101
|
Martins-Neves SR, Corver WE, Paiva-Oliveira DI, van den Akker BEWM, Briaire-de-Bruijn IH, Bovée JVMG, Gomes CMF, Cleton-Jansen AM. Osteosarcoma Stem Cells Have Active Wnt/β-catenin and Overexpress SOX2 and KLF4. J Cell Physiol 2015; 231:876-86. [DOI: 10.1002/jcp.25179] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/31/2015] [Indexed: 12/31/2022]
Affiliation(s)
- Sara R. Martins-Neves
- Department of Pathology; Leiden University Medical Center; Leiden The Netherlands
- Pharmacology and Experimental Therapeutics; Institute for Biomedical Imaging and Life Sciences (IBILI); Faculty of Medicine; University of Coimbra; Coimbra Portugal
- CNC.IBILI; University of Coimbra; Coimbra Portugal
- Center of Investigation in Environment; Genetics and Oncobiology; CIMAGO - Faculty of Medicine; University of Coimbra, Coimbra; Celas Portugal
| | - Willem E. Corver
- Department of Pathology; Leiden University Medical Center; Leiden The Netherlands
| | - Daniela I. Paiva-Oliveira
- Pharmacology and Experimental Therapeutics; Institute for Biomedical Imaging and Life Sciences (IBILI); Faculty of Medicine; University of Coimbra; Coimbra Portugal
- CNC.IBILI; University of Coimbra; Coimbra Portugal
| | | | | | | | - Célia M. F. Gomes
- Pharmacology and Experimental Therapeutics; Institute for Biomedical Imaging and Life Sciences (IBILI); Faculty of Medicine; University of Coimbra; Coimbra Portugal
- CNC.IBILI; University of Coimbra; Coimbra Portugal
- Center of Investigation in Environment; Genetics and Oncobiology; CIMAGO - Faculty of Medicine; University of Coimbra, Coimbra; Celas Portugal
| | | |
Collapse
|
102
|
Kong G, Liu Z, Wu K, Zhang Y, Deng Z, Feng W, Chen S, Wang H. Strong expression of paired-like homeodomain transcription factor 1 (PITX1) is associated with a favorable outcome in human osteosarcoma. Tumour Biol 2015; 36:7735-41. [PMID: 25936343 DOI: 10.1007/s13277-015-3512-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 04/27/2015] [Indexed: 02/05/2023] Open
Abstract
Paired-like homeodomain transcription factor 1 (PITX1) has been implicated as a tumor suppressor in various cancers. However, the biological and clinical significance of PITX1 in osteosarcoma has not been fully elucidated. Here, we studied the expression and clinical significance of PITX1 in 6 normal lower limb bone tissue specimens and 35 osteosarcoma tissue samples by immunohistochemistry. PITX1 was expressed in all normal tissues (6/6, 100 %) and in 85.7 % (30/35) of tumor tissues (P > 0.05). In addition, all normal tissue specimens showed high PITX1 expression (6/6, 100 %) while only 23.3 % (7/30) osteosarcoma tissue specimens had high PITX1 expression (P < 0.05). Patients with median overall survival (OS) >12 months had significantly higher PITX1 levels compared with those whose median OS was less than or equal to 12 months (P < 0.05 or 0.001). Furthermore, patients with lung metastasis had significantly lower PITX1 levels than patients without lung metastasis. In conclusion, PITX1 expression is downregulated in osteosarcoma and correlates with patient survival and lung metastasis.
Collapse
Affiliation(s)
- Gengbin Kong
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Zhaoyong Liu
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Kezhou Wu
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Ying Zhang
- Department of Pathology, Shantou University Medical College, No.22 Xinling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Zhihua Deng
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Weili Feng
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Shubiao Chen
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Hu Wang
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China.
| |
Collapse
|
103
|
Buddingh EP, Ruslan SEN, Reijnders CMA, Szuhai K, Kuijjer ML, Roelofs H, Hogendoorn PCW, Maarten Egeler R, Cleton-Jansen AM, Lankester AC. Mesenchymal stromal cells of osteosarcoma patients do not show evidence of neoplastic changes during long-term culture. Clin Sarcoma Res 2015; 5:16. [PMID: 26106474 PMCID: PMC4477606 DOI: 10.1186/s13569-015-0031-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/12/2015] [Indexed: 01/02/2023] Open
Abstract
Background In vitro expanded mesenchymal stromal cells (MSCs) are increasingly used as experimental cellular therapy. However, there have been concerns regarding the safety of their use, particularly with regard to possible oncogenic transformation. MSCs are the hypothesized precursor cells of high-grade osteosarcoma, a tumor with often complex karyotypes occurring mainly in adolescents and young adults. Methods To determine if MSCs from osteosarcoma patients could be predisposed to malignant transformation we cultured MSCs of nine osteosarcoma patients and five healthy donors for an average of 649 days (range 601–679 days). Also, we compared MSCs derived from osteosarcoma patients at diagnosis and from healthy donors using genome wide gene expression profiling. Results Upon increasing passage, increasing frequencies of binucleate cells were detected, but no increase in proliferation suggestive of malignant transformation occurred in MSCs from either patients or donors. Hematopoietic cell specific Lyn substrate 1 (HLCS1) was differentially expressed (fold change 0.25, P value 0.0005) between MSCs of osteosarcoma patients (n = 14) and healthy donors (n = 9). Conclusions This study shows that although HCLS1 expression was downregulated in MSCs of osteosarcoma patients and binucleate cells were present in both patient and donor derived MSCs, there was no evidence of neoplastic changes to occur during long-term culture. Electronic supplementary material The online version of this article (doi:10.1186/s13569-015-0031-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Emilie P Buddingh
- Department of Pediatrics, J6-S, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - S Eriaty N Ruslan
- Department of Pediatrics, J6-S, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | | | - Karoly Szuhai
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marieke L Kuijjer
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Helene Roelofs
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | | - R Maarten Egeler
- Department of Pediatrics, J6-S, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | | | - Arjan C Lankester
- Department of Pediatrics, J6-S, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
104
|
Maugg D, Rothenaigner I, Schorpp K, Potukuchi HK, Korsching E, Baumhoer D, Hadian K, Smida J, Nathrath M. New small molecules targeting apoptosis and cell viability in osteosarcoma. PLoS One 2015; 10:e0129058. [PMID: 26039064 PMCID: PMC4454490 DOI: 10.1371/journal.pone.0129058] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Accepted: 05/04/2015] [Indexed: 01/20/2023] Open
Abstract
Despite the option of multimodal therapy in the treatment strategies of osteosarcoma (OS), the most common primary malignant bone tumor, the standard therapy has not changed over the last decades and still involves multidrug chemotherapy and radical surgery. Although successfully applied in many patients a large number of patients eventually develop recurrent or metastatic disease in which current therapeutic regimens often lack efficacy. Thus, new therapeutic strategies are urgently needed. In this study, we performed a phenotypic high-throughput screening campaign using a 25,000 small-molecule diversity library to identify new small molecules selectively targeting osteosarcoma cells. We could identify two new small molecules that specifically reduced cell viability in OS cell lines U2OS and HOS, but affected neither hepatocellular carcinoma cell line (HepG2) nor primary human osteoblasts (hOB). In addition, the two compounds induced caspase 3 and 7 activity in the U2OS cell line. Compared to conventional drugs generally used in OS treatment such as doxorubicin, we indeed observed a greater sensitivity of OS cell viability to the newly identified compounds compared to doxorubicin and staurosporine. The p53-negative OS cell line Saos-2 almost completely lacked sensitivity to compound treatment that could indicate a role of p53 in the drug response. Taken together, our data show potential implications for designing more efficient therapies in OS.
Collapse
Affiliation(s)
- Doris Maugg
- Clinical Cooperation Group Osteosarcoma, Institute of Radiation Biology, Helmholtz Zentrum München—National Research Centre for Environmental Health, Neuherberg, Germany
- Department of Pediatrics and Children´s Cancer Research Center, Technische Universität München, Munich, Germany
- * E-mail:
| | - Ina Rothenaigner
- Assay Development and Screening Platform, Institute for Molecular Toxicology and Pharmacology, Helmholtz Zentrum München—National Research Centre for Environmental Health, Neuherberg, Germany
| | - Kenji Schorpp
- Assay Development and Screening Platform, Institute for Molecular Toxicology and Pharmacology, Helmholtz Zentrum München—National Research Centre for Environmental Health, Neuherberg, Germany
| | - Harish Kumar Potukuchi
- Lehrstuhl für Organische Chemie I and Catalysis Research Center (CRC), Technische Universität München, Garching, Germany
- Institute of Structural Biology, Helmholtz Zentrum München—National Research Centre for Environmental Health, Neuherberg, Germany
| | | | - Daniel Baumhoer
- Clinical Cooperation Group Osteosarcoma, Institute of Radiation Biology, Helmholtz Zentrum München—National Research Centre for Environmental Health, Neuherberg, Germany
- Bone Tumor Reference Center at the Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Kamyar Hadian
- Assay Development and Screening Platform, Institute for Molecular Toxicology and Pharmacology, Helmholtz Zentrum München—National Research Centre for Environmental Health, Neuherberg, Germany
| | - Jan Smida
- Clinical Cooperation Group Osteosarcoma, Institute of Radiation Biology, Helmholtz Zentrum München—National Research Centre for Environmental Health, Neuherberg, Germany
- Department of Pediatrics and Children´s Cancer Research Center, Technische Universität München, Munich, Germany
| | - Michaela Nathrath
- Clinical Cooperation Group Osteosarcoma, Institute of Radiation Biology, Helmholtz Zentrum München—National Research Centre for Environmental Health, Neuherberg, Germany
- Department of Pediatrics and Children´s Cancer Research Center, Technische Universität München, Munich, Germany
- Department of Pediatric Oncology, Klinikum Kassel, Kassel, Germany
| |
Collapse
|
105
|
Expression of α-Smooth Muscle Actin Determines the Fate of Mesenchymal Stromal Cells. Stem Cell Reports 2015; 4:1016-30. [PMID: 26028530 PMCID: PMC4471834 DOI: 10.1016/j.stemcr.2015.05.004] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 05/04/2015] [Accepted: 05/04/2015] [Indexed: 02/07/2023] Open
Abstract
Pro-fibrotic microenvironments of scars and tumors characterized by increased stiffness stimulate mesenchymal stromal cells (MSCs) to express α-smooth muscle actin (α-SMA). We investigated whether incorporation of α-SMA into contractile stress fibers regulates human MSC fate. Sorted α-SMA-positive MSCs exhibited high contractile activity, low clonogenicity, and differentiation potential limited to osteogenesis. Knockdown of α-SMA was sufficient to restore clonogenicity and adipogenesis in MSCs. Conversely, α-SMA overexpression induced YAP translocation to the nucleus and reduced the high clonogenicity and adipogenic potential of α-SMA-negative MSCs. Inhibition of YAP rescued the decreased adipogenic differentiation potential induced by α-SMA, establishing a mechanistic link between matrix stiffness, α-SMA, YAP, and MSC differentiation. Consistent with in vitro findings, nuclear localization of YAP was positively correlated in α-SMA expressing stromal cells of adiposarcoma and osteosarcoma. We propose that α-SMA mediated contraction plays a critical role in mechanically regulating MSC fate by controlling YAP/TAZ activation. The α-SMA-positive myofibroblast fraction of human MSCs exhibits low clonogenicity Formation of α-SMA stress fibers enhances nuclear translocation of YAP/TAZ in MSCs α-SMA knockdown favors adipogenesis, while overexpression promotes osteogenesis α-SMA-mediated lineage choice of MSCs is YAP dependent
Collapse
|
106
|
Transcriptional Dynamics of Immortalized Human Mesenchymal Stem Cells during Transformation. PLoS One 2015; 10:e0126562. [PMID: 25978455 PMCID: PMC4433180 DOI: 10.1371/journal.pone.0126562] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 04/03/2015] [Indexed: 12/23/2022] Open
Abstract
Comprehensive analysis of alterations in gene expression along with neoplastic transformation in human cells provides valuable information about the molecular mechanisms underlying transformation. To further address these questions, we performed whole transcriptome analysis to the human mesenchymal stem cell line, UE6E7T-3, which was immortalized with hTERT and human papillomavirus type 16 E6/E7 genes, in association with progress of transformation in these cells. At early stages of culture, UE6E7T-3 cells preferentially lost one copy of chromosome 13, as previously described; in addition, tumor suppressor genes, DNA repair genes, and apoptosis-activating genes were overexpressed. After the loss of chromosome 13, additional aneuploidy and genetic alterations that drove progressive transformation, were observed. At this stage, the cell line expressed oncogenes as well as genes related to anti-apoptotic functions, cell-cycle progression, and chromosome instability (CIN); these pro-tumorigenic changes were concomitant with a decrease in tumor suppressor gene expression. At later stages after prolong culture, the cells exhibited chromosome translocations, acquired anchorage-independent growth and tumorigenicity in nude mice, (sarcoma) and exhibited increased expression of genes encoding growth factor and DNA repair genes, and decreased expression of adhesion genes. In particular, glypican-5 (GPC5), which encodes a cell-surface proteoglycan that might be a biomarker for sarcoma, was expressed at high levels in association with transformation. Patched (Ptc1), the cell surface receptor for hedgehog (Hh) signaling, was also significantly overexpressed and co-localized with GPC5. Knockdown of GPC5 expression decreased cell proliferation, suggesting that it plays a key role in growth in U3-DT cells (transformants derived from UE6E7T-3 cells) through the Hh signaling pathway. Thus, the UE6E7T-3 cell culture model is a useful tool for assessing the functional contribution of genes showed by expression profiling to the neoplastic transformation of human fibroblasts and human mesenchymal stem cells (hMSC).
Collapse
|
107
|
Robl B, Pauli C, Botter SM, Bode-Lesniewska B, Fuchs B. Prognostic value of tumor suppressors in osteosarcoma before and after neoadjuvant chemotherapy. BMC Cancer 2015; 15:379. [PMID: 25956431 PMCID: PMC4435808 DOI: 10.1186/s12885-015-1397-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 04/29/2015] [Indexed: 11/17/2022] Open
Abstract
Background Primary bone cancers are among the deadliest cancer types in adolescents, with osteosarcomas being the most prevalent form. Osteosarcomas are commonly treated with multi-drug neoadjuvant chemotherapy and therapy success as well as patient survival is affected by the presence of tumor suppressors. In order to assess the prognostic value of tumor-suppressive biomarkers, primary osteosarcoma tissues were analyzed prior to and after neoadjuvant chemotherapy. Methods We constructed a tissue microarray from high grade osteosarcoma samples, consisting of 48 chemotherapy naïve biopsies (BXs) and 47 tumor resections (RXs) after neoadjuvant chemotherapy. We performed immunohistochemical stainings of P53, P16, maspin, PTEN, BMI1 and Ki67, characterized the subcellular localization and related staining outcome with chemotherapy response and overall survival. Binary logistic regression analysis was used to analyze chemotherapy response and Kaplan-Meier-analysis as well as the Cox proportional hazards model was applied for analysis of patient survival. Results No significant associations between biomarker expression in BXs and patient survival or chemotherapy response were detected. In univariate analysis, positive immunohistochemistry of P53 (P = 0.008) and P16 (P16; P = 0.033) in RXs was significantly associated with poor survival prognosis. In addition, presence of P16 in RXs was associated with poor survival in multivariate regression analysis (P = 0.003; HR = 0.067) while absence of P16 was associated with good chemotherapy response (P = 0.004; OR = 74.076). Presence of PTEN on tumor RXs was significantly associated with an improved survival prognosis (P = 0.022). Conclusions Positive immunohistochemistry (IHC) of P16 and P53 in RXs was indicative for poor overall patient survival whereas positive IHC of PTEN was prognostic for good overall patient survival. In addition, we found that P16 might be a marker of osteosarcoma chemotherapy resistance. Therefore, our study supports the use of tumor RXs to assess the prognostic value of biomarkers. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1397-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bernhard Robl
- Laboratory for Orthopedic Research, Department of Orthopedics, Balgrist University Hospital, Forchstrasse 340, 8008, Zurich, Switzerland.
| | - Chantal Pauli
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland.
| | - Sander Martijn Botter
- Laboratory for Orthopedic Research, Department of Orthopedics, Balgrist University Hospital, Forchstrasse 340, 8008, Zurich, Switzerland.
| | | | - Bruno Fuchs
- Laboratory for Orthopedic Research, Department of Orthopedics, Balgrist University Hospital, Forchstrasse 340, 8008, Zurich, Switzerland.
| |
Collapse
|
108
|
A shorter telomere is the key factor in preventing cultured human mesenchymal stem cells from senescence escape. Histochem Cell Biol 2015; 142:257-67. [PMID: 24658836 DOI: 10.1007/s00418-014-1210-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2014] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells (MSCs) from various animals undergo spontaneous transformation in vitro,establishing some malignant characteristics. However,this phenomenon seems seldom appearing in human (h)MSCs. To address the question whether the hMSCs really do not undergo the spontaneous transformation and why,the present study compared MSCs from two species under the same conditions, the commercialized primary hMSCs whose in vitro life span is very uniform, and the rat (r)MSCs whose spontaneous transformation in vitro is well defined.It was demonstrated that in rMSCs, there were small numbers of re-proliferating cells appearing after a substantial senescent period. These “senescence-escaped”rMSCs were highly proliferative and did not show any sign of growth arrest during the following subcultures upto observed passage 32. Whereas after entering senescence, hMSCs no longer re-proliferated and finally died from apoptosis. Compared with rMSCs, the hMSCs possessed a much shorter telomere, and lacked both telomerase reverse transcriptase expression and telomerase activity. When proliferating from pre-senescent to senescent stages,the hMSCs had a greater loss of relative telomere length(51 % in hMSC vs. 15 % in rMSC), but both cells displayed a similar average telomere shortening per population doubling (0.50 ± 0.06 kb in rMSC vs. 0.49 ± 0.06 kbin hMSC; p > 0.05), indicating that the greater relative shortening of the hMSC telomeres was due to their original shorter length, rather than lack of telomere maintenance mechanisms. In conclusion, the hMSCs do not spontaneously initiate transformation, because they cannot escape senescence. This is particularly due to their much shorter telomere.
Collapse
|
109
|
NY-ESO-1 (CTAG1B) expression in mesenchymal tumors. Mod Pathol 2015; 28:587-95. [PMID: 25412843 DOI: 10.1038/modpathol.2014.155] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 10/09/2014] [Accepted: 10/15/2014] [Indexed: 12/29/2022]
Abstract
New York esophageal squamous cell carcinoma 1 (NY-ESO-1, CTAG1B) is a cancer-testis antigen and currently a focus of several targeted immunotherapeutic strategies. We performed a large-scale immunohistochemical expression study of NY-ESO-1 using tissue microarrays of mesenchymal tumors from three institutions in an international collaboration. A total of 1132 intermediate and malignant and 175 benign mesenchymal lesions were enrolled in this study. Immunohistochemical staining was performed on tissue microarrays using a monoclonal antibody for NY-ESO-1. Among mesenchymal tumors, myxoid liposarcomas showed the highest positivity for NY-ESO-1 (88%), followed by synovial sarcomas (49%), myxofibrosarcomas (35%), and conventional chondrosarcomas (28%). Positivity of NY-ESO-1 in the remaining mesenchymal tumors was consistently low, and no immunoreactivity was observed in benign mesenchymal lesions. On the basis of these findings, nearly 90% of myxoid liposarcomas, as well as a significant proportion of synovial sarcomas, myxofibrosarcomas, and conventional chondrosarcomas are good candidates for immunotherapy targeting NY-ESO-1.
Collapse
|
110
|
Baranski Z, Booij TH, Cleton-Jansen AM, Price LS, van de Water B, Bovée JVMG, Hogendoorn PCW, Danen EHJ. Aven-mediated checkpoint kinase control regulates proliferation and resistance to chemotherapy in conventional osteosarcoma. J Pathol 2015; 236:348-59. [PMID: 25757065 DOI: 10.1002/path.4528] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 02/23/2015] [Accepted: 03/04/2015] [Indexed: 12/25/2022]
Abstract
Conventional high-grade osteosarcoma is the most common primary bone sarcoma, with relatively high incidence in young people. In this study we found that expression of Aven correlates inversely with metastasis-free survival in osteosarcoma patients and is increased in metastases compared to primary tumours. Aven is an adaptor protein that has been implicated in anti-apoptotic signalling and serves as an oncoprotein in acute lymphoblastic leukaemia. In osteosarcoma cells, silencing Aven triggered G2 cell-cycle arrest; Chk1 protein levels were attenuated and ATR-Chk1 DNA damage response signalling in response to chemotherapy was abolished in Aven-depleted osteosarcoma cells, while ATM, Chk2 and p53 activation remained intact. Osteosarcoma is notoriously difficult to treat with standard chemotherapy, and we examined whether pharmacological inhibition of the Aven-controlled ATR-Chk1 response could sensitize osteosarcoma cells to genotoxic compounds. Indeed, pharmacological inhibitors targeting Chk1/Chk2 or those selective for Chk1 synergized with standard chemotherapy in 2D cultures. Likewise, in 3D extracellular matrix-embedded cultures, Chk1 inhibition led to effective sensitization to chemotherapy. Together, these findings implicate Aven in ATR-Chk1 signalling and point towards Chk1 inhibition as a strategy to sensitize human osteosarcomas to chemotherapy.
Collapse
Affiliation(s)
- Zuzanna Baranski
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands
| | - Tijmen H Booij
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands
| | | | - Leo S Price
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands.,OcellO B.V. Leiden, The Netherlands
| | - Bob van de Water
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands
| | - Judith V M G Bovée
- Department of Pathology, Leiden University Medical Centre (LUMC), The Netherlands
| | | | - Erik H J Danen
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands
| |
Collapse
|
111
|
Liu F, Zhou D, Zhang Q, Wang B. Comments on the article of Bu, et al entitled "P16INK4a overexpression and survival in osteosarcoma patients: a meta analysis". INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:3384-3385. [PMID: 26045875 PMCID: PMC4440184 DOI: pmid/26045875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 01/28/2015] [Indexed: 02/08/2023]
Affiliation(s)
- Fanxiao Liu
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong University324 Jingwu Weiqi Road, Jinan 250021, P.R. China
| | - Dongsheng Zhou
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong University324# Jingwu Weiqi Road, Jinan 250021, P.R. China
| | - Qingyu Zhang
- Department of Orthopedics, Qilu Hospital Affiliated to Shandong UniversityJinan 250012, P.R. China
| | - Bomin Wang
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong University324 Jingwu Weiqi Road, Jinan 250021, P.R. China
| |
Collapse
|
112
|
Silva G, Aboussekhra A. p16(INK4A) inhibits the pro-metastatic potentials of osteosarcoma cells through targeting the ERK pathway and TGF-β1. Mol Carcinog 2015; 55:525-36. [PMID: 25728247 DOI: 10.1002/mc.22299] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 01/06/2015] [Accepted: 01/14/2015] [Indexed: 12/15/2022]
Abstract
Extracellular signal-regulated kinase (ERK) is a downstream component of the evolutionarily conserved mitogen-activated protein kinase-signaling pathway, which controls the expression of a plethora of genes implicated in various physiological processes. This pathway is often hyper-activated by mutations or abnormal extracellular signaling in different types of human cancer, including the most common primary malignant bone tumor osteosarcomas. p16(INK4A) is an important tumor suppressor gene frequently lost in osteosarcomas, and is associated with the progression of these malignancies. We have shown, here, that the ERK1/2 protein kinase is also activated by p16(INK4A) down-regulation in osteosarcoma cells and normal human as well as mouse cells. This inhibitory effect is associated with the suppression of the upstream kinase MEK1/2, and is mediated via the repression of miR-21-5p and the consequent up-regulation of the MEK/ERK antagonist SPRY2 in osteosarcoma cells. Furthermore, we have shown that p16(INK4) inhibits the migration/invasion abilities of these cells through miR-21-5p-dependent inhibition of ERK1/2. In addition, we present clear evidence that p16(INK4) represses the paracrine pro-migratory effect of osteosarcoma cells on stromal fibroblasts through the inhibition of the TGF-β1 expression/secretion. This effect is also ERK1/2-dependent, indicating that in addition to their cell-autonomous actions, p16(INK4) and ERK1/2 have also non-cell-autonomous cancer-related functions. Together, these results indicate that the tumor suppressor p16(INK4) protein represses the carcinogenic process of osteosarcoma cells not only as a cell cycle regulator, but also as a negative regulator of pro-carcinogenic/-metastatic pathways. This indicates that targeting the ERK pathway is of utmost therapeutic value.
Collapse
Affiliation(s)
- Gabriela Silva
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, KSA
| | - Abdelilah Aboussekhra
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, KSA
| |
Collapse
|
113
|
Alteration of histone acetylation pattern during long-term serum-free culture conditions of human fetal placental mesenchymal stem cells. PLoS One 2015; 10:e0117068. [PMID: 25671548 PMCID: PMC4324636 DOI: 10.1371/journal.pone.0117068] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 12/18/2014] [Indexed: 01/01/2023] Open
Abstract
Increasing evidence suggests that the mesenchymal stem cells (MSCs) derived from placenta of fetal origin (fPMSCs) are superior to MSCs of other sources for cell therapy. Since the initial number of isolated MSCs is limited, in vitro propagation is often required to reach sufficient numbers of cells for therapeutic applications, during which MSCs may undergo genetic and/or epigenetic alterations that subsequently increase the probability of spontaneous malignant transformation. Thus, factors that influence genomic and epigenetic stability of MSCs following long-term expansions need to be clarified before cultured MSCs are employed for clinical settings. To date, the genetic and epigenetic stability of fPMSCs after long-term in vitro expansion has not been fully investigated. In this report, alterations to histone acetylation and consequence on the expression pattern of fPMSCs following in vitro propagation under serum-free conditions were explored. The results show that fPMSCs maintain their MSC characteristics before they reached a senescent state. Furthermore, acetylation modification patterns were changed in fPMSCs along with gradually increased global histone deacetylase (HDAC) activity and expression of HDAC subtypes HDAC4, HDAC5 and HDAC6, as well as a down-regulated global histone H3/H4 acetylation during in vitro culturing. In line with the acetylation alterations, the expression of oncogenes Oct4, Sox2 and TERT were significantly decreased over the propagation period. Of note, the down-regulation of Oct4 was strongly associated with changes in acetylation. Intriguingly, telomere length in fPMSCs did not significantly change during the propagating process. These findings suggest that human fPMSCs may be a safe and reliable resource of MSCs and can be propagated under serum-free conditions with less risk of spontaneous malignancy, and warrants further validation in clinical settings.
Collapse
|
114
|
Zhu Y, Song X, Wang J, Li Y, Yang Y, Yang T, Ma H, Wang L, Zhang G, Cho WC, Liu X, Wei J. Placental mesenchymal stem cells of fetal origin deposit epigenetic alterations during long-term culture under serum-free condition. Expert Opin Biol Ther 2015; 15:163-180. [PMID: 25231124 DOI: 10.1517/14712598.2015.960837] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Fetal placental mesenchymal stem cells (fPMSCs) have shown promising cell therapy potentials. However, their genetic and epigenetic stability during in vitro propagation has not been well studied. We thus interrogated the methylation alterations and tumorigenicity of fPMSCs after in vitro expansion using serum-free medium. RESEARCH DESIGN AND METHODS The properties of fPMSCs cultured in a serum-free medium at passage 3 and passage 8 were ascertained by determining their MSC markers, proliferative capacity, chromosomal stability, activity of global DNA methyltransferases and methylation profile. Their potential of malignant transformation was also evaluated in a severe combined immunodeficiency (SCID) murine model. RESULTS The fPMSCs could maintain their MSC characteristics but quickly reached a senescent state of proliferation during in vitro expansion. 246 genes with differential DNA methylation of promoters were identified, along with a significantly downregulated global DNA methyltransferase activity. The genes associated with aging and tumorigenesis had a significantly demethylated tendency over in vitro propagation. However, the deposition of epigenetic alterations did not translate into malignant transformation in SCID mice. CONCLUSION The fPMSCs cultured in serum-free medium have a tendency to deposit methylation modifications over in vitro expansion, therefore the detection of genetic and/or epigenetic alterations is necessary for fPMSCs before they are employed for clinical uses.
Collapse
Affiliation(s)
- Yongzhao Zhu
- Ningxia Medical University, Human Stem Cell Institute of General Hospital , Yinchuan , China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Savage P. Clinical observations on chemotherapy curable malignancies: unique genetic events, frozen development and enduring apoptotic potential. BMC Cancer 2015; 15:11. [PMID: 25605631 PMCID: PMC4308945 DOI: 10.1186/s12885-015-1006-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 12/31/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND A select number of relatively rare metastatic malignancies comprising trophoblast tumours, the rare childhood cancers, germ cells tumours, leukemias and lymphomas have been routinely curable with chemotherapy for more than 30 years. However for the more common metastatic malignancies chemotherapy treatment frequently brings clinical benefits but cure is not expected. Clinically this clear divide in outcome between the tumour types can appear at odds with the classical theories of chemotherapy sensitivity and resistance that include rates of proliferation, genetic development of drug resistance and drug efflux pumps. We have looked at the clinical characteristics of the chemotherapy curable malignancies to see if they have any common factors that could explain this extreme differential sensitivity to chemotherapy. DISCUSSION It has previously been noted how the onset of malignancy can leave malignant cells fixed with some key cellular functions remaining frozen at the point in development at which malignant transformation occurred. In the chemotherapy curable malignancies the onset of malignancy is in each case closely linked to one of the unique genetic events of; nuclear fusion for molar pregnancies, choriocarcinoma and placental site trophoblast tumours, gastrulation for the childhood cancers, meiosis for testicular cancer and ovarian germ cell tumours and VDJ rearrangement and somatic hypermutation for acute leukemia and lymphoma. These processes are all linked to natural periods of supra-physiological apoptotic potential and it appears that the malignant cells arising from them usually retain this heightened sensitivity to DNA damage. To investigate this hypothesis we have examined the natural history of the healthy cells during these processes and the chemotherapy sensitivity of malignancies arising before, during and after the events. To add to the debate on chemotherapy resistance and sensitivity, we would argue that malignancies can be functionally divided into 2 groups. Firstly those that arise in cells with naturally heightened apoptotic potential as a result of their proximity to the unique genetic events, where the malignancies are generally chemotherapy curable and then the more common malignancies that arise in cells of standard apoptotic potential that are not curable with classical cytotoxic drugs.
Collapse
Affiliation(s)
- Philip Savage
- BCCA Vancouver Island, 2410 Lee Avenue, Victoria, BC, V8R 6V5, Canada.
| |
Collapse
|
116
|
Sampson VB, Yoo S, Kumar A, Vetter NS, Kolb EA. MicroRNAs and Potential Targets in Osteosarcoma: Review. Front Pediatr 2015; 3:69. [PMID: 26380245 PMCID: PMC4547013 DOI: 10.3389/fped.2015.00069] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 07/20/2015] [Indexed: 12/13/2022] Open
Abstract
Osteosarcoma is the most common bone cancer in children and young adults. Surgery and multi-agent chemotherapy are the standard treatment regimens for this disease. New therapies are being investigated to improve overall survival in patients. Molecular targets that actively modulate cell processes, such as cell-cycle control, cell proliferation, metabolism, and apoptosis, have been studied, but it remains a challenge to develop novel, effective-targeted therapies to treat this heterogeneous and complex disease. MicroRNAs (miRNAs) are small non-coding RNAs that play critical roles in regulating cell processes including growth, development, and disease. miRNAs function as oncogenes or tumor suppressors to regulate gene and protein expression. Several studies have demonstrated the involvement of miRNAs in the pathogenesis of osteosarcoma with the potential for development in disease diagnostics and therapeutics. In this review, we discuss the current knowledge on the role of miRNAs and their target genes and evaluate their potential use as therapeutic agents in osteosarcoma. We also summarize the efficacy of inhibition of oncogenic miRNAs or expression of tumor suppressor miRNAs in preclinical models of osteosarcoma. Recent progress on systemic delivery as well as current applications for miRNAs as therapeutic agents has seen the advancement of miR-34a in clinical trials for adult patients with non-resectable primary liver cancer or metastatic cancer with liver involvement. We suggest a global approach to the understanding of the pathogenesis of osteosarcoma may identify candidate miRNAs as promising biomarkers for this rare disease.
Collapse
Affiliation(s)
- Valerie B Sampson
- Nemours Center for Cancer and Blood Disorders, Alfred I. duPont Hospital for Children , Wilmington, DE , USA
| | - Soonmoon Yoo
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children , Wilmington, DE , USA
| | - Asmita Kumar
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children , Wilmington, DE , USA
| | - Nancy S Vetter
- Nemours Center for Cancer and Blood Disorders, Alfred I. duPont Hospital for Children , Wilmington, DE , USA
| | - E Anders Kolb
- Nemours Center for Cancer and Blood Disorders, Alfred I. duPont Hospital for Children , Wilmington, DE , USA
| |
Collapse
|
117
|
Riminucci M, Remoli C, Robey PG, Bianco P. Stem cells and bone diseases: new tools, new perspective. Bone 2015; 70:55-61. [PMID: 25240458 PMCID: PMC5524373 DOI: 10.1016/j.bone.2014.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 09/03/2014] [Accepted: 09/09/2014] [Indexed: 12/13/2022]
Abstract
Postnatal skeletal stem cells are a unique class of progenitors with biological properties that extend well beyond the limits of stemness as commonly defined. Skeletal stem cells sustain skeletal tissue homeostasis, organize and maintain the complex architectural structure of the bone marrow microenvironment and provide a niche for hematopoietic progenitor cells. The identification of stem cells in the human post-natal skeleton has profoundly changed our approach to the physiology and pathology of this system. Skeletal diseases have been long interpreted essentially in terms of defective function of differentiated cells and/or abnormal turnover of the matrix that they produce. The notion of a skeletal stem cell has brought forth multiple, novel concepts in skeletal biology that provide potential alternative concepts. At the same time, the recognition of the complex functions played by skeletal progenitors, such as the structural and functional organization of the bone marrow, has provided an innovative, unifying perspective for understanding bone and bone marrow changes simultaneously occurring in many disorders. Finally, the possibility to isolate and highly enrich for skeletal progenitors, enables us to reproduce perfectly normal or pathological organ miniatures. These, in turn, provide suitable models to investigate and manipulate the pathogenetic mechanisms of many genetic and non-genetic skeletal diseases. This article is part of a Special Issue entitled Stem cells and Bone.
Collapse
Affiliation(s)
- Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, Italy.
| | - Cristina Remoli
- Department of Molecular Medicine, Sapienza University of Rome, Italy
| | - Pamela G Robey
- Craniofacial and Skeletal Diseases Branch, National Institute of Craniofacial and Dental Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Paolo Bianco
- Department of Molecular Medicine, Sapienza University of Rome, Italy
| |
Collapse
|
118
|
Cai R, Kawazoe N, Chen G. Influence of surfaces modified with biomimetic extracellular matrices on adhesion and proliferation of mesenchymal stem cells and osteosarcoma cells. Colloids Surf B Biointerfaces 2014; 126:381-6. [PMID: 25516267 DOI: 10.1016/j.colsurfb.2014.11.050] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 11/27/2014] [Accepted: 11/30/2014] [Indexed: 12/21/2022]
Abstract
Preparation of surfaces modified with biomimetic extracellular matrices (ECMs) is important for investigation of the interaction between ECMs and cells. In the present study, surfaces modified with ECMs from normal somatic cells, stem cells and tumor cells were prepared by cell culture method. The ECMs derived from bone marrow-derived mesenchymal stem cells (MSCs), dermal fibroblasts (FBs), osteoblasts (OBs) and MG63 osteosarcoma cells were deposited on the surfaces of cell-culture polystyrene plates (TCPS). The ECMs from different cell types had different compositions. The effects of the ECM-deposited surfaces on the adhesion, spreading and proliferation of MSCs and MG63 human osteosarcoma cells were dependent on the type of both ECMs and cells. The surfaces deposited with ECMs from MSCs, FBs and OBs promoted cell adhesion more strongly than surfaces deposited with ECMs from MG63 cells and TCPS. Compared to TCPS, the ECM-deposited surfaces promoted proliferation of MSCs while they inhibited the proliferation of MG63 cells.
Collapse
Affiliation(s)
- Rong Cai
- Tissue Regeneration Materials Unit, International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Naoki Kawazoe
- Tissue Regeneration Materials Unit, International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Guoping Chen
- Tissue Regeneration Materials Unit, International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan.
| |
Collapse
|
119
|
Yang J, Ren Z, Du X, Hao M, Zhou W. The role of mesenchymal stem/progenitor cells in sarcoma: update and dispute. Stem Cell Investig 2014; 1:18. [PMID: 27358864 DOI: 10.3978/j.issn.2306-9759.2014.10.01] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 10/10/2014] [Indexed: 12/26/2022]
Abstract
Sarcoma is the collective name for a relatively rare, yet heterogeneous group of cancers, most probably derived from mesenchymal tissues. There are currently over 50 sarcoma subtypes described underscoring the clinical and biologic diversity of this group of malignant cancers. This wide lineage range might suggest that sarcomas originate from either many committed different cell types or from a multipotent cell. Mesenchymal stem/progenitor cells (MSCs) are able to differentiate into many cell types and these multipotent cells have been isolated from several adult human tumors, making them available for research as well as potential beneficial therapeutical agents. Recent accomplishments in the field have broadened our knowledge of MSCs in relation to sarcoma origin and sarcoma treatment in therapeutic settings. However, numerous concerns and disputes have been raised about whether they are the putative originating cells of sarcoma and their questionable role in sarcomagenesis and progression. We summarize the update and dispute about MSC investigations in sarcomas including the definition, cell origin hypothesis, functional and descriptive assays, roles in sarcomagenesis and targeted therapy, with the purpose to give a comprehensive view of the role of MSCs in sarcomas.
Collapse
Affiliation(s)
- Jilong Yang
- 1 Departments of Bone and Soft Tissue Tumor, 2 National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China ; 3 Departments of Diagnostics, Tianjin Medical University, Tianjin 30060, China
| | - Zhiwu Ren
- 1 Departments of Bone and Soft Tissue Tumor, 2 National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China ; 3 Departments of Diagnostics, Tianjin Medical University, Tianjin 30060, China
| | - Xiaoling Du
- 1 Departments of Bone and Soft Tissue Tumor, 2 National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China ; 3 Departments of Diagnostics, Tianjin Medical University, Tianjin 30060, China
| | - Mengze Hao
- 1 Departments of Bone and Soft Tissue Tumor, 2 National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China ; 3 Departments of Diagnostics, Tianjin Medical University, Tianjin 30060, China
| | - Wenya Zhou
- 1 Departments of Bone and Soft Tissue Tumor, 2 National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China ; 3 Departments of Diagnostics, Tianjin Medical University, Tianjin 30060, China
| |
Collapse
|
120
|
Sanchez-Diaz PC, Hsiao TH, Zou Y, Sugalski AJ, Heim-Hall J, Chen Y, Langevin AM, Hung JY. In silico functional analyses and discovery of survival-associated microRNA signatures in pediatric osteosarcoma. Oncoscience 2014; 1:599-608. [PMID: 25594070 PMCID: PMC4278335 DOI: 10.18632/oncoscience.85] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 09/17/2014] [Indexed: 11/25/2022] Open
Abstract
Purpose Osteosarcoma is the most common bone tumor in children, adolescents, and young adults. In contrast to other childhood malignancies, no biomarkers have been consistently identified as predictors of outcome. This study was conducted to assess the microRNAs(miRs) expression signatures in pre-treatment osteosarcoma specimens and correlate with outcome to identify biomarkers for disease relapse. Results A 42-miRs signature whose expression levels were associated with overall and relapse-free survival waas identified. There were 8 common miRs between the two sets of survival-associated miRs. Bioinformatic analyses of these survival-associated miRs suggested that they might regulate genes involved in ubiquitin proteasome system, TGFb, IGF, PTEN/AKT/mTOR, MAPK, PDGFR/RAF/MEK/ERK, and ErbB/HER pathways. Methods The cohort consisted of 27 patients of 70% Mexican-American ethnicity. High-throughput RT-qPCR approach was used to generate quantitative expression of 754 miRs in the human genome. We examined tumor recurrence status, survival time and their association with miR expression levels by Cox proportional hazard regression analysis. TargetScan was used to predict miR/genes interactions, and functional analyses using KEGG, BioCarta, Gene Ontology were applied to these potential targets to predict deregulated pathways. Conclusions Our findings suggested that these miRs might be potentially useful as prognostic biomarkers and therapeutic targets in pediatric osteosarcoma.
Collapse
Affiliation(s)
- Patricia C Sanchez-Diaz
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA ; Current address: Rosenberg School of Optometry, University of the Incarnate Word, San Antonio, Texas, USA
| | - Tzu-Hung Hsiao
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Yi Zou
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Aaron J Sugalski
- Division of Hematology and Oncology, Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Josefine Heim-Hall
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA ; Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Yidong Chen
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA ; Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA ; Department of Epidemiology and Biostatistics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Anne-Marie Langevin
- Division of Hematology and Oncology, Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA ; Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Jaclyn Y Hung
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA ; Division of Hematology and Oncology, Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA ; Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
121
|
Zhou R, Li Z, He C, Li R, Xia H, Li C, Xiao J, Chen ZY. Human umbilical cord mesenchymal stem cells and derived hepatocyte-like cells exhibit similar therapeutic effects on an acute liver failure mouse model. PLoS One 2014; 9:e104392. [PMID: 25101638 PMCID: PMC4125182 DOI: 10.1371/journal.pone.0104392] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 07/08/2014] [Indexed: 01/20/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have exhibited therapeutic effects in multiple animal models so that are promising liver substitute for transplantation treatment of end-stage liver diseases. However, it has been shown that over-manipulation of these cells increased their tumorigenic potential, and that reducing the in vitro culture time could minimize the risk. In this study, we used a D-galactosamine plus lipopolysaccharide (Gal/LPS)-induced acute liver failure mouse model, which caused death of about 50% of the mice with necrosis of more than 50% hepatocytes, to compare the therapeutic effects of human umbilical cord MSCs (hUCMSCs) before and after induction of differentiation into hepatocyte (i-Heps). Induction of hUCMSCs to become i-Heps was achieved by treatment of the cells with a group of growth factors within 4 weeks. The resulted i-Heps exhibited a panel of human hepatocyte biomarkers including cytokeratin (hCK-18), α-fetoprotein (hAFP), albumin (hALB), and hepatocyte-specific functions glycogen storage and urea metabolism. We demonstrated that transplantation of both cell types through tail vein injection rescued almost all of the Gal/LPS-intoxicated mice. Although both cell types exhibited similar ability in homing at the mouse livers, the populations of the hUCMSCs-derived cells, as judged by expressing hAFP, hCK-18 and human hepatocyte growth factor (hHGF), were small. These observations let us to conclude that the hUCMSCs was as effective as the i-Heps in treatment of the mouse acute liver failure, and that the therapeutic effects of hUCMSCs were mediated largely via stimulation of host hepatocyte regeneration, and that delivery of the cells through intravenous injection was effective.
Collapse
Affiliation(s)
- Ruiping Zhou
- Laboratory for Gene and Cell Therapy, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Department of Stomatology, Shenzhen Seventh (Yantian District) People’s Hospital, Shenzhen, China
| | - Zhuokun Li
- Laboratory for Gene and Cell Therapy, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chengyi He
- Laboratory for Gene and Cell Therapy, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ronglin Li
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Hongbin Xia
- Laboratory for Gene and Cell Therapy, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chunyang Li
- Department of Stomatology, The 5 Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Jia Xiao
- Department of Immunobiology, Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
- Department of Anatomy, The University of Hong Kong, Hong Kong, China
- * E-mail: (JX); (ZYC)
| | - Zhi-Ying Chen
- Laboratory for Gene and Cell Therapy, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- * E-mail: (JX); (ZYC)
| |
Collapse
|
122
|
Dai F, Yang S, Zhang F, Shi D, Zhang Z, Wu J, Xu J. hTERT- and hCTLA4Ig-expressing human bone marrow-derived mesenchymal stem cells: in vitro and in vivo characterization and osteogenic differentiation. J Tissue Eng Regen Med 2014; 11:400-411. [PMID: 25047146 DOI: 10.1002/term.1924] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 02/21/2014] [Accepted: 04/25/2014] [Indexed: 01/01/2023]
Abstract
Multipotent mesenchymal stem cells (MSCs) are commonly used as seed cells in studies of tissue engineering and regenerative medicine but their clinical application is limited, due to insufficient numbers of autogeneic MSCs, immune rejection of allogeneic MSCs and replicative senescence. We constructed two gene expression vectors for transfection of the human telomerase reverse transcriptase (hTERT) and cytotoxic T lymphocyte-associated antigen 4-Ig (CTLA4Ig) genes into human bone marrow-derived stem cells (hBMSCs). Successful transfection of both genes generated hTERT-CTLA4Ig hBMSCs that expressed both telomerase (shown by immunohistochemistry and a TRAPeze assay) and CTLA4Ig (demonstrated by immunocytochemistry and western blotting) without apparent mutual interference. Both hTERT BMSCs (92 population doublings) and hTERT-CTLA4Ig hBMSCs (60 population doublings) had an extended lifespan compared with hBMSCs (18 population doublings). Cell cycle analysis revealed that, compared with hBMSCs, a lower proportion of hTERT hBMSCs were in G0 /G1 phase but a higher proportion were in S phase; compared with hTERT hBMSCs, a higher proportion of hTERT-CTLA4Ig hBMSCs were in G0 /G1 phase, while a lower proportion were in S and G2 /M phases. hTERT-CTLA4Ig hBMSCs retained their capacity for osteogenic differentiation in vitro, shown by the detection of hydroxyapatite mineral deposition (labelled tetracycline fluorescence staining), calcareous nodules (alizarin red S staining), alkaline phosphatase (calcium-cobalt method) and osteocalcin (immunocytochemistry). Furthermore, subcutaneous transplantation of hTERT-CTLA4Ig hBMSCs in a rat xenotransplantation model resulted in the successful generation of bone-like tissue, confirmed using radiography and histological assessment. We propose that allogeneic hTERT-CTLA4Ig hBMSCs may be ideal seed cells for bone tissue engineering. Copyright © 2014 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Fei Dai
- National and Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Sisi Yang
- Institute of Burns Research, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Fei Zhang
- National and Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Dongwen Shi
- Institute of Burns Research, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Zehua Zhang
- National and Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Jun Wu
- Institute of Burns Research, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Jianzhong Xu
- National and Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| |
Collapse
|
123
|
Richardson RB. Age-specific bone tumour incidence rates are governed by stem cell exhaustion influencing the supply and demand of progenitor cells. Mech Ageing Dev 2014; 139:31-40. [DOI: 10.1016/j.mad.2014.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 05/29/2014] [Accepted: 06/01/2014] [Indexed: 12/21/2022]
|
124
|
Vrtačnik P, Kos Š, Bustin SA, Marc J, Ostanek B. Influence of trypsinization and alternative procedures for cell preparation before RNA extraction on RNA integrity. Anal Biochem 2014; 463:38-44. [PMID: 24983903 DOI: 10.1016/j.ab.2014.06.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 06/18/2014] [Accepted: 06/19/2014] [Indexed: 10/25/2022]
Abstract
The accuracy of techniques such as microarrays, reverse transcription polymerase chain reaction, and whole transcriptome shotgun sequencing is critically dependent on RNA quality. We have repeatedly observed extensive RNA degradation following trypsinization, a routine procedure used to dissociate adherent tissue culture cells prior to RNA extraction. This study investigated the cause of this degradation and identifies an alternative procedure that enables extraction of intact high-quality RNA. Trypsinization and several alternative procedures were used to dissociate a range of different cell lines prior to RNA extraction. The contribution of exogenous ribonucleases or induction of endogenous ribonucleases by trypsin reagent proteases to RNA degradation was examined. Trypsinization resulted in a complete degradation of RNA regardless of cell line type, differentiation stage, or passage number. This occurred when intact RNA was incubated directly with trypsin and was not suppressed by inhibiting trypsin's protease activity. Prevention of degradation by sodium hypochlorite treatment of trypsin reagent identified the presence of ribonucleases in trypsin derived from animal pancreas. Consistent extraction of high-quality RNA requires the use of direct cell lysis with a phenol guanidine-based reagent or an animal origin-free protease-based dissociation agent if enzymatic detachment prior to RNA extraction cannot be avoided.
Collapse
Affiliation(s)
- Peter Vrtačnik
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Špela Kos
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Stephen A Bustin
- Postgraduate Medical Institute, Anglia Ruskin University, Chelmsford CM1 1SQ, UK
| | - Janja Marc
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Barbara Ostanek
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana 1000, Slovenia.
| |
Collapse
|
125
|
Tian J, Li X, Si M, Liu T, Li J. CD271+ osteosarcoma cells display stem-like properties. PLoS One 2014; 9:e98549. [PMID: 24893164 PMCID: PMC4043643 DOI: 10.1371/journal.pone.0098549] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 05/05/2014] [Indexed: 11/19/2022] Open
Abstract
Cancer stem cell (CSC) theory has been proposed and verified in many cancers. The existence of osteosarcoma CSCs has been confirmed for many years and multiple surface markers have been employed to identify them. In this study, we identified CD271+ subpopulation of osteosarcoma displaying stem-like properties. CD271, known as the neural crest nerve growth factor receptor, is the marker of bone marrow mesenchymal stem cells (MSCs) and human melanoma-initiating cells. We discovered that CD271 was expressed differentially in diverse types of human osteosarcoma and stabilized cell lines. CD271+ osteosarcoma cells displayed most of the properties of CSC, such as self-renewal, differentiation, drug resistance and tumorigenicity in vivo. Nanog, Oct3/4, STAT3, DNA-PKcs, Bcl-2 and ABCG2 were more expressed in CD271+ cells compared with CD271− cells. Our study supported the osteosarcoma CSC hypothesis and, to a certain extent, revealed one of the possible mechanisms involved in maintaining CSCs properties.
Collapse
Affiliation(s)
- Jiguang Tian
- Department of Orthopedics, Qilu Hospital, Shandong University, Shandong, China
| | - Xin Li
- Department of Orthopedics, Qilu Hospital, Shandong University, Shandong, China
| | - Meng Si
- Department of Orthopedics, Qilu Hospital, Shandong University, Shandong, China
| | - Ting Liu
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Shandong, China
| | - Jianmin Li
- Department of Orthopedics, Qilu Hospital, Shandong University, Shandong, China
- * E-mail:
| |
Collapse
|
126
|
Wang L, Zhao Y, Liu Y, Akiyama K, Chen C, Qu C, Jin Y, Shi S. IFN-γ and TNF-α synergistically induce mesenchymal stem cell impairment and tumorigenesis via NFκB signaling. Stem Cells 2014; 31:1383-95. [PMID: 23553791 DOI: 10.1002/stem.1388] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 02/18/2013] [Accepted: 03/04/2013] [Indexed: 12/25/2022]
Abstract
An inflammatory microenvironment may cause organ degenerative diseases and malignant tumors. However, the precise mechanisms of inflammation-induced diseases are not fully understood. Here, we show that the proinflammatory cytokines interferon-γ (IFN-γ) and tumor necrosis factor α (TNF-α) synergistically impair self-renewal and differentiation of mesenchymal stem cells (MSCs) via nuclear factor κB (NFκB)-mediated activation of mothers against decapentaplegic homolog 7 (SMAD7) in ovariectomized (OVX) mice. More interestingly, a long-term elevated levels of IFN-γ and TNF-α result in significantly increased susceptibility to malignant transformation in MSCs through NFκB-mediated upregulation of the oncogenes c-Fos and c-Myc. Depletion of either IFN-γ or TNF-α in OVX mice abolishes MSC impairment and the tendency toward malignant transformation with no NFκB-mediated oncogene activation. Systemic administration of aspirin, which significantly reduces the levels of IFN-γ and TNF-α, results in blockage of MSC deficiency and tumorigenesis by inhibition of NFκB/SMAD7 and NFκB/c-FOS and c-MYC pathways in OVX mice. In summary, this study reveals that inflammation factors, such as IFN-γ and TNF-α, synergistically induce MSC deficiency via NFκB/SMAD7 signaling and tumorigenesis via NFκB-mediated oncogene activation.
Collapse
Affiliation(s)
- Lei Wang
- University of Southern California, Los Angeles, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
127
|
Mutsaers AJ, Walkley CR. Cells of origin in osteosarcoma: mesenchymal stem cells or osteoblast committed cells? Bone 2014; 62:56-63. [PMID: 24530473 DOI: 10.1016/j.bone.2014.02.003] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 01/14/2014] [Accepted: 02/05/2014] [Indexed: 12/14/2022]
Abstract
Osteosarcoma is a disease with many complex genetic abnormalities but few well defined genetic drivers of tumor initiation and evolution. The disease is diagnosed and defined through the observation of malignant osteoblastic cells that produce osteoid, however the exact cell of origin for this cancer remains to be definitively defined. Evidence exists to support a mesenchymal stem cell as well as committed osteoblast precursors as the cell of origin. Increasing numbers of experimental models have begun to shed light on to the likely cell population that gives rise to OS in vivo with the weight of evidence favoring an osteoblastic population as the cell of origin. As more information is gathered regarding osteosarcoma initiating cells and how they may relate to the cell of origin we will derive a better understanding of the development of this disease which may ultimately lead to clinical improvements through more personalized therapeutic approaches.
Collapse
Affiliation(s)
- Anthony J Mutsaers
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Carl R Walkley
- Stem Cell Regulation Unit and ACRF Rational Drug Discovery Centre, St. Vincent's Institute, Fitzroy, Victoria, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia.
| |
Collapse
|
128
|
Honoki K, Tsujiuchi T. Senescence bypass in mesenchymal stem cells: a potential pathogenesis and implications of pro-senescence therapy in sarcomas. Expert Rev Anticancer Ther 2014; 13:983-96. [PMID: 23984899 DOI: 10.1586/14737140.2013.820010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cellular senescence is a mechanism that limits the lifespan of somatic cells as the results of replicative proliferation and response to stresses, and that prevents undesired oncogenic changes constituting a barrier against immortalization and tumorigenesis. Mesenchymal stem cells (MSCs) reside in a variety of tissues, and participates in tissue maintenance with their multipotent differentiation ability. MSCs are also considered to be as cells of origin for certain type of sarcomas. We reviewed the mechanisms of cellular senescence in MSCs and hypothesized senescence bypass as the potential pathogenesis for sarcoma development, and proposed the possibility of senescence induction therapy for an alternative treatment strategy against sarcomas, especially cells with the resistance to conventional chemo and radiotherapy including sarcoma stem cells.
Collapse
Affiliation(s)
- Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan.
| | | |
Collapse
|
129
|
Lamoureux F, Baud’huin M, Rodriguez Calleja L, Jacques C, Berreur M, Rédini F, Lecanda F, Bradner JE, Heymann D, Ory B. Selective inhibition of BET bromodomain epigenetic signalling interferes with the bone-associated tumour vicious cycle. Nat Commun 2014; 5:3511. [DOI: 10.1038/ncomms4511] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 02/25/2014] [Indexed: 12/22/2022] Open
|
130
|
Botter SM, Neri D, Fuchs B. Recent advances in osteosarcoma. Curr Opin Pharmacol 2014; 16:15-23. [PMID: 24632219 DOI: 10.1016/j.coph.2014.02.002] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/06/2014] [Accepted: 02/07/2014] [Indexed: 01/09/2023]
Abstract
Although osteosarcoma (OS) is a rare malignancy, it is ranked among the leading causes of cancer-related death in the pediatric age group. The cancer's low prevalence and its large tumor heterogeneity make it difficult to obtain meaningful progress in patient survival. In this review we present an overview of current clinical trials which largely focus on stimulation of the immune system or rely on the inhibition of kinases such as Src and mTOR. The potential efficacy of tumor-targeted TNFalpha is discussed, as well as the importance of preclinical validation of new targets. To improve the success of future clinical trials, clinicians and basic researchers need to intensify their exchange. Finally, a case is made for individualized treatment of OS patients, based on interdisciplinary cooperation in dedicated Sarcoma Centers.
Collapse
Affiliation(s)
- Sander M Botter
- Sarcoma Center & Laboratory for Orthopedic Research, Department of Orthopedics, Balgrist University Hospital, University of Zurich, Forchstrasse 340, 8008 Zurich, Switzerland
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich Wolfgang-Pauli-Str. 10, 8093 Zurich, Switzerland
| | - Bruno Fuchs
- Sarcoma Center & Laboratory for Orthopedic Research, Department of Orthopedics, Balgrist University Hospital, University of Zurich, Forchstrasse 340, 8008 Zurich, Switzerland.
| |
Collapse
|
131
|
Zhang L, Zhou Q, Zhang N, Li W, Ying M, Ding W, Yang B, He Q. E2F1 impairs all-trans retinoic acid-induced osteogenic differentiation of osteosarcoma via promoting ubiquitination-mediated degradation of RARα. Cell Cycle 2014; 13:1277-87. [PMID: 24608861 DOI: 10.4161/cc.28190] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
All-trans retinoic acid (ATRA) is a widely used differentiation drug that can effectively induce osteogenic differentiation of osteosarcoma cells, but the underlying mechanism remains elusive, which limits the clinical application for ATRA in osteosarcoma patients. In this study, we identified E2F1 as a novel regulator involved in ATRA-induced osteogenic differentiation of osteosarcoma cells. We observed that osteosarcoma cells are coupled with individual differences in the expression levels of E2F1 in patients, and E2F1 impairs ATRA-induced differentiation of osteosarcoma cells. Moreover, remarkable anti-proliferative and differentiation-inducing effects of ATRA treatment are only observed in E2F1 low to negative expressed primary osteosarcoma cultures. These results strongly suggested that E2F1 may serve as a potent indicator for the effectiveness of ATRA treatment in osteosarcoma. Interestingly, E2F1 is found to downregulate retinoic acid receptor α (RARα), a key factor determines the effectiveness of ATRA. E2F1 specifically binds to RARα and promotes its ubiquitination-mediated degradation; as a consequence, RARα-mediated differentiation is inhibited in osteosarcoma. Therefore, our studies present E2F1 as a potent biomarker, as well as a therapeutic target for ATRA-based differentiation therapeutics, and raise the hope of using differentiation-based approaches for osteosarcoma patients.
Collapse
Affiliation(s)
- Lei Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Qian Zhou
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Ning Zhang
- Department of Orthopedics; The Second Affiliated Hospital of Zhejiang University; Zhejiang University; Hangzhou, China
| | - Weixu Li
- Department of Orthopedics; The Second Affiliated Hospital of Zhejiang University; Zhejiang University; Hangzhou, China
| | - Meidan Ying
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Wanjing Ding
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| |
Collapse
|
132
|
Amaral AT, Manara MC, Berghuis D, Ordóñez JL, Biscuola M, Lopez-García MA, Osuna D, Lucarelli E, Alviano F, Lankester A, Scotlandi K, de Álava E. Characterization of human mesenchymal stem cells from ewing sarcoma patients. Pathogenetic implications. PLoS One 2014; 9:e85814. [PMID: 24498265 PMCID: PMC3911896 DOI: 10.1371/journal.pone.0085814] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 12/02/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Ewing Sarcoma (EWS) is a mesenchymal-derived tumor that generally arises in bone and soft tissue. Intensive research regarding the pathogenesis of EWS has been insufficient to pinpoint the early events of Ewing sarcomagenesis. However, the Mesenchymal Stem Cell (MSC) is currently accepted as the most probable cell of origin. MATERIALS AND METHODS In an initial study regarding a deep characterization of MSC obtained specifically from EWS patients (MSC-P), we compared them with MSC derived from healthy donors (MSC-HD) and EWS cell lines. We evaluated the presence of the EWS-FLI1 gene fusion and EWSR1 gene rearrangements in MSC-P. The presence of the EWS transcript was confirmed by q-RT-PCR. In order to determine early events possibly involved in malignant transformation, we used a multiparameter quantitative strategy that included both MSC immunophenotypic negative/positive markers, and EWS intrinsic phenotypical features. Markers CD105, CD90, CD34 and CD45 were confirmed in EWS samples. RESULTS We determined that MSC-P lack the most prevalent gene fusion, EWSR1-FLI1 as well as EWSR1 gene rearrangements. Our study also revealed that MSC-P are more alike to MSC-HD than to EWS cells. Nonetheless, we also observed that EWS cells had a few overlapping features with MSC. As a relevant example, also MSC showed CD99 expression, hallmark of EWS diagnosis. However, we observed that, in contrast to EWS cells, MSC were not sensitive to the inhibition of CD99. CONCLUSIONS In conclusion, our results suggest that MSC from EWS patients behave like MSC-HD and are phenotypically different from EWS cells, thus raising important questions regarding MSC role in sarcomagenesis.
Collapse
MESH Headings
- 12E7 Antigen
- Antigens, CD/metabolism
- Antigens, CD34/metabolism
- Calmodulin-Binding Proteins/genetics
- Cell Adhesion Molecules/metabolism
- Cell Line
- Cells, Cultured
- Endoglin
- Flow Cytometry
- Gene Expression Regulation, Neoplastic
- Gene Rearrangement
- Humans
- In Situ Hybridization, Fluorescence
- Leukocyte Common Antigens/metabolism
- Mesenchymal Stem Cells/metabolism
- Mesenchymal Stem Cells/pathology
- Oncogene Proteins, Fusion/genetics
- Proto-Oncogene Protein c-fli-1/genetics
- RNA-Binding Protein EWS/genetics
- RNA-Binding Proteins/genetics
- Receptors, Cell Surface/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sarcoma, Ewing/genetics
- Sarcoma, Ewing/metabolism
- Sarcoma, Ewing/pathology
- Thy-1 Antigens/metabolism
Collapse
Affiliation(s)
- Ana Teresa Amaral
- Molecular Pathology Program, Institute of Biomedical Research of Salamanca-Centro de Investigación del Cáncer, Centro de Investigación del Cáncer (IBSAL-CIC), Salamanca, Spain
- Instituto de Biomedicina de Sevilla (IBiS), CSIC-Universidad de Sevilla, Department of Pathology and Biobank, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Maria Cristina Manara
- CRS Sviluppo di Terapie Biomolecolari, Oncologia Sperimentale, Istituto Ortopedico Rizzoli (IOR), Bologna, Italy
| | - Dagmar Berghuis
- Department of Pediatrics and Biobank, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - José Luis Ordóñez
- Molecular Pathology Program, Institute of Biomedical Research of Salamanca-Centro de Investigación del Cáncer, Centro de Investigación del Cáncer (IBSAL-CIC), Salamanca, Spain
| | - Michele Biscuola
- Instituto de Biomedicina de Sevilla (IBiS), CSIC-Universidad de Sevilla, Department of Pathology and Biobank, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Maria Angeles Lopez-García
- Instituto de Biomedicina de Sevilla (IBiS), CSIC-Universidad de Sevilla, Department of Pathology and Biobank, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Daniel Osuna
- Molecular Pathology Program, Institute of Biomedical Research of Salamanca-Centro de Investigación del Cáncer, Centro de Investigación del Cáncer (IBSAL-CIC), Salamanca, Spain
| | - Enrico Lucarelli
- Osteoarticolar Regeneration Laboratory, Istituto Ortopedico Rizzoli (IOR), Bologna, Italy
| | - Francesco Alviano
- Dipartimento di Istologia, Embriologia e Biologia, Istituto Ortopedico Rizzoli (IOR), Bologna, Italy
| | - Arjan Lankester
- Department of Pediatrics and Biobank, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Katia Scotlandi
- CRS Sviluppo di Terapie Biomolecolari, Oncologia Sperimentale, Istituto Ortopedico Rizzoli (IOR), Bologna, Italy
| | - Enrique de Álava
- Molecular Pathology Program, Institute of Biomedical Research of Salamanca-Centro de Investigación del Cáncer, Centro de Investigación del Cáncer (IBSAL-CIC), Salamanca, Spain
- Instituto de Biomedicina de Sevilla (IBiS), CSIC-Universidad de Sevilla, Department of Pathology and Biobank, Hospital Universitario Virgen del Rocío, Seville, Spain
| |
Collapse
|
133
|
Guijarro MV. Osteosarcoma: mouse models, cell of origin and cancer stem cell. POSTDOC JOURNAL : A JOURNAL OF POSTDOCTORAL RESEARCH AND POSTDOCTORAL AFFAIRS 2014; 2:19-30. [PMID: 27617267 DOI: 10.14304/surya.jpr.v2n2.3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Osteosarcoma (OS) is the most common non-hematologic primary tumor of bone in children and adults. High-dose cytotoxic chemotherapy and surgical resection have improved prognosis, with long-term survival for non-metastatic disease approaching 70%. However, most OS tumors are high grade and tend to rapidly develop pulmonary metastases. Despite clinical advances, patients with metastatic disease or relapse have a poor prognosis. Here the cell biology of OS is reviewed with a special emphasis on mouse models as well as the roles of the cell of origin and cancer stem cells. A better understanding of the molecular pathogenesis of human OS is essential for the development of improved prognostic and diagnostic markers as well as targeted therapies for both primary and metastatic OS.
Collapse
Affiliation(s)
- Maria V Guijarro
- Gene Therapy Lab. Dept. Orthopaedics and Rehabilitation. University of Florida. 1600 Archer Road, MSB M2-212. Gainesville, FL 32610. USA
| |
Collapse
|
134
|
Safari M, Khoshnevisan A. An overview of the role of cancer stem cells in spine tumors with a special focus on chordoma. World J Stem Cells 2014; 6:53-64. [PMID: 24567788 PMCID: PMC3927014 DOI: 10.4252/wjsc.v6.i1.53] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Revised: 08/31/2013] [Accepted: 11/05/2013] [Indexed: 02/06/2023] Open
Abstract
Primary malignant tumors of the spine are relatively rare, less than 5% of all spinal column tumors. However, these lesions are often among the most difficult to treat and encompass challenging pathologies such as chordoma and a variety of invasive sarcomas. The mechanisms of tumor recurrence after surgical intervention, as well as resistance to radiation and chemotherapy, remain a pervasive and costly problem. Recent evidence has emerged supporting the hypothesis that solid tumors contain a sub-population of cancer cells that possess characteristics normally associated with stem cells. Particularly, the potential for long-term proliferation appears to be restricted to subpopulations of cancer stem cells (CSCs) functionally defined by their capacity to self-renew and give rise to differentiated cells that phenotypically recapitulate the original tumor, thereby causing relapse and patient death. These cancer stem cells present a unique opportunity to better understand the biology of solid tumors in general, as well as targets for future therapeutics. The general objective of the current study is to discuss the fundamental concepts for understanding the role of CSCs with respect to chemoresistance, radioresistance, special cell surface markers, cancer recurrence and metastasis in tumors of the osseous spine. This discussion is followed by a specific review of what is known about the role of CSCs in chordoma, the most common primary malignant osseous tumor of the spine.
Collapse
|
135
|
Zhang H, Wu H, Zheng J, Yu P, Xu L, Jiang P, Gao J, Wang H, Zhang Y. Transforming growth factor β1 signal is crucial for dedifferentiation of cancer cells to cancer stem cells in osteosarcoma. Stem Cells 2014; 31:433-46. [PMID: 23225703 DOI: 10.1002/stem.1298] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Revised: 11/01/2012] [Accepted: 11/15/2012] [Indexed: 01/04/2023]
Abstract
Human osteosarcoma harbors a small subpopulation of cancer stem cells (CSCs) that is believed to be associated with tumor metastasis, radioresistance/chemoresistance, local invasion, and poor clinical outcome. In this study, we found that transforming growth factor β1 (TGF-β1) signaling and a hypoxic environment dramatically induced self-renewal capacity in non-stem osteosarcoma cells, which in turn promoted chemoresistance, tumorigenicity, neovasculogenesis, and metastatic potential. Furthermore, blocking the TGF-β1 signaling pathway resulted in the inhibition of the dedifferentiation and clonogenicity of osteosarcoma cells, and the reduction of CSC self-renewal capacity and hypoxia-mediated dedifferentiation. These findings demonstrate that stem cells and non-stem cells exist in a dynamic equilibrium within the osteosarcoma cell population, and that CSCs may develop de novo from differentiated cancer cells. Hierarchical models of mammalian CSCs, therefore, should be considered to serve as bidirectional interconversion between the stem and non-stem cell components of the tumor.
Collapse
Affiliation(s)
- Haixia Zhang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Mohseny AB, Hogendoorn PCW. Zebrafish as a model for human osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 804:221-36. [PMID: 24924177 DOI: 10.1007/978-3-319-04843-7_12] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
For various reasons involving biological comparativeness, expansive technological possibilities, accelerated experimental speed, and competitive costs, zebrafish has become a comprehensive model for cancer research. Hence, zebrafish embryos and full-grown fish have been instrumental for studies of leukemia, melanoma, pancreatic cancer, bone tumors, and other malignancies. Although because of its similarities to human osteogenesis zebrafish appears to be an appealing model to investigate osteosarcoma, only a few osteosarcoma specific studies have been accomplished yet. Here, we review interesting related and unrelated reports of which the findings might be extrapolated to osteosarcoma. More importantly, rational but yet unexplored applications of zebrafish are debated to expand the window of opportunities for future establishment of osteosarcoma models. Accordingly technological advances of zebrafish based cancer research, such as robotic high-throughput multicolor injection systems and advanced imaging methods are discussed. Furthermore, various use of zebrafish embryos for screening drug regimens by combinations of chemotherapy, novel drug deliverers, and immune system modulators are suggested. Concerning the etiology, the high degree of genetic similarity between zebrafish and human cancers indicates that affected regions are evolutionarily conserved. Therefore, zebrafish as a swift model system that allows for the investigation of multiple candidate gene-defects is presented.
Collapse
Affiliation(s)
- A B Mohseny
- Department of Pathology, Leiden University Medical Center, 9600, H1-Q, Leiden, The Netherlands
| | | |
Collapse
|
137
|
Yu L, Liu S, Zhang C, Zhang B, Simões BM, Eyre R, Liang Y, Yan H, Wu Z, Guo W, Clarke RB. Enrichment of human osteosarcoma stem cells based on hTERT transcriptional activity. Oncotarget 2013; 4:2326-38. [PMID: 24334332 PMCID: PMC3926830 DOI: 10.18632/oncotarget.1554] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 11/03/2013] [Indexed: 01/01/2023] Open
Abstract
Telomerase is crucial for the maintenance of stem/progenitor cells in adult tissues and is detected in most malignant cancers, including osteosarcoma. However, the relationship between telomerase expression and cancer stem cells remains unknown. We observed that sphere-derived osteosarcoma cells had higher telomerase activity, indicating that telomerase activity might be enriched in osteosarcoma stem cells. We sorted subpopulations with high or low telomerase activity (TEL) using hTERT transcriptional promoter-induced green fluorescent protein (GFP). The TELpos cells showed an increased sphere and tumor propagating capacity compared to TELneg cells, and enhanced stem cell-like properties such as invasiveness, metastatic activity and resistance to chemotherapeutic agents both in vitro and in vivo. Furthermore, the telomerase inhibitor MST312 prevented tumorigenic potential both in vitro and in vivo, preferentially targeting the TELpos cells. These data support telomerase inhibition as a potential targeted therapy for osteosarcoma stem-like cells.
Collapse
Affiliation(s)
- Ling Yu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, P.R. China
- Institute of Cancer Sciences, University of Manchester, Wilmslow Road, Manchester, UK
| | - Shiqing Liu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, P.R. China
| | - Chun Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, P.R. China
| | - Bo Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, P.R. China
| | - Bruno M. Simões
- Institute of Cancer Sciences, University of Manchester, Wilmslow Road, Manchester, UK
| | - Rachel Eyre
- Institute of Cancer Sciences, University of Manchester, Wilmslow Road, Manchester, UK
| | - Yi Liang
- State Key Laboratory of Oncology in Southern China and the Department of Experimental Research, Sun Yat-Sen University Cancer Center, Dongfeng Road, Guangzhou, Guangdong, P.R. China
| | - Huichao Yan
- Opening Laboratory for Oversea Scientists, Wuhan University School of Basic Medical Science, Donghu Road, Wuhan, Hubei, P.R. China
| | - Zheng Wu
- Department of Radiation Oncology, Tumor Hospital Xiangya School of Medicine of Central South University, Tongzipo Road, Changsha, Hunan, P.R.China
| | - Weichun Guo
- Department of Orthopedics, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, P.R. China
| | - Robert B. Clarke
- Institute of Cancer Sciences, University of Manchester, Wilmslow Road, Manchester, UK
| |
Collapse
|
138
|
Chan LH, Wang W, Yeung W, Deng Y, Yuan P, Mak KK. Hedgehog signaling induces osteosarcoma development through Yap1 and H19 overexpression. Oncogene 2013; 33:4857-66. [PMID: 24141783 DOI: 10.1038/onc.2013.433] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 09/06/2013] [Accepted: 09/13/2013] [Indexed: 02/06/2023]
Abstract
Osteosarcoma is one of the most common bone tumors. However, the genetic basis for its pathogenesis remains elusive. Here, we investigated the roles of Hedgehog (Hh) signaling in osteosarcoma development. Genetically-engineered mice with ubiquitous upregulated Hh signaling specifically in mature osteoblasts develop focal bone overgrowth, which greatly resembles the early stage of osteosarcoma. However, these mice die within three months, which prohibits further analysis of tumor progression. We therefore generated a mouse model with partial upregulated Hh signaling in mature osteoblasts and crossed it into a p53 heterozygous background to potentiate tumor development. We found that these mutant mice developed malignant osteosarcoma with high penetrance. Isolated primary tumor cells were mainly osteoblastic and highly proliferative with many characteristics of human osteosarcomas. Allograft transplantation into immunocompromised mice displayed high tumorigenic potential. More importantly, both human and mouse tumor tissues express high level of yes-associated protein 1 (Yap1), a potent oncogene that is amplified in various cancers. We show that inhibition of Hh signaling reduces Yap1 expression and knockdown of Yap1 significantly inhibits tumor progression. Moreover, long non-coding RNA H19 is aberrantly expressed and induced by upregulated Hh signaling and Yap1 overexpression. Our results demonstrate that aberrant Hh signaling in mature osteoblasts is responsible for the pathogenesis of osteoblastic osteosarcoma through Yap1 and H19 overexpression.
Collapse
Affiliation(s)
- L H Chan
- Key Laboratories for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR
| | - W Wang
- Key Laboratories for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR
| | - W Yeung
- Key Laboratories for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR
| | - Y Deng
- Key Laboratories for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR
| | - P Yuan
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong SAR
| | - K K Mak
- 1] Key Laboratories for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR [2] Stem Cell and Regeneration Thematic Research Program, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR [3] CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| |
Collapse
|
139
|
Update on Targets and Novel Treatment Options for High-Grade Osteosarcoma and Chondrosarcoma. Hematol Oncol Clin North Am 2013; 27:1021-48. [DOI: 10.1016/j.hoc.2013.07.012] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
140
|
Posthumadeboer J, Piersma SR, Pham TV, van Egmond PW, Knol JC, Cleton-Jansen AM, van Geer MA, van Beusechem VW, Kaspers GJL, van Royen BJ, Jiménez CR, Helder MN. Surface proteomic analysis of osteosarcoma identifies EPHA2 as receptor for targeted drug delivery. Br J Cancer 2013; 109:2142-54. [PMID: 24064975 PMCID: PMC3798973 DOI: 10.1038/bjc.2013.578] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/23/2013] [Accepted: 08/28/2013] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Osteosarcoma (OS) is the most common bone tumour in children and adolescents. Despite aggressive therapy regimens, treatment outcomes are unsatisfactory. Targeted delivery of drugs can provide higher effective doses at the site of the tumour, ultimately improving the efficacy of existing therapy. Identification of suitable receptors for drug targeting is an essential step in the design of targeted therapy for OS. METHODS We conducted a comparative analysis of the surface proteome of human OS cells and osteoblasts using cell surface biotinylation combined with nano-liquid chromatography - tandem mass spectrometry-based proteomics to identify surface proteins specifically upregulated on OS cells. This approach generated an extensive data set from which we selected a candidate to study for its suitability as receptor for targeted treatment delivery to OS. First, surface expression of the ephrin type-A receptor 2 (EPHA2) receptor was confirmed using FACS analysis. Ephrin type-A receptor 2 expression in human tumour tissue was tested using immunohistochemistry. Receptor targeting and internalisation studies were conducted to assess intracellular uptake of targeted modalities via EPHA2. Finally, tissue micro arrays containing cores of human OS tissue were stained using immunohistochemistry and EPHA2 staining was correlated to clinical outcome measures. RESULTS Using mass spectrometry, a total of 2841 proteins were identified of which 156 were surface proteins significantly upregulated on OS cells compared with human primary osteoblasts. Ephrin type-A receptor 2 was highly upregulated and the most abundant surface protein on OS cells. In addition, EPHA2 was expressed in a vast majority of human OS samples. Ephrin type-A receptor 2 effectively mediates internalisation of targeted adenoviral vectors into OS cells. Patients with EPHA2-positive tumours showed a trend toward inferior overall survival. CONCLUSION The results presented here suggest that the EPHA2 receptor can be considered an attractive candidate receptor for targeted delivery of therapeutics to OS.
Collapse
Affiliation(s)
- J Posthumadeboer
- Department of Orthopaedic Surgery, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
de Girolamo L, Lucarelli E, Alessandri G, Avanzini MA, Bernardo ME, Biagi E, Brini AT, D'Amico G, Fagioli F, Ferrero I, Locatelli F, Maccario R, Marazzi M, Parolini O, Pessina A, Torre ML, Italian Mesenchymal Stem Cell Group. Mesenchymal stem/stromal cells: a new ''cells as drugs'' paradigm. Efficacy and critical aspects in cell therapy. Curr Pharm Des 2013; 19:2459-73. [PMID: 23278600 PMCID: PMC3788322 DOI: 10.2174/1381612811319130015] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 12/24/2012] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) were first isolated more than 50 years ago from the bone marrow. Currently MSCs may also be isolated from several alternative sources and they have been used in more than a hundred clinical trials worldwide to treat a wide variety of diseases. The MSCs mechanism of action is undefined and currently under investigation. For in vivo purposes MSCs must be produced in compliance with good manufacturing practices and this has stimulated research on MSCs characterization and safety. The objective of this review is to describe recent developments regarding MSCs properties, physiological effects, delivery, clinical applications and possible side effects.
Collapse
Affiliation(s)
- Laura de Girolamo
- Laboratorio di Biotecnologie applicate all'Ortopedia, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Active TGF-β signaling and decreased expression of PTEN separates angiosarcoma of bone from its soft tissue counterpart. Mod Pathol 2013; 26:1211-21. [PMID: 23599148 DOI: 10.1038/modpathol.2013.56] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 01/28/2013] [Accepted: 01/28/2013] [Indexed: 12/25/2022]
Abstract
Angiosarcomas constitute a heterogeneous group of highly malignant vascular tumors. Angiosarcoma of bone is rare and poorly characterized. For angiosarcoma of soft tissue, some pathways seem to be involved in tumor development. Our aim was to evaluate the role of these pathways in angiosarcoma of bone. We collected 37 primary angiosarcomas of bone and used 20 angiosarcomas of soft tissue for comparison. Immunohistochemistry was performed on constructed tissue microarrays to evaluate expression of CDKN2A, TP53, PTEN, BCL2, CDK4, MDM2, cyclin D1, β-catenin, transforming growth factor-β (TGF-β), CD105, phospho-Smad1, phospho-Smad2, hypoxia-inducible factor-1α, plasminogen activator inhibitor type 1 (PAI-1), VEGF, CD117 and glucose transporter--1. PIK3CA was screened for hotspot mutations in 19 angiosarcomas. In nearly 55% of the angiosarcoma of bone, the retinoblastoma (Rb) pathway was affected. Loss of CDKN2A expression was associated with a significantly worse prognosis. No overexpression of TP53 or MDM2 was found, suggesting that the TP53 pathway is not important in angiosarcoma of bone. Angiosarcoma of bone showed highly active TGF-β signaling with immunoreactivity for phospho-Smad2 and PAI-1. Although the phosphatidylinositol 3-kinase (PI3K)/Akt pathway seems to be active in both tumor groups, different mechanisms were involved: 41% of angiosarcoma of bone showed a decrease in expression of PTEN, whereas in angiosarcoma of soft tissue overexpression of KIT was found (90%). PIK3CA hotspot mutations were absent. In conclusion, the Rb pathway is involved in tumorigenesis of angiosarcoma of bone. The PI3K/Akt pathway is activated in both angiosarcoma of bone and soft tissue, however, with a different cause; PTEN expression is decreased in angiosarcoma of bone, whereas angiosarcomas of soft tissue show overexpression of KIT. Our findings support that angiosarcomas are a heterogeneous group of vascular malignancies. Both angiosarcoma of bone and soft tissue may benefit from therapeutic strategies targeting the PI3K/Akt pathway. However, interference with TGF-β signaling may be specifically relevant in angiosarcoma of bone.
Collapse
|
143
|
Xiao W, Mohseny AB, Hogendoorn PCW, Cleton-Jansen AM. Mesenchymal stem cell transformation and sarcoma genesis. Clin Sarcoma Res 2013; 3:10. [PMID: 23880362 PMCID: PMC3724575 DOI: 10.1186/2045-3329-3-10] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 07/01/2013] [Indexed: 01/27/2023] Open
Abstract
MSCs are hypothesized to potentially give rise to sarcomas after transformation and therefore serve as a good model to study sarcomagenesis. Both spontaneous and induced transformation of MSCs have been reported, however, spontaneous transformation has only been convincingly shown in mouse MSCs while induced transformation has been demonstrated in both mouse and human MSCs. Transformed MSCs of both species can give rise to pleomorphic sarcomas after transplantation into mice, indicating the potential MSC origin of so-called non-translocation induced sarcomas. Comparison of expression profiles and differentiation capacities between MSCs and sarcoma cells further supports this. Deregulation of P53- Retinoblastoma-, PI3K-AKT-and MAPK pathways has been implicated in transformation of MSCs. MSCs have also been indicated as cell of origin in several types of chromosomal translocation associated sarcomas. In mouse models the generated sarcoma type depends on amongst others the tissue origin of the MSCs, the targeted pathways and genes and the differentiation commitment status of MSCs. While some insights are glowing, it is clear that more studies are needed to thoroughly understand the molecular mechanism of sarcomagenesis from MSCs and mechanisms determining the sarcoma type, which will potentially give directions for targeted therapies.
Collapse
Affiliation(s)
- Wei Xiao
- Department of Pathology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333ZA, the Netherlands.
| | | | | | | |
Collapse
|
144
|
Abstract
Cell therapy with Multipotent Mesenchymal Stromal Cells (MSC) holds enormous promise for the treatment of a large number of degenerative and immune/inflammatory diseases. Their multilineage differentiation potential, immunoprivilege and capacity of promoting recovery of damaged tissues coupled with anti-inflammatory and immunosuppressive properties are the focus of a multitude of clinical studies currently underway. The recognized clinical potential of MSC repairing/immunomodulatory effects now encompasses graft-versus-host disease, hematologic malignancies, cardiovascular diseases, neurologic and inherited diseases, autoimmune diseases, organ transplantation, refractory wounds, and bone/cartilage defects among others. However, it has been suggested that both the need of extensive ex vivo culture for MSC clinical use, and their proangiogenic, anti-apoptotic and immunomodulatory properties may act together as tumor promoters, raising significant safety concerns. This paper will review the available data on in vitro MSC maldifferentiation and the ability of MSC to sustain tumor growth in vivo, with the aim to clarify whether MSC-based therapeutic approaches may carry actual risk of malignancies.
Collapse
|
145
|
Sampson VB, Gorlick R, Kamara D, Anders Kolb E. A review of targeted therapies evaluated by the pediatric preclinical testing program for osteosarcoma. Front Oncol 2013; 3:132. [PMID: 23755370 PMCID: PMC3668267 DOI: 10.3389/fonc.2013.00132] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 05/12/2013] [Indexed: 12/21/2022] Open
Abstract
Osteosarcoma, the most common malignant bone tumor of childhood, is a high-grade primary bone sarcoma that occurs mostly in adolescence. Standard treatment consists of surgery in combination with multi-agent chemotherapy regimens. The development and approval of imatinib for Philadelphia chromosome-positive acute lymphoblastic leukemia in children and the fully human monoclonal antibody, anti-GD2, as part of an immune therapy for high-risk neuroblastoma patients have established the precedent for use of targeted inhibitors along with standard chemotherapy backbones. However, few targeted agents tested have achieved traditional clinical endpoints for osteosarcoma. Many biological agents demonstrating anti-tumor responses in preclinical and early-phase clinical testing have failed to reach response thresholds to justify randomized trials with large numbers of patients. The development of targeted therapies for pediatric cancer remains a significant challenge. To aid in the prioritization of new agents for clinical testing, the Pediatric Preclinical Testing Program (PPTP) has developed reliable and robust preclinical pediatric cancer models to rapidly screen agents for activity in multiple childhood cancers and establish pharmacological parameters and effective drug concentrations for clinical trials. In this article, we examine a range of standard and novel agents that have been evaluated by the PPTP, and we discuss the preclinical and clinical development of these for the treatment of osteosarcoma. We further demonstrate that committed resources for hypothesis-driven drug discovery and development are needed to yield clinical successes in the search for new therapies for this pediatric disease.
Collapse
Affiliation(s)
- Valerie B Sampson
- Nemours Center for Childhood Cancer and Blood Disorders, Alfred I. duPont Hospital for Children , Wilmington, DE , USA
| | | | | | | |
Collapse
|
146
|
Risk of tumorigenicity in mesenchymal stromal cell-based therapies--bridging scientific observations and regulatory viewpoints. Cytotherapy 2013; 15:753-9. [PMID: 23602595 DOI: 10.1016/j.jcyt.2013.03.005] [Citation(s) in RCA: 284] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 03/11/2013] [Accepted: 03/13/2013] [Indexed: 12/17/2022]
Abstract
In the past decade, the therapeutic value of mesenchymal stromal cells (MSCs) has been studied in various indications, thereby taking advantage of their immunosuppressive properties. Easy procurement from bone marrow, adipose tissue or other sources and conventional in vitro expansion culture have made their clinical use attractive. Bridging the gap between current scientific knowledge and regulatory prospects on the transformation potential and possible tumorigenicity of MSCs, the Cell Products Working Party and the Committee for Advanced Therapies organized a meeting with leading European experts in the field of MSCs. This meeting elucidated the risk of potential tumorigenicity related to MSC-based therapies from two angles: the scientific perspective and the regulatory point of view. The conclusions of this meeting, including the current regulatory thinking on quality, nonclinical and clinical aspects for MSCs, are presented in this review, leading to a clearer way forward for the development of such products.
Collapse
|
147
|
Kuijjer ML, Hogendoorn PCW, Cleton-Jansen AM. Genome-wide analyses on high-grade osteosarcoma: making sense of a genomically most unstable tumor. Int J Cancer 2013; 133:2512-21. [PMID: 23436697 DOI: 10.1002/ijc.28124] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 02/13/2013] [Indexed: 12/16/2022]
Abstract
High-grade osteosarcoma is an extremely genomically unstable tumor. This, together with other challenges, such as the heterogeneity within and between tumor samples, and the rarity of the disease, renders it difficult to study this tumor on a genome-wide level. Now that most laboratories change from genome-wide microarray experiments to Next-Generation Sequencing it is important to discuss the lessons we have learned from microarray studies. In this review, we discuss the challenges of high-grade osteosarcoma data analysis. We give an overview of microarray studies that have been conducted so far on both osteosarcoma tissue samples and cell lines. We discuss recent findings from integration of different data types, which is particularly relevant in a tumor with such a complex genomic profile. Finally, we elaborate on the translation of results obtained with bioinformatics into functional studies, which has lead to valuable findings, especially when keeping in mind that no new therapies with a significant impact on survival have been developed in the past decades.
Collapse
Affiliation(s)
- Marieke L Kuijjer
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | |
Collapse
|
148
|
Posthumadeboer J, van Egmond PW, Helder MN, de Menezes RX, Cleton-Jansen AM, Beliën JAM, Verheul HMW, van Royen BJ, Kaspers GJJL, van Beusechem VW. Targeting JNK-interacting-protein-1 (JIP1) sensitises osteosarcoma to doxorubicin. Oncotarget 2013; 3:1169-81. [PMID: 23045411 PMCID: PMC3717953 DOI: 10.18632/oncotarget.600] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumour in children and adolescents. Despite aggressive therapy, survival outcomes remain unsatisfactory, especially for patients with metastatic disease or patients with a poor chemotherapy response. Chemoresistance contributes to treatment failure. To increase the efficacy of conventional chemotherapy, essential survival pathways should be targeted concomitantly. Here, we performed a loss-of-function siRNA screen of the human kinome in SaOS-2 cells to identify critical survival kinases after doxorubicin treatment. Gene silencing of JNK-interacting-protein-1 (JIP1) elicited the most potent sensitisation to doxorubicin. This candidate was further explored as potential target for chemosensitisation in OS. A panel of OS cell lines and human primary osteoblasts was examined for sensitisation to doxorubicin using small molecule JIP1-inhibitor BI-78D3. JIP1 expression and JIP1-inhibitor effects on JNK-signalling were investigated by Western blot analysis. JIP1 expression in human OS tumours was assessed by immunohistochemistry on tissue micro arrays. BI-78D3 blocked JNK-signalling and sensitised three out of four tested OS cell lines, but not healthy osteoblasts, to treatment with doxorubicin. Combination treatment increased the induction of apoptosis. JIP1 was found to be expressed in two-thirds of human primary OS tissue samples. Patients with JIP1 positive tumours showed a trend to inferior overall survival. Collectively, JIP1 appears a clinically relevant novel target in OS to enhance the efficacy of doxorubicin treatment by means of RNA interference or pharmacological inhibition.
Collapse
Affiliation(s)
- Jantine Posthumadeboer
- Department of Orthopaedic Surgery, VU University Medical Center, Amsterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Szuhai K, Hogendoorn PCW. 'The chicken or the egg?' dilemma strikes back for the controlling mechanism in chordoma(#). J Pathol 2013; 228:261-5. [PMID: 22952146 DOI: 10.1002/path.4102] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Chordoma is a rare malignant tumour of bone showing notochordal differentiation with characteristic expression of the transcription factor brachyury (T). Next to giving insight into its differentiation spectrum, the expression of T can be used as an adjunct diagnostic tool. The expression of brachyury in chordoma is necessary to maintain cell proliferation in chordoma cell lines, indicating that in chordoma it might be a master regulator of oncogenesis. Identification and mapping of the full gene regulatory network in a recent work in The Journal of Pathology by Nelson and colleagues not only shed light on involved pathways but also indicated pathways for targeted therapy, including brachyury itself.
Collapse
Affiliation(s)
- Karoly Szuhai
- Department of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2330 RC, Leiden, The Netherlands
| | | |
Collapse
|
150
|
Rubio R, Gutierrez-Aranda I, Sáez-Castillo AI, Labarga A, Rosu-Myles M, Gonzalez-Garcia S, Toribio ML, Menendez P, Rodriguez R. The differentiation stage of p53-Rb-deficient bone marrow mesenchymal stem cells imposes the phenotype of in vivo sarcoma development. Oncogene 2012; 32:4970-80. [PMID: 23222711 DOI: 10.1038/onc.2012.507] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 09/25/2012] [Accepted: 09/28/2012] [Indexed: 12/15/2022]
Abstract
Increasing evidence suggests that mesenchymal stem/stromal cells (MSCs) carrying specific mutations are at the origin of some sarcomas. We have reported that the deficiency of p53 alone or in combination with Rb (Rb(-/-) p53(-/-)) in adipose-derived MSCs (ASCs) promotes leiomyosarcoma-like tumors in vivo. Here, we hypothesized that the source of MSCs and/or the cell differentiation stage could determine the phenotype of sarcoma development. To investigate whether there is a link between the source of MSCs and sarcoma phenotype, we generated p53(-/-) and Rb(-/-)p53(-/-) MSCs from bone marrow (BM-MSCs). Both genotypes of BM-MSCs initiated leiomyosarcoma formation similar to p53(-/-) and Rb(-/-)p53(-/-) ASCs. In addition, gene expression profiling revealed transcriptome similarities between p53- or Rb-p53-deficient BM-MSCs/ASCs and muscle-associated sarcomagenesis. These data suggest that the tissue source of MSC does not seem to determine the development of a particular sarcoma phenotype. To analyze whether the differentiation stage defines the sarcoma phenotype, BM-MSCs and ASCs were induced to differentiate toward the osteogenic lineage, and both p53 and Rb were excised using Cre-expressing adenovectors at different stages along osteogenic differentiation. Regardless the level of osteogenic commitment, the inactivation of Rb and p53 in BM-MSC-derived, but not in ASC-derived, osteogenic progenitors gave rise to osteosarcoma-like tumors, which could be serially transplanted. This indicates that the osteogenic differentiation stage of BM-MSCs imposes the phenotype of in vivo sarcoma development, and that BM-MSC-derived osteogenic progenitors rather than undifferentiated BM-MSCs, undifferentiated ASCs or ASC-derived osteogenic progenitors, represent the cell of origin for osteosarcoma development.
Collapse
Affiliation(s)
- R Rubio
- GENYO. Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|