151
|
Angstadt AY, Thayanithy V, Subramanian S, Modiano JF, Breen M. A genome-wide approach to comparative oncology: high-resolution oligonucleotide aCGH of canine and human osteosarcoma pinpoints shared microaberrations. Cancer Genet 2012; 205:572-87. [PMID: 23137772 DOI: 10.1016/j.cancergen.2012.09.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 08/31/2012] [Accepted: 09/24/2012] [Indexed: 11/19/2022]
Abstract
Molecular cytogenetic evaluation of human osteosarcoma (OS) has revealed the characteristically high degree of genomic reorganization that is the hallmark of this cancer. The extent of genomic disorder in OS has hindered identification of the genomic aberrations driving disease progression. With pathophysiological similarities to its human counterpart, canine OS represents an ideal model for comparison of conserved regions of genomic instability that may be disease-associated rather than genomic passengers. This study used high-resolution oligonucleotide array comparative genomic hybridization and a variety of informatics tools to aid in the identification of disease-associated genome-wide DNA copy number aberrations in canine and human OS. Our findings support and build upon the high level of cytogenetic complexity, through the identification of shared regions of microaberration (<500 kb) and functional analysis of possible orthologous OS-associated genes to pinpoint the cellular processes most commonly affected by aberration in human and canine OS. Aberrant regions contained previously reported genes such as CDC5L, MYC, RUNX2, and CDKN2A/CDKN2B, while expanding the gene of interest list to include ADAM15, CTC1, MEN1, CDK7, and others. Such regions of instability may thus have functional significance in the etiology of OS, the most common primary bone tumor in both species.
Collapse
Affiliation(s)
- Andrea Y Angstadt
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
| | | | | | | | | |
Collapse
|
152
|
β-Catenin Does Not Confer Tumorigenicity When Introduced into Partially Transformed Human Mesenchymal Stem Cells. Sarcoma 2012; 2012:164803. [PMID: 23125530 PMCID: PMC3483784 DOI: 10.1155/2012/164803] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 09/23/2012] [Accepted: 09/23/2012] [Indexed: 11/17/2022] Open
Abstract
Although osteosarcoma is the most common primary malignant bone tumor in children and adolescents, its cell of origin and the genetic alterations are unclear. Previous studies have shown that serially introducing hTERT, SV40 large TAg, and H-Ras transforms human mesenchymal stem cells into two distinct sarcomas cell populations, but they do not form osteoid. In this study, β-catenin was introduced into mesenchymal stem cells already containing hTERT and SV40 large TAg to analyze if this resulted in a model which more closely recapitulated osteosarcoma. Results. Regardless of the level of induced β-catenin expression in the stable transfectants, there were no marked differences induced in their phenotype or invasion and migration capacity. Perhaps more importantly, none of them formed tumors when injected into immunocompromised mice. Moreover, the resulting transformed cells could be induced to osteogenic and chondrogenic differentiation but not to adipogenic differentiation. Conclusions. β-catenin, although fostering osteogenic differentiation, does not induce the malignant features and tumorigenicity conveyed by oncogenic H-RAS when introduced into partly transformed mesenchymal stem cells. This may have implications for the role of β-catenin in osteosarcoma pathogenesis. It also may suggest that adipogenesis is an earlier branch point than osteogenesis and chondrogenesis in normal mesenchymal differentiation.
Collapse
|
153
|
Zuch D, Giang AH, Shapovalov Y, Schwarz E, Rosier R, O'Keefe R, Eliseev RA. Targeting radioresistant osteosarcoma cells with parthenolide. J Cell Biochem 2012; 113:1282-91. [PMID: 22109788 DOI: 10.1002/jcb.24002] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Osteosarcoma is a devastating tumor of bone, primarily affecting adolescents. Osteosarcoma tumors are notoriously radioresistant. Radioresistant cancers, including osteosarcoma, typically exhibit a considerable potential for relapse and development of metastases following treatment. Relapse and metastatic potential can, in part, be due to a specific radioresistant subpopulation of cells with stem-like characteristics, cancer stem cells, which maintain the capacity to regenerate entire tumors. In the current study, we have investigated whether in vitro treatments with parthenolide, a naturally occurring small molecule that interferes with NF-κB signaling and has various other effects, will re-sensitize cancer stem cells and the entire cell population to radiotherapy in osteosarcoma. Our results indicate that parthenolide and ionizing radiation synergistically induce cell death in LM7 osteosarcoma cells. Importantly, the combination treatment results in a significant reduction in the viability of both the overall population of osteosarcoma cells and the cancer stem cell subpopulation. This effect is dependent on the ability of parthenolide to induce oxidative stress. Therefore, as a supplement to current multimodal therapy, parthenolide may sensitize osteosarcoma tumors to radiation and greatly reduce the prevalence of relapse and metastatic progression.
Collapse
Affiliation(s)
- Daniel Zuch
- Center for Musculoskeletal Research, University of Rochester School of Medicine & Dentistry, 575 Elmwood Ave., Rochester, New York 14642, USA
| | | | | | | | | | | | | |
Collapse
|
154
|
Ng AJ, Mutsaers AJ, Baker EK, Walkley CR. Genetically engineered mouse models and human osteosarcoma. Clin Sarcoma Res 2012; 2:19. [PMID: 23036272 PMCID: PMC3523007 DOI: 10.1186/2045-3329-2-19] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 11/30/2011] [Indexed: 12/19/2022] Open
Abstract
Osteosarcoma is the most common form of bone cancer. Pivotal insight into the genes involved in human osteosarcoma has been provided by the study of rare familial cancer predisposition syndromes. Three kindreds stand out as predisposing to the development of osteosarcoma: Li-Fraumeni syndrome, familial retinoblastoma and RecQ helicase disorders, which include Rothmund-Thomson Syndrome in particular. These disorders have highlighted the important roles of P53 and RB respectively, in the development of osteosarcoma. The association of OS with RECQL4 mutations is apparent but the relevance of this to OS is uncertain as mutations in RECQL4 are not found in sporadic OS. Application of the knowledge or mutations of P53 and RB in familial and sporadic OS has enabled the development of tractable, highly penetrant murine models of OS. These models share many of the cardinal features associated with human osteosarcoma including, importantly, a high incidence of spontaneous metastasis. The recent development of these models has been a significant advance for efforts to improve our understanding of the genetics of human OS and, more critically, to provide a high-throughput genetically modifiable platform for preclinical evaluation of new therapeutics.
Collapse
Affiliation(s)
- Alvin Jm Ng
- St Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, VIC, 3065, Australia.,Department of Medicine, University of Melbourne, St. Vincent's Hospital, Fitzroy, VIC, 3065, Australia
| | - Anthony J Mutsaers
- St Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, VIC, 3065, Australia.,Department of Medicine, University of Melbourne, St. Vincent's Hospital, Fitzroy, VIC, 3065, Australia.,Ontario Veterinary College, University of Guelph, 50 Stone Road, Guelph, ON, N1G 2W1, Canada
| | - Emma K Baker
- St Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, VIC, 3065, Australia.,Department of Medicine, University of Melbourne, St. Vincent's Hospital, Fitzroy, VIC, 3065, Australia
| | - Carl R Walkley
- St Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, VIC, 3065, Australia.,Department of Medicine, University of Melbourne, St. Vincent's Hospital, Fitzroy, VIC, 3065, Australia
| |
Collapse
|
155
|
Vosdoganes P, Lim R, Moss TJM, Wallace EM. Cell therapy: a novel treatment approach for bronchopulmonary dysplasia. Pediatrics 2012; 130:727-37. [PMID: 22945412 DOI: 10.1542/peds.2011-2576] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a major cause of substantial lifelong morbidity in preterm infants. Despite a better understanding of the pathophysiology of BPD and significant research effort into its management, there remains today no effective treatment. Cell-based therapy is a novel approach that offers much promise in the prevention and treatment of BPD. Recent research supports a therapeutic role for cell transplantation in the management of a variety of acute and chronic adult and childhood lung diseases, with potential of such therapy to reduce inflammation and prevent acute lung injury. However, considerable uncertainties remain regarding cell therapies before they can be established as safe and effective clinical treatments for BPD. This review summarizes the current literature investigating cell therapies in lung disease, with particular focus on the various types of cells available and their specific properties in the context of a future therapy for BPD.
Collapse
Affiliation(s)
- Patricia Vosdoganes
- Ritchie Centre, Department of Obstetrics and Gynaecology, Monash Medical Centre, 246 Clayton Rd, Clayton, VIC, Australia 3168
| | | | | | | |
Collapse
|
156
|
Käcker C, Marx A, Mössinger K, Svehla F, Schneider U, Hogendoorn PCW, Nielsen OS, Küffer S, Sauer C, Fisher C, Hallermann C, Hartmann JT, Blay JY, Mechtersheimer G, Hohenberger P, Ströbel P. High frequency of MYC gene amplification is a common feature of radiation-induced sarcomas. Further results from EORTC STBSG TL 01/01. Genes Chromosomes Cancer 2012; 52:93-8. [PMID: 23012233 DOI: 10.1002/gcc.22009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 08/15/2012] [Accepted: 08/16/2012] [Indexed: 12/20/2022] Open
Abstract
Irradiation is a major causative factor among the small subgroup of sarcomas with a known etiology. The prognosis of radiation-induced sarcomas (RIS) is significantly worse than that of their spontaneous counterparts. The most frequent histological subtypes include undifferentiated pleomorphic sarcomas, angiosarcomas, and leiomyosarcomas. A high frequency of MYC amplifications in radiation-induced angiosarcomas, but not in primary angiosarcomas, has recently been described. To investigate whether MYC amplifications are also frequent in RIS other than angiosarcomas, we analyzed the MYC amplification status of 83 RIS and 192 sporadic sarcomas by fluorescence in situ hybridization. We found significantly higher numbers of MYC amplifications in RIS than in sporadic sarcomas (P < 0.0001), especially in angiosarcomas, undifferentiated pleomorphic sarcomas, and leiomyosarcomas. Angiosarcomas were special in that MYC amplifications were particularly frequent and always high level, while other RIS showed low-level amplifications. We conclude that MYC amplifications are a frequent feature of RIS as a group and may contribute to the biology of these tumors.
Collapse
Affiliation(s)
- Christoph Käcker
- Department of Pathology, University Medical Center Mannheim, University of Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Subramanian A, Shu-Uin G, Kae-Siang N, Gauthaman K, Biswas A, Choolani M, Bongso A, Chui-Yee F. Human umbilical cord Wharton's jelly mesenchymal stem cells do not transform to tumor-associated fibroblasts in the presence of breast and ovarian cancer cells unlike bone marrow mesenchymal stem cells. J Cell Biochem 2012; 113:1886-95. [PMID: 22234854 DOI: 10.1002/jcb.24057] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Human bone marrow mesenchymal stem cells (hBMMSCs) were shown to transform into tumor-associated fibroblasts (TAFs) when in the vicinity of breast cancer tumors and played an important role in tumor enhancement and metastasis. In early human development MSCs migrating from the yolk sac and aorta-gonad-mesonephros (AGM) via the umbilical cord to the placenta and back to the fetal bone marrow were shown to get trapped in the gelatinous Wharton's jelly of the umbilical cord. The common origin of the Wharton's jelly MSCs and the finally homed hBMMSCs prompted us to evaluate whether hWJSCs are also involved in TAF transformation. hWJSCs and hBMMSCs were grown in the presence of breast and ovarian cancer cell conditioned medium (MDA-TCM, TOV-TCM) for 30 days. No changes were observed in the hWJSCs but the hBMMSCs transformed from short to thin long fibroblasts, their proliferation rates increased and CD marker expression decreased. The transformed hBMMSCs showed positive staining for the tumor-associated markers FSP, VEGF, EGF, and Tn-C. Real-time PCR and multiplex luminex bead analysis showed upregulation of TAF-related genes (FSP, FAP, Tn-C, Tsp-1, EGF, bFGF, IL-6, α-SMA, VEGF, and TGF-β) for hBMMSCs with low expression for hWJSCs. The luciferase assay showed that hWJSCs previously exposed to MDA-TCM or TOV-TCM had no stimulatory growth effect on luciferase-tagged MDA or TOV cells unlike hBMMSCs. The results confirmed that hWJSCs do not transform to the TAF phenotype and may therefore not be associated with enhanced growth of solid tumors making them a safe MSC for cell based therapies.
Collapse
Affiliation(s)
- Arjunan Subramanian
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge 119074, Singapore
| | | | | | | | | | | | | | | |
Collapse
|
158
|
David E, Tirode F, Baud'huin M, Guihard P, Laud K, Delattre O, Heymann MF, Heymann D, Redini F, Blanchard F. Oncostatin M is a growth factor for Ewing sarcoma. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:1782-95. [PMID: 22982441 DOI: 10.1016/j.ajpath.2012.07.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 06/28/2012] [Accepted: 07/18/2012] [Indexed: 11/18/2022]
Abstract
Primary bone tumors, osteosarcomas and chondrosarcomas, derive from mesenchymal stem cells committed into osteoblasts and chondrocytes; in Ewing sarcomas (ESs), the oncogenic fusion protein EWS-FLI1 prevents mesenchymal differentiation and induces neuroectodermic features. Oncostatin M (OSM) is a cytokine from the IL-6 family that modulates proliferation and differentiation in numerous cells. The basis for inhibition versus induction of proliferation by this cytokine is obscure, although MYC was described as a potent molecular switch in OSM signaling. We show herein that, in contrast to osteosarcomas and chondrosarcomas, for which OSM was cytostatic, OSM induced proliferation of ES cell lines. Knockdown experiments demonstrated that growth induction by OSM depends on both types I [leukemia inhibitory factor receptor (LIFR)] and II [OSM receptor (OSMR)] receptors, high STAT3 activation, and induction of MYC to a high expression level. Indeed, ES cell lines, mice xenografts, and patient biopsy specimens poorly expressed LIF, precluding LIFR lysosomal degradation and OSMR transcriptional induction, thus leading to a high LIFR/OSMR ratio. Because other neuroectodermic tumors (ie, glioma, medulloblastoma, and neuroblastoma) had a similar expression profile, the main role of EWS-FLI1 could be through maintenance of stemness and neuroectodermic features, characterized by a low LIF, a high LIFR/OSMR ratio, and high MYC expression. Thus, this study on rare bone malignancies gives valuable insights on more common cancer regulatory mechanisms and could provide new therapeutic opportunities.
Collapse
|
159
|
Szuhai K, Cleton-Jansen AM, Hogendoorn PCW, Bovée JVMG. Molecular pathology and its diagnostic use in bone tumors. Cancer Genet 2012; 205:193-204. [PMID: 22682618 DOI: 10.1016/j.cancergen.2012.04.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 03/30/2012] [Accepted: 04/04/2012] [Indexed: 12/22/2022]
Abstract
Bone tumors are considered by most pathologists difficult to diagnose as they are rare, have overlapping morphology, need radiological correlation, and the usefulness of immunohistochemistry is limited, making conventional morphology the cornerstone of the diagnosis. Over the past decade, more and more has become known of the molecular background of bone tumors. Three groups of bone tumors are recognized, namely, tumors with specific translocations combined with a relatively simple karyotype involving chromosomal translocations (Ewing sarcoma, aneurysmal bone cyst), tumors with specific gene mutations or amplifications (chondrosarcoma, fibrous dysplasia, chordoma), and sarcomas with genetic instability and as a consequence complex karyotypes (osteosarcoma). Technical advancements will rapidly reveal new alterations in the more rare sarcoma subtypes for which the molecular background has remained enigmatic. Opening the archives and using new technologies, as well as refinement of existing technologies for decalcified paraffin-embedded tissue, may bring to light more specific genetic aberrations in bone tumors that can be applied in molecular diagnostics in the near future.
Collapse
Affiliation(s)
- Karoly Szuhai
- Department of Molecular Cell Biology, Leiden University Medical Center, The Netherlands
| | | | | | | |
Collapse
|
160
|
The activities of Smad and Gli mediated signalling pathways in high-grade conventional osteosarcoma. Eur J Cancer 2012; 48:3429-38. [PMID: 22868198 DOI: 10.1016/j.ejca.2012.06.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 06/08/2012] [Accepted: 06/20/2012] [Indexed: 12/22/2022]
Abstract
High-grade conventional osteosarcoma is a malignant tumour predominantly affecting adolescents and, despite multimodal intensive therapy, lethal for one third of the patients. Although there is currently detailed knowledge of normal skeletal development, this has not been integrated into research on the genesis of osteosarcoma. Recently we showed that the canonical Wnt pathway is not active in osteosarcoma and that its reactivation is disadvantageous to osteosarcoma cells. Since Wnt is regulating normal skeletogenesis together with other pathways, here we report on the activities of the bone morphogenic protein (BMP), the transforming growth factor beta (TGFβ) and the hedgehog (Hh) pathways in osteosarcoma. Human osteosarcoma samples (n=210), benign bone tumours of osteoblastic lineage called osteoblastoma (n=25) and osteosarcoma cell lines (n=19) were examined. For pathway activity luciferase transcriptional reporter assays and gene and protein expression analyses were performed. Immunohistochemical analysis of phosphorylated Smad1 and Smad2, the intracellular effectors of BMP and TGFβ, respectively, showed nuclear expression of both proteins in 70% of the osteosarcoma samples at levels comparable to osteoblastoma. Interestingly cases with lower expression showed significantly worse disease free survival. This may imply that drugs restoring impaired signalling pathways in osteosarcoma might change the tumour's aggressive clinical course, however targeted pathway modulation in vitro did not affect cell proliferation.
Collapse
|
161
|
Kido A, Yoshitani K, Shimizu T, Akahane M, Fujii H, Tsukamoto S, Kondo Y, Honoki K, Imano M, Tanaka Y. Effect of mesenchymal stem cells on hypoxia-induced desensitization of β2-adrenergic receptors in rat osteosarcoma cells. Oncol Lett 2012. [PMID: 23205094 DOI: 10.3892/ol.2012.813] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The β2-adrenergic receptor (β2AR) mediates the effects of chronic stress in several neoplasms, however, β2AR signaling is impaired by hypoxia in various tissues. While hypoxia is a common feature significant in the progression of solid tumors, little is known about the effect of hypoxia on β2AR signaling in the tumor microenvironment. Previously, it has been reported that the systemic administration of mesenchymal stem cells (MSCs) increased the engraftment and metastatic colonization of rat osteosarcoma (OS) cells. In the current study, the effect of MSCs on the hypoxia-induced desensitization of the β2AR in OS cells was investigated. Epinephrine, norepinephrine and isoproterenol increased the cellular proliferation of the rat OS cell line COS1NR and rat MSCs in a dose-dependent and β2AR antagonist-sensitive manner. While isoproterenol had significant proliferative effects on MSCs under normoxic and hypoxic conditions, COS1NR cells did not respond under hypoxic conditions. A sensitivity assay for the β2AR revealed that hypoxia impaired the sensitivity of COS1NR cells, whereas hypoxia did not affect MSCs. An immunoassay revealed no significant change in the expression of hypoxia-inducible factor-1α (HIF1α) in COS1NR cells, whilst an immunoassay demonstrated a 15% increase in MSCs following isoproterenol stimulation. In COS1NR cells co-cultured with MSCs under hypoxic conditions, isoproterenol caused a significant increase in proliferation and this effect was inhibited by an anti-interleukin (IL)-6 antibody. A tumor formation assay in syngeneic rats revealed that the systemic administration of MSCs enhances the growth of OS and the effect of MSCs was inhibited by IL-6 neutralization. In conclusion, MSCs are resistant to the hypoxia-induced desensitization to β2AR. Hypoxia caused a siginificant desensitization of the β2AR in COS1NR cells alone, whereas MSCs may support tumor progression through cellular interactions.
Collapse
|
162
|
Honoki K, Fujii H, Tohma Y, Tsujiuchi T, Kido A, Tsukamoto S, Mori T, Tanaka Y. Comparison of gene expression profiling in sarcomas and mesenchymal stem cells identifies tumorigenic pathways in chemically induced rat sarcoma model. ISRN ONCOLOGY 2012; 2012:909453. [PMID: 22852096 PMCID: PMC3407640 DOI: 10.5402/2012/909453] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 05/20/2012] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem cells (MSCs) are believed to be the cell of origin for most sarcomas including osteosarcoma and malignant fibrous histiocytoma (MFH/UPS). To identify the signaling pathways involved in sarcoma pathogenesis, we compared gene expression profiles in rat osteosarcoma and MFH cells with those in syngeneic rat MSCs. Analysis of genes that characterize MSCs such as CD44, CD105, CD73, and CD90 showed higher expression in MSCs compared to sarcomas. Pathways involved in focal and cell adhesion, cytokine-cytokine receptors, extracellular matrix receptors, chemokines, and Wnt signaling were down-regulated in both sarcomas. Meanwhile, DNA replication, cell cycle, mismatch repair, Hedgehog signaling, and metabolic pathways were upregulated in both sarcomas. Downregulation of p21Cip1 and higher expression of CDK4-cyclinD1 and CDK2-cyclinE could accelerate cell cycle in sarcomas. The current study indicated that these rat sarcomas could be a good model for their human counterparts and will provide the further insights into the molecular pathways and mechanisms involved in sarcoma pathogenesis.
Collapse
Affiliation(s)
- Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan
| | | | | | | | | | | | | | | |
Collapse
|
163
|
Sollazzo V, Galasso M, Volinia S, Carinci F. Prion proteins (PRNP and PRND) are over-expressed in osteosarcoma. J Orthop Res 2012; 30:1004-12. [PMID: 22147650 DOI: 10.1002/jor.22034] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Accepted: 11/10/2011] [Indexed: 02/04/2023]
Abstract
Although osteosarcoma is the most common bone malignancy, the molecular and cellular mechanisms influencing its pathogenesis have remained elusive. Prion proteins (PRNP and PRND), known mostly for its involvement in neurodegenerative spongiform encephalopathies, have been recently demonstrated to be involved in resistance to apoptosis, tumorigenesis, proliferation, and metastasis. The main aim of research was to study whether prion proteins were over-expressed in human osteosarcoma, and if prion proteins could have a role also in osteosarcomas. We evaluated differential gene expression between 22 cases of osteosarcoma and 40 cases of normal bone specimens through cDNA microarray analysis spanning a substantial fraction of the human genome. PRNP and PRND are significantly over-expressed in osteosarcoma. PRNP and PRND appear involved with some important genes related to tumorigenesis and apoptosis. PRNP is linked to PTK2, RBBP9, and TGFB1 while PRND is linked to TNFSF10, BCL2A1, NFKB2, and TP53RK. Increased expression on Affymetrix arrays of prion proteins seems to be associated with the development of osteosarcoma. Prions seem to induce a negative regulation of apoptosis, thus promoting osteosarcoma development and progression. Osteosarcoma is a very aggressive tumor and even after modern chemotherapy and excision of tumors efforts are needed to improve clinical outcome. Since Prion proteins seem to be related to osteosarcoma development, their inhibition could represent a new approach to the molecular treatment of osteosarcoma.
Collapse
|
164
|
Mohseny AB, Xiao W, Carvalho R, Spaink HP, Hogendoorn PCW, Cleton-Jansen AM. An osteosarcoma zebrafish model implicates Mmp-19 and Ets-1 as well as reduced host immune response in angiogenesis and migration. J Pathol 2012; 227:245-53. [PMID: 22297719 DOI: 10.1002/path.3998] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 01/06/2012] [Accepted: 01/25/2012] [Indexed: 01/14/2023]
Abstract
About 40% of osteosarcoma patients die of metastases. Novel strategies to improve treatment of metastatic patients require a better understanding of the processes involved, like angiogenesis, migration, and the immune response. However, the rarity of osteosarcoma and its heterogeneity make this neoplasm difficult to study. Recently we reported malignant transformation of mouse mesenchymal stem cells (MSCs) which formed osteosarcoma upon transplantation into mice. Here we studied these cells in zebrafish embryos and found that transformed MSCs induced angiogenesis and migrated through the bodies of the embryos, but this was never observed with non-transformed normal MSCs (progenitors of the transformed MSCs). Whole genome expression analysis of both the cells and the host showed that angiogenesis and migration-related genes matrix metalloproteinase 19 (Mmp-19) and erythroblastosis virus E26 oncogene homologue 1 (Ets-1) were overexpressed in transformed MSCs compared to normal MSCs. Investigating the host response, embryos injected with transformed MSCs showed decreased expression of immune response-related genes, especially major histocompatibility complex class 1 (mhc1ze), as compared to embryos injected with normal MSCs. These findings contribute to the identification of genetic events involved in angiogenesis, migration, and host response providing targets as well as an appropriate model for high-throughput drug screens.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Bone Neoplasms/blood supply
- Bone Neoplasms/enzymology
- Bone Neoplasms/genetics
- Bone Neoplasms/immunology
- Bone Neoplasms/pathology
- Carbocyanines/metabolism
- Cell Movement
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Cells, Cultured
- Fluorescent Dyes/metabolism
- Gene Expression Profiling/methods
- Gene Expression Regulation, Neoplastic
- Luminescent Proteins/biosynthesis
- Luminescent Proteins/genetics
- Matrix Metalloproteinases, Secreted/genetics
- Matrix Metalloproteinases, Secreted/metabolism
- Mesenchymal Stem Cell Transplantation
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Neoplasm Invasiveness
- Neovascularization, Pathologic/enzymology
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/pathology
- Oligonucleotide Array Sequence Analysis
- Osteosarcoma/blood supply
- Osteosarcoma/enzymology
- Osteosarcoma/genetics
- Osteosarcoma/immunology
- Osteosarcoma/secondary
- Proto-Oncogene Protein c-ets-1/genetics
- Proto-Oncogene Protein c-ets-1/metabolism
- Time Factors
- Tumor Escape
- Zebrafish/embryology
- Zebrafish/genetics
- Zebrafish/metabolism
- Zebrafish Proteins/genetics
- Zebrafish Proteins/metabolism
- Red Fluorescent Protein
Collapse
Affiliation(s)
- Alexander B Mohseny
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
165
|
Dani N, Olivero M, Mareschi K, van Duist MM, Miretti S, Cuvertino S, Patané S, Calogero R, Ferracini R, Scotlandi K, Fagioli F, Di Renzo MF. The MET oncogene transforms human primary bone-derived cells into osteosarcomas by targeting committed osteo-progenitors. J Bone Miner Res 2012; 27:1322-34. [PMID: 22367914 DOI: 10.1002/jbmr.1578] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The MET oncogene is aberrantly overexpressed in human osteosarcomas. We have previously converted primary cultures of human bone-derived cells into osteosarcoma cells by overexpressing MET. To determine whether MET transforms mesenchymal stem cells or committed progenitor cells, here we characterize distinct MET overexpressing osteosarcoma (MET-OS) clones using genome-wide expression profiling, cytometric analysis, and functional assays. All the MET-OS clones consistently display mesenchymal and stemness markers, but not most of the mesenchymal–stem cell-specific markers. Conversely, the MET-OS clones express genes characteristic of early osteoblastic differentiation phases, but not those of late phases. Profiling of mesenchymal stem cells induced to differentiate along osteoblast, adipocyte, and chondrocyte lineages confirms that MET-OS cells are similar to cells at an initial phase of osteoblastic differentiation. Accordingly, MET-OS cells cannot differentiate into adipocytes or chondrocytes, but can partially differentiate into osteogenic-matrix-producing cells. Moreover, in vitro MET-OS cells form self-renewing spheres enriched in cells that can initiate tumors in vivo. MET kinase inhibition abrogates the self-renewal capacity of MET-OS cells and allows them to progress toward osteoblastic differentiation. These data show that MET initiates the transformation of a cell population that has features of osteo-progenitors and suggest that MET regulates self-renewal and lineage differentiation of osteosarcoma cells.
Collapse
Affiliation(s)
- Nadia Dani
- Laboratory of Cancer Genetics, Department of Oncological Sciences University of Torino School of Medicine, Turin, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Basu-Roy U, Basilico C, Mansukhani A. Perspectives on cancer stem cells in osteosarcoma. Cancer Lett 2012; 338:158-67. [PMID: 22659734 DOI: 10.1016/j.canlet.2012.05.028] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 05/21/2012] [Accepted: 05/24/2012] [Indexed: 12/27/2022]
Abstract
Osteosarcoma is an aggressive pediatric tumor of growing bones that, despite surgery and chemotherapy, is prone to relapse. These mesenchymal tumors are derived from progenitor cells in the osteoblast lineage that have accumulated mutations to escape cell cycle checkpoints leading to excessive proliferation and defects in their ability to differentiate appropriately into mature bone-forming osteoblasts. Like other malignant tumors, osteosarcoma is often heterogeneous, consisting of phenotypically distinct cells with features of different stages of differentiation. The cancer stem cell hypothesis posits that tumors are maintained by stem cells and it is the incomplete eradication of a refractory population of tumor-initiating stem cells that accounts for drug resistance and tumor relapse. In this review we present our current knowledge about the biology of osteosarcoma stem cells from mouse and human tumors, highlighting new insights and unresolved issues in the identification of this elusive population. We focus on factors and pathways that are implicated in maintaining such cells, and differences from paradigms of epithelial cancers. Targeting of the cancer stem cells in osteosarcoma is a promising avenue to explore to develop new therapies for this devastating childhood cancer.
Collapse
Affiliation(s)
- Upal Basu-Roy
- Department of Microbiology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, United States
| | | | | |
Collapse
|
167
|
Abstract
Osteosarcoma is a primary bone malignancy with a particularly high incidence rate in children and adolescents relative to other age groups. The etiology of this often aggressive cancer is currently unknown, because complicated structural and numeric genomic rearrangements in cancer cells preclude understanding of tumour development. In addition, few consistent genetic changes that may indicate effective molecular therapeutic targets have been reported. However, high-resolution techniques continue to improve knowledge of distinct areas of the genome that are more commonly associated with osteosarcomas. Copy number gains at chromosomes 1p, 1q, 6p, 8q, and 17p as well as copy number losses at chromosomes 3q, 6q, 9, 10, 13, 17p, and 18q have been detected by numerous groups, but definitive oncogenes or tumour suppressor genes remain elusive with respect to many loci. In this paper, we examine studies of the genetics of osteosarcoma to comprehensively describe the heterogeneity and complexity of this cancer.
Collapse
|
168
|
Lan J, Liu X, Rong W, Wei F, Jiang L, Yu H, Dang G, Liu Z. Stro-1(+) stromal cells have stem-like features in giant cell tumor of bone. J Surg Oncol 2012; 106:826-36. [PMID: 22605660 DOI: 10.1002/jso.23151] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 04/17/2012] [Indexed: 11/07/2022]
Abstract
BACKGROUND Giant cell tumor of bone (GCTB) is an aggressive benign bone tumor with poor prognosis whose neoplastic component is stromal cells (SCs). Tumor stem-like cells (TSCs) have been demonstrated as precursors for tumor genesis and growth. The aim of this study is to identify TSCs in GCTB. METHODS Stro-1(+) and Stro-1(-) cells were isolated by fluorescence-activated cell sorting (FACS). Stem-like properties of both Stro-1(+) and Stro-1(-) subpopulations were assessed using MTT colorimetric assays, cell cycle analyses, sphere formation assays, and differentiation assays. Molecular profiles were analyzed by flow cytometry, immunofluorescence, and qRT-PCR. RESULTS The existence of rare Stro-1(+) cells was confirmed in vitro using FACS and in vivo by immunohistochemistry. These Stro-1(+) cells exhibited higher proliferative and cisplatin-resistant potentials than Stro-1(-) cells. In serum-free suspension cultures, Stro-1(+) SCs could form cell spheres and maintain self-renewal. Furthermore, Stro-1(+) SCs could differentiate into two mesenchymal lineage cells: osteoblasts and adipocytes. Cell surface markers CD44, CD117, and CD133 and stem cell-associated genes OCT3/4, NANOG, and ABCG2 were significantly higher in the Stro-1(+) subpopulation. CONCLUSIONS This study demonstrates that Stro-1(+) SCs in GCTB possess stem-like biological and molecular phenotypes, indicating that they are the TSCs of GCTB.
Collapse
Affiliation(s)
- Jie Lan
- Department of Orthopaedics, Peking University Third Hospital, Beijing, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
169
|
Arndt CAS, Rose PS, Folpe AL, Laack NN. Common musculoskeletal tumors of childhood and adolescence. Mayo Clin Proc 2012; 87:475-87. [PMID: 22560526 PMCID: PMC3538469 DOI: 10.1016/j.mayocp.2012.01.015] [Citation(s) in RCA: 219] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 01/07/2012] [Accepted: 01/16/2012] [Indexed: 11/23/2022]
Abstract
Osteosarcoma, Ewing sarcoma, and rhabdomyosarcoma are the most common malignant musculoskeletal tumors in children and adolescents. Today, most patients can be cured. Numerous factors have contributed to improved outcome for these patients over the past several decades. These include multidisciplinary care involving oncologists, radiation oncologists, surgeons, pathologists, and radiologists and enrollment of patients in clinical trials. Better understanding of molecular mechanisms of disease have resulted in studies using molecular targets in addition to standard chemotherapeutic agents, which hopefully will lead to better outcomes in the future. Moreover, new orthopedic techniques and devices as well as new technologies in radiation oncology hold promise for better local control of primary tumors and the potential for fewer late adverse effects. Despite this progress, patients must undergo lifelong follow-up for possible late effects of intense chemotherapy and radiation therapy. We review the diagnosis, prognosis, staging, multidisciplinary therapy, new directions in therapy, and long-term complications of treatment for these tumors. For this review, we searched MEDLINE using the terms rhabdomyosarcoma, osteosarcoma, Ewing sarcoma, biology, and humans and limited the search to articles from 2000 to September 2011. Additional references found in these articles were utilized as appropriate, as well as references from the background information in current therapeutic studies of the Children's Oncology Group. The same database and time frame were searched for articles written by leading authorities in the field.
Collapse
Key Words
- arms, alveolar rhabdomyosarcoma
- cog, children's oncology group
- efs, event-free survival
- erms, embryonal rhabdomyosarcoma
- es, ewing sarcoma
- fdg-pet, fluorodeoxyglucose positron emission tomography
- ie, ifosfamide and etoposide
- map, methotrexate, doxorubicin (adriamycin), and cisplatin
- os, osteosarcoma
- pnet, primitive neuroectodermal tumor
- rms, rhabdomyosarcoma
- vdc, vincristine, doxorubicin, and cyclophosphamide
Collapse
Affiliation(s)
- Carola A S Arndt
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | |
Collapse
|
170
|
Bennani-Baiti IM. Epigenetic and epigenomic mechanisms shape sarcoma and other mesenchymal tumor pathogenesis. Epigenomics 2012; 3:715-32. [PMID: 22126291 DOI: 10.2217/epi.11.93] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Sarcomas comprise a large number of rare, histogenetically heterogeneous, mesenchymal tumors. Cancers such as Ewing's sarcoma, liposarcoma, rhabdomyosarcoma and synovial sarcoma can be generated by the transduction of mesenchymal stem cell progenitors with sarcoma-pathognomonic oncogenic fusions, a neoplastic transformation process accompanied by profound locus-specific and pangenomic epigenetic alterations. The epigenetic activities of histone-modifying and chromatin-remodeling enzymes such as SUV39H1/KMT1A, EZH2/KMT6A and BMI1 are central to epigenetic-regulated transformation, a property we coin oncoepigenic. Sarcoma-specific oncoepigenic aberrations modulate critical signaling pathways that control cell growth and differentiation including several miRNAs, Wnt, PI3K/AKT, Sav-RASSF1-Hpo and regulators of the G1 and G2/M checkpoints of the cell cycle. Herein an overview of the current knowledge of this rapidly evolving field that will undoubtedly uncover additional oncoepigenic mechanisms and yield druggable targets in the near future is discussed.
Collapse
|
171
|
Torsvik A, Bjerkvig R. Mesenchymal stem cell signaling in cancer progression. Cancer Treat Rev 2012; 39:180-8. [PMID: 22494966 DOI: 10.1016/j.ctrv.2012.03.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 03/09/2012] [Accepted: 03/13/2012] [Indexed: 12/22/2022]
Abstract
Mesenchymal (multipotent) stem/stromal cells (MSCs) may affect cancer progression through a number of secreted factors triggering activation of various cell signaling pathways. Depending on receptor status, phosphatase and tensin homolog (PTEN) status, or Wnt activation in the cancer cells, the signals may either result in increased growth and metastasis or lead to inhibition of growth with increased cell death. Thus, MSCs can play a dual role in cancer progression depending on the cellular context wherein they reside. The phosphatidylinositol-3-kinase (PI3K)/Akt signaling pathway has a central role in regulating tumor growth, and several MSC secreted factors stimulate activation of this pathway. A comprehensive understanding of the signals regulating MSC-tumor cross-talk is highly important for the development of MSCs as potential therapeutic vehicles. Thus, the presented review focuses on factors released by MSCs and on the dual role they may have on various stages of tumorigenesis.
Collapse
Affiliation(s)
- Anja Torsvik
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, N-5020 Bergen, Norway.
| | | |
Collapse
|
172
|
Martins-Neves SR, Lopes ÁO, do Carmo A, Paiva AA, Simões PC, Abrunhosa AJ, Gomes CMF. Therapeutic implications of an enriched cancer stem-like cell population in a human osteosarcoma cell line. BMC Cancer 2012; 12:139. [PMID: 22475227 PMCID: PMC3351999 DOI: 10.1186/1471-2407-12-139] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 04/04/2012] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Osteosarcoma is a bone-forming tumor of mesenchymal origin that presents a clinical pattern that is consistent with the cancer stem cell model. Cells with stem-like properties (CSCs) have been identified in several tumors and hypothesized as the responsible for the relative resistance to therapy and tumor relapses. In this study, we aimed to identify and characterize CSCs populations in a human osteosarcoma cell line and to explore their role in the responsiveness to conventional therapies. METHODS CSCs were isolated from the human MNNG/HOS cell line using the sphere formation assay and characterized in terms of self-renewal, mesenchymal stem cell properties, expression of pluripotency markers and ABC transporters, metabolic activity and tumorigenicity. Cell's sensitivity to conventional chemotherapeutic agents and to irradiation was analyzed and related with cell cycle-induced alterations and apoptosis. RESULTS The isolated CSCs were found to possess self-renewal and multipotential differentiation capabilities, express markers of pluripotent embryonic stem cells Oct4 and Nanog and the ABC transporters P-glycoprotein and BCRP, exhibit low metabolic activity and induce tumors in athymic mice. Compared with parental MNNG/HOS cells, CSCs were relatively more resistant to both chemotherapy and irradiation. None of the treatments have induced significant cell-cycle alterations and apoptosis in CSCs. CONCLUSIONS MNNG/HOS osteosarcoma cells contain a stem-like cell population relatively resistant to conventional chemotherapeutic agents and irradiation. This resistant phenotype appears to be related with some stem features, namely the high expression of the drug efflux transporters P-glycoprotein and BCRP and their quiescent nature, which may provide a biological basis for resistance to therapy and recurrence commonly observed in osteosarcoma.
Collapse
Affiliation(s)
- Sara R Martins-Neves
- Pharmacology and Experimental Therapeutics - Institute of Biomedical Research in Light and Image (IBILI), Faculty of Medicine, University of Coimbra, Az, de Sta. Comba, Celas, Coimbra 3000-354, Portugal
| | | | | | | | | | | | | |
Collapse
|
173
|
Kuijjer ML, Rydbeck H, Kresse SH, Buddingh EP, Lid AB, Roelofs H, Bürger H, Myklebost O, Hogendoorn PCW, Meza-Zepeda LA, Cleton-Jansen AM. Identification of osteosarcoma driver genes by integrative analysis of copy number and gene expression data. Genes Chromosomes Cancer 2012; 51:696-706. [PMID: 22454324 DOI: 10.1002/gcc.21956] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 03/02/2012] [Indexed: 12/11/2022] Open
Abstract
High-grade osteosarcoma is a tumor with a complex genomic profile, occurring primarily in adolescents with a second peak at middle age. The extensive genomic alterations obscure the identification of genes driving tumorigenesis during osteosarcoma development. To identify such driver genes, we integrated DNA copy number profiles (Affymetrix SNP 6.0) of 32 diagnostic biopsies with 84 expression profiles (Illumina Human-6 v2.0) of high-grade osteosarcoma as compared with its putative progenitor cells, i.e., mesenchymal stem cells (n = 12) or osteoblasts (n = 3). In addition, we performed paired analyses between copy number and expression profiles of a subset of 29 patients for which both DNA and mRNA profiles were available. Integrative analyses were performed in Nexus Copy Number software and statistical language R. Paired analyses were performed on all probes detecting significantly differentially expressed genes in corresponding LIMMA analyses. For both nonpaired and paired analyses, copy number aberration frequency was set to >35%. Nonpaired and paired integrative analyses resulted in 45 and 101 genes, respectively, which were present in both analyses using different control sets. Paired analyses detected >90% of all genes found with the corresponding nonpaired analyses. Remarkably, approximately twice as many genes as found in the corresponding nonpaired analyses were detected. Affected genes were intersected with differentially expressed genes in osteosarcoma cell lines, resulting in 31 new osteosarcoma driver genes. Cell division related genes, such as MCM4 and LATS2, were overrepresented and genomic instability was predictive for metastasis-free survival, suggesting that deregulation of the cell cycle is a driver of osteosarcomagenesis.
Collapse
Affiliation(s)
- Marieke L Kuijjer
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Osteosarcoma models: from cell lines to zebrafish. Sarcoma 2012; 2012:417271. [PMID: 22566751 PMCID: PMC3329665 DOI: 10.1155/2012/417271] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 11/24/2011] [Indexed: 01/14/2023] Open
Abstract
High-grade osteosarcoma is an aggressive tumor most commonly affecting adolescents. The early age of onset might suggest genetic predisposition; however, the vast majority of the tumors are sporadic. Early onset, most often lack of a predisposing condition or lesion, only infrequent (<2%) prevalence of inheritance, extensive genomic instability, and a wide histological heterogeneity are just few factors to mention that make osteosarcoma difficult to study. Therefore, it is sensible to design and use models representative of the human disease. Here we summarize multiple osteosarcoma models established in vitro and in vivo, comment on their utilities, and highlight newest achievements, such as the use of zebrafish embryos. We conclude that to gain a better understanding of osteosarcoma, simplification of this extremely complex tumor is needed. Therefore, we parse the osteosarcoma problem into parts and propose adequate models to study them each separately. A better understanding of osteosarcoma provides opportunities for discovering and assaying novel effective treatment strategies. “Sometimes the model is more interesting than the original disease” PJ Hoedemaeker (1937–2007).
Collapse
|
175
|
Searching for molecular targets in sarcoma. Biochem Pharmacol 2012; 84:1-10. [PMID: 22387046 DOI: 10.1016/j.bcp.2012.02.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 02/13/2012] [Accepted: 02/15/2012] [Indexed: 12/18/2022]
Abstract
Sarcoma are about 1% of cancers. Within that 1% are widely varied tumors now divided into types and subtypes. Sarcoma occur in patients of all ages with frequency spread evenly over the human age range. Although the specific cell of origin of many sarcoma remains unclear, sarcoma are all tumors of mesenchymal origin. The mesenchymal stem cell, a pluripotent cell, which gives rise to varied differentiated cells including osteocytes, adipocytes, chondrocytes, muscle cells, fibroblasts, neural cells and stromal cells, is the most likely ultimate cell of origin for sarcoma. When mesenchymal stem cell genetics go awry and malignant transformation occurs sarcoma including osteosarcoma, Ewing's sarcoma, chondrosarcoma, rhabdomyosarcoma, synovial sarcoma fibrosarcoma, liposarcoma and many others can initiate. Our knowledge of sarcoma genetics is increasing rapidly. Two general groups, sarcoma arising from chromosomal translocations and sarcoma with very complex genetics, can be identified. Genes that are frequently mutated in sarcoma include TP53, NF1, PIK3CA, HDAC1, IDH1 and 2, KDR, KIT and MED12. Genes that are frequently amplified in sarcoma include CDK4, YEATS4, HMGA2, MDM2, JUN, DNM3, FLT4, MYCN, MAP3K5, GLI1 and the microRNAs miR-214 and miR-199a2. Genes that are upregulated in sarcoma include MUC4, CD24, FOXL1, ANGPTL2, HIF1α, MDK, cMET, TIMP-2, PRL, PCSK1, IGFR-1, TIE1, KDR, TEK, FLT1 and several microRNAs. While some alterations occur in specific subtypes of sarcoma, others cross several sarcoma types. Discovering and developing new therapeutic approaches for these relentless diseases is critical. The detailed knowledge of sarcoma genetics may allow development of sarcoma subtype-targeted therapeutics.
Collapse
|
176
|
Putative multifunctional signature of lung metastases in dedifferentiated chondrosarcoma. Sarcoma 2012; 2012:820254. [PMID: 22448124 PMCID: PMC3289931 DOI: 10.1155/2012/820254] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 10/21/2011] [Accepted: 11/03/2011] [Indexed: 12/22/2022] Open
Abstract
Chondrosarcomas are among the most malignant skeletal tumors. Dedifferentiated chondrosarcoma is a highly aggressive subtype of chondrosarcoma, with lung metastases developing within a few months of diagnosis in 90% of patients. In this paper we performed comparative analyses of the transcriptomes of five individual metastatic lung lesions that were surgically resected from a patient with dedifferentiated chondrosarcoma. We document for the first time a high heterogeneity of gene expression profiles among the individual lung metastases. Moreover, we reveal a signature of “multifunctional” genes that are expressed in all metastatic lung lesions. Also, for the first time, we document the occurrence of massive macrophage infiltration in dedifferentiated chondrosarcoma lung metastases.
Collapse
|
177
|
van Maldegem AM, Bhosale A, Gelderblom HJ, Hogendoorn PC, Hassan AB. Comprehensive analysis of published phase I/II clinical trials between 1990-2010 in osteosarcoma and Ewing sarcoma confirms limited outcomes and need for translational investment. Clin Sarcoma Res 2012; 2:5. [PMID: 22587841 PMCID: PMC3351714 DOI: 10.1186/2045-3329-2-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 01/27/2012] [Indexed: 02/01/2023] Open
Abstract
Background High grade primary bone sarcomas are rare cancers that affect mostly children and young adults. Osteosarcoma and Ewing sarcoma are the most common histological subtypes in this age group, with current multimodality treatment strategies achieving 55-70% overall survival. As there remains an urgent need to develop new therapeutic interventions, we have reviewed published phase I/II trials that have been reported for osteosarcoma and Ewing sarcoma in the last twenty years. Results We conducted a literature search for clinical trials between 1990 and 2010, either for trials enrolling bone sarcoma patients as part of a general sarcoma indication or trials specifically in osteosarcoma and Ewing sarcoma. We identified 42 clinical trials that fulfilled our search criteria for general sarcoma that enrolled these patient groups, and eight and twenty specific trials for Ewing and osteosarcoma patients, respectively. For the phase I trials which enrolled different tumour types our results were incomplete, because the sarcoma patients were not mentioned in the PubMed abstract. A total of 3,736 sarcoma patients were included in these trials over this period, 1,114 for osteosarcoma and 1,263 for Ewing sarcoma. As a proportion of the worldwide disease burden over this period, these numbers reflect a very small percentage of the potential patient recruitment, approximately 0.6% for Ewing sarcoma and 0.2% for osteosarcoma. However, these data show an increase in recent activity overall and suggest there is still much room for improvement in the current trial development structures. Conclusion Lack of resources and commercial investment will inevitably limit opportunity to develop sufficiently rapid improvements in clinical outcomes. International collaboration exists in many well founded co-operative groups for phase III trials, but progress may be more effective if there were also more investment of molecular and translational research into disease focused phase I/II clinical trials. Examples of new models for early translational and early phase trial collaboration include the European based EuroBoNeT network, the Sarcoma Alliance for Research through Collaboration network (SARC) and the new European collaborative translational trial network, EuroSarc.
Collapse
Affiliation(s)
- Annemiek M van Maldegem
- Department of Oncology, Oxford Cancer and Haematology Centre, Churchill Hospital, University of Oxford, Oxford OX3 7LJ, UK
| | - Aparna Bhosale
- Department of Oncology, Oxford Cancer and Haematology Centre, Churchill Hospital, University of Oxford, Oxford OX3 7LJ, UK
| | - Hans J Gelderblom
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, PO Box 9600, 2600 RC Leiden, The Netherlands
| | - Pancras Cw Hogendoorn
- Department of Pathology, Leiden University Medical Center, Leiden, PO Box 9600, 2600 RC Leiden, The Netherlands
| | - Andrew B Hassan
- Department of Oncology, Oxford Cancer and Haematology Centre, Churchill Hospital, University of Oxford, Oxford OX3 7LJ, UK.,Sir William Dunn School of Pathology, South Parks Road, University of Oxford, Oxford OX1 3RE, UK
| |
Collapse
|
178
|
Patel N, Klassert TE, Greco SJ, Patel SA, Munoz JL, Reddy BY, Bryan M, Campbell N, Kokorina N, Sabaawy HE, Rameshwar P. Developmental regulation of TAC1 in peptidergic-induced human mesenchymal stem cells: implication for spinal cord injury in zebrafish. Stem Cells Dev 2012; 21:308-20. [PMID: 21671725 PMCID: PMC3258436 DOI: 10.1089/scd.2011.0179] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2011] [Accepted: 06/10/2011] [Indexed: 02/06/2023] Open
Abstract
Human mesenchymal stem cells (MSCs) are easy to expand, are relatively safe, and can be transplanted in allogeneic recipients as off-the-shelf cells. MSCs can be induced to form functional peptidergic neurons and express the neurotransmitter gene, TAC1. Expression of TAC1 requires that the repressor gene, RE-1 silencing transcription factor (REST), is decreased. This study investigated the molecular pathway in TAC1 induction as MSCs differentiated into neurons and then applied the findings in a model of spinal cord injury (SCI) in zebrafish. We studied the developmental roles of the 2 cAMP response element (CRE) sites: CRE1 and CRE2. Activator protein-1 (AP-1) binding site overlaps with CRE2 (CRE2/AP-1). Reporter gene studies with the 5' regulatory region of TAC1 containing wild-type or mutant CRE sites and, parallel studies with ectopically expressed inhibitor of cAMP proteins (inducible cAMP early repressor) indicated that CRE1 and CRE2/AP-1 are activated at days 6 and 12, respectively. Studies with protein kinase-A (PKA) and Jun N-terminal kinase (JNK) inhibitors in the reporter gene studies, chromatin immunoprecipation assay, and ectopic expression of REST indicated the following pathways: Decrease of REST activated upstream c-Jun N-terminal kinase (JNK). In turn, JNK activated ATF-2 and AP-1 for interaction with CRE1 and CRE2/AP-1, respectively. To apply the finding to SCI, we transplanted 6-day-induced MSCs in transgenic HB9-GFP zebrafish larvae with SCI, in the presence or absence of JNK inhibitors. Imaging and functional studies showed significant improvement in the fish. The repair mechanism involved the activation of JNK. The findings have long-term implications for SCI repair with MSCs.
Collapse
Affiliation(s)
- Nitixa Patel
- UMDNJ–New Jersey Medical School, Newark, New Jersey
| | - Tilman E. Klassert
- Instituto de Enfermedades Tropicales y Salud Pública, Universito of La Laguna, Canary Islands, Spain
| | | | | | | | | | | | - Neil Campbell
- UMDNJ–Robert Wood Johnson Medical School and Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Natalia Kokorina
- UMDNJ–Robert Wood Johnson Medical School and Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Hatem E. Sabaawy
- UMDNJ–Robert Wood Johnson Medical School and Cancer Institute of New Jersey, New Brunswick, New Jersey
| | | |
Collapse
|
179
|
Eberle I, Moslem M, Henschler R, Cantz T. Engineered MSCs from Patient-Specific iPS Cells. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2012; 130:1-17. [PMID: 22915200 DOI: 10.1007/10_2012_156] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mesenchymal stroma/stem cells (MSCs) represent a heterogenic cell population that can be isolated from various tissues of the body or can be generated from pluripotent stem cells by in vitro differentiation. Various promising pre-clinical and clinical studies suggest that MSCs might stimulate endogenous regeneration and/or act as anti-inflammatory agents, which could be of high therapeutic relevance for a number of diseases, including graft-versus-host disease after allogeneic hematopoietic stem cell transplantation, inflammatory bowel diseases, or some forms of liver failure. Notably, conflicting results of various studies illustrated that the source of MSCs, the cultivation condition, and the way of administration have important effects on the desired clinical effect. Some of the involved molecular pathways have recently been elucidated and an artificial modulation of these pathways by engineered MSCs might result in superfunctional MSCs for enhanced endogenous regeneration or anti-inflammatory response. In this review, we summarize important findings of conventional MSCs for applications in gastroenterology and we describe the state-of-the-art for the generation of patient-derived iPS cells that eventually might provide genetically engineered superfunctional iPS cells for advanced cell therapies.
Collapse
Affiliation(s)
- Irina Eberle
- Junior Research Group Stem Cell Biology, OE 8881, Cluster-of-Excellence REBIRTH, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | | | | | | |
Collapse
|
180
|
Migratory properties of mesenchymal stem cells. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2012; 129:117-36. [PMID: 22899378 DOI: 10.1007/10_2012_144] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells raise great expectations in regenerative medicine due to their capacity to regenerate damaged tissues, thereby restoring organ tissue integrity and functionality. Even though it is not yet clear how mesenchymal stem cells are guided to injured tissue it is generally assumed that the directed migration of these cells is facilitated by the same soluble factors that also recruit immune competent cells to inflamed tissue areas. Tumor tissue represents another type of (chronically) inflamed tissue and because of that mesenchymal stem cells are highly attracted. Although some data indicate that esenchymal stem cells might have a beneficial effect on tumor growth due to anti-tumor effects the plethora of data suggest that tumor tissue recruited mesenchymal stem cells rather promote tumor growth and metastasis formation. Nonetheless, the enhanced tumor tropism of mesenchymal stem cells makes them ideal candidates for novel anti-cancer strategies. Like Trojan Horses genetically modified mesenchymal stem cells will deliver their deadly cargo, such as anti-tumor cytokines or oncolytic viruses, into cancerous tissues, thereby destroying the tumor form within. In this chapter we will summarize the current concepts of genetic modification of mesenchymal stem cells for future anti-cancer therapies.
Collapse
|
181
|
Zeng W, Wan R, Zheng Y, Singh SR, Wei Y. Hypoxia, stem cells and bone tumor. Cancer Lett 2011; 313:129-36. [PMID: 21999934 PMCID: PMC3215823 DOI: 10.1016/j.canlet.2011.09.023] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 09/20/2011] [Accepted: 09/21/2011] [Indexed: 12/26/2022]
Abstract
Normal oxygen level is critical for niches that together with other components of the niche play vital role in regulating stem or tumor cells behavior. Hypoxia plays an important role in normal development and disease progression, including the growth of solid tumors. The hypoxia inducible factors (HIFs) are the key mediators of the cellular response to hypoxia. In this review, we focused on the role of HIFs on bone tumor formation. Further, we also emphasized how hypoxia, stem cells, and its niches regulate the bone tumorigenesis.
Collapse
Affiliation(s)
- Wen Zeng
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Shanghai First People’s Hospital, Shanghai Jiaotong University, 200080, People’s Republic of China
| | - Rong Wan
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Yuehuan Zheng
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Shree Ram Singh
- Mouse Cancer Genetics Program, National Institutes of Health, National Cancer Institute at Frederick, Frederick, Maryland, 21702, USA
| | - Yiyong Wei
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, People’s Republic of China
| |
Collapse
|
182
|
Pahl JH, Ruslan SEN, Buddingh EP, Santos SJ, Szuhai K, Serra M, Gelderblom H, Hogendoorn PC, Egeler RM, Schilham MW, Lankester AC. Anti-EGFR Antibody Cetuximab Enhances the Cytolytic Activity of Natural Killer Cells toward Osteosarcoma. Clin Cancer Res 2011; 18:432-41. [DOI: 10.1158/1078-0432.ccr-11-2277] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
183
|
Russinoff S, Miran S, Gowda AL, Lucas PA. Osteosarcoma cells differentiate into phenotypes from all three dermal layers. Clin Orthop Relat Res 2011; 469:2895-904. [PMID: 21678097 PMCID: PMC3171540 DOI: 10.1007/s11999-011-1946-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 06/01/2011] [Indexed: 01/31/2023]
Abstract
BACKGROUND Osteosarcomas are the most common solid malignant bone tumors, but little is known of their origin. The embryonal rest hypothesis views cancer cells as arising from committed progenitor stem cells in each tissue. Adult tissue contains primitive stem cells that retain the ability to differentiate across dermal lines, raising the possibility that the stem cell of origin of cancers may be from a more primitive stem cell than a progenitor. QUESTIONS/PURPOSES Can osteosarcoma cells, when cultured under conditions used for multipotent stem cells, be induced to differentiate into multiple phenotypes, including those of the three different dermal lineages: mesodermal, ectodermal, and endodermal? METHODS One rat and one human osteosarcoma cell line were cultured and treated with concentrations of 0, 10(-10), 10(-9), 10(-8), 10(-7), and 10(-6) mol/L dexamethasone for 5 weeks. Seventeen phenotypes were assayed either by tissue-specific histochemical stains or antibodies to tissue-specific proteins. Each phenotype was tested across all dexamethasone concentrations for each cell line and each phenotype was tested in three separate experiments with induction by dexamethasone RESULTS Rat osteosarcoma (ROS) 17/2.8 and human osteosarcoma cell line U-2 show the appearance of cells that have markers for (1) mesodermal phenotypes such as bone, cartilage, skeletal muscle, and endothelial cells, (2) ectodermal phenotypes such as astrocytes, oligodendrocytes, neurons, and keratinocytes, and (3) an endodermal phenotype, hepatocytes. This indicates osteosarcomas are composed, at least in part, of primitive stem cells capable of differentiating into tissues from all three dermal lineages. CLINICAL RELEVANCE If osteosarcomas arise from primitive stem cells, then treatment of osteosarcomas with exogenous differentiation agents may cause the stem cells to differentiate, thus halting their proliferation and stopping tumor growth.
Collapse
Affiliation(s)
- Scott Russinoff
- Department of Orthopedic Surgery, Macy Pavillion, New York Medical College, Valhalla, NY 10595 USA
| | - Sara Miran
- Department of Orthopedic Surgery, Macy Pavillion, New York Medical College, Valhalla, NY 10595 USA
| | - Ashok L. Gowda
- Department of Orthopedic Surgery, Macy Pavillion, New York Medical College, Valhalla, NY 10595 USA
| | - Paul A. Lucas
- Department of Orthopedic Surgery, Macy Pavillion, New York Medical College, Valhalla, NY 10595 USA
| |
Collapse
|
184
|
Rodriguez R, Rubio R, Menendez P. Modeling sarcomagenesis using multipotent mesenchymal stem cells. Cell Res 2011; 22:62-77. [PMID: 21931359 DOI: 10.1038/cr.2011.157] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Because of their unique properties, multipotent mesenchymal stem cells (MSCs) represent one of the most promising adult stem cells being used worldwide in a wide array of clinical applications. Overall, compelling evidence supports the long-term safety of ex vivo expanded human MSCs, which do not seem to transform spontaneously. However, experimental data reveal a link between MSCs and cancer, and MSCs have been reported to inhibit or promote tumor growth depending on yet undefined conditions. Interestingly, solid evidence based on transgenic mice and genetic intervention of MSCs has placed these cells as the most likely cell of origin for certain sarcomas. This research area is being increasingly explored to develop accurate MSC-based models of sarcomagenesis, which will be undoubtedly valuable in providing a better understanding about the etiology and pathogenesis of mesenchymal cancer, eventually leading to the development of more specific therapies directed against the sarcoma-initiating cell. Unfortunately, still little is known about the mechanisms underlying MSC transformation and further studies are required to develop bona fide sarcoma models based on human MSCs. Here, we comprehensively review the existing MSC-based models of sarcoma and discuss the most common mechanisms leading to tumoral transformation of MSCs and sarcomagenesis.
Collapse
Affiliation(s)
- Rene Rodriguez
- Pfizer-University of Granada-Andalusian Government Centre for Genomics and Oncological Research (GENyO), Parque Tecnológico de Ciencias de la Salud, Granada, Spain.
| | | | | |
Collapse
|
185
|
Otto WR, Wright NA. Mesenchymal stem cells: from experiment to clinic. FIBROGENESIS & TISSUE REPAIR 2011; 4:20. [PMID: 21902837 PMCID: PMC3182886 DOI: 10.1186/1755-1536-4-20] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 09/08/2011] [Indexed: 02/07/2023]
Abstract
There is currently much interest in adult mesenchymal stem cells (MSCs) and their ability to differentiate into other cell types, and to partake in the anatomy and physiology of remote organs. It is now clear these cells may be purified from several organs in the body besides bone marrow. MSCs take part in wound healing by contributing to myofibroblast and possibly fibroblast populations, and may be involved in epithelial tissue regeneration in certain organs, although this remains more controversial. In this review, we examine the ability of MSCs to modulate liver, kidney, heart and intestinal repair, and we update their opposing qualities of being less immunogenic and therefore tolerated in a transplant situation, yet being able to contribute to xenograft models of human tumour formation in other contexts. However, such observations have not been replicated in the clinic. Recent studies showing the clinical safety of MSC in several pathologies are discussed. The possible opposing powers of MSC need careful understanding and control if their clinical potential is to be realised with long-term safety for patients.
Collapse
Affiliation(s)
- William R Otto
- Histopathology Laboratory, Cancer Research UK, London Research Institute, 44, Lincoln's Inn Fields, London WC2A 3LY, UK.
| | | |
Collapse
|
186
|
Skårn M, Namløs HM, Noordhuis P, Wang MY, Meza-Zepeda LA, Myklebost O. Adipocyte differentiation of human bone marrow-derived stromal cells is modulated by microRNA-155, microRNA-221, and microRNA-222. Stem Cells Dev 2011; 21:873-83. [PMID: 21756067 DOI: 10.1089/scd.2010.0503] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Human mesenchymal stromal cells (hMSCs) are capable of limited self-renewal and multilineage differentiation in vitro. Several studies have demonstrated that microRNAs (miRNAs, miRs), post-transcriptional modifiers of mRNA stability and protein translation, play crucial roles in the regulation of these complex processes. To gain knowledge regarding the role of miRNAs in human adipocyte differentiation, we examined the miRNA expression profile of the immortalized human bone marrow-derived stromal cell line hMSC-Tert20. Such a model system has the advantage of a reproducible and consistent phenotype while maintaining important properties of the primary donor cells, including the potential to differentiate to adipocytes, osteoblasts, and chondrocytes. We identified 12 miRNAs that were differentially expressed during adipogenesis, of which several have been previously shown to play important roles in adipocyte biology. Among these, the expression of miRNA-155, miRNA-221, and miRNA-222 decreased during the adipogenic program of both immortalized and primary hMSCs, suggesting that they act as negative regulators of differentiation. Interestingly, ectopic expression of the miRNAs significantly inhibited adipogenesis and repressed induction of the master regulators PPARγ and CCAAT/enhancer-binding protein alpha. Our study provides the first experimental evidence that miRNA-155, miRNA-221, and miRNA-222 have an important function in human adipocyte differentiation, and that their downregulation is necessary to relieve the repression of genes crucial for this process.
Collapse
Affiliation(s)
- Magne Skårn
- Department of Tumor Biology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | | | | | | | | | | |
Collapse
|
187
|
Mohseny AB, Machado I, Cai Y, Schaefer KL, Serra M, Hogendoorn PCW, Llombart-Bosch A, Cleton-Jansen AM. Functional characterization of osteosarcoma cell lines provides representative models to study the human disease. J Transl Med 2011; 91:1195-205. [PMID: 21519327 DOI: 10.1038/labinvest.2011.72] [Citation(s) in RCA: 146] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cancer cell lines represent in vitro models for studying malignancies, general cell biology, drug discovery and more. Whether they can be considered as exact representative models of the parental tumors remains uncertain given the acquisition of additional ex vivo changes of the cells and the lack of tissue architecture and stroma. Previously, within the EuroBoNeT consortium, we characterized a collection of bone sarcoma cell lines on genomic and proteomic level. Here, we address the phenotypical and functional characterization of the unique set of osteosarcoma cell lines (n=19) in vitro and in vivo. For functional analysis of differentiation capacity, cells were stimulated towards osteoblasts, adipocytes and chondrocytes. Furthermore, all cell lines were injected subcutaneously and intramuscularly into nude mice to assay their in vivo tumor formation capacity as well as for phenotypical analysis of the tumors. All formed tumors were further characterized histologically and immunohistochemically. Out of 19 cell lines, 17 (89%) showed adipogenic differentiation, 13/19 (68%) could differentiate towards osteoblasts and in 6/19 (32%) cell lines chondrogenic differentiation was evident. About half of the cell lines (8/19, 42%) produced tumors in vivo after subcutaneous and intramuscular injections. Several cell lines showed invasion into adjacent tissues and one tumor developed several lung metastases. The use of cell lines, especially in cancer research, is of paramount importance. Here, we identify comprehensively characterized osteosarcoma cell lines, which robustly represent clinical osteosarcoma providing researchers useful in vitro and in vivo models to study the genetics and functional characteristics of this highly malignant neoplasm.
Collapse
Affiliation(s)
- Alexander B Mohseny
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
188
|
Mohseny AB, Hogendoorn PCW. Concise review: mesenchymal tumors: when stem cells go mad. Stem Cells 2011; 29:397-403. [PMID: 21425403 DOI: 10.1002/stem.596] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Sarcomas are nonepithelial, nonhematopoietic malignant tumors that arise from the embryonic mesoderm. Despite their rarity, less than 10% of all cancers, sarcomas are accountable for relatively high morbidity and mortality especially in children and adolescents. Although there are some hereditary conditions predisposing sarcoma, such as the Li-Fraumeni and Retinoblastoma syndrome, the vast majority of these tumors are sporadic. Based on their histological morphology, sarcomas have been divided into a broad spectrum of subtypes recognized in the 2002 WHO classification of tumors. This wide lineage range suggests that sarcomas originate from either many committed different cell types or from a multipotent cell, subsequently driven into a certain lineage. Mesenchymal stem cells (MSCs) are able to differentiate into many cell types needed to create mature structures like vessels, muscle, and bone. These multipotent cells can be isolated from several adult human tissues and massively expanded in culture, making them both of use for research as well as potential beneficial therapeutical agents. For this reason MSCs are being extensively studied, however, concerns have raised about whether they are the putative originating cells of sarcoma and their questionable role in cancer progression. Recent accomplishments in the field have broadened our knowledge of MSCs in relation to sarcoma origin, sarcoma treatment and the safety of MSCs usage in therapeutic settings.
Collapse
Affiliation(s)
- Alexander B Mohseny
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
189
|
Abstract
Osteosarcoma is a highly malignant bone tumor of children and young adults. Cytotoxic chemotherapy combined with aggressive surgery only has a 60% survival rate. Historically, chemotherapy has been developed assuming that all cells within a particular cancer are clonal and near identical. Appreciating the now apparent functional heterogeneity of osteosarcoma cells within and between individual tumors will likely be critical in developing much needed novel effective therapies. The foundation for this heterogeneity may lie in the so called "cancer stem cell" or tumorigenic cell of origin. In this brief review, we will examine the evidence for the existence of this cell and its potential importance for future therapies.
Collapse
|
190
|
Alterations of cytological and karyological profile of human mesenchymal stem cells during in vitro culturing. Bull Exp Biol Med 2011; 150:125-30. [PMID: 21161070 DOI: 10.1007/s10517-010-1086-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Transplantation of human bone marrow mesenchymal stem cells is considered as a promising therapeutic approach to the therapy of many diseases. However, the problem of possible alterations of the properties of mesenchymal stem cells during their expansion in in vitro cultures before transplantation is not solved. In our study, one of two hundred examined cultures of mesenchymal stem cell cultures derived from donors without bone marrow pathologies and developed under standard culturing conditions demonstrated spontaneous disturbances in morphology, proliferation, and karyotype at early passages. The cells of this abnormal culture retained immunophenotype characteristic of normal mesenchymal stem cells, but some of them (15-25%) had numerous numerical and structural chromosome aberrations.
Collapse
|
191
|
Buddingh EP, Kuijjer ML, Duim RA, Bürger H, Agelopoulos K, Myklebost O, Serra M, Mertens F, Hogendoorn PC, Lankester AC, Cleton-Jansen AM. Tumor-Infiltrating Macrophages Are Associated with Metastasis Suppression in High-Grade Osteosarcoma: A Rationale for Treatment with Macrophage Activating Agents. Clin Cancer Res 2011; 17:2110-9. [DOI: 10.1158/1078-0432.ccr-10-2047] [Citation(s) in RCA: 281] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
192
|
Buddingh EP, Schilham MW, Ruslan SEN, Berghuis D, Szuhai K, Suurmond J, Taminiau AHM, Gelderblom H, Egeler RM, Serra M, Hogendoorn PCW, Lankester AC. Chemotherapy-resistant osteosarcoma is highly susceptible to IL-15-activated allogeneic and autologous NK cells. Cancer Immunol Immunother 2011; 60:575-86. [PMID: 21240486 PMCID: PMC3061210 DOI: 10.1007/s00262-010-0965-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2010] [Accepted: 12/22/2010] [Indexed: 01/04/2023]
Abstract
High-grade osteosarcoma occurs predominantly in adolescents and young adults and has an overall survival rate of about 60%, despite chemotherapy and surgery. Therefore, novel treatment modalities are needed to prevent or treat recurrent disease. Natural killer (NK) cells are lymphocytes with cytotoxic activity toward virus-infected or malignant cells. We explored the feasibility of autologous and allogeneic NK cell-mediated therapies for chemotherapy-resistant and chemotherapy-sensitive high-grade osteosarcoma. The expression by osteosarcoma cells of ligands for activating NK cell receptors was studied in vitro and in vivo, and their contribution to NK cell-mediated cytolysis was studied by specific antibody blockade. Chromium release cytotoxicity assays revealed chemotherapy-sensitive and chemotherapy-resistant osteosarcoma cell lines and osteosarcoma primary cultures to be sensitive to NK cell-mediated cytolysis. Cytolytic activity was strongly enhanced by IL-15 activation and was dependent on DNAM-1 and NKG2D pathways. Autologous and allogeneic activated NK cells lysed osteosarcoma primary cultures equally well. Osteosarcoma patient-derived NK cells were functionally and phenotypically unimpaired. In conclusion, osteosarcoma cells, including chemoresistant variants, are highly susceptible to lysis by IL-15-induced NK cells from both allogeneic and autologous origin. Our data support the exploitation of NK cells or NK cell-activating agents in patients with high-grade osteosarcoma.
Collapse
Affiliation(s)
- Emilie P Buddingh
- Department of Pediatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Josse C, Schoemans R, Niessen NA, Delgaudine M, Hellin AC, Herens C, Delvenne P, Bours V. Systematic chromosomal aberrations found in murine bone marrow-derived mesenchymal stem cells. Stem Cells Dev 2011; 19:1167-73. [PMID: 20109032 DOI: 10.1089/scd.2009.0264] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are studied as a cellular source for the treatment of various diseases. In this work, we isolated and cultivated murine bone marrow-derived MSCs. After a first observation of a solid tumor in a mouse injected with these cells, we systematically explored their chromosomal stability. We observed in all the cytogenetically analyzed cases gross chromosomal alterations every time the MSCs went through the senescence crisis while the lymphocytes from the same animals showed a normal chromosome count. This observation was confirmed in different mouse strains, with different culture protocols, and even in short-term cultures after a hematopoietic cell negative immunodepletion performed in order to accelerate the isolation procedure. Therefore, we conclude that murine MSCs display high chromosomal instability and can generate tumors, and that care must be taken before using them for the evaluation of MSC therapeutic potential.
Collapse
Affiliation(s)
- Claire Josse
- Department of Human Genetics, Liège University, Liège, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
194
|
David E, Guihard P, Brounais B, Riet A, Charrier C, Battaglia S, Gouin F, Ponsolle S, Bot RL, Richards CD, Heymann D, Rédini F, Blanchard F. Direct anti-cancer effect of oncostatin M on chondrosarcoma. Int J Cancer 2011; 128:1822-35. [PMID: 21344373 DOI: 10.1002/ijc.25776] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Accepted: 10/22/2010] [Indexed: 12/24/2022]
Abstract
The cytokine Oncostatin M (OSM) is cytostatic, pro-apoptotic and induces differentiation of osteosarcoma cells into osteocytes, suggesting new adjuvant treatment for these bone-forming sarcomas. However, OSM systemic over-expression could lead to adverse side effects such as generalized inflammation, neoangiogenesis and osteolysis. We determine here the effect of OSM on chondrosarcoma, another primary bone sarcoma characterized by the production of cartilage matrix and altered bone remodelling. Chondrosarcomas are resistant to conventional chemotherapy and radiotherapy, and wide surgical excision remains the only available treatment. We found that OSM blocked the cell cycle in four of five chondrosarcoma cell lines, independently of p53 and presumably through the JAK3/STAT1 pathway. In two tested cell lines, OSM induced a hypertrophic chondrocyte differentiation, with an induced Cbfa1/SOX9 ratio and induced Coll10, matrix metalloproteinase 13 (MMP13) and RANKL expression. Adenoviral gene transfer of OSM (AdOSM) in the Swarm rat chondrosarcoma (SRC) model indicated that local intra-tumoral OSM over-expression reduces chondrosarcoma development not only with reduced tumor proliferation and enhanced apoptosis but also with enhanced RANKL expression, osteoclast formation and reduced bone volumes. Flu-like symptoms were induced by the AdOSM, but there was no effect on tumor angiogenesis. Therefore, OSM could be considered as a new adjuvant anti-cancer agent for chondrosarcomas. A local application of this cytokine is presumably needed to overcome the poor vascularization of these tumors and to limit the deleterious effect on other tissues. Its side effect on bone remodeling could be managed with anti-resorption agents, thus offering potential new lines of therapeutic interventions.
Collapse
|
195
|
Abstract
Osteosarcoma is an aggressive but ill-understood cancer of bone that predominantly affects adolescents. Its rarity and biological heterogeneity have limited studies of its molecular basis. In recent years, an important role has emerged for the RUNX2 "platform protein" in osteosarcoma oncogenesis. RUNX proteins are DNA-binding transcription factors that regulate the expression of multiple genes involved in cellular differentiation and cell-cycle progression. RUNX2 is genetically essential for developing bone and osteoblast maturation. Studies of osteosarcoma tumours have revealed that the RUNX2 DNA copy number together with RNA and protein levels are highly elevated in osteosarcoma tumors. The protein is also important for metastatic bone disease of prostate and breast cancers, while RUNX2 may have both tumor suppressive and oncogenic roles in bone morphogenesis. This paper provides a synopsis of the current understanding of the functions of RUNX2 and its potential role in osteosarcoma and suggests directions for future study.
Collapse
|
196
|
Abstract
Osteosarcoma is the most common type of solid bone cancer and the second leading cause of cancer-related death in pediatric patients. Many patients are not cured by the current osteosarcoma therapy consisting of combination chemotherapy along with surgery and thus new treatments are urgently needed. In the last decade, cancer stem cells have been identified in many tumors such as leukemia, brain, breast, head and neck, colon, skin, pancreatic, and prostate cancers and these cells are proposed to play major roles in drug resistance, tumor recurrence, and metastasis. Recent studies have shown evidence that osteosarcoma also possesses cancer stem cells. This review summarizes the current knowledge about the osteosarcoma cancer stem cell including the methods used for its isolation, its properties, and its potential as a new target for osteosarcoma treatment.
Collapse
Affiliation(s)
- Valerie A Siclari
- Department of Orthopaedic Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.
| | | |
Collapse
|
197
|
Alison MR, Lim SML, Nicholson LJ. Cancer stem cells: problems for therapy? J Pathol 2010; 223:147-61. [PMID: 21125672 DOI: 10.1002/path.2793] [Citation(s) in RCA: 212] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2010] [Revised: 09/13/2010] [Accepted: 09/21/2010] [Indexed: 12/20/2022]
Abstract
Many, if not all, tumours contain a sub-population of self-renewing and expanding stem cells known as cancer stem cells (CSCs). The symmetric division of CSCs is one mechanism enabling expansion in their numbers as tumours grow, while epithelial-mesenchymal transition (EMT) is an increasingly recognized mechanism to generate further CSCs endowed with a more invasive and metastatic phenotype. Putative CSCs are prospectively isolated using methods based on either a surface marker or an intracellular enzyme activity and then assessed by a 'sphere-forming' assay in non-adherent culture and/or by their ability to initiate new tumour growth when xenotransplanted into immunocompromised mice-hence, these cells are often referred to as tumour-propagating cells (TPCs). Cell sub-populations enriched for tumour-initiating ability have also been found in murine tumours, countering the argument that xenografting human cells merely select human cells with an ability to grow in mice. Cancer progression can be viewed as an evolutionary process that generates new/multiple clones with a fresh identity; this may be a major obstacle to successful cancer stem cell eradication if treatment targets only a single type of stem cell. In this review, we first briefly discuss evidence that cancer can originate from normal stem cells or closely related descendants. We then outline the attributes of CSCs and review studies in which they have been identified in various cancers. Finally, we discuss the implications of these findings for successful cancer therapies, concentrating on the self-renewal pathways (Wnt, Notch, and Hedgehog), aldehyde dehydrogenase activity, EMT, miRNAs, and other epigenetic modifiers as potential targets for therapeutic manipulation.
Collapse
Affiliation(s)
- Malcolm R Alison
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | | | | |
Collapse
|
198
|
Picci P, Sieberova G, Alberghini M, Balladelli A, Vanel D, Hogendoorn PCW, Mercuri M. Late sarcoma development after curettage and bone grafting of benign bone tumors. Eur J Radiol 2010; 77:19-25. [PMID: 20828957 DOI: 10.1016/j.ejrad.2010.06.053] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 06/15/2010] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND AIM Rarely sarcomas develop in previous benign lesions, after a long term disease free interval. We report the experience on these rare cases observed at a single Institution. PATIENTS AND METHODS 12 cases curetted and grafted, without radiotherapy developed sarcomas, between 1970 and 2005, 6.5-28 years from curettage (median 18, average 19). Age ranged from 13 to 55 years (median 30, average 32) at first diagnosis; tumors were located in the extremities (9 GCT, benign fibrous histiocytoma, ABC, and solitary bone cyst). Radiographic and clinic documentation, for the benign and malignant lesions, were available. Histology was available for 7 benign and all malignant lesions. RESULTS To fill cavities, autogenous bone was used in 4 cases, allograft in 2, allograft and tricalcium-phosphate/hydroxyapatite in 1, autogenous/allograft in 1, heterogenous in 1. For 3 cases the origin was not reported. Secondary sarcomas, all high grade, were 8 osteosarcoma, 3 malignant fibrous histiocytoma, and 1 fibrosarcoma. CONCLUSIONS Recurrences with progression from benign tumors are possible, but the very long intervals here reported suggest a different cancerogenesis for these sarcomas. This condition is extremely rare accounting for only 0.26% of all malignant bone sarcomas treated in the years 1970-2005 and represents only 8.76% of all secondary bone sarcomas treated in the same years. This incidence is the same as that of sarcomas arising on fibrous dysplasia, and is lower than those arising on bone infarcts or on Paget's disease. This possible event must be considered during follow-up of benign lesions.
Collapse
Affiliation(s)
- Piero Picci
- Bone Tumor Center, Istituto Ortopedico Rizzoli, Bologna, Italy.
| | | | | | | | | | | | | |
Collapse
|
199
|
Mohseny AB, Tieken C, van der Velden PA, Szuhai K, de Andrea C, Hogendoorn PCW, Cleton-Jansen AM. Small deletions but not methylation underlie CDKN2A/p16 loss of expression in conventional osteosarcoma. Genes Chromosomes Cancer 2010; 49:1095-103. [DOI: 10.1002/gcc.20817] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
200
|
Torsvik A, Røsland GV, Svendsen A, Molven A, Immervoll H, McCormack E, Lønning PE, Primon M, Sobala E, Tonn JC, Goldbrunner R, Schichor C, Mysliwietz J, Lah TT, Motaln H, Knappskog S, Bjerkvig R. Spontaneous malignant transformation of human mesenchymal stem cells reflects cross-contamination: putting the research field on track - letter. Cancer Res 2010; 70:6393-6. [PMID: 20631079 DOI: 10.1158/0008-5472.can-10-1305] [Citation(s) in RCA: 212] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|