Skip to main content

Sarcoma Stem Cell Heterogeneity

  • Chapter
  • First Online:
Stem Cells Heterogeneity - Novel Concepts

Abstract

Sarcomas represent an extensive group of divergent malignant diseases, with the only common characteristic of being derived from mesenchymal cells. As such, sarcomas are by definition very heterogeneous, and this heterogeneity does not manifest only upon intertumoral comparison on a bulk tumor level but can be extended to intratumoral level. Whereas part of this intratumoral heterogeneity could be understood in terms of clonal genetic evolution, an essential part includes a hierarchical relationship between sarcoma cells, governed by both genetic and epigenetic influences, signals that sarcoma cells are exposed to, and intrinsic developmental programs derived from sarcoma cells of origin. The notion of this functional hierarchy operating within each tumor implies the existence of sarcoma stem cells, which may originate from mesenchymal stem cells, and indeed, mesenchymal stem cells have been used to establish several crucial experimental sarcoma models and to trace down their respective stem cell populations. Mesenchymal stem cells themselves are heterogeneous, and, moreover, there are alternative possibilities for sarcoma cells of origin, like neural crest-derived stem cells, or mesenchymal committed precursor cells, or – in rhabdomyosarcoma – muscle satellite cells. These various origins result in substantial heterogeneity in possible sarcoma initiation. Genetic and epigenetic changes associated with sarcomagenesis profoundly impact the biology of sarcoma stem cells. For pediatric sarcomas featuring discrete reciprocal translocations and largely stable karyotypes, the translocation-activated oncogenes could be crucial factors that confer stemness, principally by modifying transcriptome and interfering with normal epigenetic regulation; the most extensively studied examples of this process are myxoid/round cell liposarcoma, Ewing sarcoma, and synovial sarcoma. For adult sarcomas, which have typically complex and unstable karyotypes, stemness might be defined more operationally, as a reflection of actual assembly of genetically and epigenetically conditioned stemness factors, with dedifferentiated liposarcoma providing a most thoroughly studied example. Alternatively, stemness can be imposed by tumor microenvironment, as extensively documented in osteosarcoma. In spite of this heterogeneity in both sarcoma initiation and underlying stemness biology, some of the molecular mechanisms of stemness might be remarkably similar in diverse sarcoma types, like abrogation of classical tumor suppressors pRb and p53, activation of Sox-2, or inhibition of canonical Wnt/β-catenin signaling. Moreover, even some stem cell markers initially characterized for their stem cell enrichment capacity in various carcinomas or leukemias seem to function quite similarly in various sarcomas. Understanding the biology of sarcoma stem cells could significantly improve sarcoma patient clinical care, leading to both better patient stratification and, hopefully, development of more effective therapeutic options.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Skubitz KM, D’Adamo DR (2007) Sarcoma. Mayo Clin Proc 82(11):1409–1432

    Article  CAS  PubMed  Google Scholar 

  2. Penzel R, Schirmacher P, Renner M, Mechtersheimer G (2011) Molekularpathologie maligner Weichgewebetumoren. In: Schlag PM, Hartmann JT, Budach V (eds) Weichgewebetumoren: Interdisziplinäres Management. Springer, Berlin Heidelberg, Berlin, Heidelberg, pp 23–35

    Chapter  Google Scholar 

  3. Oda Y, Yamamoto H, Kohashi K, Yamada Y, Iura K, Ishii T, Maekawa A, Bekki H (2017) Soft tissue sarcomas: from a morphological to a molecular biological approach. Pathol Int 67(9):435–446

    Article  CAS  PubMed  Google Scholar 

  4. Taylor BS, Barretina J, Maki RG, Antonescu CR, Singer S, Ladanyi M (2011a) Advances in sarcoma genomics and new therapeutic targets. Nat Rev Cancer 11(8):541–557

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Henze J, Bauer S (2013) Liposarcomas. Hematol Oncol Clin North Am 27(5):939–955

    Article  PubMed  Google Scholar 

  6. Papenfuss AT, Thomas DM (2015) The life history of neochromosomes revealed. Mol Cell Oncol 2(4):e1000698

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Frenette PS, Pinho S, Lucas D, Scheiermann C (2013) Mesenchymal stem cell: keystone of the hematopoietic stem cell niche and a stepping-stone for regenerative medicine. Annu Rev Immunol 31:285–316

    Article  PubMed  Google Scholar 

  8. Schäfer R, Northoff H (2008) Characteristics of mesenchymal stem cells—new stars in regenerative medicine or unrecognized old fellows in autologous regeneration? Transfus Med Hemotherapy 35(3):154–159

    Article  Google Scholar 

  9. Guneta V, Tan NS, Chan SKJ, Tanavde V, Lim TC, Wong TCM, Choong C (2016) Comparative study of adipose-derived stem cells and bone marrow-derived stem cells in similar microenvironmental conditions. Exp Cell Res 348(2):155–164

    Article  CAS  PubMed  Google Scholar 

  10. Meyer MB, Benkusky NA, Sen B, Rubin J, Pike JW (2016) Epigenetic plasticity drives adipogenic and osteogenic differentiation of marrow-derived mesenchymal stem cells. J Biol Chem 291(34):17,829–17,847

    Article  CAS  Google Scholar 

  11. Bianco P, Robey PG (2015) Skeletal stem cells. Development 142(6):1023–1027

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Dinulovic I, Furrer R, Handschin C (2017) Plasticity of the muscle stem cell microenvironment. Adv Exp Med Biol 1041:141–169

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Shakhova O, Sommer L (2010) Neural crest-derived stem cells. In: The Stem Cell Research Community (ed) StemBook. Harvard Stem Cell Institute, Cambridge, MA

    Google Scholar 

  14. Almalki SG, Agrawal DK (2016) Key transcription factors in the differentiation of mesenchymal stem cells. Differ Res Biol Divers 92(1–2):41–51

    Article  CAS  Google Scholar 

  15. Rivlin N, Koifman G, Rotter V (2015) p53 orchestrates between normal differentiation and cancer. Semin Cancer Biol 32:10–17

    Article  CAS  PubMed  Google Scholar 

  16. Chen WY, Wang DH, Yen RC, Luo J, Gu W, Baylin SB (2005) Tumor suppressor HIC1 directly regulates SIRT1 to modulate p53-dependent DNA-damage responses. Cell 123(3):437–448

    Article  CAS  PubMed  Google Scholar 

  17. Yamakuchi M, Lowenstein CJ (2009) miR-34, SIRT1 and p53: the feedback loop. Cell Cycle 8:712–715

    Article  CAS  PubMed  Google Scholar 

  18. Goeman F, Strano S, Blandino G (2017) MicroRNAs as key effectors in the p53 network. Int Rev Cell Mol Biol 333:51–90

    Google Scholar 

  19. Luo Z, Cui R, Tili E, Croce C (2018) Friend or foe: microRNAs in the p53 network. Cancer Lett 419:96–102

    Article  CAS  PubMed  Google Scholar 

  20. Riggi N, Suvà M-L, De Vito C, Provero P, Stehle J-C, Baumer K, Cironi L, Janiszewska M, Petricevic T, Suvà D, Tercier S, Joseph J-M, Guillou L, Stamenkovic I (2010) EWS-FLI-1 modulates miRNA145 and SOX2 expression to initiate mesenchymal stem cell reprogramming toward Ewing sarcoma cancer stem cells. Genes Dev 24(9):916–932

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Han M-K, Song E-K, Guo Y, Ou X, Mantel C, Broxmeyer HE (2008) SIRT1 regulates apoptosis and Nanog expression in mouse embryonic stem cells by controlling p53 subcellular localization. Cell Stem Cell 2(3):241–251

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yoon DS, Choi Y, Jang Y, Lee M, Choi WJ, Kim S-H, Lee JW (2014) SIRT1 directly regulates SOX2 to maintain self-renewal and multipotency in bone marrow-derived mesenchymal stem cells. Stem Cells 32(12):3219–3231

    Article  CAS  PubMed  Google Scholar 

  23. Vilas JM, Ferreirós A, Carneiro C, Morey L, Silva-Álvarez SD, Fernandes T, Abad M, Croce LD, García-Caballero T, Serrano M, Rivas C, Vidal A, Collado M (2014) Transcriptional regulation of Sox2 by the retinoblastoma family of pocket proteins. Oncotarget 6(5):2992–3002

    PubMed Central  Google Scholar 

  24. Boregowda SV, Krishnappa V, Strivelli J, Haga CL, Booker CN, Phinney DG (2018) Basal p53 expression is indispensable for mesenchymal stem cell integrity. Cell Death Differ 25(4):677–690

    CAS  PubMed Central  Google Scholar 

  25. Artigas N, Gámez B, Cubillos-Rojas M, Sánchez-de Diego C, Valer JA, Pons G, Rosa JL, Ventura F (2017) p53 inhibits SP7/Osterix activity in the transcriptional program of osteoblast differentiation. Cell Death Differ 24(12):2022–2031

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Martinez-Sanchez A, Dudek KA, Murphy CL (2012) Regulation of human chondrocyte function through direct inhibition of cartilage master regulator SOX9 by microRNA-145 (miRNA-145). J Biol Chem 287(2):916–924

    Article  CAS  PubMed  Google Scholar 

  27. Honoki K, Tsujiuchi T (2013) Senescence bypass in mesenchymal stem cells: a potential pathogenesis and implications of pro-senescence therapy in sarcomas. Expert Rev Anticancer Ther 13(8):983–996

    Article  CAS  PubMed  Google Scholar 

  28. Matushansky I, Hernando E, Socci ND, Mills JE, Matos TA, Edgar MA, Singer S, Maki RG, Cordon-Cardo C (2007) Derivation of sarcomas from mesenchymal stem cells via inactivation of the Wnt pathway. J Clin Invest 117(11):3248–3257

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kim JR, Moon YJ, Kwon KS, Bae JS, Wagle S, Yu TK, Kim KM, Park HS, Lee J-H, Moon WS, Lee H, Chung MJ, Jang KY (2013) Expression of SIRT1 and DBC1 is associated with poor prognosis of soft tissue sarcomas. PLoS One 8(9):e74738

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Skoda J, Nunukova A, Loja T, Zambo I, Neradil J, Mudry P, Zitterbart K, Hermanova M, Hampl A, Sterba J, Veselska R (2016) Cancer stem cell markers in pediatric sarcomas: Sox2 is associated with tumorigenicity in immunodeficient mice. Tumour Biol 37(7):9535–9548

    Article  CAS  PubMed  Google Scholar 

  31. Plon SE, Malkin D (2010) Childhood cancer and heredity. In: Principles and practice of pediatric oncology, Sixth edition. LWW, Philadelphia, PA, pp 17–37

    Google Scholar 

  32. Kanojia D, Nagata Y, Garg M, Lee DH, Sato A, Yoshida K, Sato Y, Sanada M, Mayakonda A, Bartenhagen C, Klein H-U, Doan NB, Said JW, Mohith S, Gunasekar S, Shiraishi Y, Chiba K, Tanaka H, Miyano S, Myklebost O, Yang H, Dugas M, Meza-Zepeda LA, Silberman AW, Forscher C, Tyner JW, Ogawa S, Koeffler HP (2015) Genomic landscape of liposarcoma. Oncotarget 6(40):42,429–42,444

    Article  Google Scholar 

  33. Hatina J, Hájková L, Peychl J, Rudolf E, Fínek J, Cervinka M, Reischig J (2003) Establishment and characterization of clonal cell lines derived from a fibrosarcoma of the H2-K/V-JUN transgenic mouse. A model of H2-K/V-JUN mediated tumorigenesis. Tumour Biol 24(4):176–184

    Article  PubMed  Google Scholar 

  34. Mariani O, Brennetot C, Coindre J-M, Gruel N, Ganem C, Delattre O, Stern M-H, Aurias A (2007) JUN oncogene amplification and overexpression block adipocytic differentiation in highly aggressive sarcomas. Cancer Cell 11(4):361–374

    Article  CAS  PubMed  Google Scholar 

  35. Endo M, Nishita M, Fujii M, Minami Y (2015) Insight into the role of Wnt5a-induced signaling in normal and cancer cells. Int Rev Cell Mol Biol 314:117–148

    Google Scholar 

  36. Lye KL, Nordin N, Vidyadaran S, Thilakavathy K (2016) Mesenchymal stem cells: from stem cells to sarcomas. Cell Biol Int 40(6):610–618

    Article  PubMed  Google Scholar 

  37. Gaebler M, Silvestri A, Haybaeck J, Reichardt P, Lowery CD, Stancato LF, Zybarth G, Regenbrecht CRA (2017) Three-dimensional patient-derived in vitro sarcoma models: Promising tools for improving clinical tumor management. Front Oncol 7:203

    Google Scholar 

  38. Xiao W, Mohseny AB, Hogendoorn PCW, Cleton-Jansen A-M (2013) Mesenchymal stem cell transformation and sarcoma genesis. Clin Sarcoma Res 3(1):10

    Article  PubMed  PubMed Central  Google Scholar 

  39. Hahn WC, Counter CM, Lundberg AS, Beijersbergen RL, Brooks MW, Weinberg RA (1999) Creation of human tumour cells with defined genetic elements. Nature 400(6743):464–468

    Article  CAS  PubMed  Google Scholar 

  40. Rubio R, Gutierrez-Aranda I, Sáez-Castillo AI, Labarga A, Rosu-Myles M, Gonzalez-Garcia S, Toribio ML, Menendez P, Rodriguez R (2013) The differentiation stage of p53-Rb-deficient bone marrow mesenchymal stem cells imposes the phenotype of in vivo sarcoma development. Oncogene 32(41):4970–4980

    Article  CAS  PubMed  Google Scholar 

  41. Tao J, Jiang M-M, Jiang L, Salvo JS, Zeng H-C, Dawson B, Bertin TK, Rao PH, Chen R, Donehower LA, Gannon F, Lee BH (2014) Notch activation as a driver of osteogenic sarcoma. Cancer Cell 26(3):390–401

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Engström K, Willén H, Kåbjörn-Gustafsson C, Andersson C, Olsson M, Göransson M, Järnum S, Olofsson A, Warnhammar E, Aman P (2006) The myxoid/round cell liposarcoma fusion oncogene FUS-DDIT3 and the normal DDIT3 induce a liposarcoma phenotype in transfected human fibrosarcoma cells. Am J Pathol 168(5):1642–1653

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Morena D, Maestro N, Bersani F, Forni PE, Lingua MF, Foglizzo V, Šćepanović P, Miretti S, Morotti A, Shern JF, Khan J, Ala U, Provero P, Sala V, Crepaldi T, Gasparini P, Casanova M, Ferrari A, Sozzi G, Chiarle R, Ponzetto C, Taulli R (2016) Hepatocyte growth factor-mediated satellite cells niche perturbation promotes development of distinct sarcoma subtypes. Elife 5:pii: e12116

    Article  Google Scholar 

  44. Rubin BP, Nishijo K, Chen H-IH, Yi X, Schuetze DP, Pal R, Prajapati SI, Abraham J, Arenkiel BR, Chen Q-R, Davis S, McCleish AT, Capecchi MR, Michalek JE, Zarzabal LA, Khan J, Yu Z, Parham DM, Barr FG, Meltzer PS, Chen Y, Keller C (2011) Evidence for an unanticipated relationship between undifferentiated pleomorphic sarcoma and embryonal rhabdomyosarcoma. Cancer Cell 19(2):177–191

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Boeuf S, Kunz P, Hennig T, Lehner B, Hogendoorn P, Bovée J, Richter W (2008) A chondrogenic gene expression signature in mesenchymal stem cells is a classifier of conventional central chondrosarcoma. J Pathol 216(2):158–166

    Article  CAS  PubMed  Google Scholar 

  46. Matushansky I, Hernando E, Socci ND, Matos T, Mills J, Edgar MA, Schwartz GK, Singer S, Cordon-Cardo C, Maki RG (2008) A developmental model of sarcomagenesis defines a differentiation-based classification for liposarcomas. Am J Pathol 172(4):1069–1080

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Borgo C, Milan G, Favaretto F, Stasi F, Fabris R, Salizzato V, Cesaro L, Belligoli A, Sanna M, Foletto M, Prevedello L, Vindigni V, Bardini R, Donella-Deana A, Vettor R (2017) CK2 modulates adipocyte insulin-signaling and is up-regulated in human obesity. Sci Rep 7(1):17,569

    Article  CAS  Google Scholar 

  48. Funes JM, Quintero M, Henderson S, Martinez D, Qureshi U, Westwood C, Clements MO, Bourboulia D, Pedley RB, Moncada S, Boshoff C (2007) Transformation of human mesenchymal stem cells increases their dependency on oxidative phosphorylation for energy production. Proc Natl Acad Sci U S A 104(15):6223–6228

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Rodriguez R, Rubio R, Menendez P (2012) Modeling sarcomagenesis using multipotent mesenchymal stem cells. Cell Res 22(1):62–77

    Article  CAS  PubMed  Google Scholar 

  50. Schulz W (2005) Molecular biology of human cancers: an advanced student’s textbook. Springer, Netherlands

    Google Scholar 

  51. Rodriguez R, Tornin J, Suarez C, Astudillo A, Rubio R, Yauk C, Williams A, Rosu-Myles M, Funes JM, Boshoff C, Menendez P (2013) Expression of FUS-CHOP fusion protein in immortalized/transformed human mesenchymal stem cells drives mixoid liposarcoma formation. Stem Cells 31(10):2061–2072

    Article  CAS  PubMed  Google Scholar 

  52. Li H, Fan X, Kovi RC, Jo Y, Moquin B, Konz R, Stoicov C, Kurt-Jones E, Grossman SR, Lyle S, Rogers AB, Montrose M, Houghton J (2007) Spontaneous expression of embryonic factors and p53 point mutations in aged mesenchymal stem cells: a model of age-related tumorigenesis in mice. Cancer Res 67(22):10,889–10,898

    Article  CAS  Google Scholar 

  53. Shimizu T, Ishikawa T, Sugihara E, Kuninaka S, Miyamoto T, Mabuchi Y, Matsuzaki Y, Tsunoda T, Miya F, Morioka H, Nakayama R, Kobayashi E, Toyama Y, Kawai A, Ichikawa H, Hasegawa T, Okada S, Ito T, Ikeda Y, Suda T, Saya H (2010) c-MYC overexpression with loss of Ink4a/Arf transforms bone marrow stromal cells into osteosarcoma accompanied by loss of adipogenesis. Oncogene 29(42):5687–5699

    Article  CAS  PubMed  Google Scholar 

  54. Lin PP, Pandey MK, Jin F, Raymond AK, Akiyama H, Lozano G (2009) Targeted mutation of p53 and Rb in mesenchymal cells of the limb bud produces sarcomas in mice. Carcinogenesis 30(10):1789–1795

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Lin PP, Pandey MK, Jin F, Xiong S, Deavers M, Parant JM, Lozano G (2008) EWS-FLI1 induces developmental abnormalities and accelerates sarcoma formation in a transgenic mouse model. Cancer Res 68(21):8968–8975

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Riggi N, Suvà M-L, Suvà D, Cironi L, Provero P, Tercier S, Joseph J-M, Stehle J-C, Baumer K, Kindler V, Stamenkovic I (2008) EWS-FLI-1 expression triggers a Ewing’s sarcoma initiation program in primary human mesenchymal stem cells. Cancer Res 68(7):2176–2185

    Article  CAS  PubMed  Google Scholar 

  57. von Levetzow C, Jiang X, Gwye Y, von Levetzow G, Hung L, Cooper A, Hsu JH-R, Lawlor ER (2011) Modeling initiation of Ewing sarcoma in human neural crest cells. PLoS One 6(4):e19305

    Article  CAS  Google Scholar 

  58. Mihály D, Matula Z, Changchien Y-C, Papp G, Tátrai P, Sápi Z (2017) First cloned human immortalized adipose derived mesenchymal stem-cell line with chimeric SS18-SSX1 gene (SS-iASC). Cancer Genet 216-217:52–60

    Article  CAS  PubMed  Google Scholar 

  59. Tamaki S, Fukuta M, Sekiguchi K, Jin Y, Nagata S, Hayakawa K, Hineno S, Okamoto T, Watanabe M, Woltjen K, Ikeya M, Jr TK, Toguchida J (2015) SS18-SSX, the oncogenic fusion protein in synovial sarcoma, is a cellular context-dependent epigenetic modifier. PLoS One 10(11):e0142991

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Dela Cruz FS (2013) Cancer stem cells in pediatric sarcomas. Front Oncol 3:168

    Google Scholar 

  61. Skoda J, Veselska R (2018) Cancer stem cells in sarcomas: getting to the stemness core. Biochim Biophys Acta 1862(10):2134–2139

    Article  CAS  Google Scholar 

  62. Veselska R, Skoda J, Neradil J (2012) Detection of cancer stem cell markers in sarcomas. Klin Onkol 25(Suppl 2):2S16–2S20

    PubMed  Google Scholar 

  63. Wu C, Wei Q, Utomo V, Nadesan P, Whetstone H, Kandel R, Wunder JS, Alman BA (2007) Side population cells isolated from mesenchymal neoplasms have tumor initiating potential. Cancer Res 67(17):8216–8222

    Article  CAS  PubMed  Google Scholar 

  64. Zhou Y, Zhou Y, Chen D, Chen D, Qi Y, Qi Y, Liu R, Liu R, Li S, Li S, Zou H, Zou H, Lan J, Lan J, Ju X, Ju X, Jiang J, Jiang J, Liang W, Liang W, Shen Y, Shen Y, Pang L, Pang L, Li F, Li F (2017) Evaluation of expression of cancer stem cell markers and fusion gene in synovial sarcoma: insights into histogenesis and pathogenesis. Oncol Rep 37(6):3351–3360

    Article  CAS  PubMed  Google Scholar 

  65. Lohberger B, Rinner B, Stuendl N, Absenger M, Liegl-Atzwanger B, Walzer SM, Windhager R, Leithner A (2012) Aldehyde dehydrogenase 1, a potential marker for cancer stem cells in human sarcoma. PLoS One 7(8):e43664

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Martinez-Cruzado L, Tornin J, Santos L, Rodriguez A, García-Castro J, Morís F, Rodriguez R (2016) Aldh1 expression and activity increase during tumor evolution in sarcoma cancer stem cell populations. Sci Rep 6:27,878

    Article  CAS  Google Scholar 

  67. Siclari VA, Qin L (2010) Targeting the osteosarcoma cancer stem cell. J Orthop Surg 5(1):78

    Article  Google Scholar 

  68. Kimura T, Wang L, Tabu K, Tsuda M, Tanino M, Maekawa A, Nishihara H, Hiraga H, Taga T, Oda Y, Tanaka S (2015) Identification and analysis of CXCR4-positive synovial sarcoma-initiating cells. Oncogene 35(30):3932–3943

    Article  CAS  PubMed  Google Scholar 

  69. Adhikari AS, Agarwal N, Wood BM, Porretta C, Ruiz B, Pochampally RR, Iwakuma T (2010) CD117 and Stro-1 identify osteosarcoma tumor-initiating cells associated with metastasis and drug resistance. Cancer Res 70(11):4602–4612

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Tirino V, Desiderio V, Paino F, De Rosa A, Papaccio F, Fazioli F, Pirozzi G, Papaccio G (2011) Human primary bone sarcomas contain CD133+ cancer stem cells displaying high tumorigenicity in vivo. FASEB J 25(6):2022–2030

    Article  CAS  PubMed  Google Scholar 

  71. Suvà M-L, Riggi N, Stehle J-C, Baumer K, Tercier S, Joseph J-M, Suvà D, Clément V, Provero P, Cironi L, Osterheld M-C, Guillou L, Stamenkovic I (2009) Identification of cancer stem cells in Ewing’s sarcoma. Cancer Res 69(5):1776–1781

    Article  CAS  PubMed  Google Scholar 

  72. Walter D, Satheesha S, Albrecht P, Bornhauser BC, D’Alessandro V, Oesch SM, Rehrauer H, Leuschner I, Koscielniak E, Gengler C, Moch H, Bernasconi M, Niggli FK, Schäfer BW, CWS Study Group (2011) CD133 positive embryonal rhabdomyosarcoma stem-like cell population is enriched in rhabdospheres. PLoS One 6(5):e19506

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Levings PP, McGarry SV, Currie TP, Nickerson DM, McClellan S, Ghivizzani SC, Steindler DA, Gibbs CP (2009) Expression of an exogenous human Oct-4 promoter identifies tumor-initiating cells in osteosarcoma. Cancer Res 69(14):5648–5655

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Saini V, Hose CD, Monks A, Nagashima K, Han B, Newton DL, Millione A, Shah J, Hollingshead MG, Hite KM, Burkett MW, Delosh RM, Silvers TE, Scudiero DA, Shoemaker RH (2012) Identification of CBX3 and ABCA5 as putative biomarkers for tumor stem cells in osteosarcoma. PLoS One 7(8):e41401

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Yang J, Ren Z, Du X, Hao M, Zhou W (2014) The role of mesenchymal stem/progenitor cells in sarcoma: update and dispute. Stem Cell Investig 1:18

    PubMed  PubMed Central  Google Scholar 

  76. Zhang L, Wang C (2007) Identification of a new class of PAX3-FKHR target promoters: a role of the Pax3 paired box DNA binding domain. Oncogene 26(11):1595–1605

    Article  CAS  PubMed  Google Scholar 

  77. Gibbs CP, Kukekov VG, Reith JD, Tchigrinova O, Suslov ON, Scott EW, Ghivizzani SC, Ignatova TN, Steindler DA (2005) Stem-like cells in bone sarcomas: implications for tumorigenesis. Neoplasia 7(11):967–976

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Wei Q, Tang YJ, Voisin V, Sato S, Hirata M, Whetstone H, Han I, Ailles L, Bader GD, Wunder J, Alman BA (2015) Identification of CD146 as a marker enriched for tumor-propagating capacity reveals targetable pathways in primary human sarcoma. Oncotarget 6(37):40,283–40,294

    Article  Google Scholar 

  79. Trucco M, Loeb D (2012) Sarcoma stem cells: do we know what we are looking for? Sarcoma 2012:291705

    Google Scholar 

  80. Wang M-Y, Nestvold J, Rekdal Ø, Kvalheim G, Fodstad Ø (2017) A novel rat fibrosarcoma cell line from transformed bone marrow-derived mesenchymal stem cells with maintained in vitro and in vivo stemness properties. Exp Cell Res 352(2):218–224

    Article  CAS  PubMed  Google Scholar 

  81. Fujiwara T, Kawai A, Yoshida A, Ozaki T, Ochiya T (2013) Cancer stem cells of sarcoma. In: Role of cancer stem cells in cancer biology and therapy. CRC Press, Boca Raton, FL, pp 23–78

    Chapter  Google Scholar 

  82. Hatina J, Fernandes MI, Hoffmann MJ, Zeimet AG (2013) Cancer stem cells – basic biological properties and experimental approaches. Encyclopedia of Life Sciences. Chichester, John Wiley & Sons. https://doi.org/10.1002/9780470015902.a0021164.pub2

  83. Krause U, Ryan DM, Clough BH, Gregory CA (2014) An unexpected role for a Wnt-inhibitor: Dickkopf-1 triggers a novel cancer survival mechanism through modulation of aldehyde-dehydrogenase-1 activity. Cell Death Dis 5:e1093

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Martinez-Cruzado L, Tornin J, Rodriguez A, Santos L, Allonca E, Fernandez-Garcia MT, Astudillo A, Garcia-Pedrero JM, Rodriguez R (2017) Trabectedin and campthotecin synergistically eliminate cancer stem cells in cell-of-origin sarcoma models. Neoplasia 19(6):460–470

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Stacchiotti S, Van Tine BA (2017) Synovial sarcoma: current concepts and future perspectives. J Clin Oncol 36(2):180–187

    Article  PubMed  Google Scholar 

  86. Naka N, Takenaka S, Araki N, Miwa T, Hashimoto N, Yoshioka K, Joyama S, Hamada K-I, Tsukamoto Y, Tomita Y, Ueda T, Yoshikawa H, Itoh K (2010) Synovial sarcoma is a stem cell malignancy. Stem Cells 28(7):1119–1131

    CAS  PubMed  Google Scholar 

  87. Saito T, Nagai M, Ladanyi M (2006) SYT-SSX1 and SYT-SSX2 interfere with repression of E-cadherin by snail and slug: a potential mechanism for aberrant mesenchymal to epithelial transition in human synovial sarcoma. Cancer Res 66(14):6919–6927

    Google Scholar 

  88. Eid JE, Garcia CB (2015) Reprogramming of mesenchymal stem cells by oncogenes. Semin Cancer Biol 32:18–31

    Article  CAS  PubMed  Google Scholar 

  89. Zöllner SK, Rössig C, Toretsky JA (2015) Synovial sarcoma is a gateway to the role of chromatin remodeling in cancer. Cancer Metastasis Rev 34(3):417–428

    Article  CAS  PubMed  Google Scholar 

  90. Jedlicka P (2010) Ewing Sarcoma, an enigmatic malignancy of likely progenitor cell origin, driven by transcription factor oncogenic fusions. Int J Clin Exp Pathol 3(4):338–347

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Svoboda LK, Harris A, Bailey NJ, Schwentner R, Tomazou E, von Levetzow C, Magnuson B, Ljungman M, Kovar H, Lawlor ER (2014) Overexpression of HOX genes is prevalent in Ewing sarcoma and is associated with altered epigenetic regulation of developmental transcription programs. Epigenetics 9(12):1613–1625

    Article  PubMed  Google Scholar 

  92. Beird HC, Wu C-C, Ingram DR, Wang W-L, Alimohamed A, Gumbs C, Little L, Song X, Feig BW, Roland CL, Zhang J, Benjamin RS, Hwu P, Lazar AJ, Futreal PA, Somaiah N (2018) Genomic profiling of dedifferentiated liposarcoma compared to matched well-differentiated liposarcoma reveals higher genomic complexity and a common origin. Cold Spring Harb Mol Case Stud 4(2):pii:a002386

    Article  CAS  Google Scholar 

  93. Mayr C, Hemann MT, Bartel DP (2007) Disrupting the pairing between let-7 and Hmga2 enhances oncogenic transformation. Science 315(5818):1576–1579

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Henriksen J, Stabell M, Meza-Zepeda LA, Lauvrak SA, Kassem M, Myklebost O (2010) Identification of target genes for wild type and truncated HMGA2 in mesenchymal stem-like cells. BMC Cancer 10:329

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Taylor BS, DeCarolis PL, Angeles CV, Brenet F, Schultz N, Antonescu CR, Scandura JM, Sander C, Viale AJ, Socci ND, Singer S (2011b) Frequent alterations and epigenetic silencing of differentiation pathway genes in structurally rearranged liposarcomas. Cancer Discov 1(7):587–597

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Guan M, Wu X, Chu P, Chow WA (2017) Fatty acid synthase reprograms the epigenome in uterine leiomyosarcomas. PLoS One 12(6):e0179692

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Di Pompo G, Salerno M, Rotili D, Valente S, Zwergel C, Avnet S, Lattanzi G, Baldini N, Mai A (2015) Novel histone deacetylase inhibitors induce growth arrest, apoptosis, and differentiation in sarcoma cancer stem cells. J Med Chem 58(9):4073–4079

    Article  CAS  PubMed  Google Scholar 

  98. Abarrategi A, Tornin J, Martinez-Cruzado L, Hamilton A, Martinez-Campos E, Rodrigo JP, González MV, Baldini N, Garcia-Castro J, Rodriguez R (2016) Osteosarcoma: cells-of-origin, cancer stem cells, and targeted therapies. Stem Cells Int 2016:3631764

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Alfranca A, Martinez-Cruzado L, Tornin J, Abarrategi A, Amaral T, de Alava E, Menendez P, Garcia-Castro J, Rodriguez R (2015) Bone microenvironment signals in osteosarcoma development. Cell Mol Life Sci 72(16):3097–3113

    Article  CAS  PubMed  Google Scholar 

  100. Plaks V, Kong N, Werb Z (2015) The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 16(3):225–238

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Rubio R, Abarrategi A, Garcia-Castro J, Martinez-Cruzado L, Suarez C, Tornin J, Santos L, Astudillo A, Colmenero I, Mulero F, Rosu-Myles M, Menendez P, Rodriguez R (2014) Bone environment is essential for osteosarcoma development from transformed mesenchymal stem cells. Stem Cells 32(5):1136–1148

    Article  CAS  PubMed  Google Scholar 

  102. Heymann M-F, Lézot F, Heymann D (2017) The contribution of immune infiltrates and the local microenvironment in the pathogenesis of osteosarcoma. Cell Immunol (17):30,189–30,182

    Google Scholar 

  103. Basu-Roy U, Seo E, Ramanathapuram L, Rapp TB, Perry JA, Orkin SH, Mansukhani A, Basilico C (2012) Sox2 maintains self renewal of tumor-initiating cells in osteosarcomas. Oncogene 31(18):2270–2282

    Article  CAS  PubMed  Google Scholar 

  104. Zhang H, Wu H, Zheng J, Yu P, Xu L, Jiang P, Gao J, Wang H, Zhang Y (2013) Transforming growth factor β1 signal is crucial for dedifferentiation of cancer cells to cancer stem cells in osteosarcoma. Stem Cells 31(3):433–446

    Article  CAS  PubMed  Google Scholar 

  105. Basu-Roy U, Bayin NS, Rattanakorn K, Han E, Placantonakis DG, Mansukhani A, Basilico C (2015) Sox2 antagonizes the Hippo pathway to maintain stemness in cancer cells. Nat Commun 6:6411

    Article  CAS  PubMed  Google Scholar 

  106. Wang L, Park P, Zhang H, La Marca F, Claeson A, Valdivia J, Lin C-Y (2011) BMP-2 inhibits the tumorigenicity of cancer stem cells in human osteosarcoma OS99-1 cell line. Cancer Biol Ther 11(5):457–463

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Avnet S, Cortini M (2016) Role of pericellular matrix in the regulation of cancer stemness. Stem Cell Rev 12(4):464–475

    Article  CAS  Google Scholar 

  108. Avnet S, Di Pompo G, Chano T, Errani C, Ibrahim-Hashim A, Gillies RJ, Donati DM, Baldini N (2017) Cancer-associated mesenchymal stroma fosters the stemness of osteosarcoma cells in response to intratumoral acidosis via NF-κB activation. Int J Cancer 140(6):1331–1345

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Tu B, Zhu J, Liu S, Wang L, Fan Q, Hao Y, Fan C, Tang T-T (2016) Mesenchymal stem cells promote osteosarcoma cell survival and drug resistance through activation of STAT3. Oncotarget 7(30):48296–48308

    Article  PubMed  PubMed Central  Google Scholar 

  110. Kuhn NZ, Tuan RS (2010) Regulation of stemness and stem cell niche of mesenchymal stem cells: implications in tumorigenesis and metastasis. J Cell Physiol 222(2):268–277

    Article  CAS  PubMed  Google Scholar 

  111. Zeng W, Wan R, Zheng Y, Singh SR, Wei Y (2011) Hypoxia, stem cells and bone tumor. Cancer Lett 313(2):129–136

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Cathomas R, Rothermundt C, Bode B, Fuchs B, von Moos R, Schwitter M (2015) RANK ligand blockade with denosumab in combination with sorafenib in chemorefractory osteosarcoma: a possible step forward? Oncology 88(4):257–260

    Article  CAS  PubMed  Google Scholar 

  113. Moriceau G, Ory B, Gobin B, Verrecchia F, Gouin F, Blanchard F, Redini F, Heymann D (2010) Therapeutic approach of primary bone tumours by bisphosphonates. Curr Pharm Des 16(27):2981–2987

    Article  CAS  PubMed  Google Scholar 

  114. Sampson VB, Gorlick R, Kamara D, Anders Kolb E (2013) A review of targeted therapies evaluated by the pediatric preclinical testing program for osteosarcoma. Front Oncol 3:132

    Article  PubMed  PubMed Central  Google Scholar 

  115. Rainusso N, Brawley VS, Ghazi A, Hicks MJ, Gottschalk S, Rosen JM, Ahmed N (2012) Immunotherapy targeting HER2 with genetically modified T cells eliminates tumor-initiating cells in osteosarcoma. Cancer Gene Ther 19(3):212–217

    Article  CAS  PubMed  Google Scholar 

  116. Tarek N, Lee DA (2014) Natural killer cells for osteosarcoma. Adv Exp Med Biol 804:341–353

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

The original studies cited are supported by the Czech Science Foundation project No. 17-17636S (J.H., M.K., K.H., J.K., J.S., O.S.); by the project CZ.02.1.01/0.0/0.0/16_019/0000787 provided by the European Regional Development Fund and the Ministry of Education, Youth and Sports of the Czech Republic (M.K. and J.K); by Charles University in Prague Specific Student Research Projects No. 260394/2017 and No. 260393/2017 (J.H., M.K., K.H, M.P., J.K.); and by Spanish Plan Nacional de I+D+I 2013-2016: ISCIII (CPII16/00049), CIBERONC (CB16/12/00390), and MINECO/FEDER (SAF2016-75286-R) (R.R.).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jiri Hatina .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2019 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Hatina, J. et al. (2019). Sarcoma Stem Cell Heterogeneity. In: Birbrair, A. (eds) Stem Cells Heterogeneity - Novel Concepts. Advances in Experimental Medicine and Biology, vol 1123. Springer, Cham. https://doi.org/10.1007/978-3-030-11096-3_7

Download citation

Publish with us

Policies and ethics