期刊论文详细信息
BMC Veterinary Research
HES1, a target of Notch signaling, is elevated in canine osteosarcoma, but reduced in the most aggressive tumors
Dawn L Duval1  Thora J Jonasdottir2  Barbara E Powers3  Douglas H Thamm4  Tina B Bønsdorff2  J Brad Charles3  EJ Ehrhart3  Liza E Pfaff5  Kristin P Anfinsen2  Deanna D Dailey5 
[1] Department of Clinical Sciences, Animal Cancer Center, Colorado State University, 300 West Drake Road, Fort Collins, CO 80523-1620, USA;Department of Companion Animal Clinical Sciences, Norwegian School of Veterinary Science, P.O. Box 8416 Dep., Oslo, NO-0033, Norway;Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA;University of Colorado Comprehensive Cancer Center, Aurora, CO, USA;Cell and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, USA
关键词: Microarray;    Canine;    Immunohistochemistry;    RT-qPCR;    RT-PCR;    Osteosarcoma;    Notch;    HES1;    Hes-1;   
Others  :  1119508
DOI  :  10.1186/1746-6148-9-130
 received in 2012-08-23, accepted in 2013-06-24,  发布年份 2013
PDF
【 摘 要 】

Background

Hairy and enhancer of split 1 (HES1), a basic helix-loop-helix transcriptional repressor, is a downstream target of Notch signaling. Notch signaling and HES1 expression have been linked to growth and survival in a variety of human cancer types and have been associated with increased metastasis and invasiveness in human osteosarcoma cell lines. Osteosarcoma (OSA) is an aggressive cancer demonstrating both high metastatic rate and chemotherapeutic resistance. The current study examined expression of Notch signaling mediators in primary canine OSA tumors and canine and human osteosarcoma cell lines to assess their role in OSA development and progression.

Results

Reverse transcriptase - quantitative PCR (RT-qPCR) was utilized to quantify HES1, HEY1, NOTCH1 and NOTCH2 gene expression in matched tumor and normal metaphyseal bone samples taken from dogs treated for appendicular OSA at the Colorado State University Veterinary Teaching Hospital. Gene expression was also assessed in tumors from dogs with a disease free interval (DFI) of <100 days compared to those with a DFI > 300 days following treatment with surgical amputation followed by standard chemotherapy. Immunohistochemistry was performed to confirm expression of HES1. Data from RT-qPCR and immunohistochemical (IHC) experiments were analyzed using REST2009 software and survival analysis based on IHC expression employed the Kaplan-Meier method and log rank analysis. Unbiased clustered images were generated from gene array analysis data for Notch/HES1 associated genes.

Gene array analysis of Notch/HES1 associated genes suggested alterations in the Notch signaling pathway may contribute to the development of canine OSA. HES1 mRNA expression was elevated in tumor samples relative to normal bone, but decreased in tumor samples from dogs with a DFI < 100 days relative to those with a DFI > 300 days. NOTCH2 and HEY1 mRNA expression was also elevated in tumors relative to normal bone, but was not differentially expressed between the DFI tumor groups. Survival analysis confirmed an association between decreased HES1 immunosignal and shorter DFI.

Conclusions

Our findings suggest that activation of Notch signaling occurs and may contribute to the development of canine OSA. However, association of low HES1 expression and shorter DFI suggests that mechanisms that do not alter HES1 expression may drive the most aggressive tumors.

【 授权许可】

   
2013 Dailey et al.; licensee BioMed Central Ltd.

【 预 览 】
附件列表
Files Size Format View
20150208073313356.pdf 1366KB PDF download
Figure 7. 44KB Image download
Figure 6. 123KB Image download
Figure 5. 41KB Image download
Figure 4. 50KB Image download
Figure 3. 38KB Image download
Figure 2. 38KB Image download
Figure 1. 158KB Image download
【 图 表 】

Figure 1.

Figure 2.

Figure 3.

Figure 4.

Figure 5.

Figure 6.

Figure 7.

【 参考文献 】
  • [1]Mirabello L, Troisi RJ, Savage SA: Osteosarcoma incidence and survival rates from 1973 to 2004: data from the Surveillance, Epidemiology, and End Results Program. Cancer 2009, 115(7):1531-1543.
  • [2]Dernell WS, Ehrhart NP, Straw RC, Vail DM: Tumors of the Skeletal System. In Withrow & MacEwen's Small Animal Clinical Oncology. 4th edition. St. Louis, Mo: Saunders Elsevier; 2007:540-567.
  • [3]Mueller F, Fuchs B, Kaser-Hotz B: Comparative biology of human and canine osteosarcoma. Anticancer Res 2007, 27(1A):155-164.
  • [4]Paoloni M, Davis S, Lana S, Withrow S, Sangiorgi L, Picci P, Hewitt S, Triche T, Meltzer P, Khanna C: Canine tumor cross-species genomics uncovers targets linked to osteosarcoma progression. BMC Genomics 2009, 10:625. BioMed Central Full Text
  • [5]De Maria R, Miretti S, Iussich S, Olivero M, Morello E, Bertotti A, Christensen JG, Biolatti B, Levine RA, Buracco P, et al.: Met oncogene activation qualifies spontaneous canine osteosarcoma as a suitable pre-clinical model of human osteosarcoma. J Pathol 2009, 218(3):399-408.
  • [6]Morello E, Martano M, Buracco P: Biology, diagnosis and treatment of canine appendicular osteosarcoma: similarities and differences with human osteosarcoma. Vet J 2011, 189(3):268-277.
  • [7]Maniscalco L, Iussich S, Morello E, Martano M, Biolatti B, Riondato F, Della Salda L, Romanucci M, Malatesta D, Bongiovanni L, et al.: PDGFs and PDGFRs in canine osteosarcoma: new targets for innovative therapeutic strategies in comparative oncology. Vet J 2013, 195(1):41-47.
  • [8]Jaffe N: Osteosarcoma: review of the past, impact on the future. The American experience. Cancer Treat Res 2009, 152:239-262.
  • [9]Iso T, Kedes L, Hamamori Y: HES and HERP families: multiple effectors of the Notch signaling pathway. J Cell Physiol 2003, 194(3):237-255.
  • [10]Fischer A, Gessler M: Delta-Notch–and then? Protein interactions and proposed modes of repression by Hes and Hey bHLH factors. Nucleic Acids Res 2007, 35(14):4583-4596.
  • [11]Coglievina M, Guarnaccia C, Pintar A, Pongor S: Different degrees of structural order in distinct regions of the transcriptional repressor HES-1. Biochim Biophys Acta 2010, 1804(12):2153-2161.
  • [12]Takebayashi K, Sasai Y, Sakai Y, Watanabe T, Nakanishi S, Kageyama R: Structure, chromosomal locus, and promoter analysis of the gene encoding the mouse helix-loop-helix factor HES-1. Negative autoregulation through the multiple N box elements. J Biol Chem 1994, 269(7):5150-5156.
  • [13]Ingram WJ, McCue KI, Tran TH, Hallahan AR, Wainwright BJ: Sonic Hedgehog regulates Hes1 through a novel mechanism that is independent of canonical Notch pathway signalling. Oncogene 2008, 27(10):1489-1500.
  • [14]Wall DS, Wallace VA: Hedgehog to Hes1: the heist of a Notch target. Cell Cycle 2009, 8(9):1301-1302.
  • [15]Bennani-Baiti IM, Aryee DN, Ban J, Machado I, Kauer M, Muhlbacher K, Amann G, Llombart-Bosch A, Kovar H: Notch signalling is off and is uncoupled from HES1 expression in Ewing's sarcoma. J Pathol 2011, 225(3):353-363.
  • [16]Shih Ie M, Wang TL: Notch signaling, gamma-secretase inhibitors, and cancer therapy. Cancer Res 2007, 67(5):1879-1882.
  • [17]Weng AP, Aster JC: Multiple niches for Notch in cancer: context is everything. Curr Opin Genet Dev 2004, 14(1):48-54.
  • [18]Liu J, Ye F, Chen H, Lu W, Zhou C, Xie X: Expression of differentiation associated protein Hes1 and Hes5 in cervical squamous carcinoma and its precursors. Int J Gynecol Canc 2007, 17(6):1293-1299.
  • [19]Wang X, Fu Y, Chen X, Ye J, Lu B, Ye F, Lu W, Xie X: The expressions of bHLH gene HES1 and HES5 in advanced ovarian serous adenocarcinomas and their prognostic significance: a retrospective clinical study. J Cancer Res Clin Oncol 2010, 136(7):989-996.
  • [20]Kannan S, Fang W, Song G, Mullighan CG, Hammitt R, McMurray J, Zweidler-McKay PA: Notch/HES1-mediated PARP1 activation: a cell type-specific mechanism for tumor suppression. Blood 2011, 117(10):2891-2900.
  • [21]Viatour P, Ehmer U, Saddic LA, Dorrell C, Andersen JB, Lin C, Zmoos AF, Mazur PK, Schaffer BE, Ostermeier A, et al.: Notch signaling inhibits hepatocellular carcinoma following inactivation of the RB pathway. J Exp Med 2011, 208(10):1963-1976.
  • [22]Zage PE, Nolo R, Fang W, Stewart J, Garcia-Manero G, Zweidler-McKay PA: Notch pathway activation induces neuroblastoma tumor cell growth arrest. Pediatr Blood Cancer 2012, 58(5):682-689.
  • [23]Klinakis A, Lobry C, Abdel-Wahab O, Oh P, Haeno H, Buonamici S, van De Walle I, Cathelin S, Trimarchi T, Araldi E, et al.: A novel tumour-suppressor function for the Notch pathway in myeloid leukaemia. Nature 2011, 473(7346):230-233.
  • [24]Zhang P, Yang Y, Zweidler-McKay PA, Hughes DP: Critical role of notch signaling in osteosarcoma invasion and metastasis. Clin Cancer Res 2008, 14(10):2962-2969.
  • [25]Tanaka M, Setoguchi T, Hirotsu M, Gao H, Sasaki H, Matsunoshita Y, Komiya S: Inhibition of Notch pathway prevents osteosarcoma growth by cell cycle regulation. Br J Cancer 2009, 100(12):1957-1965.
  • [26]Engin F, Bertin T, Ma O, Jiang MM, Wang L, Sutton RE, Donehower LA, Lee B: Notch signaling contributes to the pathogenesis of human osteosarcomas. Hum Mol Genet 2009, 18(8):1464-1470.
  • [27]Hughes DP: How the NOTCH pathway contributes to the ability of osteosarcoma cells to metastasize. Cancer Treat Res 2009, 152:479-496.
  • [28]Zhang P, Yang Y, Nolo R, Zweidler-McKay PA, Hughes DP: Regulation of NOTCH signaling by reciprocal inhibition of HES1 and Deltex 1 and its role in osteosarcoma invasiveness. Oncogene 2010, 29(20):2916-2926.
  • [29]Modder UI, Oursler MJ, Khosla S, Monroe DG: Wnt10b activates the Wnt, notch, and NFkappaB pathways in U2OS osteosarcoma cells. J Cell Biochem 2011, 112(5):1392-1402.
  • [30]Lin GL, Hankenson KD: Integration of BMP, Wnt, and notch signaling pathways in osteoblast differentiation. J Cell Biochem 2011, 112(12):3491-3501.
  • [31]Day TF, Yang Y: Wnt and hedgehog signaling pathways in bone development. J Bone Joint Surg Am 2008, 90(Suppl 1):19-24.
  • [32]O'Donoghue LE, Ptitsyn AA, Kamstock DA, Siebert J, Thomas RS, Duval DL: Expression profiling in canine osteosarcoma: identification of biomarkers and pathways associated with outcome. BMC Cancer 2010, 10:506. BioMed Central Full Text
  • [33]Moore AS, Dernell WS, Ogilvie GK, Kristal O, Elmslie R, Kitchell B, Susaneck S, Rosenthal R, Klein MK, Obradovich J, et al.: Doxorubicin and BAY 12–9566 for the treatment of osteosarcoma in dogs: a randomized, double-blind, placebo-controlled study. J Vet Intern Med 2007, 21:783-790.
  • [34]O'Donoghue LE, Rivest JP, Duval DL: Polymerase chain reaction-based species verification and microsatellite analysis for canine cell line validation. J Vet Diagn Invest 2011, 23(4):780-785.
  • [35]Su Y, Luo X, He BC, Wang Y, Chen L, Zuo GW, Liu B, Bi Y, Huang J, Zhu GH, et al.: Establishment and characterization of a new highly metastatic human osteosarcoma cell line. Clin Exp Metastasis 2009, 26(7):599-610.
  • [36]Thomson SA, Kennerly E, Olby N, Mickelson JR, Hoffmann DE, Dickinson PJ, Gibson G, Breen M: Microarray analysis of differentially expressed genes of primary tumors in the canine central nervous system. Vet Pathol 2005, 42(5):550-558.
  • [37]de Jonge HJ, Fehrmann RS, de Bont ES, Hofstra RM, Gerbens F, Kamps WA, de Vries EG, van der Zee AG, te Meerman GJ, ter Elst A: Evidence based selection of housekeeping genes. PLoS One 2007, 2(9):e898.
  • [38]Pfaffl MW, Horgan GW, Dempfle L: Relative expression software tool (REST) for group-wise comparison and statistical analysis of relative expression results in real-time PCR. Nucleic Acids Res 2002, 30(9):e36.
  • [39]Livak KJ, Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001, 25(4):402-408.
  • [40]Dallas PB, Gottardo NG, Firth MJ, Beesley AH, Hoffmann K, Terry PA, Freitas JR, Boag JM, Cummings AJ, Kees UR: Gene expression levels assessed by oligonucleotide microarray analysis and quantitative real-time RT-PCR – how well do they correlate? BMC Genomics 2005, 6:59. BioMed Central Full Text
  • [41]Herr F, Schreiner I, Baal N, Pfarrer C, Zygmunt M: Expression patterns of Notch receptors and their ligands Jagged and Delta in human placenta. Placenta 2011, 32(8):554-563.
  • [42]Katoh M, Katoh M: Integrative genomic analyses on HES/HEY family: Notch-independent HES1, HES3 transcription in undifferentiated ES cells, and Notch-dependent HES1, HES5, HEY1, HEY2, HEYL transcription in fetal tissues, adult tissues, or cancer. Int J Oncol 2007, 31(2):461-466.
  • [43]Johansson T, Lejonklou MH, Ekeblad S, Stalberg P, Skogseid B: Lack of nuclear expression of hairy and enhancer of split-1 (HES1) in pancreatic endocrine tumors. Horm Metab Res 2008, 40(5):354-359.
  • [44]Morimoto M, Liu Z, Cheng HT, Winters N, Bader D, Kopan R: Canonical Notch signaling in the developing lung is required for determination of arterial smooth muscle cells and selection of Clara versus ciliated cell fate. J Cell Sci 2010, 123(Pt 2):213-224.
  • [45]Shimojo H, Ohtsuka T, Kageyama R: Oscillations in notch signaling regulate maintenance of neural progenitors. Neuron 2008, 58(1):52-64.
  • [46]Wang C, Yao N, Lu CL, Li D, Ma X: Mouse microRNA-124 regulates the expression of Hes1 in P19 cells. Front Biosci (Elite Ed) 2010, 2:127-132.
  • [47]Kimura H, Kawasaki H, Taira K: Mouse microRNA-23b regulates expression of Hes1 gene in P19 cells. Nucleic Acids Symp Ser (Oxf) 2004, (48):213-214.
  • [48]Ju BG, Solum D, Song EJ, Lee KJ, Rose DW, Glass CK, Rosenfeld MG: Activating the PARP-1 sensor component of the groucho/TLE1 corepressor complex mediates a CaMKinase IIdelta-dependent neurogenic gene activation pathway. Cell 2004, 119(6):815-829.
  • [49]Hilton MJ, Tu X, Wu X, Bai S, Zhao H, Kobayashi T, Kronenberg HM, Teitelbaum SL, Ross FP, Kopan R, et al.: Notch signaling maintains bone marrow mesenchymal progenitors by suppressing osteoblast differentiation. Nat Med 2008, 14(3):306-314.
  • [50]Engin F, Yao Z, Yang T, Zhou G, Bertin T, Jiang MM, Chen Y, Wang L, Zheng H, Sutton RE, et al.: Dimorphic effects of Notch signaling in bone homeostasis. Nat Med 2008, 14(3):299-305.
  • [51]Lee JS, Thomas DM, Gutierrez G, Carty SA, Yanagawa S, Hinds PW: HES1 cooperates with pRb to activate RUNX2-dependent transcription. J Bone Min Res 2006, 21(6):921-933.
  • [52]Sang L, Roberts JM, Coller HA: Hijacking HES1: how tumors co-opt the anti-differentiation strategies of quiescent cells. Trends Mol Med 2010, 16(1):17-26.
  文献评价指标  
  下载次数:13次 浏览次数:16次