer cell lines and over 1,850 primary human breast cancers, (5) the AHR interacts directly with Sox2, a master regulator of self-renewal; AHR ligands increase this interaction and nuclear SOX2 translocation, (6) AHR knockdown inhibits tumorsphere formation in low adherence conditions, (7) AHR inhibition blocks the rapid migration of ALDHhigh cells and reduces ALDHhigh cell chemoresistance, (8) ALDHhigh cells are highly efficient at initiating tumors in orthotopic xenografts, and (9) AHR knockdown inhibits tumor initiation and reduces tumor Aldh1a1, Sox2, and Cyp1b1 expression in vivo. Conclusions These data suggest that the AHR plays an important role in development of cells with cancer stem cell-like qualities and that environmental AHR ligands may exacerbate breast cancer by enhancing expression of these properties. Electronic supplementary material The online version of this article (doi:10.1186/s12915-016-0240-y) contains supplementary material, which is available to authorized users.
- Published
- 2016
43. Role of SH3GLB1 in the regulation of CD133 expression in GBM cells.
- Author
-
Chien, Chia-Hung, Lai, Chien-Cheng, Chuang, Jian-Ying, Chu, Jui-Mei, Liu, Chan-Chuan, and Chang, Kwang-Yu
- Subjects
RNA interference ,GLIOBLASTOMA multiforme ,BRAIN tumors ,OXIDATIVE phosphorylation ,SURVIVAL rate ,METHYLGUANINE - Abstract
Background: Glioblastoma (GBM), a malignant brain tumor, has poor survival outcomes due to recurrence or drug resistance. We found that SH3GLB1 is a crucial factor for cells to evade temozolomide (TMZ) cytotoxicity through autophagy-mediated oxidative phosphorylation, which is associated with CD133 levels. Therefore, we propose that SH3GLB1 participate in the impact on tumor-initiating cells (TICs). Methods: The parental, the derived resistant cell lines and their CD133
+ cells were used, and the levels of the proteins were compared by western blotting. Then RNA interference was applied to observe the effects of the target protein on TIC-related features. Finally, in vitro transcription assays were used to validate the association between SH3GLB1 and CD133. Results: The CD133+ cells from resistant cells with enhanced SH3GLB1 levels more easily survived cytotoxic treatment than those from the parental cells. Inhibition of SH3GLB1 attenuated frequency and size of spheroid formation, and the levels of CD133 and histone 4 lysine 5 (H4K5) acetylation can be simultaneously regulated by SH3GLB1 modification. The H4K5 acetylation of the CD133 promoter was later suggested to be the mediating mechanism of SH3GLB1. Conclusions: These data indicate that SH3GLB1 can regulate CD133 expression, suggesting that the protein plays a crucial role in TICs. Our findings on the effects of SH3GLB1 on the cells will help explain tumor resistance formation. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
44. TSPO acts as an immune resistance gene involved in the T cell mediated immune control of glioblastoma.
- Author
-
Menevse, Ayse N., Ammer, Laura-Marie, Vollmann-Zwerenz, Arabel, Kupczyk, Marcell, Lorenz, Julia, Weidner, Lorraine, Hussein, Abir, Sax, Julian, Mühlbauer, Jasmin, Heuschneider, Nicole, Rohrmus, Celine, Mai, Laura S., Jachnik, Birgit, Stamova, Slava, Volpin, Valentina, Durst, Franziska C., Sorrentino, Antonio, Xydia, Maria, Milenkovic, Vladimir M., and Bader, Stefanie
- Subjects
CYTOTOXIC T cells ,T cells ,TUMOR-infiltrating immune cells ,TRANSLOCATOR proteins ,GENE expression ,CELL-mediated cytotoxicity ,GLIOBLASTOMA multiforme ,T cell receptors - Abstract
Glioblastoma (GB) IDH-wildtype is the most malignant primary brain tumor. It is particularly resistant to current immunotherapies. Translocator protein 18 kDa (TSPO) is upregulated in GB and correlates with malignancy and poor prognosis, but also with increased immune infiltration. Here, we studied the role of TSPO in the regulation of immune resistance of human GB cells. The role of TSPO in tumor immune resistance was experimentally determined in primary brain tumor initiating cells (BTICs) and cell lines through genetic manipulation of TSPO expression and subsequent cocultures with antigen specific cytotoxic T cells and autologous tumor-infiltrating T cells. Death inducing intrinsic and extrinsic apoptotic pathways affected by TSPO were investigated. TSPO-regulated genes mediating apoptosis resistance in BTICs were identified through gene expression analysis and subsequent functional analyses. TSPO transcription in primary GB cells correlated with CD8
+ T cell infiltration, cytotoxic activity of T cell infiltrate, expression of TNFR and IFNGR and with the activity of their downstream signalling pathways, as well as with the expression of TRAIL receptors. Coculture of BTICs with tumor reactive cytotoxic T cells or with T cell-derived factors induced TSPO up-regulation through T cell derived TNFα and IFNγ. Silencing of TSPO sensitized BTICs against T cell-mediated cytotoxicity. TSPO selectively protected BTICs against TRAIL-induced apoptosis by regulating apoptosis pathways. TSPO also regulated the expression of multiple genes associated with resistance against apoptosis. We conclude that TSPO expression in GB is induced through T cell-derived cytokines TNFα and IFNγ and that TSPO expression protects GB cells against cytotoxic T cell attack through TRAIL. Our data thereby provide an indication that therapeutic targeting of TSPO may be a suitable approach to sensitize GB to immune cell-mediated cytotoxicity by circumventing tumor intrinsic TRAIL resistance. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
45. Phase 2 study of AV-GBM-1 (a tumor-initiating cell targeted dendritic cell vaccine) in newly diagnosed Glioblastoma patients: safety and efficacy assessment.
- Author
-
Bota, Daniela A., Taylor, Thomas H., Piccioni, David E., Duma, Christopher M., LaRocca, Renato V., Kesari, Santosh, Carrillo, Jose A., Abedi, Mehrdad, Aiken, Robert D., Hsu, Frank P. K., Kong, Xiao-Tang, Hsieh, Candace, Bota, Peter G., Nistor, Gabriel I., Keirstead, Hans S., and Dillman, Robert O.
- Subjects
DENDRITIC cells ,GRANULOCYTE-macrophage colony-stimulating factor ,GLIOBLASTOMA multiforme ,RADIOTHERAPY ,PATIENT safety ,DIAGNOSIS - Abstract
Background: Vaccine immunotherapy may improve survival in Glioblastoma (GBM). A multicenter phase II trial was designed to determine: (1) the success rate of manufacturing the Aivita GBM vaccine (AV-GBM-1), (2) Adverse Events (AE) associated with AV-GBM-1 administration, and (3) survival. Methods: Fresh suspected glioblastoma tissue was collected during surgery, and patients with pathology-confirmed GBM enrolled before starting concurrent Radiation Therapy and Temozolomide (RT/TMZ) with Intent to Treat (ITT) after recovery from RT/TMZ. AV-GBM-1 was made by incubating autologous dendritic cells with a lysate of irradiated autologous Tumor-Initiating Cells (TICs). Eligible patients were adults (18 to 70 years old) with a Karnofsky Performance Score (KPS) of 70 or greater, a successful TIC culture, and sufficient monocytes collected. A cryopreserved AV-GBM-1 dose was thawed and admixed with 500 μg of Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) before every subcutaneous (s.c.) administration. Results: Success rates were 97% for both TIC production and monocyte collection. AV-GBM-1 was manufactured for 63/63 patients; 60 enrolled per ITT; 57 started AV-GBM-1. The most common AEs attributed to AV-GBM-1 were local injection site reactions (16%) and flu-like symptoms (10%). Treatment-emergent AEs included seizures (33%), headache (37%), and focal neurologic symptoms (28%). One patient discontinued AV-GBM-1 because of seizures. Median Progression-Free Survival (mPFS) and median Overall Survival (mOS) from ITT enrollment were 10.4 and 16.0 months, respectively. 2-year Overall Survival (OS) is 27%. Conclusions: AV-GBM-1 was reliably manufactured. Treatment was well-tolerated, but there were numerous treatment-emergent central nervous system AEs. mPFS was longer than historical benchmarks, though no mOS improvement was noted. Trial registration: NCT, NCT03400917, Registered 10 January 2018, [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
46. Small interfering RNA library screen identified polo-like kinase-1 (PLK1) as a potential therapeutic target for breast cancer that uniquely eliminates tumor-initiating cells.
- Author
-
Hu, Kaiji, Law, Jennifer H, Fotovati, Abbas, and Dunn, Sandra E
- Subjects
PROTEIN metabolism ,PROTEINS ,DOXORUBICIN ,HETEROCYCLIC compounds ,CELL cycle proteins ,SMALL interfering RNA ,ANTINEOPLASTIC agents ,APOPTOSIS ,CELL physiology ,FLUOROURACIL ,GENE expression ,DRUG therapy ,STEM cells ,TRANSFERASES ,CELLS ,GENES ,RESEARCH funding ,GENETIC techniques ,PACLITAXEL ,BREAST tumors ,ANTIGENS ,PHARMACODYNAMICS ,CHEMICAL inhibitors - Abstract
Introduction: Triple-negative breast cancer (TNBC) high rate of relapse is thought to be due to the presence of tumor-initiating cells (TICs), molecularly defined as being CD44high/CD24-/low. TICs are resilient to chemotherapy and radiation. However, no currently accepted molecular target exists against TNBC and, moreover, TICs. Therefore, we sought the identification of kinase targets that inhibit TNBC growth and eliminate TICs.Methods: A genome-wide human kinase small interfering RNA (siRNA) library (691 kinases) was screened against the TNBC cell line SUM149 for growth inhibition. Selected siRNAs were then tested on four different breast cancer cell lines to confirm the spectrum of activity. Their effect on the CD44high subpopulation and sorted CD44high/CD24-/low cells of SUM149 also was studied. Further studies were focused on polo-like kinase 1 (PLK1), including its expression in breast cancer cell lines, effect on the CD44high/CD24-/low TIC subpopulation, growth inhibition, mammosphere formation, and apoptosis, as well as the activity of the PLK1 inhibitor, BI 2536.Results: Of the 85 kinases identified in the screen, 28 of them were further silenced by siRNAs on MDA-MB-231 (TNBC), BT474-M1 (ER+/HER2+, a metastatic variant), and HR5 (ER+/HER2+, a trastuzumab-resistant model) cells and showed a broad spectrum of growth inhibition. Importantly, 12 of 28 kinases also reduced the CD44high subpopulation compared with control in SUM149. Further tests of these 12 kinases directly on a sorted CD44high/CD24-/low TIC subpopulation of SUM149 cells confirmed their effect. Blocking PLK1 had the greatest growth inhibition on breast cancer cells and TICs by about 80% to 90% after 72 hours. PLK1 was universally expressed in breast cancer cell lines, representing all of the breast cancer subtypes, and was positively correlated to CD44. The PLK1 inhibitor BI 2536 showed similar effects on growth, mammosphere formation, and apoptosis as did PLK1 siRNAs. Finally, whereas paclitaxel, doxorubicin, and 5-fluorouracil enriched the CD44high/CD24-/low population compared with control in SUM149, subsequent treatment with BI 2536 killed the emergent population, suggesting that it could potentially be used to prevent relapse.Conclusion: Inhibiting PLK1 with siRNA or BI 2536 blocked growth of TNBCs including the CD44high/CD24-/low TIC subpopulation and mammosphere formation. Thus, PLK1 could be a potential therapeutic target for the treatment of TNBC as well as other subtypes of breast cancer. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
47. Generation of tumor-initiating cells by exogenous delivery of OCT4 transcription factor.
- Author
-
Beltran, Adriana S, Rivenbark, Ashley G, Richardson, Bryan T, Yuan, Xinni, Quian, Haili, Hunt, John P, Zimmerman, Eric, Graves, Lee M, and Blancafort, Pilar
- Abstract
Introduction: Tumor-initiating cells (TIC) are being extensively studied for their role in tumor etiology, maintenance and resistance to treatment. The isolation of TICs has been limited by the scarcity of this population in the tissue of origin and because the molecular signatures that characterize these cells are not well understood. Herein, we describe the generation of TIC-like cell lines by ectopic expression of the OCT4 transcription factor (TF) in primary breast cell preparations.Methods: OCT4 cDNA was over-expressed in four different primary human mammary epithelial (HMEC) breast cell preparations from reduction mammoplasty donors. OCT4-transduced breast cells (OTBCs) generated colonies (frequency ~0.01%) in self-renewal conditions (feeder cultures in human embryonic stem cell media). Differentiation assays, immunofluorescence, immunohistochemistry, and flow cytometry were performed to investigate the cell of origin of OTBCs. Serial dilutions of OTBCs were injected in nude mice to address their tumorigenic capabilities. Gene expression microarrays were performed in OTBCs, and the role of downstream targets of OCT4 in maintaining self-renewal was investigated by knock-down experiments.Results: OTBCs overcame senescence, overexpressed telomerase, and down-regulated p16INK4A. In differentiation conditions, OTBCs generated populations of both myoepithelial and luminal cells at low frequency, suggesting that the cell of origin of some OTBCs was a bi-potent stem cell. Injection of OTBCs in nude mice generated poorly differentiated breast carcinomas with colonization capabilities. Gene expression microarrays of OTBC lines revealed a gene signature that was over-represented in the claudin-low molecular subtype of breast cancer. Lastly, siRNA-mediated knockdown of OCT4 or downstream embryonic targets of OCT4, such as NANOG and ZIC1, suppressed the ability of OTBCs to self-renew.Conclusions: Transduction of OCT4 in normal breast preparations led to the generation of cell lines possessing tumor-initiating and colonization capabilities. These cells developed high-grade, poorly differentiated breast carcinomas in nude mice. Genome-wide analysis of OTBCs outlined an embryonic TF circuitry that could be operative in TICs, resulting in up-regulation of oncogenes and loss of tumor suppressive functions. These OTBCs represent a patient-specific model system for the discovery of novel oncogenic targets in claudin-low tumors. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
48. The emerging role of histone lysine demethylases in prostate cancer
- Author
-
Antonello Mai, Cheryl D. Helgason, Francesco Crea, Romano Danesi, Yan Ting Chiang, Lei Sun, and William L. Farrar
- Subjects
Male ,Cancer Research ,genetics/metabolism ,Review ,urologic and male genital diseases ,Epigenesis, Genetic ,0302 clinical medicine ,Histone demethylation ,Histone methylation ,androgen receptor ,epigenetics ,histone demethylase ,prostate cancer ,tumor-initiating cells ,Enzyme Inhibitors ,Tumor Markers ,Regulation of gene expression ,Histone demethylase ,Histone Demethylases ,0303 health sciences ,Prostate cancer ,biology ,Tumor-initiating cells ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Prognosis ,3. Good health ,Gene Expression Regulation, Neoplastic ,Androgen receptor ,Histone ,Oncology ,030220 oncology & carcinogenesis ,Histone methyltransferase ,Molecular Medicine ,Epigenetics ,drug therapy/enzymology/genetics ,lcsh:RC254-282 ,antagonists /&/ inhibitors/genetics/metabolism ,03 medical and health sciences ,Genetic ,Biomarkers, Tumor ,chemistry/pharmacology/therapeutic use, Epigenesis ,Genetic, Gene Expression Regulation ,Neoplastic, Histone Demethylases ,antagonists /&/ inhibitors/genetics/metabolism, Humans, Male, Prognosis, Prostatic Neoplasms ,drug therapy/enzymology/genetics, Tumor Markers ,Biological ,Humans ,chemistry/pharmacology/therapeutic use ,030304 developmental biology ,Neoplastic ,Prostatic Neoplasms ,KDM1A ,Gene Expression Regulation ,Cancer research ,biology.protein ,Epigenesis - Abstract
Early prostate cancer (PCa) is generally treatable and associated with good prognosis. After a variable time, PCa evolves into a highly metastatic and treatment-refractory disease: castration-resistant PCa (CRPC). Currently, few prognostic factors are available to predict the emergence of CRPC, and no curative option is available. Epigenetic gene regulation has been shown to trigger PCa metastasis and androgen-independence. Most epigenetic studies have focused on DNA and histone methyltransferases. While DNA methylation leads to gene silencing, histone methylation can trigger gene activation or inactivation, depending on the target amino acid residues and the extent of methylation (me1, me2, or me3). Interestingly, some histone modifiers are essential for PCa tumor-initiating cell (TIC) self-renewal. TICs are considered the seeds responsible for metastatic spreading and androgen-independence. Histone Lysine Demethylases (KDMs) are a novel class of epigenetic enzymes which can remove both repressive and activating histone marks. KDMs are currently grouped into 7 major classes, each one targeting a specific methylation site. Since their discovery, KDM expression has been found to be deregulated in several neoplasms. In PCa, KDMs may act as either tumor suppressors or oncogenes, depending on their gene regulatory function. For example, KDM1A and KDM4C are essential for PCa androgen-dependent proliferation, while PHF8 is involved in PCa migration and invasion. Interestingly, the possibility of pharmacologically targeting KDMs has been demonstrated. In the present paper, we summarize the emerging role of KDMs in regulating the metastatic potential and androgen-dependence of PCa. In addition, we speculate on the possible interaction between KDMs and other epigenetic effectors relevant for PCa TICs. Finally, we explore the role of KDMs as novel prognostic factors and therapeutic targets. We believe that studies on histone demethylation may add a novel perspective in our efforts to prevent and cure advanced PCa.
- Published
- 2012
49. "A novel in vivo model for the study of human breast cancer metastasis using primary breast tumor-initiating cells from patient biopsies".
- Author
-
Marsden, Carolyn G, Wright, Mary Jo, Carrier, Latonya, Moroz, Krzysztof, Pochampally, Radhika, and Rowan, Brian G
- Abstract
Background: The study of breast cancer metastasis depends on the use of established breast cancer cell lines that do not accurately represent the heterogeneity and complexity of human breast tumors. A tumor model was developed using primary breast tumor-initiating cells isolated from patient core biopsies that would more accurately reflect human breast cancer metastasis.Methods: Tumorspheres were isolated under serum-free culture conditions from core biopsies collected from five patients with clinical diagnosis of invasive ductal carcinoma (IDC). Isolated tumorspheres were transplanted into the mammary fat pad of NUDE mice to establish tumorigenicity in vivo. Tumors and metastatic lesions were analyzed by hematoxylin and eosin (H+E) staining and immunohistochemistry (IHC).Results: Tumorspheres were successfully isolated from all patient core biopsies, independent of the estrogen receptor α (ERα)/progesterone receptor (PR)/Her2/neu status or tumor grade. Each tumorsphere was estimated to contain 50-100 cells. Transplantation of 50 tumorspheres (1-5 × 103 cells) in combination with Matrigel into the mammary fat pad of NUDE mice resulted in small, palpable tumors that were sustained up to 12 months post-injection. Tumors were serially transplanted three times by re-isolation of tumorspheres from the tumors and injection into the mammary fat pad of NUDE mice. At 3 months post-injection, micrometastases to the lung, liver, kidneys, brain and femur were detected by measuring content of human chromosome 17. Visible macrometastases were detected in the lung, liver and kidneys by 6 months post-injection. Primary tumors variably expressed cytokeratins, Her2/neu, cytoplasmic E-cadherin, nuclear β catenin and fibronectin but were negative for ERα and vimentin. In lung and liver metastases, variable redistribution of E-cadherin and β catenin to the membrane of tumor cells was observed. ERα was re-expressed in lung metastatic cells in two of five samples.Conclusions: Tumorspheres isolated under defined culture conditions from patient core biopsies were tumorigenic when transplanted into the mammary fat pad of NUDE mice, and metastasized to multiple mouse organs. Micrometastases in mouse organs demonstrated a dormancy period prior to outgrowth of macrometastases. The development of macrometastases with organ-specific phenotypic distinctions provides a superior model for the investigation of organ-specific effects on metastatic cancer cell survival and growth. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
50. Expression of Six1 in luminal breast cancers predicts poor prognosis and promotes increases in tumor initiating cells by activation of extracellular signal-regulated kinase and transforming growth factor-beta signaling pathways.
- Author
-
Iwanaga, Ritsuko, Wang, Chu-An, Micalizzi, Douglas S, Harrell, J Chuck, Jedlicka, Paul, Sartorius, Carol A, Kabos, Peter, Farabaugh, Susan M, Bradford, Andrew P, and Ford, Heide L
- Subjects
BREAST cancer prognosis ,GENETICS of breast cancer ,TRANSFORMING growth factors ,CELLULAR control mechanisms ,CANCER cells ,CELL populations ,GENE expression ,LABORATORY mice - Abstract
Introduction: Mammary-specific overexpression of Six1 in mice induces tumors that resemble human breast cancer, some having undergone epithelial to mesenchymal transition (EMT) and exhibiting stem/progenitor cell features. Six1 overexpression in human breast cancer cells promotes EMT and metastatic dissemination. We hypothesized that Six1 plays a role in the tumor initiating cell (TIC) population specifically in certain subtypes of breast cancer, and that by understanding its mechanism of action, we could potentially develop new means to target TICs.Methods: We examined gene expression datasets to determine the breast cancer subtypes with Six1 overexpression, and then examined its expression in the CD24low/CD44+ putative TIC population in human luminal breast cancers xenografted through mice and in luminal breast cancer cell lines. Six1 overexpression, or knockdown, was performed in different systems to examine how Six1 levels affect TIC characteristics, using gene expression and flow cytometric analysis, tumorsphere assays, and in vivo TIC assays in immunocompromised and immune-competent mice. We examined the molecular pathways by which Six1 influences TICs using genetic/inhibitor approaches in vitro and in vivo. Finally, we examined the expression of Six1 and phosphorylated extracellular signal-regulated kinase (p-ERK) in human breast cancers.Results: High levels of Six1 are associated with adverse outcomes in luminal breast cancers, particularly the luminal B subtype. Six1 levels are enriched in the CD24low/CD44+ TIC population in human luminal breast cancers xenografted through mice, and in tumorsphere cultures in MCF7 and T47D luminal breast cancer cells. When overexpressed in MCF7 cells, Six1expands the TIC population through activation of transforming growth factor-beta (TGF-β) and mitogen activated protein kinase (MEK)/ERK signaling. Inhibition of ERK signaling in MCF7-Six1 cells with MEK1/2 inhibitors, U0126 and AZD6244, restores the TIC population of luminal breast cancer cells back to that observed in control cells. Administration of AZD6244 dramatically inhibits tumor formation efficiency and metastasis in cells that express high levels of Six1 ectopically or endogenously. Finally, we demonstrate that Six1 significantly correlates with phosphorylated ERK in human breast cancers.Conclusions: Six1 plays an important role in the TIC population in luminal breast cancers and induces a TIC phenotype by enhancing both TGF-β and ERK signaling. MEK1/2 kinase inhibitors are potential candidates for targeting TICs in breast tumors. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.