Semin Reprod Med 2013; 31(01): 005-013
DOI: 10.1055/s-0032-1331792
Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

Cells, Stem Cells, and Cancer Stem Cells

Cristóbal Aguilar-Gallardo
1   Valencia Node of the Spanish Stem Cell Bank, Prince Felipe Research Centre (CIPF)
,
Carlos Simón
1   Valencia Node of the Spanish Stem Cell Bank, Prince Felipe Research Centre (CIPF)
2   Fundación IVI-Instituto Universitario IVI, INCLIVA, Valencia University, Valencia, Spain
› Author Affiliations
Further Information

Publication History

Publication Date:
17 January 2013 (online)

Abstract

The stem cell field owes a great deal to the previous work conducted by embryologists and researchers devoted to reproductive medicine. The time is coming when this emerging field will pay off in the reproductive sciences by offering new avenues of understanding gametogenesis and early embryonic development. Human embryonic stem cells are pluripotent cells that proliferate in vitro while maintaining an undifferentiated state, and they are capable of differentiating into most cell types under appropriate conditions. Embryo-friendly approaches have been developed as new methods of obtaining human embryonic stem cells without destroying the embryo. Somatic stem cells have been identified and isolated from numerous adult organs and tissues to create a multipotent and autologous source of cells with established medical indications. Cell reprogramming is now a scientific fact, and induced pluripotent cells, a new pluripotent cell type, have been generated by the overexpression of specific genes from a myriad of differentiated adult cell types. Cancer is now considered a stem cell disease. Cancer stem cells share numerous features with normal stem cells including hallmarks properties such as self-renewal and undifferentiation. Therefore, the actual focus of ovarian cancer research on the cancer stem cell model should generate efficient and personalized treatment designs to improve treatment efficiency.

 
  • References

  • 1 Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006; 126 (4) 663-676
  • 2 Thomson JA, Itskovitz-Eldor J, Shapiro SS , et al. Embryonic stem cell lines derived from human blastocysts. Science 1998; 282 (5391) 1145-1147
  • 3 Turetsky T, Aizenman E, Gil Y , et al. Laser-assisted derivation of human embryonic stem cell lines from IVF embryos after preimplantation genetic diagnosis. Hum Reprod 2008; 23 (1) 46-53
  • 4 Valbuena D, Galán A, Sánchez E , et al. Derivation and characterization of three new Spanish human embryonic stem cell lines (VAL -3 -4 -5) on human feeder and in serum-free conditions. Reprod Biomed Online 2006; 13 (6) 875-886
  • 5 Klimanskaya I, Chung Y, Meisner L, Johnson J, West MD, Lanza R. Human embryonic stem cells derived without feeder cells. Lancet 2005; 365 (9471) 1636-1641
  • 6 Klimanskaya I, Chung Y, Becker S, Lu SJ, Lanza R. Human embryonic stem cell lines derived from single blastomeres. Nature 2006; 444 (7118) 481-485
  • 7 Chung Y, Klimanskaya I, Becker S , et al. Embryonic and extraembryonic stem cell lines derived from single mouse blastomeres. Nature 2006; 439 (7073) 216-219
  • 8 Forbes S, Vig P, Poulsom R, Thomas H, Alison M. Hepatic stem cells. J Pathol 2002; 197 (4) 510-518
  • 9 Pittenger MF, Mackay AM, Beck SC , et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999; 284 (5411) 143-147
  • 10 Zuk PA, Zhu M, Mizuno H , et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng 2001; 7 (2) 211-228
  • 11 Bieback K, Kern S, Klüter H, Eichler H. Critical parameters for the isolation of mesenchymal stem cells from umbilical cord blood. Stem Cells 2004; 22 (4) 625-634
  • 12 Jiang Y, Vaessen B, Lenvik T, Blackstad M, Reyes M, Verfaillie CM. Multipotent progenitor cells can be isolated from postnatal murine bone marrow, muscle, and brain. Exp Hematol 2002; 30 (8) 896-904
  • 13 Zhou S, Schuetz JD, Bunting KD , et al. The ABC transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype. Nat Med 2001; 7 (9) 1028-1034
  • 14 Brazelton TR, Rossi FM, Keshet GI, Blau HM. From marrow to brain: expression of neuronal phenotypes in adult mice. Science 2000; 290 (5497) 1775-1779
  • 15 Yoon J, Choi SC, Park CY , et al. Bone marrow-derived side population cells are capable of functional cardiomyogenic differentiation. Mol Cells 2008; 25 (2) 216-223
  • 16 Sadek HA, Martin CM, Latif SS, Garry MG, Garry DJ. Bone-marrow-derived side population cells for myocardial regeneration. J Cardiovasc Transl Res 2009; 2 (2) 173-181
  • 17 Krause DS, Theise ND, Collector MI , et al. Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell. Cell 2001; 105 (3) 369-377
  • 18 Horwitz EM, Prockop DJ, Fitzpatrick LA , et al. Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta. Nat Med 1999; 5 (3) 309-313
  • 19 Wang L, Menendez P, Cerdan C, Bhatia M. Hematopoietic development from human embryonic stem cell lines. Exp Hematol 2005; 33 (9) 987-996
  • 20 Trounson A. The production and directed differentiation of human embryonic stem cells. Endocr Rev 2006; 27 (2) 208-219
  • 21 Takahashi K, Tanabe K, Ohnuki M , et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007; 131 (5) 861-872
  • 22 Yu J, Vodyanik MA, Smuga-Otto K , et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 2007; 318 (5858) 1917-1920
  • 23 Szabo E, Rampalli S, Risueño RM , et al. Direct conversion of human fibroblasts to multilineage blood progenitors. Nature 2010; 468 (7323) 521-526
  • 24 Kim JB, Greber B, Araúzo-Bravo MJ , et al. Direct reprogramming of human neural stem cells by OCT4. Nature 2009; 461 (7264) 649-3
  • 25 Marión RM, Strati K, Li H , et al. A p53-mediated DNA damage response limits reprogramming to ensure iPS cell genomic integrity. Nature 2009; 460 (7259) 1149-1153
  • 26 Hong H, Takahashi K, Ichisaka T , et al. Suppression of induced pluripotent stem cell generation by the p53-p21 pathway. Nature 2009; 460 (7259) 1132-1135
  • 27 Kawamura T, Suzuki J, Wang YV , et al. Linking the p53 tumour suppressor pathway to somatic cell reprogramming. Nature 2009; 460 (7259) 1140-1144
  • 28 Soldner F, Hockemeyer D, Beard C , et al. Parkinson's disease patient-derived induced pluripotent stem cells free of viral reprogramming factors. Cell 2009; 136 (5) 964-977
  • 29 Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S. Generation of mouse induced pluripotent stem cells without viral vectors. Science 2008; 322 (5903) 949-953
  • 30 Kaji K, Norrby K, Paca A, Mileikovsky M, Mohseni P, Woltjen K. Virus-free induction of pluripotency and subsequent excision of reprogramming factors. Nature 2009; 458 (7239) 771-775
  • 31 Yu J, Hu K, Smuga-Otto K , et al. Human induced pluripotent stem cells free of vector and transgene sequences. Science 2009; 324 (5928) 797-801
  • 32 Marro S, Pang ZP, Yang N , et al. Direct lineage conversion of terminally differentiated hepatocytes to functional neurons. Cell Stem Cell 2011; 9 (4) 374-382
  • 33 Carvajal-Vergara X, Sevilla A, D'Souza SL , et al. Patient-specific induced pluripotent stem-cell-derived models of LEOPARD syndrome. Nature 2010; 465 (7299) 808-812
  • 34 Raya A, Rodríguez-Pizà I, Guenechea G , et al. Disease-corrected haematopoietic progenitors from Fanconi anaemia induced pluripotent stem cells. Nature 2009; 460 (7251) 53-59
  • 35 Urbach A, Bar-Nur O, Daley GQ, Benvenisty N. Differential modeling of fragile X syndrome by human embryonic stem cells and induced pluripotent stem cells. Cell Stem Cell 2010; 6 (5) 407-411
  • 36 Wunderlich V. JMM—past and present. Chromosomes and cancer: Theodor Boveri's predictions 100 years later. J Mol Med (Berl) 2002; 80 (9) 545-548
  • 37 Clarke MF, Dick JE, Dirks PB , et al. Cancer stem cells—perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res 2006; 66 (19) 9339-9344
  • 38 Ruiz-Vela A, Aguilar-Gallardo C, Simón C. Building a framework for embryonic microenvironments and cancer stem cells. Stem Cell Rev 2009; 5 (4) 319-327
  • 39 Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 1997; 3 (7) 730-737
  • 40 Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 2003; 100 (7) 3983-3988
  • 41 Bapat SA, Mali AM, Koppikar CB, Kurrey NK. Stem and progenitor-like cells contribute to the aggressive behavior of human epithelial ovarian cancer. Cancer Res 2005; 65 (8) 3025-3029
  • 42 Zhang S, Balch C, Chan MW , et al. Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer Res 2008; 68 (11) 4311-4320
  • 43 Ginestier C, Hur MH, Charafe-Jauffret E , et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 2007; 1 (5) 555-567
  • 44 Haraguchi N, Utsunomiya T, Inoue H , et al. Characterization of a side population of cancer cells from human gastrointestinal system. Stem Cells 2006; 24 (3) 506-513
  • 45 Ishii H, Iwatsuki M, Ieta K , et al. Cancer stem cells and chemoradiation resistance. Cancer Sci 2008; 99 (10) 1871-1877
  • 46 Clarke AR. Wnt signalling in the mouse intestine. Oncogene 2006; 25 (57) 7512-7521
  • 47 Wang S, Yang D, Lippman ME. Targeting Bcl-2 and Bcl-XL with nonpeptidic small-molecule antagonists. Semin Oncol 2003; 30 (5) (Suppl. 16) 133-142
  • 48 Wicha MS, Liu S, Dontu G. Cancer stem cells: an old idea—a paradigm shift. Cancer Res 2006; 66 (4) 1883-1890 ; discussion 1895–1896
  • 49 Hosonuma S, Kobayashi Y, Kojo S , et al. Clinical significance of side population in ovarian cancer cells. Hum Cell 2011; 24 (1) 9-12
  • 50 Lu L, Katsaros D, Wiley A, Rigault de la Longrais IA, Puopolo M, Yu H. Expression of MDR1 in epithelial ovarian cancer and its association with disease progression. Oncol Res 2007; 16 (8) 395-403
  • 51 Ferlay J, Parkin DM, Steliarova-Foucher E. Estimates of cancer incidence and mortality in Europe in 2008. Eur J Cancer 2010; 46 (4) 765-781
  • 52 Piek JM, van Diest PJ, Verheijen RH. Ovarian carcinogenesis: an alternative hypothesis. Adv Exp Med Biol 2008; 622: 79-87
  • 53 Kurrey NK, Jalgaonkar SP, Joglekar AV , et al. Snail and slug mediate radioresistance and chemoresistance by antagonizing p53-mediated apoptosis and acquiring a stem-like phenotype in ovarian cancer cells. Stem Cells 2009; 27 (9) 2059-2068
  • 54 Fraser M, Leung BM, Yan X, Dan HC, Cheng JQ, Tsang BK. p53 is a determinant of X-linked inhibitor of apoptosis protein/Akt-mediated chemoresistance in human ovarian cancer cells. Cancer Res 2003; 63 (21) 7081-7088
  • 55 Fraser M, Bai T, Tsang BK. Akt promotes cisplatin resistance in human ovarian cancer cells through inhibition of p53 phosphorylation and nuclear function. Int J Cancer 2008; 122 (3) 534-546
  • 56 Levine AJ, Puzio-Kuter AM. The control of the metabolic switch in cancers by oncogenes and tumor suppressor genes. Science 2010; 330 (6009) 1340-1344
  • 57 Gustafsson MV, Zheng X, Pereira T , et al. Hypoxia requires notch signaling to maintain the undifferentiated cell state. Dev Cell 2005; 9 (5) 617-628
  • 58 Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 2011; 474 (7353) 609-615
  • 59 Shayesteh L, Lu Y, Kuo WL , et al. PIK3CA is implicated as an oncogene in ovarian cancer. Nat Genet 1999; 21 (1) 99-102
  • 60 Obata K, Morland SJ, Watson RH , et al. Frequent PTEN/MMAC mutations in endometrioid but not serous or mucinous epithelial ovarian tumors. Cancer Res 1998; 58 (10) 2095-2097
  • 61 Esteller M, Silva JM, Dominguez G , et al. Promoter hypermethylation and BRCA1 inactivation in sporadic breast and ovarian tumors. J Natl Cancer Inst 2000; 92 (7) 564-569
  • 62 Kwong J, Lee JY, Wong KK , et al. Candidate tumor-suppressor gene DLEC1 is frequently downregulated by promoter hypermethylation and histone hypoacetylation in human epithelial ovarian cancer. Neoplasia 2006; 8 (4) 268-278
  • 63 Barton CA, Clark SJ, Hacker NF, O'Brien PM. Epigenetic markers of ovarian cancer. Adv Exp Med Biol 2008; 622: 35-51
  • 64 Woloszynska-Read A, James SR, Link PA, Yu J, Odunsi K, Karpf AR. DNA methylation-dependent regulation of BORIS/CTCFL expression in ovarian cancer. Cancer Immun 2007; 7: 21
  • 65 Bram EE, Stark M, Raz S, Assaraf YG. Chemotherapeutic drug-induced ABCG2 promoter demethylation as a novel mechanism of acquired multidrug resistance. Neoplasia 2009; 11 (12) 1359-1370
  • 66 Boylan KL, Misemer B, Derycke MS , et al. Claudin 4 is differentially expressed between ovarian cancer subtypes and plays a role in spheroid formation. Int J Mol Sci 2011; 12 (2) 1334-1358