DOI QR코드

DOI QR Code

Senescence Effects of Angelica sinensis Polysaccharides on Human Acute Myelogenous Leukemia Stem and Progenitor Cells

  • Liu, Jun (Histology and Embryology, Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University) ;
  • Xu, Chun-Yan (Histology and Embryology, Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University) ;
  • Cai, Shi-Zhong (Histology and Embryology, Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University) ;
  • Zhou, Yue (Histology and Embryology, Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University) ;
  • Li, Jing (Histology and Embryology, Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University) ;
  • Jiang, Rong (Histology and Embryology, Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University) ;
  • Wang, Ya-Ping (Histology and Embryology, Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University)
  • Published : 2013.11.30

Abstract

Leukemia stem cells (LSCs) play important roles in leukemia initiation, progression and relapse, and thus represent a critical target for therapeutic intervention. Hence, it is extremely urgent to explore new therapeutic strategies directly targeting LSCs for acute myelogenous leukemia (AML) therapy. We show here that Angelica sinensis polysaccharide (ASP), a major active component in Dong quai (Chinese Angelica sinensis), effectively inhibited human AML $CD34^+CD38^-$ cell proliferation in vitro culture in a dose-dependent manner while sparing normal hematopoietic stem and progenitor cells at physiologically achievable concentrations. Furthermore, ASP exerted cytotoxic effects on AML K562 cells, especially LSC-enriched $CD34^+CD38^-$ cells. Colony formation assays further showed that ASP significantly suppressed the formation of colonies derived from AML $CD34^+CD38^-$ cells but not those from normal $CD34^+CD38^-$ cells. Examination of the underlying mechanisms revealed that ASP induced $CD34^+CD38^-$ cell senescence, which was strongly associated with a series of characteristic events, including up-regulation of p53, p16, p21, and Rb genes and changes of related cell cycle regulation proteins P16, P21, cyclin E and CDK4, telomere end attrition as well as repression of telomerase activity. On the basis of these findings, we propose that ASP represents a potentially important agent for leukemia stem cell-targeted therapy.

Keywords

References

  1. Ben-Porath I and RA Weinberg (2005). The signals and pathways activating cellular senescence. Int J Biochem Cell Biol, 37, 961-76. https://doi.org/10.1016/j.biocel.2004.10.013
  2. Blair A, Hogge DE, Ailles LE, et al (1997). Lack of expression of Thy-1 (CD90) on acute myeloid leukemia cells with long-term proliferative ability in vitro and in vivo. Blood, 89, 3104-12.
  3. Blasco MA (2007). The epigenetic regulation of mammalian telomeres. Nature Rev Gene, 8, 299-309. https://doi.org/10.1038/nrg2047
  4. Bonnet D and JE Dick (1997). Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nature med, 3, 730-7. https://doi.org/10.1038/nm0797-730
  5. Cao W, Li XQ, Wang X, et al (2010). A novel polysaccharide, isolated from Angelica sinensis (Oliv.) Diels induces the apoptosis of cervical cancer HeLa cells through an intrinsic apoptotic pathway. Phytomedicine, 17, 598-605. https://doi.org/10.1016/j.phymed.2009.12.014
  6. Cao W, Li XQ, Wang X, et al (2010). Characterizations and anti-tumor activities of three acidic polysaccharides fromAngelica sinensis (Oliv.) Diels. Int J Biol Macromol, 46, 115-22. https://doi.org/10.1016/j.ijbiomac.2009.11.005
  7. Carnero, A (2013). Markers of Cellular Senescence. Cell Senescence, Springer, 63-81.
  8. Dimri GP, Lee X, Basile G, et al (1995). A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci U S A, 92, 9363-7. https://doi.org/10.1073/pnas.92.20.9363
  9. Gunes C and KL Rudolph (2013). The Role of Telomeres in Stem Cells and Cancer. Cell, 152, 390-3. https://doi.org/10.1016/j.cell.2013.01.010
  10. Gunn EJ, Williams JT, Huynh DT, et al (2011). The natural products parthenolide and andrographolide exhibit anti-cancer stem cell activity in multiple myeloma. Leuk Lymphoma, 52, 1085-97. https://doi.org/10.3109/10428194.2011.555891
  11. Guzman ML, Rossi RM, Karnischky L et al (2005). The sesquiterpene lactone parthenolide induces apoptosis of human acute myelogenous leukemia stem and progenitor cells. Blood, 105, 4163-9. https://doi.org/10.1182/blood-2004-10-4135
  12. Guzman ML, Rossi RM, Neelakantan S et al (2007). An orally bioavailable parthenolide analog selectively eradicates acute myelogenous leukemia stem and progenitor cells. Blood, 110, 4427-35. https://doi.org/10.1182/blood-2007-05-090621
  13. Herbig U, Jobling WA, Chen BP, et al (2004). Telomere shortening triggers senescence of human cells through a pathway involving ATM, p53, and p21 (CIP1), but not p16 (INK4a). Mol Cell, 14, 501-13. https://doi.org/10.1016/S1097-2765(04)00256-4
  14. Hoffmeyer K, A Raggioli, Rudloff S et al (2012). Wnt/{beta}-Catenin Signaling Regulates Telomerase in Stem Cells and Cancer Cells. Science, 336, 1549-54. https://doi.org/10.1126/science.1218370
  15. Hwang ES, Yoon G, Kang HT (2009). A comparative analysis of the cell biology of senescence and aging. Cell Mol Life Sci, 66, 2503-24. https://doi.org/10.1007/s00018-009-0034-2
  16. Insinga A, Cicalese A, Faretta M, et al (2013). DNA damage in stem cells activates p21, inhibits p53, and induces symmetric self-renewing divisions. Proc Natl Acad Sci U S A, 110, 3931-6. https://doi.org/10.1073/pnas.1213394110
  17. Janzen V, Forkert R, Fleming HE et al (2006). Stem-cell ageing modified by the cyclin-dependent kinase inhibitor p16INK4a. Nature, 443, 421-6. https://doi.org/10.1038/nature05159
  18. Jordan CT, Upchurch D, Szilvassy SJ et al (2000). The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells. Leukemia, 14, 1777-84. https://doi.org/10.1038/sj.leu.2401903
  19. Liu J, Cai SZ, Zhou Y, et al (2012). Senescence as A Consequence of Ginsenoside Rg1 response on k562 human leukemia cell line. Asian Pac J Cancer Prev, 13, 6191-6. https://doi.org/10.7314/APJCP.2012.13.12.6191
  20. Martinez P and MA Blasco (2011). Telomeric and extra-telomeric roles for telomerase and the telomere-binding proteins. Nat Rev Cancer, 11, 161-76. https://doi.org/10.1038/nrc3025
  21. Masgras I and S Macip (2013). p21 Mediates Senescence by a Mechanism Involving Accumulation of Reactive Oxygen Species. Springer, 153-67.
  22. Narita M, N.nez S, Heard E, et al (2003). Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell, 113, 703-16. https://doi.org/10.1016/S0092-8674(03)00401-X
  23. Phalke S, Mzoughi S, Bezzi M, et al (2012). p53-Independent regulation of p21Waf1/Cip1 expression and senescence by PRMT6. Nucleic Acids Res, 40, 9534-42. https://doi.org/10.1093/nar/gks858
  24. Provinciali M, Cardelli M, Marchegiani F, Pierpaoli E (2013). Impact of Cellular Senescence in Aging and Cancer. Curr Pharm Des, 19, 1699-709.
  25. Quelle DE, Cheng M, Ashmun RA, Sherr CJ (1997). Cancer-associated mutations at the INK4a locus cancel cell cycle arrest by p16INK4a but not by the alternative reading frame protein p19ARF. Proc Natl Acad Sci U S A, 94, 669-73. https://doi.org/10.1073/pnas.94.2.669
  26. Rhee SG (2006). Cell signaling. H202, a necessary evil for ceil signalling. Sciene, 312, 1882-3. https://doi.org/10.1126/science.1130481
  27. Severino J, Allen RG, Balin S, et al (2000). Is $\beta$-Galactosidase Staining a Marker of Senescence in vitro and in vivo?. Exp Cell Res, 257, 162-71. https://doi.org/10.1006/excr.2000.4875
  28. Shang P, Qian AR, Yang TH, et al (2003). Experimental study of anti-tumor effects of polysaccharides from Angelica sinensis. World J Gastroenterol, 9, 1963-7. https://doi.org/10.3748/wjg.v9.i9.1963
  29. Sherr, CJ (2012). Ink4. Arf locus in cancer and aging. Wiley Interdiscip Rev Dev Biol, 1, 731-41. https://doi.org/10.1002/wdev.40
  30. Signer RA and SJ Morrison (2013). Mechanisms that Regulate Stem Cell Aging and Life Span. Cell Stem Cell, 12, 152-65. https://doi.org/10.1016/j.stem.2013.01.001
  31. Song Z, J Zhang, et al (2012). Telomere dysfunctional environment induces loss of quiescence and inherent impairments of hematopoietic stem cell function. Aging Cell, 11, 449-55 https://doi.org/10.1111/j.1474-9726.2012.00802.x
  32. Sperka T, J Wang, et al (2012). DNA damage checkpoints in stem cells, ageing and cancer. Nat Rev Mol Cell Biol, 13, 579-90. https://doi.org/10.1038/nrm3420
  33. Tsai NM, SZ Lin, et al (2005). The antitumor effects of Angelica sinensis on malignant brain tumors in vitro and in vivo. Clin Cancer Res, 11, 3475-84. https://doi.org/10.1158/1078-0432.CCR-04-1827
  34. Zhang H, JQ Mi, et al (2013). Preferential eradication of acute myelogenous leukemia stem cells by fenretinide. Proc Natl Acad Sci U S A, 110, 5606-11 https://doi.org/10.1073/pnas.1302352110
  35. Zong A, H Cao, et al (2012). Anticancer polysaccharides from natural resources: A review of recent research. Carbohydr Polym, 90, 1395-410 https://doi.org/10.1016/j.carbpol.2012.07.026

Cited by

  1. Elevated Serum Ferritin Levels in Patients with Hematologic Malignancies vol.15, pp.15, 2014, https://doi.org/10.7314/APJCP.2014.15.15.6099
  2. Shelterin Proteins and Cancer vol.16, pp.8, 2015, https://doi.org/10.7314/APJCP.2015.16.8.3085
  3. Advances on Bioactive Polysaccharides from Medicinal Plants vol.56, pp.sup1, 2016, https://doi.org/10.1080/10408398.2015.1069255
  4. Systems-Based Interactome Analysis for the Hematopoiesis Effect of Angelicae Sinensis Radix: Regulated Network of Cell Proliferation towards Hemopoiesis pp.1993-0402, 2018, https://doi.org/10.1007/s11655-018-3003-5
  5. on some related indexes of free radical and energy metabolism after strenuous exercise in rats vol.218, pp.1755-1315, 2019, https://doi.org/10.1088/1755-1315/218/1/012147