1
|
Jo JH, Park SB, Chung J, Oh T, Lee HS, Chung MJ, Park JY, Bang S, Park SW, Jung DE, Song SY. Transgelin-2, a novel cancer stem cell-related biomarker, is a diagnostic and therapeutic target for biliary tract cancer. BMC Cancer 2024; 24:357. [PMID: 38509504 PMCID: PMC10953140 DOI: 10.1186/s12885-024-12082-3] [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: 06/10/2023] [Accepted: 03/04/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Biliary tract cancer (BTC) is a relatively rare but aggressive gastrointestinal cancer with a high mortality rate. Cancer stem cell (CSC) populations play crucial roles in tumor biology and are responsible for the low response to anti-cancer treatment and the high recurrence rate. This study investigated the role of Transgelin-2 (TAGLN2), overexpressed in CSC in BTC cells, and analyzed its expression in patient tissues and serum to identify potential new targets for BTC. METHODS TAGLN2 expression was suppressed by small-interfering or short hairpin RNAs, and its effects on tumor biology were assessed in several BTC cell lines. Furthermore, the effects of TAGLN2 silencing on gemcitabine-resistant BTC cells, differentially expressed genes, proteins, and sensitivity to therapeutics or radiation were assessed. TAGLN2 expression was also assessed using western blotting and immunohistochemistry in samples obtained from patients with BTC to validate its clinical application. RESULTS Suppression of TAGLN2 in BTC cell lines decreased cell proliferation, migration, invasion, and tumor size, in addition to a reduction in CSC features, including clonogenicity, radioresistance, and chemoresistance. TAGLN2 was highly expressed in BTC tissues, especially in cancer-associated fibroblasts in the stroma. Patients with a low stromal immunohistochemical index had prolonged disease-free survival compared to those with a high stromal immunohistochemical index (11.5 vs. 7.4 months, P = 0.013). TAGLN2 expression was higher in the plasma of patients with BTC than that in those with benign diseases. TAGLN2 had a higher area under the curve (0.901) than CA19-9, a validated tumor biomarker (0.799; P < 0.001). CONCLUSION TAGLN2 plays a critical role in promoting BTC cell growth and motility and is involved in regulating BTC stemness. Silencing TAGLN2 expression enhanced cell sensitivity to radiation and chemotherapeutic drugs. The expression of TAGLN2 in patient tissue and plasma suggests its potential to serve as a secretory biomarker for BTC. Overall, targeting TAGLN2 could be an appropriate therapeutic strategy against advanced cancer following chemotherapy failure.
Collapse
Affiliation(s)
- Jung Hyun Jo
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Soo Been Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Joowon Chung
- Department of Internal Medicine, Nowon Eulji Medical Center, Eulji University School of Medicine, Seoul, Korea
| | - Taeyun Oh
- Cowell Biodigm Co., Ltd., Seoul, Korea
| | - Hee Seung Lee
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Moon Jae Chung
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jeong Youp Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Seungmin Bang
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Woo Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Dawoon E Jung
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea.
| | - Si Young Song
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.
- Cowell Biodigm Co., Ltd., Seoul, Korea.
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
2
|
Blackmore DG, Waters MJ. The multiple roles of GH in neural ageing and injury. Front Neurosci 2023; 17:1082449. [PMID: 36960169 PMCID: PMC10027725 DOI: 10.3389/fnins.2023.1082449] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/17/2023] [Indexed: 03/09/2023] Open
Abstract
Advanced age is typically associated with a decrease in cognitive function including impairment in the formation and retention of new memories. The hippocampus is critical for learning and memory, especially spatial learning, and is particularly affected by ageing. With advanced age, multiple neural components can be detrimentally affected including a reduction in the number of neural stem and precursor cells, a decrease in the formation of adult born neurons (neurogenesis), and deficits in neural circuitry, all of which ultimately contribute to impaired cognitive function. Importantly, physical exercise has been shown to ameliorate many of these impairments and is able to improve learning and memory. Relevantly, growth hormone (GH) is an important protein hormone that decreases with ageing and increases following physical exercise. Originally described due to its role in longitudinal growth, GH has now been identified to play several additional key roles, especially in relation to the brain. Indeed, the regular decrease in GH levels following puberty is one of the most well documented components of neuroendocrine ageing. Growth hormone deficiency (GHD) has been described to have adverse effects on brain function, which can be ameliorated via GH replacement therapy. Physical exercise has been shown to increase circulating GH levels. Furthermore, we recently demonstrated the increase in exercise-mediated GH is critical for improved cognitive function in the aged mouse. Here we examine the multiple roles that GH plays, particularly in the aged brain and following trauma, irradiation and stroke, and how increasing GH levels can ameliorate deficits in cognition.
Collapse
Affiliation(s)
- Daniel G. Blackmore
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Michael J. Waters
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- *Correspondence: Michael J. Waters,
| |
Collapse
|
3
|
Banerjee D, Singh YP, Datta P, Ozbolat V, O'Donnell A, Yeo M, Ozbolat IT. Strategies for 3D bioprinting of spheroids: A comprehensive review. Biomaterials 2022; 291:121881. [DOI: 10.1016/j.biomaterials.2022.121881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/04/2022] [Accepted: 10/23/2022] [Indexed: 11/17/2022]
|
4
|
Chen J, Qiao K, Zhang C, Zhou X, Du Q, Deng Y, Cao L. VRK2 activates TNFα/NF-κB signaling by phosphorylating IKKβ in pancreatic cancer. Int J Biol Sci 2022; 18:1288-1302. [PMID: 35173553 PMCID: PMC8771851 DOI: 10.7150/ijbs.66313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/31/2021] [Indexed: 11/05/2022] Open
Abstract
NF-κB signaling is active in more than 50% of patients with pancreatic cancer and plays an important role in promoting the progression of pancreatic cancer. Revealing the activation mechanism of NF-κB signaling is important for the treatment of pancreatic cancer. In this study, the regulation of TNFα/NF-κB signaling by VRK2 (vaccinia-related kinase 2) was investigated. The levels of VRK2 protein were examined by immunohistochemistry (IHC). The functions of VRK2 in the progression of pancreatic cancer were examined using CCK8 assay, anchorage-independent assay, EdU assay and tumorigenesis assay. The regulation of VRK2 on the NF-κB signaling was investigated by immunoprecipitation and invitro kinase assay. It was discovered in this study that the expression of VRK2 was upregulated in pancreatic cancer and that the VRK2 expression level was significantly correlated with the pathological characteristics and the survival time of patients. VRK2 promoted the growth, sphere formation and subcutaneous tumorigenesis of pancreatic carcinoma cells as well as the organoid growth derived from the pancreatic cancer mouse model. Investigation of the molecular mechanism indicated that VRK2 interacts with IKKβ, phosphorylating its Ser177 and Ser181 residues and thus activating the TNFα/NF-κB signaling pathway. An IKKβ inhibitors abolished the promotive effect of VRK2 on the growth of organoids. The findings of this study indicate that VRK2 promotes the progression of pancreatic cancer by activating the TNFα/NF-κB signaling pathway, suggesting that VRK2 is a potential therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Jionghuang Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kexiong Qiao
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chaolei Zhang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyang Zhou
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qian Du
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuezhen Deng
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
| | - Liping Cao
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
5
|
Scalise M, Torella M, Marino F, Ravo M, Giurato G, Vicinanza C, Cianflone E, Mancuso T, Aquila I, Salerno L, Nassa G, Agosti V, De Angelis A, Urbanek K, Berrino L, Veltri P, Paolino D, Mastroroberto P, De Feo M, Viglietto G, Weisz A, Nadal-Ginard B, Ellison-Hughes GM, Torella D. Atrial myxomas arise from multipotent cardiac stem cells. Eur Heart J 2021; 41:4332-4345. [PMID: 32330934 PMCID: PMC7735815 DOI: 10.1093/eurheartj/ehaa156] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/22/2019] [Accepted: 03/03/2020] [Indexed: 12/11/2022] Open
Abstract
Aims Cardiac myxomas usually develop in the atria and consist of an acid-mucopolysaccharide-rich myxoid matrix with polygonal stromal cells scattered throughout. These human benign tumours are a valuable research model because of the rarity of cardiac tumours, their clinical presentation and uncertain origin. Here, we assessed whether multipotent cardiac stem/progenitor cells (CSCs) give rise to atrial myxoma tissue. Methods and results Twenty-three myxomas were collected and analysed for the presence of multipotent CSCs. We detected myxoma cells positive for c-kit (c-kitpos) but very rare Isl-1 positive cells. Most of the c-kitpos cells were blood lineage-committed CD45pos/CD31pos cells. However, c-kitpos/CD45neg/CD31neg cardiac myxoma cells expressed stemness and cardiac progenitor cell transcription factors. Approximately ≤10% of the c-kitpos/CD45neg/CD31neg myxoma cells also expressed calretinin, a characteristic of myxoma stromal cells. In vitro, the c-kitpos/CD45neg/CD31neg myxoma cells secrete chondroitin-6-sulfate and hyaluronic acid, which are the main components of gelatinous myxoma matrix in vivo. In vitro, c-kitpos/CD45neg/CD31neg myxoma cells have stem cell properties being clonogenic, self-renewing, and sphere forming while exhibiting an abortive cardiac differentiation potential. Myxoma-derived CSCs possess a mRNA and microRNA transcriptome overall similar to normal myocardium-derived c-kitpos/CD45neg/CD31negCSCs , yet showing a relatively small and relevant fraction of dysregulated mRNA/miRNAs (miR-126-3p and miR-335-5p, in particular). Importantly, myxoma-derived CSCs but not normal myocardium-derived CSCs, seed human myxoma tumours in xenograft’s in immunodeficient NOD/SCID mice. Conclusion Myxoma-derived c-kitpos/CD45neg/CD31neg CSCs fulfill the criteria expected of atrial myxoma-initiating stem cells. The transcriptome of these cells indicates that they belong to or are derived from the same lineage as the atrial multipotent c-kitpos/CD45neg/CD31neg CSCs. Taken together the data presented here suggest that human myxomas could be the first-described CSC-related human heart disease. ![]()
Collapse
Affiliation(s)
- Mariangela Scalise
- Department of Experimental and Clinical Medicine, Molecular and Cellular Cardiology, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - Michele Torella
- Department of Translational Medical Sciences, AORN dei Colli/Monaldi Hospital, University of Campania "L. Vanvitelli", Via Leonardo Bianchi, 80131 Naples, Italy
| | - Fabiola Marino
- Department of Experimental and Clinical Medicine, Molecular and Cellular Cardiology, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - Maria Ravo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Laboratory of Molecular Medicine and Genomics, University of Salerno, Via Salvador Allende, 84081 Baronissi (Salerno), Italy.,Genomix4Life, Spin-Off of the Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry, University of Salerno, Via Salvador Allende, 84081 Baronissi (Salerno), Italy
| | - Giorgio Giurato
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Laboratory of Molecular Medicine and Genomics, University of Salerno, Via Salvador Allende, 84081 Baronissi (Salerno), Italy.,Genomix4Life, Spin-Off of the Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry, University of Salerno, Via Salvador Allende, 84081 Baronissi (Salerno), Italy
| | - Carla Vicinanza
- Department of Experimental and Clinical Medicine, Molecular and Cellular Cardiology, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - Eleonora Cianflone
- Department of Experimental and Clinical Medicine, Molecular and Cellular Cardiology, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy.,Department of Medical and Surgical Sciences, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - Teresa Mancuso
- Department of Experimental and Clinical Medicine, Molecular and Cellular Cardiology, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - Iolanda Aquila
- Department of Experimental and Clinical Medicine, Molecular and Cellular Cardiology, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - Luca Salerno
- Department of Experimental and Clinical Medicine, Molecular and Cellular Cardiology, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - Giovanni Nassa
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Laboratory of Molecular Medicine and Genomics, University of Salerno, Via Salvador Allende, 84081 Baronissi (Salerno), Italy
| | - Valter Agosti
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - Antonella De Angelis
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - Konrad Urbanek
- Department of Experimental and Clinical Medicine, Molecular and Cellular Cardiology, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy.,Department of Experimental Medicine, University of Campania "L. Vanvitelli", Via Santa Maria di Costantinopoli, 80138 Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Via Santa Maria di Costantinopoli, 80138 Naples, Italy
| | - Pierangelo Veltri
- Department of Medical and Surgical Sciences, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - Pasquale Mastroroberto
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - Marisa De Feo
- Department of Translational Medical Sciences, AORN dei Colli/Monaldi Hospital, University of Campania "L. Vanvitelli", Via Leonardo Bianchi, 80131 Naples, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - Alessandro Weisz
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Laboratory of Molecular Medicine and Genomics, University of Salerno, Via Salvador Allende, 84081 Baronissi (Salerno), Italy.,Genomix4Life, Spin-Off of the Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry, University of Salerno, Via Salvador Allende, 84081 Baronissi (Salerno), Italy
| | - Bernardo Nadal-Ginard
- Department of Experimental and Clinical Medicine, Molecular and Cellular Cardiology, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - Georgina M Ellison-Hughes
- Centre for Human and Applied Physiological Sciences and Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, Guys Campus - Great Maze Pond rd, SE1 1UL London, UK
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Molecular and Cellular Cardiology, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| |
Collapse
|
6
|
Liu SC, Wu YC, Huang CM, Hsieh MS, Huang TY, Huang CS, Hsu TN, Huang MS, Lee WH, Yeh CT, Lin CS. Inhibition of Bruton's tyrosine kinase as a therapeutic strategy for chemoresistant oral squamous cell carcinoma and potential suppression of cancer stemness. Oncogenesis 2021; 10:20. [PMID: 33640903 PMCID: PMC7914253 DOI: 10.1038/s41389-021-00308-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 02/03/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023] Open
Abstract
Locally advanced oral squamous cell carcinoma (OSCC) requires multimodal therapy, including surgery and concurrent chemoradiotherapy (CCRT). CCRT-resistant and recurrent cancer has a poor prognosis. We investigated the effects of Bruton's tyrosine kinase (BTK) on CCRT-resistant OSCC tissues. The effect of ibrutinib, a first-in-class BTK inhibitor, was tested on stem cell-like OSCC tumorspheres. A tissue array was constructed using tissue samples from 70 patients with OSCC. Human OSCC cell lines, SAS, TW2.6 and HSC-3, were examined. Wound healing, Matrigel invasion, and tumorsphere formation assays, as well as immunofluorescence analysis and flow cytometry, were used to investigate the effects of BTK knockdown (shBTK), ibrutinib, cisplatin, and ibrutinib/cisplatin combination on OSCC cells. We demonstrated that BTK was aberrantly highly expressed in the clinical CCRT-resistant OSCC tissue array, which resulted in poor overall survival in our local Tri-Service General Hospital and freely accessible TCGA OSCC cohorts. shBTK significantly downregulated the stemness markers Nanog, CD133, T cell immunoglobulin-3 (TIM-3), and Krüppel-like factor 4 (KLF4) in SAS tumorspheres and attenuated OSCC cell migration and colony formation. Ibrutinib reduced the number of aldehyde dehydrogenase (ALDH)-rich OSCC cells and reduced tumorsphere formation, migration, and invasion in a dose-dependent manner. Compared with ibrutinib or cisplatin monotherapy, the ibrutinib/cisplatin combination significantly reduced the formation of ALDH + OSCC tumorspheres and enhanced apoptosis. These results demonstrate that ibrutinib effectively inhibits the CSCs-like phenotype of OSCC cells through dysregulation of BTK/CD133 signaling. The ibrutinib/cisplatin combination may be considered for future clinical use.
Collapse
Affiliation(s)
- Shao-Cheng Liu
- grid.260565.20000 0004 0634 0356Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114 Taiwan
| | - Yang-Che Wu
- grid.412896.00000 0000 9337 0481School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei City, 110 Taiwan ,grid.412955.e0000 0004 0419 7197Department of Dentistry, Taipei Medical University—Shuang Ho Hospital, New Taipei City, 235 Taiwan
| | - Chih-Ming Huang
- grid.413593.90000 0004 0573 007XDepartment of Otolaryngology, Taitung Mackay Memorial Hospital, Taipei City, Taiwan
| | - Ming-Shou Hsieh
- grid.412896.00000 0000 9337 0481School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei City, 110 Taiwan ,grid.412955.e0000 0004 0419 7197Department of Dentistry, Taipei Medical University—Shuang Ho Hospital, New Taipei City, 235 Taiwan
| | - Ting-Yi Huang
- grid.412955.e0000 0004 0419 7197Department of Hematology and Oncology, Cancer Center, Taipei Medical University—Shuang Ho Hospital, New Taipei City, 235 Taiwan ,grid.412955.e0000 0004 0419 7197Department of Medical Research & Education, Taipei Medical University—Shuang Ho Hospital, New Taipei City, 235 Taiwan
| | - Chin-Sheng Huang
- grid.412896.00000 0000 9337 0481School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei City, 110 Taiwan ,grid.412955.e0000 0004 0419 7197Department of Dentistry, Taipei Medical University—Shuang Ho Hospital, New Taipei City, 235 Taiwan
| | - Tung-Nien Hsu
- grid.412896.00000 0000 9337 0481School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei City, 110 Taiwan ,grid.412955.e0000 0004 0419 7197Department of Dentistry, Taipei Medical University—Shuang Ho Hospital, New Taipei City, 235 Taiwan
| | - Mao-Suan Huang
- grid.412896.00000 0000 9337 0481School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei City, 110 Taiwan ,grid.412955.e0000 0004 0419 7197Department of Dentistry, Taipei Medical University—Shuang Ho Hospital, New Taipei City, 235 Taiwan
| | - Wei-Hwa Lee
- grid.412955.e0000 0004 0419 7197Department of Medical Research & Education, Taipei Medical University—Shuang Ho Hospital, New Taipei City, 235 Taiwan ,grid.412955.e0000 0004 0419 7197Department of Pathology, Taipei Medical University—Shuang Ho Hospital, New Taipei City, 235 Taiwan
| | - Chi-Tai Yeh
- grid.412955.e0000 0004 0419 7197Department of Medical Research & Education, Taipei Medical University—Shuang Ho Hospital, New Taipei City, 235 Taiwan ,grid.412955.e0000 0004 0419 7197Department of Pathology, Taipei Medical University—Shuang Ho Hospital, New Taipei City, 235 Taiwan ,grid.413051.20000 0004 0444 7352Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu City, 30015 Taiwan
| | - Chun-Shu Lin
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114 Taiwan
| |
Collapse
|
7
|
Abstract
Sphere assays are widely used in vitro techniques to enrich and evaluate the stem-like cell behavior of both normal and cancer cells. Utilizing three-dimensional in vitro sphere culture conditions provide a better representation of tumor growth in vivo than the more common monolayer cultures. We describe how to perform primary and secondary sphere assays, used for the enrichment and self-renewability studies of melanoma/melanocyte stem-like cells. Spheres are generated by growing melanoma cells at low density in nonadherent conditions with stem cell media. We provide protocols for preparing inexpensive and versatile polyHEMA-coated plates, setting up primary and secondary sphere assays in almost any tissue culture format and quantification methods using standard inverted microscopy. Our protocol is easily adaptable to laboratories with basic cell culture capabilities, without the need for expensive fluidic instruments.
Collapse
|
8
|
Reynolds BA, Oli MW, Oli MK. Eco-oncology: Applying ecological principles to understand and manage cancer. Ecol Evol 2020; 10:8538-8553. [PMID: 32884638 PMCID: PMC7452771 DOI: 10.1002/ece3.6590] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 12/25/2022] Open
Abstract
Cancer is a disease of single cells that expresses itself at the population level. The striking similarities between initiation and growth of tumors and dynamics of biological populations, and between metastasis and ecological invasion and community dynamics suggest that oncology can benefit from an ecological perspective to improve our understanding of cancer biology. Tumors can be viewed as complex, adaptive, and evolving systems as they are spatially and temporally heterogeneous, continually interacting with each other and with the microenvironment and evolving to increase the fitness of the cancer cells. We argue that an eco-evolutionary perspective is essential to understand cancer biology better. Furthermore, we suggest that ecologically informed therapeutic approaches that combine standard of care treatments with strategies aimed at decreasing the evolutionary potential and fitness of neoplastic cells, such as disrupting cell-to-cell communication and cooperation, and preventing successful colonization of distant organs by migrating cancer cells, may be effective in managing cancer as a chronic condition.
Collapse
Affiliation(s)
- Brent A. Reynolds
- Department of NeurosurgeryCollege of MedicineUniversity of FloridaGainesvilleFLUSA
| | - Monika W. Oli
- Department of Microbiology and Cell ScienceInstitute of Food and Agricultural SciencesUniversity of FloridaGainesvilleFLUSA
| | - Madan K. Oli
- Department of Wildlife Ecology and ConservationInstitute of Food and Agricultural SciencesUniversity of FloridaGainesvilleFLUSA
| |
Collapse
|
9
|
Kaneda S, Kawada J, Shinohara M, Kumemura M, Ueno R, Kawamoto T, Suzuki K, Kim B, Ikeuchi Y, Sakai Y, Collard D, Fujita H, Fujii T. Boyden chamber-based compartmentalized tumor spheroid culture system to implement localized anticancer drug treatment. BIOMICROFLUIDICS 2019; 13:054111. [PMID: 31893010 PMCID: PMC6932857 DOI: 10.1063/1.5125650] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 10/04/2019] [Indexed: 06/10/2023]
Abstract
In anticancer drug development, it is important to simultaneously evaluate both the effect of drugs on cell proliferation and their ability to penetrate tissues. To realize such an evaluation process, here, we present a compartmentalized tumor spheroid culture system utilizing a thin membrane with a through-hole to conduct localized anticancer treatment of tumor spheroids and monitor spheroid dimensions as an indicator of cell proliferation. The system is based on a commercialized Boyden chamber plate; a through-hole was bored through a porous membrane of the chamber, and the pre-existing 0.4 μm membrane pores were filled with parylene C. A HepG2 spheroid was immobilized onto the through-hole, separating the upper and lower compartments. Fluorescein (to verify the isolation between the compartments) and tirapazamine (TPZ; to treat only the lower part of the spheroid) were added to the upper and lower compartments, respectively. Since the transportation of fluorescein was blocked during treatment, i.e., the upper and lower compartments were isolated, it was confirmed that localized TPZ treatment was successfully conducted using the developed system. The effect of localized TPZ treatment on cell proliferation was estimated by measuring the maximum horizontal cross-sectional areas in the upper and lower parts of the spheroid by microscopic observations. This system can, thus, be used to perform localized anticancer drug treatment of tumor spheroids and evaluate the effect of drugs on cell proliferation.
Collapse
Affiliation(s)
- Shohei Kaneda
- Authors to whom correspondence should be addressed: and
| | - Jiro Kawada
- Center for International Research on Integrative Biomedical Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 156-0041, Japan
| | - Marie Shinohara
- Center for International Research on Integrative Biomedical Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 156-0041, Japan
| | | | - Ryohei Ueno
- LIMMS-CNRS/IIS, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 156-0041, Japan
| | - Tomoaki Kawamoto
- Department of Mechanical Systems Engineering, Faculty of Engineering, Kogakuin University, 2665-1 Nakano-machi, Hachioji-shi, Tokyo 192-0015, Japan
| | - Kenji Suzuki
- Department of Mechanical Systems Engineering, Faculty of Engineering, Kogakuin University, 2665-1 Nakano-machi, Hachioji-shi, Tokyo 192-0015, Japan
| | | | | | | | - Dominique Collard
- LIMMS-CNRS/IIS, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 156-0041, Japan
| | - Hiroyuki Fujita
- Center for Interdisciplinary Research on Micro-Nano Methods, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 156-0041, Japan
| | - Teruo Fujii
- Center for International Research on Integrative Biomedical Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 156-0041, Japan
| |
Collapse
|
10
|
Bagheri V, Abbaszadegan MR, Memar B, Motie MR, Asadi M, Mahmoudian RA, Gholamin M. Induction of T cell-mediated immune response by dendritic cells pulsed with mRNA of sphere-forming cells isolated from patients with gastric cancer. Life Sci 2019; 219:136-143. [PMID: 30641083 DOI: 10.1016/j.lfs.2019.01.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/29/2018] [Accepted: 01/10/2019] [Indexed: 12/19/2022]
Abstract
Gastric cancer (GC) as the third most common cause of cancer-associated mortality worldwide is one of the cancers with very high heterogeneity. Cancer stem cells (CSCs) as a small subset of cancer cells in solid tumors with the self-renewal, differentiation and tumorigenic ability are responsible for tumor initiation, progression, recurrence, metastasis, and resistance to current treatments. Therefore, eradication of CSCs is very vital to cure cancer. Here, we first isolated and identified sphere-forming cells in tumor tissue from four GC patients and then analyzed T cell responses induced by monocyte-derived dendritic cells (DCs) loaded with total mRNA of sphere-forming cells in terms of interferon-gamma (IFN-γ) gene expression and specific cytotoxicity. Spheroid colonies were formed in serum-free media. Sphere-forming cells dissociated from tumorspheres heterogeneously expressed CD44, CD54, and epithelial cell adhesion molecule (EpCAM) markers and generated one tumor in nude mice. These results demonstrated that gastric CSCs were enriched in tumorspheres. Cytokine-matured DCs loaded with mRNA of sphere-forming cells were able to induce IFN-γ gene expression in T-lymphocytes after a 12-day co-culture. mRNA level of IFN-γ gene in these lymphocytes was more highly expressed compared to stimulated T-lymphocytes by DCs transfected with normal tissue (6.4-9.39 folds). Cytotoxic activity of primed T-lymphocytes with antigens of sphere-forming cells was significantly higher than normal tissue antigens and mock DCs (P ≤ 0.0001). Taken together, DCs loaded with mRNA of sphere-forming cells that elicit effectively specific T cell-mediated immune responses in vitro, may be considered as a promising therapeutic vaccination in GC patients in future.
Collapse
Affiliation(s)
- Vahid Bagheri
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Bahram Memar
- Surgical Oncology Research Center, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Motie
- Surgical Oncology Research Center, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahdi Asadi
- Surgical Oncology Research Center, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mehran Gholamin
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Laboratory Sciences, School of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Alhawarat FM, Hammad HM, Hijjawi MS, Sharab AS, Abuarqoub DA, Al Shhab MA, Zihlif MA. The effect of cycling hypoxia on MCF-7 cancer stem cells and the impact of their microenvironment on angiogenesis using human umbilical vein endothelial cells (HUVECs) as a model. PeerJ 2019; 7:e5990. [PMID: 30729067 PMCID: PMC6361090 DOI: 10.7717/peerj.5990] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/22/2018] [Indexed: 12/21/2022] Open
Abstract
Background Breast cancer is the most common type of cancer among females. Hypoxia mediates cancer hallmarks and results from reduced oxygen level due to irregularities in tumor vascularization or when the tumor size prevents oxygen diffusion and triggers angiogenesis to compensate for low oxygen. Cancer stem cells (CSCs) are a rare subpopulation, able to self-renew and to give rise to tumor-initiating cells. It is proposed that CSCs’ secretions help to recruit endothelial cells via angiogenic factors to establish tumor vascularization. In the tumor microenvironment, the effect of hypoxia on CSCs and the impact of their secretions on triggering angiogenesis and tumor vascularization remain questionable. In this study, three-dimensional (3D) CSCs derived from MCF-7 were directly exposed to repetitive long-term cycles of hypoxia to assess its effect on CSCs and then to evaluate the role of the hypoxic CSCs’ (CSCsHYP) secretions in angiogenesis using (HUVECs) as a model for tumor neovascularization response. Methods CSCs derived from MCF-7 cell-line were expanded under repetitive, strictly optimized, long-term/continuous and intermittent hypoxic shots for almost four months to assess hypoxic effect on CSCs, sorted based on CD44+/CD24− biomarkers. Hypoxic phenotype of CSCsHYP was evaluated by assessing the acquired chemoresistance using MTT assay and elevated stemness properties were assessed by flow cytometry. To evaluate the effect of the secretions from CSCsHYP on angiogenesis, HUVECs were exposed to CSCsHYP conditioned-medium (CdM)—in which CSCs had been previously grown—to mimic the tumor microenvironment and to assess the effect of the secretions from CSCsHYP on the HUVECs’ capability of tube formation, migration and wound healing. Additionally, co-culture of CSCsHYP with HUVECs was performed. Results CSCsHYP acquired higher chemoresistance, increased stemness properties and obtained greater propagation, migration, and wound healing capacities, when compared to CSCs in normoxic condition (CSCsNOR). HUVECs’ tube formation and migration abilities were mediated by hypoxic (CSCs) conditioned media (CdM). Discussion This study demonstrates that chemoresistant and migrational properties of CSCs are enhanced under hypoxia to a certain extent. The microenvironment of CSCsHYP contributes to tumor angiogenesis and migration. Hypoxia is a key player in tumor angiogenesis mediated by CSCs.
Collapse
Affiliation(s)
- Fuad M Alhawarat
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| | - Hana M Hammad
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| | - Majd S Hijjawi
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Ahmad S Sharab
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Duaa A Abuarqoub
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| | - Mohammad A Al Shhab
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Malek A Zihlif
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman, Jordan
| |
Collapse
|
12
|
Morrison BJ, Steel JC, Morris JC. Reduction of MHC-I expression limits T-lymphocyte-mediated killing of Cancer-initiating cells. BMC Cancer 2018; 18:469. [PMID: 29699516 PMCID: PMC5918869 DOI: 10.1186/s12885-018-4389-3] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 04/17/2018] [Indexed: 12/15/2022] Open
Abstract
Background It has been proposed that cancer establishment, maintenance, and recurrence may be attributed to a unique population of tumor cells termed cancer-initiating cells (CICs) that may include characteristics of putative cancer stem cell-like cells. Studies in lung cancer have shown that such cells can be enriched and propagated in vitro by culturing tumor cells in serum-free suspension as tumorspheres. CICs have been characterized for their phenotype, stem cell-like qualities, and their role in establishing tumor and maintaining tumor growth. Less is known about the interaction of CICs with the immune system. Methods We established CIC-enriched tumorspheres from murine TC-1 lung cancer cells, expressing human papillomavirus 16 (HPV-16) E6/E7 antigens, and evaluated their susceptibility to antitumor immune responses both in vitro and in vivo. Results TC-1 CICs demonstrated reduced expression of surface major histocompatibility complex (MHC)-I molecules compared to non-CICs. We similarly determined decreased MHC-I expression in five of six human lung cancer cell lines cultured under conditions enriching for CICs. In vivo, TC-1 cells enriched for CICs were resistant to human papillomavirus 16 E6/E7 peptide vaccine-mediated killing. We found that vaccinated mice challenged with CIC enriched tumorspheres demonstrated shorter survivals and showed significantly fewer CD8+ tumor infiltrating lymphocytes compared to CIC non-enriched challenged mice. Furthermore, cultured cytotoxic T lymphocytes (CTLs) from vaccinated mice demonstrated reduced capacity to lyse TC-1 cells enriched for CICs compared to non-enriched TC-1 cells. Following treatment with IFN-γ, both CIC enriched and non-enriched TC-1 cells expressed similar levels of MHC-I, and the increased MHC-I expression on CICs resulted in greater CTL-mediated tumor lysis and improved tumor-free survival in mice. Conclusions These results suggest that the attenuated expression of MHC-I molecules by CICs represents a potential strategy of CICs to escape immune recognition, and that the development of successful immunotherapy strategies targeting CICs may decrease their resistance to T cell-mediated immune detection by enhancing CIC MHC-I expression. Electronic supplementary material The online version of this article (10.1186/s12885-018-4389-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Brian J Morrison
- Viral and Rickettsial Diseases Department, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, Maryland, 20910, USA.
| | - Jason C Steel
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, Queensland, Australia
| | - John C Morris
- Division of Hematology-Oncology, University of Cincinnati Cancer Institute, University of Cincinnati, Cincinnati, Ohio, 45267, USA
| |
Collapse
|
13
|
Nangia-Makker P, Hogan V, Raz A. Galectin-3 and cancer stemness. Glycobiology 2018; 28:172-181. [PMID: 29315388 PMCID: PMC6279147 DOI: 10.1093/glycob/cwy001] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 12/20/2017] [Accepted: 01/02/2018] [Indexed: 02/07/2023] Open
Abstract
Over the last few decades galectin-3, a carbohydrate binding protein, with affinity for N-acetyllactosamine residues, has been unique due to the regulatory roles it performs in processes associated with tumor progression and metastasis such as cell proliferation, homotypic/heterotypic aggregation, dynamic cellular transformation, migration and invasion, survival and apoptosis. Structure-function association of galectin-3 reveals that it consists of a short amino terminal motif, which regulates its nuclear-cytoplasmic shuttling; a collagen α-like domain, susceptible to cleavage by matrix metalloproteases and prostate specific antigen; accountable for its oligomerization and lattice formation, and a carbohydrate-recognition/binding domain containing the anti-death motif of the Bcl2 protein family. This structural complexity permits galectin-3 to associate with numerous molecules utilizing protein-protein and/or protein-carbohydrate interactions in the extra-cellular as well as intracellular milieu and regulate diverse signaling pathways, a number of which appear directed towards epithelial-mesenchymal transition and cancer stemness. Self-renewal, differentiation, long-term culturing and drug-resistance potential characterize cancer stem cells (CSCs), a small cell subpopulation within the tumor that is thought to be accountable for heterogeneity, recurrence and metastasis of tumors. Despite the fact that association of galectin-3 to the tumor stemness phenomenon is still in its infancy, there is sufficient direct evidence of its regulatory roles in CSC-associated phenotypes and signaling pathways. In this review, we have highlighted the available data on galectin-3 regulated functions pertinent to cancer stemness and explored the opportunities of its exploitation as a CSC marker and a therapeutic target.
Collapse
Affiliation(s)
- Pratima Nangia-Makker
- Department of Oncology, School of Medicine, Wayne State University, Karmanos Cancer Institute, 421 East Canfield, Detroit, MI, USA
- Karmanos Cancer Institute, 421 East Canfield, Wayne State University, Detroit, MI, USA
| | - Victor Hogan
- Department of Oncology, School of Medicine, Wayne State University, Karmanos Cancer Institute, 421 East Canfield, Detroit, MI, USA
| | - Avraham Raz
- Department of Oncology, School of Medicine, Wayne State University, Karmanos Cancer Institute, 421 East Canfield, Detroit, MI, USA
- Karmanos Cancer Institute, 421 East Canfield, Wayne State University, Detroit, MI, USA
- Department of Pathology, School of Medicine, 540 East Canfield, Wayne State University, Detroit, MI, USA
| |
Collapse
|
14
|
Potter EA, Dolgova EV, Proskurina AS, Minkevich AM, Efremov YR, Taranov OS, Omigov VV, Nikolin VP, Popova NA, Bayborodin SI, Ostanin AA, Chernykh ER, Kolchanov NA, Shurdov MA, Bogachev SS. A strategy to eradicate well-developed Krebs-2 ascites in mice. Oncotarget 2017; 7:11580-94. [PMID: 26872383 PMCID: PMC4905495 DOI: 10.18632/oncotarget.7311] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 01/26/2016] [Indexed: 11/25/2022] Open
Abstract
We describe the strategy, which allows curing experimental mice engrafted with Krebs-2 ascites. The strategy is based on the facts that i) Krebs-2 tumor-initiating stem cells (TISCs) are naturally capable of internalizing fragments of extracellular double-stranded DNA (dsDNA); ii) upon delivery into TISCs, these dsDNA fragments interfere with the on-going DNA repair process so that TISCs either die or lose their tumorigenic potential. The following 3-step regimen of therapeutic procedures leading to eradication of Krebs-2 ascites is considered. Firstly, three timed injections of cyclophosphamide (CP) exactly matching the interstrand cross-link (ICL) repair phases that lead to synchronization of ascites cells in late S/G2/M. Secondly, additional treatment of ascites 18 hours post each CP injection (at NER/HR transition timepoint) with a composite dsDNA-based preparation interfering with the NER and HR repair pathways, so that tumorigenic properties of ascites cells are compromised. Thirdly, final treatment of mice with a combination of CP and dsDNA injections as ascites cells undergo apoptotic destruction, and the surviving TAMRA+ TISCs arrested in late S/G2/M phases massively enter into G1/S, when they regain sensitivity to CP+dsDNA treatment. Thus, this regimen assures that no viable cells, particularly Krebs-2 TISCs, remain.
Collapse
Affiliation(s)
- Ekaterina A Potter
- Institute of Cytology and Genetics, Siberian Branch of The Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Evgenia V Dolgova
- Institute of Cytology and Genetics, Siberian Branch of The Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Anastasia S Proskurina
- Institute of Cytology and Genetics, Siberian Branch of The Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Alexandra M Minkevich
- Institute of Cytology and Genetics, Siberian Branch of The Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Yaroslav R Efremov
- Institute of Cytology and Genetics, Siberian Branch of The Russian Academy of Sciences, Novosibirsk 630090, Russia.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Oleg S Taranov
- The State Research Center of Virology and Biotechnology VECTOR, Novosibirsk 630559, Russia
| | - Vladimir V Omigov
- The State Research Center of Virology and Biotechnology VECTOR, Novosibirsk 630559, Russia
| | - Valeriy P Nikolin
- Institute of Cytology and Genetics, Siberian Branch of The Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Nelly A Popova
- Institute of Cytology and Genetics, Siberian Branch of The Russian Academy of Sciences, Novosibirsk 630090, Russia.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Sergey I Bayborodin
- Institute of Cytology and Genetics, Siberian Branch of The Russian Academy of Sciences, Novosibirsk 630090, Russia.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Alexander A Ostanin
- Institute of Clinical Immunology, Siberian Branch of The Russian Academy of Medical Sciences, Novosibirsk 630099, Russia
| | - Elena R Chernykh
- Institute of Clinical Immunology, Siberian Branch of The Russian Academy of Medical Sciences, Novosibirsk 630099, Russia
| | - Nikolay A Kolchanov
- Institute of Cytology and Genetics, Siberian Branch of The Russian Academy of Sciences, Novosibirsk 630090, Russia
| | | | - Sergey S Bogachev
- Institute of Cytology and Genetics, Siberian Branch of The Russian Academy of Sciences, Novosibirsk 630090, Russia
| |
Collapse
|
15
|
Abstract
Cancer stem cells (CSC) are a prominent component of the tumor bulk and extensive research has now identified them as the subpopulation responsible for tumor relapse and resistance to anti-cancer treatments. Surrounding the bulk formed of tumor cells, an extracellular matrix contributes to cancer growth; the main component of the tumor micro-environment is hyaluronan, a large disaccharide forming a molecular network surrounding the cells. The hyaluronan-dependent coat can regulate cell division and motility in cancer progression and metastasis. One of the receptors of hyaluronan is CD44, a surface protein frequently used as a CSC marker. Indeed, tumor cells with high levels of CD44 appear to exhibit CSC properties and are characterized by elevated relapse rate. The CD44-hyaluronan-dependent interactions are Janus-faced: on one side, they have been shown to be crucial in both malignancy and resistance to therapy; on the other, they represent a potential value for future therapies, as disturbing the CD44-hyaluronan axis would not only impair the pericellular matrix but also the subpopulation of self-renewing oncogenic cells. Here, we will review the key roles of HA and CD44 in CSC maintenance and propagation and will show that CSC-like spheroids from a rabdhomyosarcoma cell line, namely RD, have a prominent pericellular coat necessary for sphere formation and for elevated migration. Thus, a better understanding of the hyaluronan-CD44 interactions holds the potential for ameliorating current cancer therapies and eradicating CSC.
Collapse
|
16
|
Jayachandran A, Shrestha R, Dhungel B, Huang IT, Vasconcelos MYK, Morrison BJ, Ramlogan-Steel CA, Steel JC. Murine hepatocellular carcinoma derived stem cells reveal epithelial-to-mesenchymal plasticity. World J Stem Cells 2017; 9:159-168. [PMID: 29026462 PMCID: PMC5620425 DOI: 10.4252/wjsc.v9.i9.159] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 06/29/2017] [Accepted: 07/17/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To establish a model to enrich and characterize stem-like cells from murine normal liver and hepatocellular carcinoma (HCC) cell lines and to further investigate stem-like cell association with epithelial-to-mesenchymal transition (EMT).
METHODS In this study, we utilized a stem cell conditioned serum-free medium to enrich stem-like cells from mouse HCC and normal liver cell lines, Hepa 1-6 and AML12, respectively. We isolated the 3-dimensional spheres and assessed their stemness characteristics by evaluating the RNA levels of stemness genes and a cell surface stem cell marker by quantitative reverse transcriptase-PCR (qRT-PCR). Next, we examined the relationship between stem cells and EMT using qRT-PCR.
RESULTS Three-dimensional spheres were enriched by culturing murine HCC and normal hepatocyte cell lines in stem cell conditioned serum-free medium supplemented with epidermal growth factor, basic fibroblast growth factor and heparin sulfate. The 3-dimensional spheres had enhanced stemness markers such as Klf4 and Bmi1 and hepatic cancer stem cell (CSC) marker Cd44 compared to parental cells grown as adherent cultures. We report that epithelial markers E-cadherin and ZO-1 were downregulated, while mesenchymal markers Vimentin and Fibronectin were upregulated in 3-dimensional spheres. The 3-dimensional spheres also exhibited changes in expression of Snai, Zeb and Twist family of EMT transcription factors.
CONCLUSION Our novel method successfully enriched stem-like cells which possessed an EMT phenotype. The isolation and characterization of murine hepatic CSCs could establish a precise target for the development of more effective therapies for HCC.
Collapse
Affiliation(s)
- Aparna Jayachandran
- the University of Queensland School of Medicine and the Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, QLD 4120, Australia
| | - Ritu Shrestha
- the University of Queensland School of Medicine and the Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, QLD 4120, Australia
| | - Bijay Dhungel
- the University of Queensland School of Medicine and the Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, QLD 4120, Australia
| | - I-Tao Huang
- the University of Queensland School of Medicine and the Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, QLD 4120, Australia
| | - Marianna Yumi Kawashima Vasconcelos
- the University of Queensland School of Medicine and the Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, QLD 4120, Australia
| | - Brian J Morrison
- Viral and Rickettsial Diseases Department, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, MD 20910, United States
| | - Charmaine A Ramlogan-Steel
- the University of Queensland School of Medicine and the Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, QLD 4120, Australia
| | - Jason C Steel
- the University of Queensland School of Medicine and the Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, QLD 4120, Australia
| |
Collapse
|
17
|
Aberrantly activated Cox-2 and Wnt signaling interact to maintain cancer stem cells in glioblastoma. Oncotarget 2017; 8:82217-82230. [PMID: 29137258 PMCID: PMC5669884 DOI: 10.18632/oncotarget.19283] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 06/16/2017] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma recurrence after aggressive therapy typically occurs within six months, and patients inevitably succumb to their disease. Tumor recurrence is driven by a subpopulation of cancer stem cells in glioblastoma (glioblastoma stem-like cells, GSCs), which exhibit resistance to cytotoxic therapies, compared to their non-stem-cell counterparts. Here, we show that the Cox-2 and Wnt signaling pathways are aberrantly activated in GSCs and interact to maintain the cancer stem cell identity. Cox-2 stimulates GSC self-renewal and proliferation through prostaglandin E2 (PGE2), which in turn activates the Wnt signaling pathway. Wnt signaling underlies PGE2-induced GSC self-renewal and independently directs GSC self-renewal and proliferation. Inhibition of PGE2 enhances the effect of temozolomide on GSCs, but affords only a modest survival advantage in a xenograft model in the setting of COX-independent Wnt activation. Our findings uncover an aberrant positive feedback interaction between the Cox-2/PGE2 and Wnt pathways that mediates the stem-like state in glioblastoma.
Collapse
|
18
|
Human Cytomegalovirus-Infected Glioblastoma Cells Display Stem Cell-Like Phenotypes. mSphere 2017; 2:mSphere00137-17. [PMID: 28656174 PMCID: PMC5480031 DOI: 10.1128/msphere.00137-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 06/07/2017] [Indexed: 12/27/2022] Open
Abstract
A role for HCMV in GBMs remains controversial for several reasons. Some studies find HCMV in GBM tumors, while others do not. Few cells within a GBM may harbor HCMV, making it unclear how the virus could be contributing to the tumor phenotype without infecting every cell. Finally, HCMV does not overtly transform cells in vitro. However, tumors induced by other viruses can be treated with antiviral remedies, and initial results indicate that this may be true for anti-HCMV therapies and GBMs. With such a poor prognosis for GBM patients, any potential new intervention deserves exploration. Our work here describes an evidence-based model for how HCMV could contribute to GBM biology while infecting very few cells and without transforming them. It also illuminates why anti-HCMV treatments may be beneficial to GBM patients. Our observations provide blueprints for future in vitro studies examining how HCMV manipulates stem cell-specific pathways and future clinical studies of anti-HCMV measures as GBM therapeutics. Glioblastoma multiforme (GBM) is the most common brain tumor in adults. Human cytomegalovirus (HCMV) genomes are present in GBM tumors, yielding hope that antiviral treatments could prove therapeutic and improve the poor prognosis of GBM patients. We discovered that GBM cells infected in vitro with HCMV display properties of cancer stem cells. HCMV-infected GBM cells grow more slowly than mock-infected controls, demonstrate a higher capacity for self-renewal determined by a sphere formation assay, and display resistance to the chemotherapeutic drug temozolomide. Our data suggest that HCMV, while present in only a minority of the cells within a tumor, could contribute to the pathogenesis of GBMs by promoting or prolonging stem cell-like phenotypes, thereby perpetuating tumors in the face of chemotherapy. Importantly, we show that temozolomide sensitivity is restored by the antiviral drug ganciclovir, indicating a potential mechanism underlying the positive effects observed in GBM patients treated with antiviral therapy. IMPORTANCE A role for HCMV in GBMs remains controversial for several reasons. Some studies find HCMV in GBM tumors, while others do not. Few cells within a GBM may harbor HCMV, making it unclear how the virus could be contributing to the tumor phenotype without infecting every cell. Finally, HCMV does not overtly transform cells in vitro. However, tumors induced by other viruses can be treated with antiviral remedies, and initial results indicate that this may be true for anti-HCMV therapies and GBMs. With such a poor prognosis for GBM patients, any potential new intervention deserves exploration. Our work here describes an evidence-based model for how HCMV could contribute to GBM biology while infecting very few cells and without transforming them. It also illuminates why anti-HCMV treatments may be beneficial to GBM patients. Our observations provide blueprints for future in vitro studies examining how HCMV manipulates stem cell-specific pathways and future clinical studies of anti-HCMV measures as GBM therapeutics.
Collapse
|
19
|
Colamaio M, Tosti N, Puca F, Mari A, Gattordo R, Kuzay Y, Federico A, Pepe A, Sarnataro D, Ragozzino E, Raia M, Hirata H, Gemei M, Mimori K, Del Vecchio L, Battista S, Fusco A. HMGA1 silencing reduces stemness and temozolomide resistance in glioblastoma stem cells. Expert Opin Ther Targets 2016; 20:1169-79. [PMID: 27486901 DOI: 10.1080/14728222.2016.1220543] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Glioblastoma multiforme (GBM) develops from a small subpopulation of stem-like cells, which are endowed with the ability to self-renew, proliferate and give rise to progeny of multiple neuroepithelial lineages. These cells are resistant to conventional chemo- and radiotherapy and are hence also responsible for tumor recurrence. HMGA1 overexpression has been shown to correlate with proliferation, invasion, and angiogenesis of GBMs and to affect self-renewal of cancer stem cells from colon cancer. The role of HMGA1 in GBM tumor stem cells is not completely understood. RESEARCH DESIGN AND METHODS We have investigated the role of HMGA1 in brain tumor stem cell (BTSC) self-renewal, stemness and resistance to temozolomide by shRNA- mediated HMGA1 silencing. RESULTS We first report that HMGA1 is overexpressed in a subset of BTSC lines from human GBMs. Then, we show that HMGA1 knockdown reduces self-renewal, sphere forming efficiency and stemness, and sensitizes BTSCs to temozolomide. Interestingly, HMGA1 silencing also leads to reduced tumor initiation ability in vivo. CONCLUSIONS These results demonstrate a pivotal role of HMGA1 in cancer stem cell gliomagenesis and endorse HMGA1 as a suitable target for CSC-specific GBM therapy.
Collapse
Affiliation(s)
- Marianna Colamaio
- a Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche , Università degli Studi di Napoli 'Federico II,' Naples , Italy
| | - Nadia Tosti
- a Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche , Università degli Studi di Napoli 'Federico II,' Naples , Italy.,b Molecular Pathology Unit, Institute of Pathology , University Hospital Basel , Basel , Switzerland
| | - Francesca Puca
- a Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche , Università degli Studi di Napoli 'Federico II,' Naples , Italy
| | - Alessia Mari
- a Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche , Università degli Studi di Napoli 'Federico II,' Naples , Italy
| | - Rosaria Gattordo
- a Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche , Università degli Studi di Napoli 'Federico II,' Naples , Italy
| | - Yalçın Kuzay
- a Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche , Università degli Studi di Napoli 'Federico II,' Naples , Italy
| | - Antonella Federico
- a Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche , Università degli Studi di Napoli 'Federico II,' Naples , Italy
| | - Anna Pepe
- c Dipartimento di Medicina Molecolare e Biotecnologie Mediche , Università degli Studi di Napoli 'Federico II,' Naples , Italy
| | | | - Elvira Ragozzino
- a Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche , Università degli Studi di Napoli 'Federico II,' Naples , Italy
| | | | - Hidenari Hirata
- e Department of Surgery , Kyushu University Beppu Hospital , Beppu , Japan
| | - Marica Gemei
- d CEINGE-Biotecnologie Avanzate , Naples , Italy
| | - Koshi Mimori
- e Department of Surgery , Kyushu University Beppu Hospital , Beppu , Japan
| | | | - Sabrina Battista
- a Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche , Università degli Studi di Napoli 'Federico II,' Naples , Italy
| | - Alfredo Fusco
- a Istituto di Endocrinologia ed Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche , Università degli Studi di Napoli 'Federico II,' Naples , Italy.,f Programa de Carcinogênese Molecular , Instituto Nacional de Câncer - INCA , Rio de Janeiro , Brazil
| |
Collapse
|
20
|
Bassani B, Bartolini D, Pagani A, Principi E, Zollo M, Noonan DM, Albini A, Bruno A. Fenretinide (4-HPR) Targets Caspase-9, ERK 1/2 and the Wnt3a/β-Catenin Pathway in Medulloblastoma Cells and Medulloblastoma Cell Spheroids. PLoS One 2016; 11:e0154111. [PMID: 27367907 PMCID: PMC4930187 DOI: 10.1371/journal.pone.0154111] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/08/2016] [Indexed: 12/11/2022] Open
Abstract
Medulloblastoma (MB), a neuroectodermal tumor arising in the cerebellum, represents the most frequent childhood brain malignancy. Current treatments for MB combine radiation and chemotherapy and are often associated with relevant side effects; novel therapeutic strategies are urgently needed. N-(4-Hydroxyphenyl) retinamide (4-HPR, fenretinide), a synthetic analogue of all-trans retinoic acid, has emerged as a promising and well-tolerated cancer chemopreventive and chemotherapeutic agent for various neoplasms, from breast cancer to neuroblastoma. Here we investigated the effects of 4-HPR on MB cell lines and identified the mechanism of action for a potential use in therapy of MB. Flow cytometry analysis was performed to evaluate 4-HPR induction of apoptosis and oxygen reactive species (ROS) production, as well as cell cycle effects. Functional analysis to determine 4-HPR ability to interfere with MB cell migration and invasion were performed. Western Blot analysis were used to investigate the crucial molecules involved in selected signaling pathways associated with apoptosis (caspase-9 and PARP-1), cell survival (ERK 1/2) and tumor progression (Wnt3a and β-catenin). We show that 4-HPR induces caspase 9-dependent cell death in DAOY and ONS-76 cells, associated with increased ROS generation, suggesting that free radical intermediates might be directly involved. We observed 4-HPR induction of cell cycle arrest in G1/S phase, inactivated β-catenin, and inhibition of MB cell migration and invasion. We also evaluated the ability of 4-HPR to target MB cancer-stem/cancer-initiating cells, using an MB spheroids model, followed by flow cytometry and quantitative real-time PCR. 4-HPR treatment reduced DAOY and ONS-76 spheroid formation, in term of number and size. Decreased expression of the surface markers CD133+ and ABCG2+ as well as Oct-4 and Sox-2 gene expression were observed on BTICs treated with 4-HPR further reducing BITIC invasive activities. Finally, we analyzed 4-HPR ability to inhibit MB tumor cell growth in vivo in nude mice. Taken together, our data suggest that 4-HPR targets both parental and MB tumor stem/initiating cell-like populations. Since 4-HPR exerts low toxicity, it could represent a valid compound in the treatment of human MB.
Collapse
Affiliation(s)
- Barbara Bassani
- Scientific and Technological Pole, IRCCS MultiMedica, Milano, Italy
| | | | - Arianna Pagani
- Scientific and Technological Pole, IRCCS MultiMedica, Milano, Italy
| | - Elisa Principi
- Scientific and Technological Pole, IRCCS MultiMedica, Milano, Italy
| | - Massimo Zollo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
- Ceinge Biotecnologie Avanzate, Naples, Italy
| | - Douglas M. Noonan
- Scientific and Technological Pole, IRCCS MultiMedica, Milano, Italy
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Adriana Albini
- Scientific and Technological Pole, IRCCS MultiMedica, Milano, Italy
- * E-mail:
| | - Antonino Bruno
- Scientific and Technological Pole, IRCCS MultiMedica, Milano, Italy
| |
Collapse
|
21
|
Zucha MA, Wu ATH, Lee WH, Wang LS, Lin WW, Yuan CC, Yeh CT. Bruton's tyrosine kinase (Btk) inhibitor ibrutinib suppresses stem-like traits in ovarian cancer. Oncotarget 2016; 6:13255-68. [PMID: 26036311 PMCID: PMC4537012 DOI: 10.18632/oncotarget.3658] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 02/28/2015] [Indexed: 01/06/2023] Open
Abstract
According to a Prognoscan database, upregulation of Bruton's tyrosine kinase (Btk) is associated with low overall survival in ovarian cancer patients. We found that spheroids-forming ovarian cancer cell, which highly expressed cancer stem-like cell (CSC) markers and Btk, were cisplatin resistant. We next treated CSCs and non-CSCs by a combination of ibrutinib and cisplatin. We found that chemoresistance was dependent on Btk and JAK2/STAT3, which maintained CSC by inducing Sox-2 and prosurvival genes. We suggest that addition of ibrutinib to cisplatin may improve treatment outcome in ovarian cancer.
Collapse
Affiliation(s)
- Muhammad Ary Zucha
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan.,Department of Obstetrics and Gynecology, Gadjah Mada University-Sardjito Central Hospital, Yogyakarta, Indonesia
| | - Alexander T H Wu
- Graduate Institute of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Translational Research Laboratory, Cancer Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Wei-Hwa Lee
- Department of Pathology, Taipei Medical University-Shuang Ho Hospital, Taipei, Taiwan
| | - Liang-Shun Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wan-Wan Lin
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | - Chiou-Chung Yuan
- Obstetrics and Gynecology Department, Shuang-Ho Hospital, Taipei, Taiwan
| | - Chi-Tai Yeh
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Medical Research and Education, Taipei Medical University-Shuang Ho Hospital, Taipei, Taiwan
| |
Collapse
|
22
|
A CREB-Sirt1-Hes1 Circuitry Mediates Neural Stem Cell Response to Glucose Availability. Cell Rep 2016; 14:1195-1205. [PMID: 26804914 DOI: 10.1016/j.celrep.2015.12.092] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 08/21/2015] [Accepted: 12/20/2015] [Indexed: 01/21/2023] Open
Abstract
Adult neurogenesis plays increasingly recognized roles in brain homeostasis and repair and is profoundly affected by energy balance and nutrients. We found that the expression of Hes-1 (hairy and enhancer of split 1) is modulated in neural stem and progenitor cells (NSCs) by extracellular glucose through the coordinated action of CREB (cyclic AMP responsive element binding protein) and Sirt-1 (Sirtuin 1), two cellular nutrient sensors. Excess glucose reduced CREB-activated Hes-1 expression and results in impaired cell proliferation. CREB-deficient NSCs expanded poorly in vitro and did not respond to glucose availability. Elevated glucose also promoted Sirt-1-dependent repression of the Hes-1 promoter. Conversely, in low glucose, CREB replaced Sirt-1 on the chromatin associated with the Hes-1 promoter enhancing Hes-1 expression and cell proliferation. Thus, the glucose-regulated antagonism between CREB and Sirt-1 for Hes-1 transcription participates in the metabolic regulation of neurogenesis.
Collapse
|
23
|
Lee SB, Frattini V, Bansal M, Castano AM, Sherman D, Hutchinson K, Bruce JN, Califano A, Liu G, Cardozo T, Iavarone A, Lasorella A. An ID2-dependent mechanism for VHL inactivation in cancer. Nature 2016; 529:172-7. [PMID: 26735018 PMCID: PMC5384647 DOI: 10.1038/nature16475] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 11/23/2015] [Indexed: 01/19/2023]
Abstract
Mechanisms that maintain cancer stem cells are crucial to tumour progression. The ID2 protein supports cancer hallmarks including the cancer stem cell state. HIFα transcription factors, most notably HIF2α (also known as EPAS1), are expressed in and required for maintenance of cancer stem cells (CSCs). However, the pathways that are engaged by ID2 or drive HIF2α accumulation in CSCs have remained unclear. Here we report that DYRK1A and DYRK1B kinases phosphorylate ID2 on threonine 27 (Thr27). Hypoxia downregulates this phosphorylation via inactivation of DYRK1A and DYRK1B. The activity of these kinases is stimulated in normoxia by the oxygen-sensing prolyl hydroxylase PHD1 (also known as EGLN2). ID2 binds to the VHL ubiquitin ligase complex, displaces VHL-associated Cullin 2, and impairs HIF2α ubiquitylation and degradation. Phosphorylation of Thr27 of ID2 by DYRK1 blocks ID2-VHL interaction and preserves HIF2α ubiquitylation. In glioblastoma, ID2 positively modulates HIF2α activity. Conversely, elevated expression of DYRK1 phosphorylates Thr27 of ID2, leading to HIF2α destabilization, loss of glioma stemness, inhibition of tumour growth, and a more favourable outcome for patients with glioblastoma.
Collapse
Affiliation(s)
- Sang Bae Lee
- Institute for Cancer Genetics, Columbia University Medical Center, New York
| | - Veronique Frattini
- Institute for Cancer Genetics, Columbia University Medical Center, New York
| | - Mukesh Bansal
- Department of Systems Biology, Columbia University Medical Center, New York
- Center for Computational Biology and Bioinformatics, Columbia University Medical Center, New York
| | | | - Dan Sherman
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York
| | - Keino Hutchinson
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York
| | - Jeffrey N. Bruce
- Department of Neurosurgery, Columbia University Medical Center, New York
| | - Andrea Califano
- Department of Systems Biology, Columbia University Medical Center, New York
- Center for Computational Biology and Bioinformatics, Columbia University Medical Center, New York
| | - Guangchao Liu
- Institute for Cancer Genetics, Columbia University Medical Center, New York
| | - Timothy Cardozo
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York
| | - Antonio Iavarone
- Institute for Cancer Genetics, Columbia University Medical Center, New York
- Department of Neurology, Columbia University Medical Center, New York
- Department of Pathology, Columbia University Medical Center, New York
| | - Anna Lasorella
- Institute for Cancer Genetics, Columbia University Medical Center, New York
- Department of Pathology, Columbia University Medical Center, New York
- Department of Pediatrics, Columbia University Medical Center, New York
| |
Collapse
|
24
|
Martuscello RT, Vedam-Mai V, McCarthy DJ, Schmoll ME, Jundi MA, Louviere CD, Griffith BG, Skinner CL, Suslov O, Deleyrolle LP, Reynolds BA. A Supplemented High-Fat Low-Carbohydrate Diet for the Treatment of Glioblastoma. Clin Cancer Res 2015; 22:2482-95. [PMID: 26631612 DOI: 10.1158/1078-0432.ccr-15-0916] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 11/12/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased interest in metabolic therapies. Glioblastoma (GB) is highly malignant, very metabolically active, and typically resistant to current therapies. Dietary treatment options based on glucose deprivation have been explored using a restrictive ketogenic diet (KD), with positive anticancer reports. However, negative side effects and a lack of palatability make the KD difficult to implement in an adult population. Hence, we developed a less stringent, supplemented high-fat low-carbohydrate (sHFLC) diet that mimics the metabolic and antitumor effects of the KD, maintains a stable nutritional profile, and presents an alternative clinical option for diverse patient populations. EXPERIMENTAL DESIGN The dietary paradigm was tested in vitro and in vivo, utilizing multiple patient-derived gliomasphere lines. Cellular proliferation, clonogenic frequency, and tumor stem cell population effects were determined in vitro using the neurosphere assay (NSA). Antitumor efficacy was tested in vivo in preclinical xenograft models and mechanistic regulation via the mTOR pathway was explored. RESULTS Reducing glucose in vitro to physiologic levels, coupled with ketone supplementation, inhibits proliferation of GB cells and reduces tumor stem cell expansion. In vivo, while maintaining animal health, the sHFLC diet significantly reduces the growth of tumor cells in a subcutaneous model of tumor progression and increases survival in an orthotopic xenograft model. Dietary-mediated anticancer effects correlate with the reduction of mTOR effector expression. CONCLUSIONS We demonstrate that the sHFLC diet is a viable treatment alternative to the KD, and should be considered for clinical testing. Clin Cancer Res; 22(10); 2482-95. ©2015 AACR.
Collapse
Affiliation(s)
- Regina T Martuscello
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida. Interdisciplinary Program in Biomedical Sciences, Neuroscience, College of Medicine, University of Florida, Gainesville, Florida
| | - Vinata Vedam-Mai
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida. Center for Movement Disorders and Neuro-restoration, University of Florida, Gainesville, Florida
| | - David J McCarthy
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida
| | - Michael E Schmoll
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida
| | - Musa A Jundi
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida
| | - Christopher D Louviere
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida
| | - Benjamin G Griffith
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida
| | - Colby L Skinner
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida
| | - Oleg Suslov
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida
| | - Loic P Deleyrolle
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida.
| | - Brent A Reynolds
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida. Interdisciplinary Program in Biomedical Sciences, Neuroscience, College of Medicine, University of Florida, Gainesville, Florida.
| |
Collapse
|
25
|
Ghanbari A, Esmaeilpour T, Bahmanpour S, Golmohammadi MG, Sharififar S, Azari H. Depletion of neural stem cells from the subventricular zone of adult mouse brain using cytosine b-Arabinofuranoside. Brain Behav 2015; 5:e00404. [PMID: 26664789 PMCID: PMC4667764 DOI: 10.1002/brb3.404] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 08/02/2015] [Accepted: 09/02/2015] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Neural stem cells (NSCs) reside along the ventricular axis of the mammalian brain. They divide infrequently to maintain themselves and the down-stream progenitors. Due to the quiescent property of NSCs, attempts to deplete these cells using antimitotic agents such as cytosine b-Aarabinofuranoside (Ara-C) have not been successful. We hypothesized that implementing infusion gaps in Ara-C kill paradigms would recruit the quiescent NSCs and subsequently eliminate them from their niches in the subventricular zone (SVZ). METHODS We infused the right lateral ventricle of adult mice brain with 2% Ara-C using four different paradigms--1: one week; 2: two weeks; 3, 4: two weeks with an infusion gap of 6 and 12 h on day 7. Neurosphere assay (NSA), neural colony-forming cell assay (N-CFCA) and immunofluorescent staining were used to assess depletion of NSCs from the SVZ. RESULTS Neurosphere formation dramatically decreased in all paradigms immediately after Ara-C infusion. Reduction in neurosphere formation was more pronounced in the 3rd and 4th paradigms. Interestingly 1 week after Ara-C infusion, neurosphere formation recovered toward control values implying the presence of NSCs in the harvested SVZ tissue. Unexpectedly, N-CFCA in the 3rd paradigm, as one of the most effective paradigms, did not result in formation of NSC-derived colonies (colonies >2 mm) even from SVZs harvested 1 week after completion of Ara-C infusion. However, formation of big colonies with serial passaging capability, again confirmed the presence of NSCs. CONCLUSIONS Overall, these data suggest Ara-C kill paradigms with infusion gaps deplete NSCs in the SVZ more efficiently but the niches would repopulate even after the most vigorous kill paradigm used in this study.
Collapse
Affiliation(s)
- Amir Ghanbari
- Neural Stem Cell and Regenerative Neuroscience Laboratory Department of Anatomical Sciences Shiraz School of Medicine Shiraz University of Medical Sciences Shiraz Iran
| | - Tahereh Esmaeilpour
- Neural Stem Cell and Regenerative Neuroscience Laboratory Department of Anatomical Sciences Shiraz School of Medicine Shiraz University of Medical Sciences Shiraz Iran
| | - Soghra Bahmanpour
- Neural Stem Cell and Regenerative Neuroscience Laboratory Department of Anatomical Sciences Shiraz School of Medicine Shiraz University of Medical Sciences Shiraz Iran
| | | | - Sharareh Sharififar
- Department of Physical Therapy College of Public Health and Health Professions University of Florida Gainesville Florida
| | - Hassan Azari
- Neural Stem Cell and Regenerative Neuroscience Laboratory Department of Anatomical Sciences Shiraz School of Medicine Shiraz University of Medical Sciences Shiraz Iran ; Neural Stem Cell and Regenerative Neuroscience Laboratory Shiraz Stem Cell Institute Shiraz University of Medical Sciences Shiraz Iran
| |
Collapse
|
26
|
Norton KA, Popel AS. An agent-based model of cancer stem cell initiated avascular tumour growth and metastasis: the effect of seeding frequency and location. J R Soc Interface 2015; 11:20140640. [PMID: 25185580 DOI: 10.1098/rsif.2014.0640] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
It is very important to understand the onset and growth pattern of breast primary tumours as well as their metastatic dissemination. In most cases, it is the metastatic disease that ultimately kills the patient. There is increasing evidence that cancer stem cells are closely linked to the progression of the metastatic tumour. Here, we investigate stem cell seeding to an avascular tumour site using an agent-based stochastic model of breast cancer metastatic seeding. The model includes several important cellular features such as stem cell symmetric and asymmetric division, migration, cellular quiescence, senescence, apoptosis and cell division cycles. It also includes external features such as stem cell seeding frequency and location. Using this model, we find that cell seeding rate and location are important features for tumour growth. We also define conditions in which the tumour growth exhibits decremented and exponential growth patterns. Overall, we find that seeding, senescence and division limit affect not only the number of stem cells, but also their spatial and temporal distribution.
Collapse
Affiliation(s)
- Kerri-Ann Norton
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21205, USA
| |
Collapse
|
27
|
Rahman M, Reyner K, Deleyrolle L, Millette S, Azari H, Day BW, Stringer BW, Boyd AW, Johns TG, Blot V, Duggal R, Reynolds BA. Neurosphere and adherent culture conditions are equivalent for malignant glioma stem cell lines. Anat Cell Biol 2015; 48:25-35. [PMID: 25806119 PMCID: PMC4371178 DOI: 10.5115/acb.2015.48.1.25] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 01/07/2015] [Accepted: 01/07/2015] [Indexed: 12/13/2022] Open
Abstract
Certain limitations of the neurosphere assay (NSA) have resulted in a search for alternative culture techniques for brain tumor-initiating cells (TICs). Recently, reports have described growing glioblastoma (GBM) TICs as a monolayer using laminin. We performed a side-by-side analysis of the NSA and laminin (adherent) culture conditions to compare the growth and expansion of GBM TICs. GBM cells were grown using the NSA and adherent culture conditions. Comparisons were made using growth in culture, apoptosis assays, protein expression, limiting dilution clonal frequency assay, genetic affymetrix analysis, and tumorigenicity in vivo. In vitro expansion curves for the NSA and adherent culture conditions were virtually identical (P=0.24) and the clonogenic frequencies (5.2% for NSA vs. 5.0% for laminin, P=0.9) were similar as well. Likewise, markers of differentiation (glial fibrillary acidic protein and beta tubulin III) and proliferation (Ki67 and MCM2) revealed no statistical difference between the sphere and attachment methods. Several different methods were used to determine the numbers of dead or dying cells (trypan blue, DiIC, caspase-3, and annexin V) with none of the assays noting a meaningful variance between the two methods. In addition, genetic expression analysis with microarrays revealed no significant differences between the two groups. Finally, glioma cells derived from both methods of expansion formed large invasive tumors exhibiting GBM features when implanted in immune-compromised animals. A detailed functional, protein and genetic characterization of human GBM cells cultured in serum-free defined conditions demonstrated no statistically meaningful differences when grown using sphere (NSA) or adherent conditions. Hence, both methods are functionally equivalent and remain suitable options for expanding primary high-grade gliomas in tissue culture.
Collapse
Affiliation(s)
- Maryam Rahman
- Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Karina Reyner
- Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Loic Deleyrolle
- Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | | | - Hassan Azari
- Department of Neurosurgery, University of Florida, Gainesville, FL, USA. ; Department of Anatomical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bryan W Day
- Brain Cancer Research Unit, Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Brett W Stringer
- Brain Cancer Research Unit, Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Andrew W Boyd
- Brain Cancer Research Unit, Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Terrance G Johns
- Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Vincent Blot
- CovX Research, Pfizer Worldwide Research and Development, San Diego, CA, USA
| | | | - Brent A Reynolds
- Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| |
Collapse
|
28
|
Demir M, Laywell ED. Neurotoxic effects of AZT on developing and adult neurogenesis. Front Neurosci 2015; 9:93. [PMID: 25852464 PMCID: PMC4367529 DOI: 10.3389/fnins.2015.00093] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 03/05/2015] [Indexed: 11/18/2022] Open
Abstract
Azidothymidine (AZT) is a synthetic, chain-terminating nucleoside analog used to treat HIV-1 infection. While AZT is not actively transported across the blood brain barrier, it does accumulate at high levels in cerebrospinal fluid, and subsequently diffuses into the overlying parenchyma. Due to the close anatomical proximity of the neurogenic niches to the ventricular system, we hypothesize that diffusion from CSF exposes neural stem/progenitor cells and their progeny to biologically relevant levels of AZT sufficient to perturb normal cell functions. We employed in vitro and in vivo models of mouse neurogenesis in order to assess the effects of AZT on developing and adult neurogenesis. Using in vitro assays we show that AZT reduces the population expansion potential of neural stem/progenitor cells by inducing senescence. Additionally, in a model of in vitro neurogenesis AZT severely attenuates neuroblast production. These effects are mirrored in vivo by clinically-relevant animal models. We show that in utero AZT exposure perturbs both population expansion and neurogenesis among neural stem/progenitor cells. Additionally, a short-term AZT regimen in adult mice suppresses subependymal zone neurogenesis. These data reveal novel negative effects of AZT on neural stem cell biology. Given that the sequelae of HIV infection often include neurologic deficits—subsumed under AIDS Dementia Complex (Brew, 1999)—it is important to determine to what extent AZT negatively affects neurological function in ways that contribute to, or exacerbate, ADC in order to avoid attributing iatrogenic drug effects to the underlying disease process, and thereby skewing the risk/benefit analysis of AZT therapy.
Collapse
Affiliation(s)
- Meryem Demir
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida Gainesville, FL, USA
| | - Eric D Laywell
- Department of Biomedical Sciences, College of Medicine, Florida State University Tallahassee, FL, USA
| |
Collapse
|
29
|
Zhang X, Lou Y, Wang H, Zheng X, Dong Q, Sun J, Han B. Wnt signaling regulates the stemness of lung cancer stem cells and its inhibitors exert anticancer effect on lung cancer SPC-A1 cells. Med Oncol 2015; 32:95. [PMID: 25731617 DOI: 10.1007/s12032-014-0462-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 12/15/2014] [Indexed: 10/23/2022]
Abstract
Wnt signaling plays an important role in regulating the activity of cancer stem cells (CSCs) in a variety of cancers. In this study, we explored the role of Wnt signaling in the lung cancer stem cells (LCSCs). LCSCs were obtained by sphere culture, for which human lung adenocarcinoma cell line SPC-A1 was treated with IGF, EGF and FGF-10. The stemness of LCSCs was confirmed by immunofluorescence, and pathway analysis was performed by functional genome screening and RT-PCR. The relationship between the identified signaling pathway and the expression of the stemness genes was explored by agonist/antagonist assay. Moreover, the effects of different signaling molecule inhibitors on sphere formation, cell viability and colony formation were also analyzed. The results showed that LCSCs were successfully generated as they expressed pluripotent stem cell markers Nanog and Oct 4, and lung distal epithelial markers CCSP and SP-C, by which the phenotype characterization of stem cells can be confirmed. The involvement of Wnt pathway in LCSCs was identified by functional genome screening and verified by RT-PCR. The expression of Wnt signaling components was closely related to the expression of the Nanog and Oct 4. Furthermore, targeting Wnt signaling pathway by using different signaling molecule inhibitors can exert anticancer effects. In conclusion, Wnt signaling pathway is involved in the stemness regulation of LCSCs and might be considered as a potential therapeutic target in lung adenocarcinoma.
Collapse
Affiliation(s)
- Xueyan Zhang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai, 200000, China
| | | | | | | | | | | | | |
Collapse
|
30
|
Pham K, Luo D, Siemann DW, Law BK, Reynolds BA, Hothi P, Foltz G, Harrison JK. VEGFR inhibitors upregulate CXCR4 in VEGF receptor-expressing glioblastoma in a TGFβR signaling-dependent manner. Cancer Lett 2015; 360:60-7. [PMID: 25676691 DOI: 10.1016/j.canlet.2015.02.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 02/03/2015] [Accepted: 02/03/2015] [Indexed: 11/16/2022]
Abstract
The failure of standard treatment for patients diagnosed with glioblastoma (GBM) coupled with the highly vascularized nature of this solid tumor has led to the consideration of agents targeting VEGF or VEGFRs, as alternative therapeutic strategies for this disease. Despite modest achievements in survival obtained with such treatments, failure to maintain an enduring survival benefit and more invasive relapsing tumors are evident. Our study suggests a potential mechanism by which anti-VEGF/VEGFR therapies regulate the enhanced invasive phenotype through a pathway that involves TGFβR and CXCR4. VEGFR signaling inhibitors (Cediranib and Vandetanib) elevated the expression of CXCR4 in VEGFR-expressing GBM cell lines and tumors, and enhanced the in vitro migration of these lines toward CXCL12. The combination of VEGFR inhibitor and CXCR4 antagonist provided a greater survival benefit to tumor-bearing animals. The upregulation of CXCR4 by VEGFR inhibitors was dependent on TGFβ/TGFβR, but not HGF/MET, signaling activity, suggesting a mechanism of crosstalk among VEGF/VEGFR, TGFβ/TGFβR, and CXCL12/CXCR4 pathways in the malignant phenotype of recurrent tumors after anti-VEGF/VEGFR therapies. Thus, the combination of VEGFR, CXCR4, and TGFβR inhibitors could provide an alternative strategy to halt GBM progression.
Collapse
Affiliation(s)
- Kien Pham
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Defang Luo
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Dietmar W Siemann
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Department of Radiation Oncology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Brian K Law
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Brent A Reynolds
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Parvinder Hothi
- The Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA 98122, USA
| | - Gregory Foltz
- The Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA 98122, USA
| | - Jeffrey K Harrison
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
31
|
HMGA1 silencing restores normal stem cell characteristics in colon cancer stem cells by increasing p53 levels. Oncotarget 2015; 5:3234-45. [PMID: 24833610 PMCID: PMC4102806 DOI: 10.18632/oncotarget.1914] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
High-mobility group A1 (HMGA1) proteins are architectural chromatinic proteins, abundantly expressed during embryogenesis and in most cancer tissues, but expressed at low levels or absent in normal adult tissues. Several studies have demonstrated that HMGA1 proteins play a causal role in neoplastic cell transformation. The aim of this study was to investigate the role of these proteins in the control of cancer stem cells (CSCs), which have emerged as a preferred target in cancer therapy, because of their role in cancer recurrence. We observed that HMGA1 is overexpressed in colon tumour stem cell (CTSC) lines compared to normal and colon cancer tissues. We demonstrated that HMGA1 silencing in CTSCs increases stem cell quiescence and reduces self-renewal and sphere-forming efficiency (SFE). The latter, together with the upregulation and asymmetric distribution of NUMB, is indicative of the recovery of an asymmetric division pattern, typical of normal stem cells. We further found that HMGA1 transcriptionally regulates p53, which is known to control the balance between symmetric and asymmetric divisions in CSCs. Therefore, our data indicate a critical role for HMGA1 in regulating both self-renewal and the symmetric/asymmetric division ratio in CSCs, suggesting that blocking HMGA1 function may be an effective anti-cancer therapy.
Collapse
|
32
|
Abstract
This review discusses quantitative modeling studies of stem and non-stem cancer cell interactions and the fraction of cancer stem cells.
Collapse
Affiliation(s)
- Heiko Enderling
- Department of Integrated Mathematical Oncology
- H. Lee Moffitt Cancer Center & Research Institute
- Tampa
- USA
| |
Collapse
|
33
|
Impairment of lysosomal activity as a therapeutic modality targeting cancer stem cells of embryonal rhabdomyosarcoma cell line RD. PLoS One 2014; 9:e110340. [PMID: 25329465 PMCID: PMC4203792 DOI: 10.1371/journal.pone.0110340] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 09/21/2014] [Indexed: 12/23/2022] Open
Abstract
Rhabdomyosarcoma is the most frequent soft tissue sarcoma in children and adolescents, with a high rate of relapse that dramatically affects the clinical outcome. Multiagent chemotherapy, in combination with surgery and/or radiation therapy, is the treatment of choice. However, the relapse rate is disappointingly high and identification of new therapeutic tools is urgently needed. Under this respect, the selective block of key features of cancer stem cells (CSC) appears particularly promising. In this study, we isolated rhabdomyosarcoma CSC with stem-like features (high expression of NANOG and OCT3/4, self-renewal ability, multipotency). Rhabdomyosarcoma CSC showed higher invasive ability and a reduced cytotoxicity to doxorubicin in comparison to native cells, through a mechanism unrelated to the classical multidrug resistance process. This was dependent on a high level of lysosome acidity mediated by a high expression of vacuolar ATPase (V-ATPase). Since it was not associated with other paediatric cancers, like Ewing’s sarcoma and neuroblastoma, V-ATPase higher expression in CSC was rhabdomyosarcoma specific. Inhibition of lysosomal acidification by the V-ATPase inhibitor omeprazole, or by specific siRNA silencing, significantly enhanced doxorubicin cytoxicity. Unexpectedly, lysosomal targeting also blocked cell growth and reduced the invasive potential of rhabdomyosarcoma CSC, even at very low doses of omeprazole (10 and 50 µM, respectively). Based on these observations, we propose lysosome acidity as a valuable target to enhance chemosensitivity of rhabdomyosarcoma CSC, and suggest the use of anti-V-ATPase agents in combination with standard regimens as a promising tool for the eradication of minimal residual disease or the prevention of metastatic disease.
Collapse
|
34
|
Spina R, Filocamo G, Iaccino E, Scicchitano S, Lupia M, Chiarella E, Mega T, Bernaudo F, Pelaggi D, Mesuraca M, Pazzaglia S, Semenkow S, Bar EE, Kool M, Pfister S, Bond HM, Eberhart CG, Steinkühler C, Morrone G. Critical role of zinc finger protein 521 in the control of growth, clonogenicity and tumorigenic potential of medulloblastoma cells. Oncotarget 2014; 4:1280-92. [PMID: 23907569 PMCID: PMC3787157 DOI: 10.18632/oncotarget.1176] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The stem cell-associated transcription co-factor ZNF521 has been implicated in the control of hematopoietic, osteo-adipogenic and neural progenitor cells. ZNF521 is highly expressed in cerebellum and in particular in the neonatal external granule layer that contains candidate medulloblastoma cells-of-origin, and in the majority of human medulloblastomas. Here we have explored its involvement in the control of human and murine medulloblastoma cells. The effect of ZNF521 on growth and tumorigenic potential of human medulloblastoma cell lines as well as primary Ptc1−/+ mouse medulloblastoma cells was investigated in a variety of in vitro and in vivo assays, by modulating its expression using lentiviral vectors carrying the ZNF521 cDNA, or shRNAs that silence its expression. Enforced overexpression of ZNF521 in DAOY medulloblastoma cells significantly increased their proliferation, growth as spheroids and ability to generate clones in single-cell cultures and semisolid media, and enhanced their migratory ability in wound-healing assays. Importantly, ZNF521-expressing cells displayed a greatly enhanced tumorigenic potential in nude mice. All these activities required the ZNF521 N-terminal motif that recruits the nucleosome remodeling and histone deacetylase complex, which might therefore represent an appealing therapeutic target. Conversely, silencing of ZNF521 in human UW228 medulloblastoma cells that display high baseline expression decreased their proliferation, clonogenicity, sphere formation and wound-healing ability. Similarly, Zfp521 silencing in mouse Ptc1−/+ medulloblastoma cells drastically reduced their growth and tumorigenic potential. Our data strongly support the notion that ZNF521, through the recruitment of the NuRD complex, contributes to the clonogenic growth, migration and tumorigenicity of medulloblastoma cells.
Collapse
Affiliation(s)
- Raffaella Spina
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Dept. of Experimental and Clinical Medicine, University of Catanzaro Magna Græcia, Catanzaro, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Liu X, Johnson S, Liu S, Kanojia D, Yue W, Singh UP, Singn U, Wang Q, Wang Q, Nie Q, Chen H. Nonlinear growth kinetics of breast cancer stem cells: implications for cancer stem cell targeted therapy. Sci Rep 2014; 3:2473. [PMID: 23959163 PMCID: PMC3747506 DOI: 10.1038/srep02473] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 08/05/2013] [Indexed: 01/01/2023] Open
Abstract
Cancer stem cells (CSCs) have been identified in primary breast cancer tissues and cell lines. The CSC population varies widely among cancerous tissues and cell lines, and is often associated with aggressive breast cancers. Despite of intensive research, how the CSC population is regulated within a tumor is still not well understood so far. In this paper, we present a mathematical model to explore the growth kinetics of CSC population both in vitro and in vivo. Our mathematical models and supporting experiments suggest that there exist non-linear growth kinetics of CSCs and negative feedback mechanisms to control the balance between the population of CSCs and that of non-stem cancer cells. The model predictions can help us explain a few long-standing questions in the field of cancer stem cell research, and can be potentially used to predict the efficicacy of anti-cancer therapy.
Collapse
Affiliation(s)
- Xinfeng Liu
- Department of Mathematics, University of South Carolina, Columbia, SC. 29208
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Zanini C, Forni M. The cell block technique revisited for cells cultured in adherence and as "spheres". Histochem Cell Biol 2013; 140:685-90. [PMID: 24013649 DOI: 10.1007/s00418-013-1139-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2013] [Indexed: 12/01/2022]
Abstract
A simple modification of the cell block technique for cultured cells grown in different conditions and suitable for the construction of tissue micro arrays (TMA) is described. The application of mechanical stirring during clot formation allows the uniform dispersion of cells in the fibrin mesh, thus increasing the final volume of the embedded material of evenly distributed cells. This technique is easily applied to spheres-obtained from cell lines cultured under appropriate conditions-that are enriched in stem cells. The possibility of constructing TMA using cell lines (grown in adherence and as spheres) and samples of the corresponding tumors or normal tissues may allow the direct comparison of original tumors with in vitro-expanded cell lines.
Collapse
Affiliation(s)
- Cristina Zanini
- EuroClone S.p.A Research Laboratory, Molecular Biotechnology Centre (MBC), University of Turin, Turin, Italy,
| | | |
Collapse
|
37
|
Vik-Mo EO, Nyakas M, Mikkelsen BV, Moe MC, Due-Tønnesen P, Suso EMI, Sæbøe-Larssen S, Sandberg C, Brinchmann JE, Helseth E, Rasmussen AM, Lote K, Aamdal S, Gaudernack G, Kvalheim G, Langmoen IA. Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunol Immunother 2013; 62:1499-509. [PMID: 23817721 PMCID: PMC3755221 DOI: 10.1007/s00262-013-1453-3] [Citation(s) in RCA: 229] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 06/17/2013] [Indexed: 01/31/2023]
Abstract
Background The growth and recurrence of several cancers appear to be driven by a population of cancer stem cells (CSCs). Glioblastoma, the most common primary brain tumor, is invariably fatal, with a median survival of approximately 1 year. Although experimental data have suggested the importance of CSCs, few data exist regarding the potential relevance and importance of these cells in a clinical setting. Methods We here present the first seven patients treated with a dendritic cell (DC)-based vaccine targeting CSCs in a solid tumor. Brain tumor biopsies were dissociated into single-cell suspensions, and autologous CSCs were expanded in vitro as tumorspheres. From these, CSC-mRNA was amplified and transfected into monocyte-derived autologous DCs. The DCs were aliquoted to 9–18 vaccines containing 107 cells each. These vaccines were injected intradermally at specified intervals after the patients had received a standard 6-week course of post-operative radio-chemotherapy. The study was registered with the ClinicalTrials.gov identifier NCT00846456. Results Autologous CSC cultures were established from ten out of eleven tumors. High-quality RNA was isolated, and mRNA was amplified in all cases. Seven patients were able to be weaned from corticosteroids to receive DC immunotherapy. An immune response induced by vaccination was identified in all seven patients. No patients developed adverse autoimmune events or other side effects. Compared to matched controls, progression-free survival was 2.9 times longer in vaccinated patients (median 694 vs. 236 days, p = 0.0018, log-rank test). Conclusion These findings suggest that vaccination against glioblastoma stem cells is safe, well-tolerated, and may prolong progression-free survival. Electronic supplementary material The online version of this article (doi:10.1007/s00262-013-1453-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Einar Osland Vik-Mo
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research, University of Oslo, Oslo, Norway.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Smart CE, Morrison BJ, Saunus JM, Vargas AC, Keith P, Reid L, Wockner L, Askarian-Amiri M, Sarkar D, Simpson PT, Clarke C, Schmidt CW, Reynolds BA, Lakhani SR, Lopez JA. In vitro analysis of breast cancer cell line tumourspheres and primary human breast epithelia mammospheres demonstrates inter- and intrasphere heterogeneity. PLoS One 2013; 8:e64388. [PMID: 23750209 PMCID: PMC3672101 DOI: 10.1371/journal.pone.0064388] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 04/12/2013] [Indexed: 12/19/2022] Open
Abstract
Mammosphere and breast tumoursphere culture have gained popularity as in vitro assays for propagating and analysing normal and cancer stem cells. Whether the spheres derived from different sources or parent cultures themselves are indeed single entities enriched in stem/progenitor cells compared to other culture formats has not been fully determined. We surveyed sphere-forming capacity across 26 breast cell lines, immunophenotyped spheres from six luminal- and basal-like lines by immunohistochemistry and flow cytometry and compared clonogenicity between sphere, adherent and matrigel culture formats using in vitro functional assays. Analyses revealed morphological and molecular intra- and inter-sphere heterogeneity, consistent with adherent parental cell line phenotypes. Flow cytometry showed sphere culture does not universally enrich for markers previously associated with stem cell phenotypes, although we found some cell-line specific changes between sphere and adherent formats. Sphere-forming efficiency was significantly lower than adherent or matrigel clonogenicity and constant over serial passage. Surprisingly, self-renewal capacity of sphere-derived cells was similar/lower than other culture formats. We observed significant correlation between long-term-proliferating-cell symmetric division rates in sphere and adherent cultures, suggesting functional overlap between the compartments sustaining them. Experiments with normal primary human mammary epithelia, including sorted luminal (MUC1+) and basal/myoepithelial (CD10+) cells revealed distinct luminal-like, basal-like and mesenchymal entities amongst primary mammospheres. Morphological and colony-forming-cell assay data suggested mammosphere culture may enrich for a luminal progenitor phenotype, or induce reversion/relaxation of the basal/mesenchymal in vitro selection occurring with adherent culture. Overall, cell line tumourspheres and primary mammospheres are not homogenous entities enriched for stem cells, suggesting a more cautious approach to interpreting data from these assays and careful consideration of its limitations. Sphere culture may represent an alternative 3-dimensional culture system which rather than universally ‘enriching’ for stem cells, has utility as one of a suite of functional assays that provide a read-out of progenitor activity.
Collapse
Affiliation(s)
- Chanel E Smart
- UQ Centre for Clinical Research (UQCCR), The University of Queensland, Brisbane, Queensland, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Zanini C, Ercole E, Mandili G, Salaroli R, Poli A, Renna C, Papa V, Cenacchi G, Forni M. Medullospheres from DAOY, UW228 and ONS-76 cells: increased stem cell population and proteomic modifications. PLoS One 2013; 8:e63748. [PMID: 23717474 PMCID: PMC3663798 DOI: 10.1371/journal.pone.0063748] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 04/05/2013] [Indexed: 12/18/2022] Open
Abstract
Background Medulloblastoma (MB) is an aggressive pediatric tumor of the Central Nervous System (CNS) usually treated according to a refined risk stratification. The study of cancer stem cells (CSC) in MB is a promising approach aimed at finding new treatment strategies. Methodology/Principal Findings The CSC compartment was studied in three characterized MB cell lines (DAOY, UW228 and ONS-76) grown in standard adhesion as well as being grown as spheres, which enables expansion of the CSC population. MB cell lines, grown in adherence and as spheres, were subjected to morphologic analysis at the light and electron microscopic level, as well as cytofluorimetric determinations. Medullospheres (MBS) were shown to express increasingly immature features, along with the stem cells markers: CD133, Nestin and β-catenin. Proteomic analysis highlighted the differences between MB cell lines, demonstrating a unique protein profile for each cell line, and minor differences when grown as spheres. In MBS, MALDI-TOF also identified some proteins, that have been linked to tumor progression and resistance, such as Nucleophosmin (NPM). In addition, immunocytochemistry detected Sox-2 as a stemness marker of MBS, as well as confirming high NPM expression. Conclusions/Significance Culture conditioning based on low attachment flasks and specialized medium may provide new data on the staminal compartment of CNS tumors, although a proteomic profile of CSC is still elusive for MB.
Collapse
Affiliation(s)
- Cristina Zanini
- EuroClone S.p.A Research Laboratory, Molecular Biotechnology Centre-MBC, University of Turin, Turin, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Salerno M, Avnet S, Bonuccelli G, Eramo A, De Maria R, Gambarotti M, Gamberi G, Baldini N. Sphere-forming cell subsets with cancer stem cell properties in human musculoskeletal sarcomas. Int J Oncol 2013; 43:95-102. [PMID: 23636271 DOI: 10.3892/ijo.2013.1927] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 03/04/2013] [Indexed: 11/05/2022] Open
Abstract
Musculoskeletal sarcomas are aggressive malignancies often characterized by an adverse prognosis despite the use of intense multiagent chemotherapy or molecular targeted therapy in combination to surgery and radiotherapy. Stem-like cells identified within solid tumors have been recently implicated in drug resistance, metastasis and local relapse. Here, we report the identification of putative cancer stem cells (CSCs) in sarcomas using a sphere culture system. These sarcospheres, able to grow in anchorage-independent and serum-starved conditions, express the pluripotent embryonic stem cell marker genes OCT3/4, Nanog and SOX2. Expression levels of these genes were greater in sarcospheres than in the parental tumor cultures. Importantly, the isolated tumor spheres transplanted into mice were tumorigenic and capable of recapitulating the human disease. Finally, we demonstrated that low (1%) O2 conditions, reproducing those found within the tumor microenvironment, significantly increase the number and the size of sarcospheres. The sphere formation assay is, therefore, a valuable method for the isolation of putative CSCs from human sarcomas and its efficiency is improved by controlling oxygen availability. This method provides a reliable preclinical model that can be used for future studies aimed at investigating crucial aspects of sarcoma biology, such as resistance to treatments and relapse.
Collapse
Affiliation(s)
- Manuela Salerno
- Department of Orthopaedic Pathophysiology and Regenerative Medicine, Istituto Ortopedico Rizzoli, Bologna, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Lung cancer-initiating cells: a novel target for cancer therapy. Target Oncol 2013; 8:159-172. [PMID: 23314952 PMCID: PMC3763165 DOI: 10.1007/s11523-012-0247-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 12/20/2012] [Indexed: 12/20/2022]
Abstract
Lung cancer is a major public health problem causing more deaths than any other cancer. A better understanding of the biology of this disease and improvements in treatment are greatly needed. Increasing evidence supports the concept that a rare and specialized population of cancer cells, so-called cancer-initiating cells with stem cell-like characteristics, is responsible for tumor growth, maintenance, and recurrence. Cancer-initiating cells also exhibit characteristics that render them resistant to both radiation and chemotherapy, and therefore they are believed to play a role in treatment failure. This has led to the hypothesis that traditional therapies that indiscriminately kill tumor cells will not be as effective as therapies that selectively target cancer-initiating cells. Investigating putative cancer-initiating cells in lung cancer will greatly benefit the understanding of the origins of this disease and may lead to novel approaches to therapy by suggesting markers for use in either further isolating this population for study or for selectively targeting these cells. This review will discuss (1) lung cancer, (2) stem cells, and the role of cancer-initiating cells in tumorigenesis; (3) markers and functional characteristics associated with lung cancer-initiating cells; and (4) the potential to selectively target this subpopulation of tumor cells.
Collapse
|
42
|
Louis SA, Mak CKH. Enumerating stem cell frequency: neural colony forming cell assay. Methods Mol Biol 2013; 1059:117-32. [PMID: 23934839 DOI: 10.1007/978-1-62703-574-3_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Recent reports have highlighted several parameters of the neurosphere culture or assay system which render it unreliable as a quantitative in vitro assay for measuring neural stem cell (NSC) frequency. The single-step semi-solid based assay, the Neural Colony Forming Cell (NCFC) assay is an assay which was developed to overcome some of the limitations of the neurospheres assay in terms of accurately measuring NSC numbers. The NCFC assay allows the discrimination between NSCs and progenitors by the size of colonies they produce (i.e. their proliferative potential). The NCFC assay and other improved tissue culture tools offer further advances in the promising application of NSCs for therapeutic use.
Collapse
|
43
|
Morrison BJ, Hastie ML, Grewal YS, Bruce ZC, Schmidt C, Reynolds BA, Gorman JJ, Lopez JA. Proteomic comparison of mcf-7 tumoursphere and monolayer cultures. PLoS One 2012; 7:e52692. [PMID: 23285151 PMCID: PMC3527578 DOI: 10.1371/journal.pone.0052692] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 11/21/2012] [Indexed: 01/09/2023] Open
Abstract
Breast cancer is a heterogenous disease, composed of tumour cells with differing gene expressions and phenotypes. Very few antigens have been identified and a better understanding of tumour initiating-cells as targets for therapy is critically needed. Recently, a rare subpopulation of cells within tumours has been described with the ability to: (i) initiate and sustain tumour growth; (ii) resist traditional therapies and allow for secondary tumour dissemination; and (iii) display some of the characteristics of stem cells such as self-renewal. These cells are termed tumour-initiating cells or cancer stem cells, or alternatively, in the case of breast cancer, breast cancer stem cells. Previous studies have demonstrated that breast cancer stem cells can be enriched for in “tumoursphere” culture. Proteomics represents a novel way to investigate protein expression between cells. We hypothesise that characterisation of the proteome of the breast cancer line MCF-7 tumourspheres compared to adherent/differentiated cells identifies proteins of novel interest for further isolating or targeting breast cancer stem cells. We present evidence that: (i) the proteome of adherent cells is different to the proteome of cells grown in sphere medium from either early passage (passage 2) or late passage (passage 5) spheres; (ii) that spheres are enriched in expression of a variety of tumour-relevant proteins (including MUC1 and Galectin-3); and (iii) that targeting of one of these identified proteins (galectin-3) using an inhibitor (N-acetyllactosamine) decreases sphere formation/self-renewal of MCF-7 cancer stem cells in vitro and tumourigenicity in vivo. Hence, proteomic analysis of tumourspheres may find use in identifying novel targets for future therapy. The therapeutic targeting of breast cancer stem cells, a highly clinically relevant sub-population of tumour cells, has the potential to eliminate residual disease and may become an important component of a multi-modality treatment of cancer.
Collapse
Affiliation(s)
- Brian J. Morrison
- School of Biomolecular and Physical Sciences, Griffith University, Brisbane, Queensland, Australia
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Marcus L. Hastie
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Yadveer S. Grewal
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Zara C. Bruce
- School of Biomolecular and Physical Sciences, Griffith University, Brisbane, Queensland, Australia
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Chris Schmidt
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Brent A. Reynolds
- McKnight Brain Institute, University of Florida, Gainesville, Florida, United States of America
| | - Jeffrey J. Gorman
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - J. Alejandro Lopez
- School of Biomolecular and Physical Sciences, Griffith University, Brisbane, Queensland, Australia
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
- * E-mail:
| |
Collapse
|
44
|
Mehta G, Hsiao AY, Ingram M, Luker GD, Takayama S. Opportunities and challenges for use of tumor spheroids as models to test drug delivery and efficacy. J Control Release 2012; 164:192-204. [PMID: 22613880 PMCID: PMC3436947 DOI: 10.1016/j.jconrel.2012.04.045] [Citation(s) in RCA: 849] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 04/24/2012] [Accepted: 04/29/2012] [Indexed: 12/14/2022]
Abstract
Multicellular spheroids are three dimensional in vitro microscale tissue analogs. The current article examines the suitability of spheroids as an in vitro platform for testing drug delivery systems. Spheroids model critical physiologic parameters present in vivo, including complex multicellular architecture, barriers to mass transport, and extracellular matrix deposition. Relative to two-dimensional cultures, spheroids also provide better target cells for drug testing and are appropriate in vitro models for studies of drug penetration. Key challenges associated with creation of uniformly sized spheroids, spheroids with small number of cells and co-culture spheroids are emphasized in the article. Moreover, the assay techniques required for the characterization of drug delivery and efficacy in spheroids and the challenges associated with such studies are discussed. Examples for the use of spheroids in drug delivery and testing are also emphasized. By addressing these challenges with possible solutions, multicellular spheroids are becoming an increasingly useful in vitro tool for drug screening and delivery to pathological tissues and organs.
Collapse
Affiliation(s)
- Geeta Mehta
- Department of Biomedical Engineering, University of Michigan School of Dentistry, Ann Arbor, MI, 48109-2099
- Department of Periodontics & Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, 48109-2099
| | - Amy Y. Hsiao
- Department of Biomedical Engineering, University of Michigan School of Dentistry, Ann Arbor, MI, 48109-2099
| | - Marylou Ingram
- Huntington Medical Research Institutes, 99 North El Molino Avenue, Pasadena, CA, 91101-1830
| | - Gary D. Luker
- Department of Radiology, University of Michigan Medical School, Ann Arbor, MI, 48109-2099
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109-2099
| | - Shuichi Takayama
- Department of Biomedical Engineering, University of Michigan School of Dentistry, Ann Arbor, MI, 48109-2099
- Department of Macromolecular Science and Engineering, The University of Michigan, Ann Arbor, MI, 48109-2099
- Division of Nano-Bio and Chemical Engineering, WCU Project, UNIST, Ulsan, Republic of Korea
| |
Collapse
|
45
|
Morrison BJ, Steel JC, Morris JC. Sphere culture of murine lung cancer cell lines are enriched with cancer initiating cells. PLoS One 2012; 7:e49752. [PMID: 23152931 PMCID: PMC3496706 DOI: 10.1371/journal.pone.0049752] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 10/15/2012] [Indexed: 02/06/2023] Open
Abstract
Cancer initiating cells (CICs) represent a unique cell population essential for the maintenance and growth of tumors. Most in vivo studies of CICs utilize human tumor xenografts in immunodeficient mice. These models provide limited information on the interaction of CICs with the host immune system and are of limited value in assessing therapies targeting CICs, especially immune-based therapies. To assess this, a syngeneic cancer model is needed. We examined the sphere-forming capacity of thirteen murine lung cancer cell lines and identified TC-1 and a metastatic subclone of Lewis lung carcinoma (HM-LLC) as cell lines that readily formed and maintained spheres over multiple passages. TC-1 tumorspheres were not enriched for expression of CD133 or CD44, putative CIC markers, nor did they demonstrate Hoechst 33342 side population staining or Aldefluor activity compared to adherent TC-1 cells. However, in tumorsphere culture, these cells exhibited self-renewal and long-term symmetric division capacity and expressed more Oct-4 compared to adherent cells. HM-LLC sphere-derived cells exhibited increased Oct-4, CD133, and CD44 expression, demonstrated a Hoechst 33342 side population and Aldefluor activity compared to adherent cells or a low metastatic subclone of LLC (LM-LLC). In syngeneic mice, HM-LLC sphere-derived cells required fewer cells to initiate tumorigenesis compared to adherent or LM-LLC cells. Similarly TC-1 sphere-derived cells were more tumorigenic than adherent cells in syngeneic mice. In contrast, in immunocompromised mice, less than 500 sphere or adherent TC-1 cells and less than 1,000 sphere or adherent LLC cells were required to initiate a tumor. We suggest that no single phenotypic marker can identify CICs in murine lung cancer cell lines. Tumorsphere culture may provide an alternative approach to identify and enrich for murine lung CICs. Furthermore, we propose that assessing tumorigenicity of murine lung CICs in syngeneic mice better models the interaction of CICs with the host immune system.
Collapse
MESH Headings
- AC133 Antigen
- Animals
- Antigens, CD/metabolism
- Benzimidazoles/metabolism
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/pathology
- Cell Adhesion
- Cell Membrane/metabolism
- Cell Transformation, Neoplastic/pathology
- Clone Cells
- Gene Expression Regulation, Neoplastic
- Glycoproteins/metabolism
- Humans
- Hyaluronan Receptors/metabolism
- Immunocompetence
- Immunocompromised Host
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Mice
- Mice, Inbred C57BL
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Octamer Transcription Factor-3/genetics
- Octamer Transcription Factor-3/metabolism
- Peptides/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Spheroids, Cellular/metabolism
- Spheroids, Cellular/pathology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Brian J. Morrison
- Division of Hematology-Oncology, Department of Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Jason C. Steel
- Division of Hematology-Oncology, Department of Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - John C. Morris
- Division of Hematology-Oncology, Department of Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
46
|
Lim YC, Roberts TL, Day BW, Harding A, Kozlov S, Kijas AW, Ensbey KS, Walker DG, Lavin MF. A role for homologous recombination and abnormal cell-cycle progression in radioresistance of glioma-initiating cells. Mol Cancer Ther 2012; 11:1863-72. [PMID: 22772423 DOI: 10.1158/1535-7163.mct-11-1044] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common form of brain tumor with a poor prognosis and resistance to radiotherapy. Recent evidence suggests that glioma-initiating cells play a central role in radioresistance through DNA damage checkpoint activation and enhanced DNA repair. To investigate this in more detail, we compared the DNA damage response in nontumor forming neural progenitor cells (NPC) and glioma-initiating cells isolated from GBM patient specimens. As observed for GBM tumors, initial characterization showed that glioma-initiating cells have long-term self-renewal capacity. They express markers identical to NPCs and have the ability to form tumors in an animal model. In addition, these cells are radioresistant to varying degrees, which could not be explained by enhanced nonhomologous end joining (NHEJ). Indeed, NHEJ in glioma-initiating cells was equivalent, or in some cases reduced, as compared with NPCs. However, there was evidence for more efficient homologous recombination repair in glioma-initiating cells. We did not observe a prolonged cell cycle nor enhanced basal activation of checkpoint proteins as reported previously. Rather, cell-cycle defects in the G(1)-S and S-phase checkpoints were observed by determining entry into S-phase and radioresistant DNA synthesis following irradiation. These data suggest that homologous recombination and cell-cycle checkpoint abnormalities may contribute to the radioresistance of glioma-initiating cells and that both processes may be suitable targets for therapy.
Collapse
Affiliation(s)
- Yi Chieh Lim
- Queensland Institute of Medical Research, University of Queensland Centre for Clinical Research, Royal Brisbane Hospital Campus, Herston, Queensland, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Clément-Schatlo V, Marino D, Burkhardt K, Teta P, Leyvraz F, Schatlo B, Frank S, Schaller K, Castella V, Radovanovic I. Quantification, self-renewal, and genetic tracing of FL1⁺ tumor-initiating cells in a large cohort of human gliomas. Neuro Oncol 2012; 14:720-35. [PMID: 22584872 DOI: 10.1093/neuonc/nos084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Evidence has emerged that the initiation and growth of gliomas is sustained by a subpopulation of cancer-initiating cells (CICs). Because of the difficulty of using markers to tag CICs in gliomas, we have previously exploited more robust phenotypic characteristics, including a specific morphology and intrincic autofluorescence, to identify and isolate a subpopulation of glioma CICs, called FL1(+). The objective of this study was to further validate our method in a large cohort of human glioma and a mouse model of glioma. Seventy-four human gliomas of all grades and the GFAP-V(12)HA-ras B8 mouse model were analyzed for in vitro self-renewal capacity and their content of FL1(+). Nonneoplastic brain tissue and embryonic mouse brain were used as control. Genetic traceability along passages was assessed with microsatellite analysis. We found that FL1(+) cells from low-grade gliomas and from control nonneoplasic brain tissue show a lower level of autofluorescence and undergo a restricted number of cell divisions before dying in culture. In contrast, we found that FL1(+) cells derived from many but not all high-grade gliomas acquire high levels of autofluorescence and can be propagated in long-term cultures. Moreover, FL1(+) cells show a remarkable traceability over time in vitro and in vivo. Our results show that FL1(+) cells can be found in all specimens of a large cohort of human gliomas of different grades and in a model of genetically induced mouse glioma as well as nonneoplastic brain. However, their self-renewal capacity is variable and seems to be dependent on the tumor grade.
Collapse
Affiliation(s)
- Virginie Clément-Schatlo
- Department of Clinical Neurosciences, Service of Neurosurgery, Rue Gabrielle-Perret-Gentil, 4, CH-1211 Geneva, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Blackmore DG, Reynolds BA, Golmohammadi MG, Large B, Aguilar RM, Haro L, Waters MJ, Rietze RL. Growth hormone responsive neural precursor cells reside within the adult mammalian brain. Sci Rep 2012; 2:250. [PMID: 22355762 PMCID: PMC3274722 DOI: 10.1038/srep00250] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 01/16/2012] [Indexed: 12/02/2022] Open
Abstract
The detection of growth hormone (GH) and its receptor in germinal regions of the mammalian brain prompted our investigation of GH and its role in the regulation of endogenous neural precursor cell activity. Here we report that the addition of exogenous GH significantly increased the expansion rate in long-term neurosphere cultures derived from wild-type mice, while neurospheres derived from GH null mice exhibited a reduced expansion rate. We also detected a doubling in the frequency of large (i.e. stem cell-derived) colonies for up to 120 days following a 7-day intracerebroventricular infusion of GH suggesting the activation of endogenous stem cells. Moreover, gamma irradiation induced the ablation of normally quiescent stem cells in GH-infused mice, resulting in a decline in olfactory bulb neurogenesis. These results suggest that GH activates populations of resident stem and progenitor cells, and therefore may represent a novel therapeutic target for age-related neurodegeneration and associated cognitive decline.
Collapse
Affiliation(s)
- Daniel G Blackmore
- Queensland Brain Institute, The University of Queensland, Brisbane Queensland, 4072, Australia.
| | | | | | | | | | | | | | | |
Collapse
|