1
|
Kato Y, Fukazawa T, Tanimoto K, Kanawa M, Kojima M, Saeki I, Kurihara S, Touge R, Hirohashi N, Okada S, Hiyama E. Achaete-scute family bHLH transcription factor 2 activation promotes hepatoblastoma progression. Cancer Sci 2024; 115:847-858. [PMID: 38183173 PMCID: PMC10921009 DOI: 10.1111/cas.16051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 11/16/2023] [Accepted: 12/08/2023] [Indexed: 01/07/2024] Open
Abstract
Achaete-scute family bHLH transcription factor 2 (ASCL2) is highly expressed in hepatoblastoma (HB) tissues, but its role remains unclear. Thus, biological changes in the HB cell line HepG2 in response to induced ASCL2 expression were assessed. ASCL2 expression was induced in HepG2 cells using the Tet-On 3G system, which includes doxycycline. Cell viability, proliferation activity, mobility, and stemness were evaluated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, colony-formation, migration, invasion, and sphere-formation assays. Quantitative reverse-transcription polymerase chain reaction was used to assess the expression of markers for proliferation (CCND1 and MYC), epithelial-mesenchymal transition (EMT; SNAI1, TWIST1, and ZEB1), mesenchymal-epithelial transition (CDH1), and stemness (KLF4, POU5F1, and SOX9). Compared with the non-induced HepG2 cells, cells with induced ASCL2 expression showed significant increases in viability, colony number, migration area (%), and sphere number on days 7, 14, 8, and 7, respectively, and invasion area (%) after 90 h. Furthermore, induction of ASCL2 expression significantly upregulated CCND1, MYC, POU5F1, SOX9, and KLF4 expression on days 2, 2, 3, 3, and 5, respectively, and increased the ratios of SNAI1, TWIST1, and ZEB1 to CDH1 on day 5. ASCL2 promoted the formation of malignant phenotypes in HepG2 cells, which may be correlated with the upregulation of the Wnt signaling pathway-, EMT-, and stemness-related genes. ASCL2 activation may therefore be involved in the progression of HB.
Collapse
Affiliation(s)
- Yutaka Kato
- Department of Pediatrics, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Takahiro Fukazawa
- Natural Science Center for Basic Research and DevelopmentHiroshima UniversityHiroshimaJapan
- Division of Medical Research Support, Advanced Research Support CenterEhime UniversityToonJapan
| | - Keiji Tanimoto
- Department of Radiation Disaster Medicine, Research Institute for Radiation Biology and MedicineHiroshima UniversityHiroshimaJapan
| | - Masami Kanawa
- Natural Science Center for Basic Research and DevelopmentHiroshima UniversityHiroshimaJapan
| | - Masato Kojima
- Natural Science Center for Basic Research and DevelopmentHiroshima UniversityHiroshimaJapan
- Department of Surgery, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
- Department of Pediatric SurgeryHiroshima University HospitalHiroshimaJapan
| | - Isamu Saeki
- Department of Surgery, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
- Department of Pediatric SurgeryHiroshima University HospitalHiroshimaJapan
| | - Sho Kurihara
- Department of Surgery, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
- Department of Pediatric SurgeryHiroshima University HospitalHiroshimaJapan
| | - Ryo Touge
- Department of Surgery, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
- Department of Pediatric SurgeryHiroshima University HospitalHiroshimaJapan
| | - Nobuyuki Hirohashi
- Department of Radiation Disaster Medicine, Research Institute for Radiation Biology and MedicineHiroshima UniversityHiroshimaJapan
| | - Satoshi Okada
- Department of Pediatrics, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Eiso Hiyama
- Natural Science Center for Basic Research and DevelopmentHiroshima UniversityHiroshimaJapan
- Department of Surgery, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
- Department of Pediatric SurgeryHiroshima University HospitalHiroshimaJapan
| |
Collapse
|
2
|
Shibahara Y, Espin-Garcia O, Conner J, Weiss J, Derouet M, Allen J, Allison F, Kalimuthu S, Yeung JC, Darling GE. Intestinal Stem Cell Marker ASCL2 is a Novel Prognostic Predictor in Esophageal Adenocarcinoma. Cureus 2022; 14:e21021. [PMID: 35154991 PMCID: PMC8818334 DOI: 10.7759/cureus.21021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2022] [Indexed: 11/25/2022] Open
Abstract
Purpose Intestinal stem cell markers play a significant role in esophageal adenocarcinoma carcinogenesis via Barrett’s esophagus; however, its utility as a prognostic biomarker has not been established. Methods We analyzed the immunohistochemical expression of intestinal stem cell markers, ASCL2 and LGR5, using whole slides (35 cases) and tissue microarray (TMA; 64 cases). On TMA slides, adjacent normal squamous epithelium, metaplastic glandular epithelium (Barrett's esophagus), and dysplastic glandular epithelium were inserted when applicable. Two pathologists semi-quantitatively scored stained slides independently, and the results were correlated with clinicopathologic factors and outcomes. Results In whole slides, 51% and 57% expressed high ASCL2 and high LGR5; in TMA, 69% and 88% expressed high ASCL2 and high LGR5, respectively. In TMA, high ASCL2 and low LGR5 expression significantly correlated to a higher number of involved lymph nodes (p=0.027 and p=0.0039), and LGR5 expression significantly correlated to the pathological stage (p=0.0032). Kaplan-Meier analysis showed a negative impact of high ASCL2 expression on overall survival (OS; WS p=0.0168, TMA p=0.0276) as well as progression-free survival (PFS; WS p=0.000638, TMA p=0.0466) but not LGR5. Multivariate Cox regression analysis revealed that ASCL2 expression is an independent prognostic factor for esophageal adenocarcinoma (OS; WS p=0.25, TMA p=0.011. PFS; WS p=0.012, TMA p=0.038). Analysis of the TCGA dataset showed that ASCL2 mRNA levels were correlated to nodal status but not overall survival. Conclusion High expression of the intestinal stem cell marker ASCL2 may predict unfavorable outcomes in surgically resected esophageal adenocarcinoma.
Collapse
|
3
|
Standardization of esophageal adenocarcinoma in vitro model and its applicability for model drug testing. Sci Rep 2021; 11:6664. [PMID: 33758229 PMCID: PMC7988140 DOI: 10.1038/s41598-021-85530-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/25/2021] [Indexed: 01/11/2023] Open
Abstract
FLO-1 cell line represents an important tool in esophageal adenocarcinoma (EAC) research as a verified and authentic cell line to study the disease pathophysiology and antitumor drug screenings. Since in vitro characteristics of cells depend on the microenvironment and culturing conditions, we performed a thorough characterization of the FLO-1 cell line under different culturing conditions with the aim of (1) examining the effect of serum-free growth medium and air–liquid interface (A–L) culturing, which better reflect physiological conditions in vivo and (2) investigating the differentiation potential of FLO-1 cells to mimic the properties of the in vivo esophageal epithelium. Our study shows that the composition of the media influenced the morphological, ultrastructural and molecular characteristics of FLO-1 cells, such as the expression of junctional proteins. Importantly, FLO-1 cells formed spheres at the A–L interface, recapitulating key elements of tumors in the esophageal tube, i.e., direct contact with the gas phase and three-dimensional architecture. On the other hand, FLO-1 models exhibited high permeability to model drugs and zero permeability markers, and low transepithelial resistance, and therefore poorly mimicked normal esophageal epithelium. In conclusion, the identified effect of culture conditions on the characteristics of FLO-1 cells should be considered for standardization, data reproducibility and validity of the in vitro EAC model. Moreover, the sphere-forming ability of FLO-1 cells at the A–L interface should be considered in EAC tumor biology and anticancer drug studies as a reliable and straightforward model with the potential to increase the predictive efficiency of the current in vitro approaches.
Collapse
|
4
|
Markers of Stem Cells. Stem Cells 2021. [DOI: 10.1007/978-981-16-1638-9_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
5
|
Chowdhury S, Ghosh S. Sources, Isolation and culture of stem cells? Stem Cells 2021. [DOI: 10.1007/978-981-16-1638-9_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
6
|
Khosravi A, Jafari SM, Asadi J. Knockdown of TAZ decrease the cancer stem properties of ESCC cell line YM-1 by modulation of Nanog, OCT-4 and SOX2. Gene 2020; 769:145207. [PMID: 33031893 DOI: 10.1016/j.gene.2020.145207] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/07/2020] [Accepted: 09/29/2020] [Indexed: 12/23/2022]
Abstract
Cancer stem cells are a rare population in tumors with high metastatic potential and resistance to treatment. Recent strategies in cancer treatment have focused on targeting important signaling pathways that have an important role in maintaining CSC populations. TAZ (transcriptional co-activator with PDZ-binding motif) is a key downstream of the Hippo pathway which plays a fundamental role in the survival of CSCs from different origins, however, no data on the role of TAZ in esophageal cancer are available. Our findings showed that esophageal CSCs enriched from the YM-1 cell line have stemness properties. We found that TAZ was strongly expressed in esophageal CSCs and knockdown of TAZ in esophageal CSCs results in reduced colony formation and cell migration. Moreover, this data indicated that TAZ knockdown reduces the expression of SOX-2, OCT-4, and Nanong in esophageal CSCs. Taken together, the results of the current study suggested that TAZ has a crucial role in the biology of esophageal CSCs.
Collapse
Affiliation(s)
- Ayyoob Khosravi
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Department of Molecular Medicine, Faculty of Advanced Medical Technologies, Golestan University of Medical Sciences, Gorgan, Iran
| | - Seyyed Mehdi Jafari
- Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Department of Biochemistry and Biophysics, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Jahanbakhsh Asadi
- Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
7
|
Bigoni-Ordóñez GD, Czarnowski D, Parsons T, Madlambayan GJ, Villa-Diaz LG. Integrin α6 (CD49f), The Microenvironment and Cancer Stem Cells. Curr Stem Cell Res Ther 2019; 14:428-436. [PMID: 30280675 DOI: 10.2174/1574888x13666181002151330] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/20/2018] [Accepted: 09/02/2018] [Indexed: 12/16/2022]
Abstract
Cancer is a highly prevalent and potentially terminal disease that affects millions of individuals worldwide. Here, we review the literature exploring the intricacies of stem cells bearing tumorigenic characteristics and collect evidence demonstrating the importance of integrin α6 (ITGA6, also known as CD49f) in cancer stem cell (CSC) activity. ITGA6 is commonly used to identify CSC populations in various tissues and plays an important role sustaining the self-renewal of CSCs by interconnecting them with the tumorigenic microenvironment.
Collapse
Affiliation(s)
- Gabriele D Bigoni-Ordóñez
- Division de Investigacion Basica, Instituto Nacional de Cancerologia, Secretaria de Salud, Mexico City, Mexico.,Programa de Maestría y Doctorado en Ciencias Bioquímicas, Facultad de Química, UNAM, Mexico City, Mexico
| | - Daniel Czarnowski
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, United States
| | - Tyler Parsons
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, United States
| | - Gerard J Madlambayan
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, United States
| | - Luis G Villa-Diaz
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, United States
| |
Collapse
|
8
|
Bayat Mokhtari R, Baluch N, Morgatskaya E, Kumar S, Sparaneo A, Muscarella LA, Zhao S, Cheng HL, Das B, Yeger H. Human bronchial carcinoid tumor initiating cells are targeted by the combination of acetazolamide and sulforaphane. BMC Cancer 2019; 19:864. [PMID: 31470802 PMCID: PMC6716820 DOI: 10.1186/s12885-019-6018-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 08/06/2019] [Indexed: 12/19/2022] Open
Abstract
Background Bronchial carcinoids are neuroendocrine tumors that present as typical (TC) and atypical (AC) variants, the latter being more aggressive, invasive and metastatic. Studies of tumor initiating cell (TIC) biology in bronchial carcinoids has been hindered by the lack of appropriate in-vitro and xenograft models representing the bronchial carcinoid phenotype and behavior. Methods Bronchial carcinoid cell lines (H727, TC and H720, AC) were cultured in serum-free growth factor supplemented medium to form 3D spheroids and serially passaged up to the 3rd generation permitting expansion of the TIC population as verified by expression of stemness markers, clonogenicity in-vitro and tumorigenicity in both subcutaneous and orthotopic (lung) models. Acetazolamide (AZ), sulforaphane (SFN) and the AZ + SFN combination were evaluated for targeting TIC in bronchial carcinoids. Results Data demonstrate that bronchial carcinoid cell line 3rd generation spheroid cells show increased drug resistance, clonogenicity, and tumorigenic potential compared with the parental cells, suggesting selection and expansion of a TIC fraction. Gene expression and immunolabeling studies demonstrated that the TIC expressed stemness factors Oct-4, Sox-2 and Nanog. In a lung orthotopic model bronchial carcinoid, cell line derived spheroids, and patient tumor derived 3rd generation spheroids when supported by a stroma, showed robust tumor formation. SFN and especially the AZ + SFN combination were effective in inhibiting tumor cell growth, spheroid formation and in reducing tumor formation in immunocompromised mice. Conclusions Human bronchial carcinoid tumor cells serially passaged as spheroids contain a higher fraction of TIC exhibiting a stemness phenotype. This TIC population can be effectively targeted by the combination of AZ + SFN. Our work portends clinical relevance and supports the therapeutic use of the novel AZ+ SFN combination that may target the TIC population of bronchial carcinoids.
Collapse
Affiliation(s)
- Reza Bayat Mokhtari
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada. .,Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, Canada. .,The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay St., Rm 15.9714, Toronto, Ontario, M5G 0A4, Canada.
| | - Narges Baluch
- Department of Pediatrics, Queen's University, 76 Stuart St, Kingston, ON, K7L 2V7, Canada
| | - Evgeniya Morgatskaya
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sushil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Williams Science Hall 3035, Department of Pharmaceutical Sciences 601 S. Saddle Creek Rd, Omaha, NE, 68106, USA
| | - Angelo Sparaneo
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, viale Cappuccini, 71013, San Giovanni Rotondo, FG, Italy
| | - Lucia Anna Muscarella
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, viale Cappuccini, 71013, San Giovanni Rotondo, FG, Italy
| | - Sheyun Zhao
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Hai-Ling Cheng
- Institute of Biomaterials & Biomedical Engineering, University of Toronto, 164 College Street, Rosebrugh Building, Room 407, Toronto, ON, M5S 3G9, Canada
| | - Bikul Das
- Thoreau Laboratory for Global Health, M2D2, University of Massachusetts-Lowell, Innovation Hub, 110 Canal St, Lowell, MA, 01852, USA.,KaviKrishna Laboratory, Indian Institute of Technology Complex, Guwahati, India
| | - Herman Yeger
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay St., Rm 15.9714, Toronto, Ontario, M5G 0A4, Canada
| |
Collapse
|
9
|
Etiology, cancer stem cells and potential diagnostic biomarkers for esophageal cancer. Cancer Lett 2019; 458:21-28. [PMID: 31125642 DOI: 10.1016/j.canlet.2019.05.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/10/2019] [Accepted: 05/15/2019] [Indexed: 12/19/2022]
Abstract
Esophageal cancer (EC) has been a leading cause of cancer death worldwide in part due to late detection and lack of precision treatment. EC includes two major malignancies, esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC). Recent studies reveal that ESCC and EAC have distinct cell of origin and contain cancer stem cells (also known as tumor initiating cells) expressing different cell surface markers. These biomarkers have potentially important values for both early detection and finding effective therapy. In this review we summarize the updated findings for cell of origin and provide an overview of cancer cell biomarkers that have been tested for ESCC and EAC. In addition, we also discuss recent progress in the study of molecular mechanisms leading to these malignancies.
Collapse
|
10
|
Yang W, Han Y, Zhao X, Duan L, Zhou W, Wang X, Shi G, Che Y, Zhang Y, Liu J, Zhang H, Zhao Q, Hong L, Fan D. Advances in prognostic biomarkers for esophageal cancer. Expert Rev Mol Diagn 2018; 19:109-119. [PMID: 30582379 DOI: 10.1080/14737159.2019.1563485] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Wanli Yang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, China
| | - Yu Han
- Department of Otolaryngology, Xijing Hospital, Air Force Military Medical University, Xi’an, China
| | - Xinhui Zhao
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, China
| | - Lili Duan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, China
| | - Wei Zhou
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, China
| | - Xiaoqian Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, China
| | - Gaokai Shi
- The First Brigade of Student, Air Force Military Medical University, Xi’an, China
| | - Yinggang Che
- The First Brigade of Student, Air Force Military Medical University, Xi’an, China
| | - Yujie Zhang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, China
| | - Jinqiang Liu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, China
| | - Hongwei Zhang
- Department of Digestive Surgery, Xijing Hospital, Air Force Military Medical University, Xi’an, China
| | - Qingchuan Zhao
- Department of Digestive Surgery, Xijing Hospital, Air Force Military Medical University, Xi’an, China
| | - Liu Hong
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, China
| |
Collapse
|
11
|
Senthebane DA, Jonker T, Rowe A, Thomford NE, Munro D, Dandara C, Wonkam A, Govender D, Calder B, Soares NC, Blackburn JM, Parker MI, Dzobo K. The Role of Tumor Microenvironment in Chemoresistance: 3D Extracellular Matrices as Accomplices. Int J Mol Sci 2018; 19:E2861. [PMID: 30241395 PMCID: PMC6213202 DOI: 10.3390/ijms19102861] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The functional interplay between tumor cells and their adjacent stroma has been suggested to play crucial roles in the initiation and progression of tumors and the effectiveness of chemotherapy. The extracellular matrix (ECM), a complex network of extracellular proteins, provides both physical and chemicals cues necessary for cell proliferation, survival, and migration. Understanding how ECM composition and biomechanical properties affect cancer progression and response to chemotherapeutic drugs is vital to the development of targeted treatments. METHODS 3D cell-derived-ECMs and esophageal cancer cell lines were used as a model to investigate the effect of ECM proteins on esophageal cancer cell lines response to chemotherapeutics. Immunohistochemical and qRT-PCR evaluation of ECM proteins and integrin gene expression was done on clinical esophageal squamous cell carcinoma biopsies. Esophageal cancer cell lines (WHCO1, WHCO5, WHCO6, KYSE180, KYSE 450 and KYSE 520) were cultured on decellularised ECMs (fibroblasts-derived ECM; cancer cell-derived ECM; combinatorial-ECM) and treated with 0.1% Dimethyl sulfoxide (DMSO), 4.2 µM cisplatin, 3.5 µM 5-fluorouracil and 2.5 µM epirubicin for 24 h. Cell proliferation, cell cycle progression, colony formation, apoptosis, migration and activation of signaling pathways were used as our study endpoints. RESULTS The expression of collagens, fibronectin and laminins was significantly increased in esophageal squamous cell carcinomas (ESCC) tumor samples compared to the corresponding normal tissue. Decellularised ECMs abrogated the effect of drugs on cancer cell cycling, proliferation and reduced drug induced apoptosis by 20⁻60% that of those plated on plastic. The mitogen-activated protein kinase-extracellular signal-regulated kinase (MEK-ERK) and phosphoinositide 3-kinase-protein kinase B (PI3K/Akt) signaling pathways were upregulated in the presence of the ECMs. Furthermore, our data show that concomitant addition of chemotherapeutic drugs and the use of collagen- and fibronectin-deficient ECMs through siRNA inhibition synergistically increased cancer cell sensitivity to drugs by 30⁻50%, and reduced colony formation and cancer cell migration. CONCLUSION Our study shows that ECM proteins play a key role in the response of cancer cells to chemotherapy and suggest that targeting ECM proteins can be an effective therapeutic strategy against chemoresistant tumors.
Collapse
Affiliation(s)
- Dimakatso Alice Senthebane
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Campus, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Tina Jonker
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Campus, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Arielle Rowe
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Campus, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Nicholas Ekow Thomford
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Daniella Munro
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Collet Dandara
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Ambroise Wonkam
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Dhirendra Govender
- Division of Anatomical Pathology, Faculty of Health Sciences, University of Cape Town, NHLS-Groote Schuur Hospital, Cape Town 7925, South Africa.
| | - Bridget Calder
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.
| | - Nelson C Soares
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.
| | - Jonathan M Blackburn
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.
| | - M Iqbal Parker
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Kevin Dzobo
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Campus, Anzio Road, Observatory, Cape Town 7925, South Africa.
| |
Collapse
|
12
|
Whelan KA, Muir AB, Nakagawa H. Esophageal 3D Culture Systems as Modeling Tools in Esophageal Epithelial Pathobiology and Personalized Medicine. Cell Mol Gastroenterol Hepatol 2018; 5:461-478. [PMID: 29713660 PMCID: PMC5924738 DOI: 10.1016/j.jcmgh.2018.01.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 01/11/2018] [Indexed: 12/13/2022]
Abstract
The stratified squamous epithelium of the esophagus shows a proliferative basal layer of keratinocytes that undergo terminal differentiation in overlying suprabasal layers. Esophageal pathologies, including eosinophilic esophagitis, gastroesophageal reflux disease, Barrett's esophagus, squamous cell carcinoma, and adenocarcinoma, cause perturbations in the esophageal epithelial proliferation-differentiation gradient. Three-dimensional (3D) culture platforms mimicking in vivo esophageal epithelial tissue architecture ex vivo have emerged as powerful experimental tools for the investigation of esophageal biology in the context of homeostasis and pathology. Herein, we describe types of 3D culture that are used to model the esophagus, including organotypic, organoid, and spheroid culture systems. We discuss the development and optimization of various esophageal 3D culture models; highlight the applications, strengths, and limitations of each method; and summarize how these models have been used to evaluate the esophagus under homeostatic conditions as well as under the duress of inflammation and precancerous/cancerous conditions. Finally, we present future perspectives regarding the use of esophageal 3D models in basic science research as well as translational studies with the potential for personalized medicine.
Collapse
Key Words
- 3D, 3-dimensional
- BE, Barrett’s esophagus
- COX, cyclooxygenase
- CSC, cancer stem cell
- EADC, esophageal adenocarcinoma
- EGF, epidermal growth factor
- EGFR, epidermal growth factor receptor
- EMT, epithelial-mesenchymal transition
- ESCC, esophageal squamous cell carcinoma
- EoE, eosinophilic esophagitis
- Esophageal Disease
- FEF3, primary human fetal esophageal fibroblast
- GERD, gastroesophageal reflux disease
- OTC, organotypic 3-dimensional culture
- Organoid
- Organotypic Culture
- STAT3, signal transducer and activator of transcription-3
- Spheroid Culture
Collapse
Affiliation(s)
- Kelly A. Whelan
- Pathology and Laboratory Medicine, Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Amanda B. Muir
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Correspondence Address correspondence to: Amanda B. Muir, MD, Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Abramson Research Center 902E, Philadelphia, Pennsylvania 19103. fax: (267) 426–7814.
| | - Hiroshi Nakagawa
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
13
|
Abstract
Cancer stem cells (CSCs) are a subpopulation of cancer cells that have the ability to self-renew and to generate differentiated cells of various lineages. Due to their specific morphological and biological features, they are often resistant to therapy and in turn lead to metastasis and cancer recurrence. Because of their crucial roles in carcinogenesis and patient prognosis, identification and isolation of CSCs have become an important part of improved cancer management regime. Isolation, characterization, and development of targeted therapy against CSCs have potential efficacy in treating esophageal cancer. In addition, CSCs can act as a predictive tool for chemoradiotherapy response in esophageal adenocarcinoma. Different methods including functional assays, cell sorting using various intracellular, and cell surface markers and xenotransplantation techniques are used for the identification and separation of CSCs in different cancers. None of these methods solely can guarantee complete isolation of CSC population, thus a combination of methods could be used for reliable detection and isolation of CSCs. Here, we describe the identification and isolation of CSCs from esophageal adenocarcinoma cells by cell sorting after Hoechst 33342 staining followed by in vitro functional assays, and in vivo xenograft techniques.
Collapse
Affiliation(s)
- Farhadul Islam
- Cancer Molecular Pathology of School of Medicine, Griffith University, Gold Coast, Australia
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh
| | - Vinod Gopalan
- Cancer Molecular Pathology of School of Medicine, Griffith University, Gold Coast, Australia
| | - Alfred K Lam
- Cancer Molecular Pathology of School of Medicine, Griffith University, Gold Coast, Australia.
| |
Collapse
|
14
|
Krebsbach PH, Villa-Diaz LG. The Role of Integrin α6 (CD49f) in Stem Cells: More than a Conserved Biomarker. Stem Cells Dev 2017; 26:1090-1099. [PMID: 28494695 DOI: 10.1089/scd.2016.0319] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Stem cells have the capacity for self-renewal and differentiation into specialized cells that form and repopulated all tissues and organs, from conception to adult life. Depending on their capacity for differentiation, stem cells are classified as totipotent (ie, zygote), pluripotent (ie, embryonic stem cells), multipotent (ie, neuronal stem cells, hematopoietic stem cells, epithelial stem cells, etc.), and unipotent (ie, spermatogonial stem cells). Adult or tissue-specific stem cells reside in specific niches located in, or nearby, their organ or tissue of origin. There, they have microenvironmental support to remain quiescent, to proliferate as undifferentiated cells (self-renewal), and to differentiate into progenitors or terminally differentiated cells that migrate from the niche to perform specialized functions. The presence of proteins at the cell surface is often used to identify, classify, and isolate stem cells. Among the diverse groups of cell surface proteins used for these purposes, integrin α6, also known as CD49f, may be the only biomarker commonly found in more than 30 different populations of stem cells, including some cancer stem cells. This broad expression among stem cell populations indicates that integrin α6 may play an important and conserved role in stem cell biology, which is reaffirmed by recent demonstrations of its role maintaining self-renewal of pluripotent stem cells and breast and glioblastoma cancer stem cells. Therefore, this review intends to highlight and synthesize new findings on the importance of integrin α6 in stem cell biology.
Collapse
Affiliation(s)
- Paul H Krebsbach
- 1 School of Dentistry, University of California , Los Angeles, California
| | - Luis G Villa-Diaz
- 2 Department of Biological Sciences, Oakland University , Rochester, Michigan
| |
Collapse
|
15
|
Jiménez P, Chueca E, Arruebo M, Strunk M, Solanas E, Serrano T, García-González MA, Lanas Á. CD24 Expression Is Increased in 5-Fluorouracil-Treated Esophageal Adenocarcinoma Cells. Front Pharmacol 2017; 8:321. [PMID: 28611669 PMCID: PMC5447731 DOI: 10.3389/fphar.2017.00321] [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: 02/02/2017] [Accepted: 05/15/2017] [Indexed: 01/20/2023] Open
Abstract
The cancer stem cell (CSC) model suggests that there are subsets of cells within a tumor with increased proliferation and self-renewal capacity, which play a key role in therapeutic resistance. The importance of cyclooxygenase-2 (COX-2) in carcinogenesis has been previously established and the use of COX-2 inhibitors as celecoxib has been shown to exert antitumor effects. The present study investigated whether treatment of esophageal adenocarcinoma (EAC) cells with 5-fluorouracil (5-FU) or the growth of tumor spheres increased the proportion of CSCs and also if treatment with celecoxib was able to reduce the putative CSC markers in this tumor. OE19 and OE33 EAC cells surviving 5-FU exposure exhibited an increase in CSC markers CD24 and ABCG2 and also an increased resistance to apoptosis. EAC cell lines had the capacity to form multiple spheres displaying typical CSC functionalities such as self-renewal and increased CD24 levels. In addition, after the induction of differentiation, cancer cells reached levels of CD24 similar to those observed in the parental cells. Treatment with celecoxib alone or in combination with 5-FU also resulted in a reduction of CD24 expression. Moreover, celecoxib inhibited the growth of tumor spheres. These findings showing a reduction in CSC markers induced by celecoxib suggest that the COX-2 inhibitor might be a candidate for combined chemotherapy in the treatment of EAC. However, additional clinical and experimental studies are needed.
Collapse
Affiliation(s)
| | - Eduardo Chueca
- CIBERehdMadrid, Spain.,Instituto de Investigación Sanitaria Aragón (IIS Aragón)Zaragoza, Spain
| | - María Arruebo
- Instituto de Investigación Sanitaria Aragón (IIS Aragón)Zaragoza, Spain
| | - Mark Strunk
- Centro de Investigación Biomédica de Aragón, IACS Aragón, Instituto Aragonés de Ciencias de la Salud, Servicio de Secuenciación y Genómica FuncionalZaragoza, Spain
| | - Estela Solanas
- Instituto de Investigación Sanitaria Aragón (IIS Aragón)Zaragoza, Spain
| | - Trinidad Serrano
- Instituto de Investigación Sanitaria Aragón (IIS Aragón)Zaragoza, Spain.,Department of Gastroenterology, Hospital Clínico Universitario Lozano BlesaZaragoza, Spain
| | - María A García-González
- CIBERehdMadrid, Spain.,Instituto de Investigación Sanitaria Aragón (IIS Aragón)Zaragoza, Spain.,Instituto Aragonés de Ciencias de la Salud (IACS)Zaragoza, Spain
| | - Ángel Lanas
- CIBERehdMadrid, Spain.,Instituto de Investigación Sanitaria Aragón (IIS Aragón)Zaragoza, Spain.,Department of Gastroenterology, Hospital Clínico Universitario Lozano BlesaZaragoza, Spain.,Department of Medicine, University of ZaragozaZaragoza, Spain
| |
Collapse
|
16
|
A novel intraperitoneal metastatic xenograft mouse model for survival outcome assessment of esophageal adenocarcinoma. PLoS One 2017; 12:e0171824. [PMID: 28225784 PMCID: PMC5321464 DOI: 10.1371/journal.pone.0171824] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 01/26/2017] [Indexed: 01/15/2023] Open
Abstract
Esophageal adenocarcinoma (EAC) has become the dominant type of esophageal cancer in United States. The 5-year survival rate of EAC is below 20% and most patients present with locally advanced or widespread metastatic disease, where current treatment is largely ineffective. Therefore, new therapeutic approaches are urgently needed. Improvement of EAC patient outcome requires well-characterized animal models in which to evaluate novel therapeutics. In this study we aimed to establish a peritoneal dissemination xenograft mouse model of EAC that would support survival outcome analyses. To find the best candidate cell line from 7 human EAC cell lines of different origin named ESO26, OE33, ESO51, SK-GT-2, OE19, OACM5.1C and Flo-1 were injected intraperitoneally/subcutaneously into SCID mice. The peritoneal/xenograft tumor formation and mouse survival were compared among different groups. All cell lines injected subcutaneously formed tumors within 3 months at variable rates. All cell lines except OACM5.1C formed intraperitoneal tumors within 3 months at variable rates. Median animal survival with peritoneal dissemination was 108 days for ESO26 cells (5X106), 65 days for OE33 cells (5X106), 88 days for ESO51 cells (5X106), 76 days for SK-GT-2 cells (5X106), 55 days for OE19 cells (5X106), 45 days for OE19 cells (10X106) and 82 days for Flo-1 cells (5X106). Interestingly, only in the OE19 model all mice (7/7 for 5X106 and 5/5 for10X106) developed bloody ascites with liver metastasis after intraperitoneal injection. The median survival time of these animals was the shortest (45 days for 10X106 cells). In addition, median survival was significantly increased after paclitaxel treatment compared with the control group (57 days versus 45 days, p = 0.0034) along with a significant decrease of the relative subcutaneous tumor volume (p = 0.00011). Thus peritoneal dissemination mouse xenograft model for survival outcome assessment after intraperitoneal injection of OE19 cells will be very useful for the evaluation of cancer therapeutics.
Collapse
|
17
|
Dynamic Microenvironment Induces Phenotypic Plasticity of Esophageal Cancer Cells Under Flow. Sci Rep 2016; 6:38221. [PMID: 27910892 PMCID: PMC5133540 DOI: 10.1038/srep38221] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 11/07/2016] [Indexed: 12/29/2022] Open
Abstract
Cancer microenvironment is a remarkably heterogeneous composition of cellular and non-cellular components, regulated by both external and intrinsic physical and chemical stimuli. Physical alterations driven by increased proliferation of neoplastic cells and angiogenesis in the cancer microenvironment result in the exposure of the cancer cells to elevated levels of flow-based shear stress. We developed a dynamic microfluidic cell culture platform utilizing eshopagael cancer cells as model cells to investigate the phenotypic changes of cancer cells upon exposure to fluid shear stress. We report the epithelial to hybrid epithelial/mesenchymal transition as a result of decreasing E-Cadherin and increasing N-Cadherin and vimentin expressions, higher clonogenicity and ALDH positive expression of cancer cells cultured in a dynamic microfluidic chip under laminar flow compared to the static culture condition. We also sought regulation of chemotherapeutics in cancer microenvironment towards phenotypic control of cancer cells. Such in vitro microfluidic system could potentially be used to monitor how the interstitial fluid dynamics affect cancer microenvironment and plasticity on a simple, highly controllable and inexpensive bioengineered platform.
Collapse
|
18
|
Dai JY, Tapsoba JDD, Buas MF, Risch HA, Vaughan TL. Constrained Score Statistics Identify Genetic Variants Interacting with Multiple Risk Factors in Barrett's Esophagus. Am J Hum Genet 2016; 99:352-65. [PMID: 27486777 DOI: 10.1016/j.ajhg.2016.06.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 06/20/2016] [Indexed: 12/25/2022] Open
Abstract
Few gene-environment interactions (G × E) have been discovered in cancer epidemiology thus far, in part due to the large number of possible G × E to be investigated and inherent low statistical power of traditional analytic methods for discovering G × E. We consider simultaneously testing for interactions between several related exposures and a genetic variant in a genome-wide study. To improve power, constrained testing strategies are proposed for multivariate gene-environment interactions at two levels: interactions that have the same direction (one-sided or bidirectional hypotheses) or are proportional to respective exposure main effects (a variant of Tukey's one-degree test). Score statistics were developed to expedite the genome-wide computation. We conducted extensive simulations to evaluate validity and power performance of the proposed statistics, applied them to the genetic and environmental exposure data for esophageal adenocarcinoma and Barrett's esophagus from the Barretts Esophagus and Esophageal Adenocarcinoma Consortium (BEACON), and discovered three loci simultaneously interacting with gastresophageal reflux, obesity, and tobacco smoking with genome-wide significance. These findings deepen understanding of the genetic and environmental architecture of Barrett's esophagus and esophageal adenocarcinoma.
Collapse
Affiliation(s)
- James Y Dai
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Biostatistics, University of Washington, Seattle, WA 98195, USA.
| | - Jean de Dieu Tapsoba
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Matthew F Buas
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Harvey A Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT 06520, USA
| | - Thomas L Vaughan
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Epidemiology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
19
|
Xu DD, Zhou PJ, Wang Y, Zhang L, Fu WY, Ruan BB, Xu HP, Hu CZ, Tian L, Qin JH, Wang S, Wang X, Li YC, Liu QY, Ren Z, Zhang R, Wang YF. Reciprocal activation between STAT3 and miR-181b regulates the proliferation of esophageal cancer stem-like cells via the CYLD pathway. Mol Cancer 2016; 15:40. [PMID: 27189061 PMCID: PMC4869372 DOI: 10.1186/s12943-016-0521-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/05/2016] [Indexed: 02/07/2023] Open
Abstract
Background Recent studies have suggested that cancer cells contain subpopulations that can initiate tumor growth, self-renew, and maintain tumor cell growth. However, for esophageal cancer cells, the relationship between STAT3, microRNAs and cancer stem cells remains unclear. Methods Serum-free culture was used to enrich esophageal cancer stem-like cells (ECSLC). Flow cytometry determined the proportion of ECSLC. qPCR were performed to examine expression level of stemness factors, mesenchymal markers, ATP-binding cassette (ABC) transporters, STAT3, miR-181b, CYLD. Western blot were performed to analyze the expression of STAT3, p-STAT3 and CYLD (cylindromatosis). BALB/c mice xenograft studies were conducted to evaluate the tumorigenicity of enriched ECSLC. Sphere formation assay and colony formation assays were employed to analyze the relationship between STAT3 and miR-181b. Luciferase assays were used to evaluate activity which CYLD is a target of miR-181b. Results Sphere formation cells (SFCs) with properties of ECSLC were enriched. Enriched SFCs in serum-free suspension culture exhibited cancer stem-like cell properties and increased single-positive CD44 + CD24-, stemness factor, mesenchymal marker expression ABC transporters and tumorigenicity in vivo compared with the parental cells. Additionally, we found that reciprocal activation between STAT3 and miR-181b regulated SFCs proliferation. Moreover, STAT3 directly activated miR-181b transcription in SFCs and miR-181b then potentiated p-STAT3 activity. Luciferase assays indicated that CYLD was a direct and functional target of miR-181b. Conclusion The mutual regulation between STAT3 and miR-181b in SFCs was required for proliferation and apoptosis resistance. STAT3 and miR-181b control each other’s expression in a positive feedback loop that regulates SFCs via CYLD pathway. These findings maybe is helpful for targeting ECSLC and providing approach for esophageal cancer treatments. Electronic supplementary material The online version of this article (doi:10.1186/s12943-016-0521-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dan-Dan Xu
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R. China.,Key laboratory of Bioengineering medicine of Guangdong Province, Jinan University, 510632, Guangzhou, P.R. China
| | - Peng-Jun Zhou
- Department of Pathogen Biology and Immunology, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou, 510006, P.R. China
| | - Ying Wang
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R. China.,Faculty of Environmental and Biological Engineering, Guangdong University of Petrochemical Technology, 525000, Maoming, P.R. China
| | - Li Zhang
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R. China
| | - Wu-Yu Fu
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, P.R. China
| | - Bi-Bo Ruan
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, P.R. China
| | - Hai-Peng Xu
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R. China
| | - Chao-Zhi Hu
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R. China
| | - Lu Tian
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R. China
| | - Jin-Hong Qin
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R. China
| | - Sheng Wang
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R. China
| | - Xiao Wang
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R. China
| | - Yi-Cheng Li
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R. China
| | - Qiu-Ying Liu
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R. China
| | - Zhe Ren
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R. China
| | - Rong Zhang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510632, Guangzhou, P.R. China.
| | - Yi-Fei Wang
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R. China. .,Key laboratory of Bioengineering medicine of Guangdong Province, Jinan University, 510632, Guangzhou, P.R. China.
| |
Collapse
|
20
|
Jang SJ, Kang JH, Lee YJ, Kim KI, Lee TS, Choe JG, Lim SM. Detection of metastatic tumors after γ-irradiation using longitudinal molecular imaging and gene expression profiling of metastatic tumor nodules. Int J Oncol 2016; 48:1361-8. [PMID: 26892334 PMCID: PMC4777593 DOI: 10.3892/ijo.2016.3384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 01/11/2016] [Indexed: 12/26/2022] Open
Abstract
A few recent reports have indicated that metastatic growth of several human cancer cells could be promoted by radiotherapy. C6-L cells expressing the firefly luciferase (fLuc) gene were implanted subcutaneously into the right thigh of BALB/c nu/nu mice. C6-L xenograft mice were treated locally with 50-Gy γ-irradiation (γ-IR) in five 10-Gy fractions. Metastatic tumors were evaluated after γ-IR by imaging techniques. Total RNA from non-irradiated primary tumor (NRPT), γ-irradiated primary tumor (RPT), and three metastatic lung nodule was isolated and analyzed by microarray. Metastatic lung nodules were detected by BLI and PET/CT after 6–9 weeks of γ-IR in 6 (17.1%) of the 35 mice. The images clearly demonstrated high [18F]FLT and [18F]FDG uptake into metastatic lung nodules. Whole mRNA expression patterns were analyzed by microarray to elucidate the changes among NRPT, RPT and metastatic lung nodules after γ-IR. In particular, expression changes in the cancer stem cell markers were highly significant in RPT. We observed the metastatic tumors after γ-IR in a tumor-bearing animal model using molecular imaging methods and analyzed the gene expression profile to elucidate genetic changes after γ-IR.
Collapse
Affiliation(s)
- Su Jin Jang
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 139-706, Republic of Korea
| | - Joo Hyun Kang
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 139-706, Republic of Korea
| | - Yong Jin Lee
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 139-706, Republic of Korea
| | - Kwang Il Kim
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 139-706, Republic of Korea
| | - Tae Sup Lee
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 139-706, Republic of Korea
| | - Jae Gol Choe
- Department of Nuclear Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul 136-705, Republic of Korea
| | - Sang Moo Lim
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 139-706, Republic of Korea
| |
Collapse
|
21
|
Qureshi-Baig K, Ullmann P, Rodriguez F, Frasquilho S, Nazarov PV, Haan S, Letellier E. What Do We Learn from Spheroid Culture Systems? Insights from Tumorspheres Derived from Primary Colon Cancer Tissue. PLoS One 2016; 11:e0146052. [PMID: 26745821 PMCID: PMC4706382 DOI: 10.1371/journal.pone.0146052] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 12/11/2015] [Indexed: 01/20/2023] Open
Abstract
Due to their self-renewal and tumorigenic properties, tumor-initiating cells (TICs) have been hypothesized to be important targets for colorectal cancer (CRC). However the study of TICs is hampered by the fact that the identification and culturing of TICs is still a subject of extensive debate. Floating three-dimensional spheroid cultures (SC) that grow in serum-free medium supplemented with growth factors are supposed to be enriched in TICs. We generated SC from fresh clinical tumor specimens and compared them to SC isolated from CRC cell-lines as well as to adherent differentiated counterparts. Patient-derived SC display self-renewal capacity and can induce serial transplantable tumors in immuno-deficient mice, which phenotypically resemble the tumor of origin. In addition, the original tumor tissue and established SC retain several similar CRC-relevant mutations. Primary SC express key stemness proteins such as SOX2, OCT4, NANOG and LGR5 and importantly show increased chemoresistance ability compared to their adherent differentiated counterparts and to cell line-derived SC. Strikingly, cells derived from spheroid or adherent differentiating culture conditions displayed similar self-renewal capacity and equally formed tumors in immune-deficient mice, suggesting that self-renewal and tumor-initiation capacity of TICs is not restricted to phenotypically immature spheroid cells, which we describe to be highly plastic and able to reacquire stem-cell traits even after long differentiation processes. Finally, we identified two genes among a sphere gene expression signature that predict disease relapse in CRC patients. Here we propose that SC derived from fresh patient tumor tissue present interesting phenotypic features that may have clinical relevance for chemoresistance and disease relapse and therefore represent a valuable tool to test for new CRC-therapies that overcome drug resistance.
Collapse
Affiliation(s)
- Komal Qureshi-Baig
- Molecular Disease Mechanisms Group, Life Sciences Research Unit, University of Luxembourg, 6 Avenue du Swing, L-4367, Campus Belval, Luxembourg
| | - Pit Ullmann
- Molecular Disease Mechanisms Group, Life Sciences Research Unit, University of Luxembourg, 6 Avenue du Swing, L-4367, Campus Belval, Luxembourg
| | - Fabien Rodriguez
- Molecular Disease Mechanisms Group, Life Sciences Research Unit, University of Luxembourg, 6 Avenue du Swing, L-4367, Campus Belval, Luxembourg
| | - Sónia Frasquilho
- Integrated Biobank of Luxembourg, 6 rue Nicolas Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Petr V. Nazarov
- Luxembourg Institute of Health, Genomics Research Unit, 84 Val Fleuri, L-1526, Luxembourg, Luxembourg
| | - Serge Haan
- Molecular Disease Mechanisms Group, Life Sciences Research Unit, University of Luxembourg, 6 Avenue du Swing, L-4367, Campus Belval, Luxembourg
| | - Elisabeth Letellier
- Molecular Disease Mechanisms Group, Life Sciences Research Unit, University of Luxembourg, 6 Avenue du Swing, L-4367, Campus Belval, Luxembourg
| |
Collapse
|
22
|
The role of Dickkopf-3 overexpression in esophageal adenocarcinoma. J Thorac Cardiovasc Surg 2015; 150:377-385.e2. [PMID: 26093488 DOI: 10.1016/j.jtcvs.2015.05.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/31/2015] [Accepted: 05/02/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Ninety percent of patients with esophageal adenocarcinoma ultimately die of their disease, highlighting the need for novel therapeutic targets. The goal of this study was to define the functional significance of overexpression of Dickkopf-3 (DKK3) in esophageal adenocarcinoma. METHODS DKK3 expression was analyzed by real-time polymerase chain reaction in 95 chemonaive and 21 chemoresistant esophageal adenocarcinomas. The esophageal adenocarcinoma cell line OE33 was stably transfected with DKK3 (OE33/DKK3) and evaluated using WST-1 (Roche, Basel, Switzerland), Matrigel (BD Biosciences, San Jose, Calif), endothelial tube formation, and chemosensitivity assays. Tumorigenesis was evaluated by injecting 1 × 10(6) OE33/DKK3 and vector cells in NOD/SCIDγ mice. RESULTS DKK3 was overexpressed (>2-fold) in 75.8% (72/95) of esophageal adenocarcinomas. DKK3 protein was present at moderate to high levels in 46.8% (29/62) of esophageal adenocarcinomas on tissue microarray. Stable transfection of DKK3 significantly increased proliferation (P < .05) and Matrigel invasion (P < .001). Levels of SMAD4, a key mediator of the transforming growth factor-ß pathway, increased after activin treatment of OE33/DKK3, and siSMAD4 significantly decreased Matrigel invasion, suggesting that DKK3 acts through the transforming growth factor-β pathway. OE33/DKK3 cells increased endothelial tube formation and were significantly more resistant to 5-fluorouracil and cisplatin, and DKK3 expression was significantly higher in chemoresistant esophageal adenocarcinomas (P < .005). In NOD/SCIDγ mice, OE33/DKK3 cells resulted in tumors at all sites (8/8), whereas vector cells grew in only 1 of 8 sites. Nodal metastases were also significantly increased in patients with esophageal adenocarcinomas highly overexpressing DKK3, 28 of 32 (88%) versus 42 of 63 (68%) (P < .05). CONCLUSIONS These findings suggest that DKK3 may be important in mediating invasion in esophageal adenocarcinoma and could be a novel target in the treatment and prevention of metastatic disease.
Collapse
|
23
|
Boonstra JJ, Tilanus HW, Dinjens WNM. Translational research on esophageal adenocarcinoma: from cell line to clinic. Dis Esophagus 2015; 28:90-6. [PMID: 23795680 DOI: 10.1111/dote.12095] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human esophageal adenocarcinoma (EAC) cell lines have made a substantial contribution to elucidating mechanisms of carcinogenesis and drug discovery. Model research on EAC relies almost entirely on a relatively small set of established tumor cell lines because appropriate animal models are lacking. Nowadays, more than 20% of all fundamental translational research studies regarding EAC are partially or entirely based on these cell lines. The ready availability of these cell lines to investigators worldwide have resulted in more than 250 publications, including many examples of important biomedical discoveries. The high genomic similarities (but certainly not completely identical) between the EAC cell lines and their original tumors provide rational for their use. Recently, in a collaborative effort all available EAC cell lines have been verified resulting in the establishment of a reliable panel of 10 EAC cell lines. It could be expected that the value of these cell lines increases as unlimited source of tumor material because new biomedical techniques require more tumor cells and the supply of viable tumor cells is diminishing because of neoadjuvant chemo(radio)therapy of patients with EAC. Here, we review the history of the EAC cell lines and their utility in translational research and biomedical discovery.
Collapse
Affiliation(s)
- J J Boonstra
- Department of Pathology, Josephine Nefkens Institute, University Medical Center, Rotterdam, The Netherlands; Department of Surgery, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | | | | |
Collapse
|
24
|
Allegra A, Alonci A, Penna G, Innao V, Gerace D, Rotondo F, Musolino C. The cancer stem cell hypothesis: a guide to potential molecular targets. Cancer Invest 2014; 32:470-95. [PMID: 25254602 DOI: 10.3109/07357907.2014.958231] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Common cancer theories hold that tumor is an uncontrolled somatic cell proliferation caused by the progressive addition of random mutations in critical genes that control cell growth. Nevertheless, various contradictions related to the mutation theory have been reported previously. These events may be elucidated by the persistence of residual tumor cells, called Cancer Stem Cells (CSCs) responsible for tumorigenesis, tumor maintenance, tumor spread, and tumor relapse. Herein, we summarize the current understanding of CSCs, with a focus on the possibility to identify specific markers of CSCs, and discuss the clinical application of targeting CSCs for cancer treatment.
Collapse
|
25
|
Pavlov K, Meijer C, van den Berg A, Peters FTM, Kruyt FAE, Kleibeuker JH. Embryological signaling pathways in Barrett's metaplasia development and malignant transformation; mechanisms and therapeutic opportunities. Crit Rev Oncol Hematol 2014; 92:25-37. [PMID: 24935219 DOI: 10.1016/j.critrevonc.2014.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 03/24/2014] [Accepted: 05/02/2014] [Indexed: 01/07/2023] Open
Abstract
Barrett's metaplasia of the esophagus (BE) is the precursor lesion of esophageal adenocarcinoma (EAC), a deadly disease with a 5-year overall survival of less than 20%. The molecular mechanisms of BE development and its transformation to EAC are poorly understood and current surveillance and treatment strategies are of limited efficacy. Increasing evidence suggests that aberrant signaling through pathways active in the embryological development of the esophagus contributes to BE development and progression to EAC. We discuss the role that the Bone morphogenetic protein, Hedgehog, Wingless-Type MMTV Integration Site Family (WNT) and Retinoic acid signaling pathways play during embryological development of the esophagus and their contribution to BE development and malignant transformation. Modulation of these pathways provides new therapeutic opportunities. By integrating findings in developmental biology with those from translational research and clinical trials, this review provides a platform for future studies aimed at improving current management of BE and EAC.
Collapse
Affiliation(s)
- K Pavlov
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - C Meijer
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - A van den Berg
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - F T M Peters
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - F A E Kruyt
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - J H Kleibeuker
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
26
|
Yang L, Ren Y, Yu X, Qian F, Bian BSJ, Xiao HL, Wang WG, Xu SL, Yang J, Cui W, Liu Q, Wang Z, Guo W, Xiong G, Yang K, Qian C, Zhang X, Zhang P, Cui YH, Bian XW. ALDH1A1 defines invasive cancer stem-like cells and predicts poor prognosis in patients with esophageal squamous cell carcinoma. Mod Pathol 2014; 27:775-83. [PMID: 24201124 DOI: 10.1038/modpathol.2013.189] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 08/29/2013] [Accepted: 09/01/2013] [Indexed: 12/16/2022]
Abstract
Invasion and metastasis are the major cause of deaths in patients with esophageal cancer. In this study, we isolated cancer stem-like cells from an esophageal squamous cell carcinoma cell line EC109 based on aldehyde dehydrogenase 1A1 (ALDH1A1), and found that ALDH1A1(high) cells possessed the capacities of self-renewal, differentiation and tumor initiation, indications of stem cell properties. To support their stemness, ALDH1A1(high) cells exhibited increased potential of invasion and metastasis as compared with ALDH1A1(low) cells. ALDH1A1(high) esophageal squamous cell carcinoma cells expressed increased levels of mRNA for vimentin, matrix metalloproteinase 2, 7 and 9 (MMP2, MMP7 and MMP9), but decreased the level of E-cadherin mRNA, suggesting that epithelial-mesenchymal transition and secretary MMPs may be attributed to the high invasive and metastatic capabilities of ALDH1A1(high) cells. Furthermore, we examined esophageal squamous cell carcinoma specimens from 165 patients and found that ALDH1A1(high) cells were associated with esophageal squamous dysplasia and the grades, differentiation and invasion depth, lymph node metastasis and UICC stage of esophageal squamous cell carcinoma, as well as poor prognosis of patients. Our results provide the strong evidence that ALDH1A1(high) cancer stem-like cells contribute to the invasion, metastasis and poor outcome of human esophageal squamous cell carcinoma.
Collapse
Affiliation(s)
- Lang Yang
- Institute of Pathology and Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yong Ren
- Institute of Pathology and Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xi Yu
- Institute of Pathology and Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Feng Qian
- Department of General Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Bai-Shi-Jiao Bian
- 1] Institute of Pathology and Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Third Military Medical University, Chongqing, China [2] Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Hua-liang Xiao
- Department of Pathology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Wei-guang Wang
- Research Institute in Healthcare Science, University of Wolverhampton, Wolverhampton, UK
| | - Sen-lin Xu
- Institute of Pathology and Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jing Yang
- Institute of Pathology and Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Wei Cui
- Institute of Pathology and Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Qiang Liu
- Institute of Pathology and Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Zhe Wang
- Institute of Pathology and Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Wei Guo
- Department of Thoracic Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Gang Xiong
- Department of Thoracic Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Kang Yang
- Department of Thoracic Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Cheng Qian
- Institute of Pathology and Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xia Zhang
- Institute of Pathology and Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Peng Zhang
- Institute of Pathology and Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - You-hong Cui
- Institute of Pathology and Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xiu-wu Bian
- Institute of Pathology and Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
27
|
The culture of cancer cell lines as tumorspheres does not systematically result in cancer stem cell enrichment. PLoS One 2014; 9:e89644. [PMID: 24586931 PMCID: PMC3933663 DOI: 10.1371/journal.pone.0089644] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 01/24/2014] [Indexed: 01/01/2023] Open
Abstract
Cancer stem cells (CSC) have raised great excitement during the last decade and are promising targets for an efficient treatment of tumors without relapses and metastases. Among the various methods that enable to enrich cancer cell lines in CSC, tumorspheres culture has been predominantly used. In this report, we attempted to generate tumorspheres from several murine and human cancer cell lines: B16-F10, HT-29, MCF-7 and MDA-MB-231 cells. Tumorspheres were obtained with variable efficiencies from all cell lines except from MDA-MB-231 cells. Then, we studied several CSC characteristics in both tumorspheres and adherent cultures of the B16-F10, HT-29 and MCF-7 cells. Unexpectedly, tumorspheres-forming cells were less clonogenic and, in the case of B16-F10, less proliferative than attached cells. In addition, we did not observe any enrichment in the population expressing CSC surface markers in tumorspheres from B16-F10 (CD133, CD44 and CD24 markers) or MCF-7 (CD44 and CD24 markers) cells. On the contrary, tumorspheres culture of HT-29 cells appeared to enrich in cells expressing colon CSC markers, i.e. CD133 and CD44 proteins. For the B16-F10 cell line, when 1 000 cells were injected in syngenic C57BL/6 mice, tumorspheres-forming cells displayed a significantly lower tumorigenic potential than adherent cells. Finally, tumorspheres culture of B16-F10 cells induced a down-regulation of vimentin which could explain, at least partially, the lower tumorigenicity of tumorspheres-forming cells. All these results, along with the literature, indicate that tumorspheres culture of cancer cell lines can induce an enrichment in CSC but in a cell line-dependent manner. In conclusion, extensive characterization of CSC properties in tumorspheres derived from any cancer cell line or cancer tissue must be performed in order to ensure that the generated tumorspheres are actually enriched in CSC.
Collapse
|