Blood Res.  2015 Dec;50(4):194-203. 10.5045/br.2015.50.4.194.

Hematopoietic stem cell expansion and generation: the ways to make a breakthrough

Affiliations
  • 1Department of Bioscience and Biotechnology, Sejong University, Korea. changkim@sejong.ac.kr
  • 2Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Korea. hema2170@skku.edu
  • 3Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University, Seoul, Korea.

Abstract

Hematopoietic stem cell transplantation (HSCT) is the first field where human stem cell therapy was successful. Flooding interest on human stem cell therapy to cure previously incurable diseases is largely indebted to HSCT success. Allogeneic HSCT has been an important modality to cure various diseases including hematologic malignancies, various non-malignant hematologic diseases, primary immunodeficiency diseases, and inborn errors of metabolism, while autologous HSCT is generally performed to rescue bone marrow aplasia following high-dose chemotherapy for solid tumors or multiple myeloma. Recently, HSCs are also spotlighted in the field of regenerative medicine for the amelioration of symptoms caused by neurodegenerative diseases, heart diseases, and others. Although the demand for HSCs has been growing, their supply often fails to meet the demand of the patients needing transplant due to a lack of histocompatible donors or a limited cell number. This review focuses on the generation and large-scale expansion of HSCs, which might overcome current limitations in the application of HSCs for clinical use. Furthermore, current proof of concept to replenish hematological homeostasis from non-hematological origin will be covered.

Keyword

HSCT; Stem cell; HLA; Blood generation; HSC expansion

MeSH Terms

Bone Marrow
Cell Count
Drug Therapy
Heart Diseases
Hematologic Diseases
Hematologic Neoplasms
Hematopoietic Stem Cell Transplantation
Hematopoietic Stem Cells*
Homeostasis
Humans
Metabolism, Inborn Errors
Multiple Myeloma
Neurodegenerative Diseases
Regenerative Medicine
Stem Cells
Tissue Donors

Figure

  • Fig. 1 HSC proliferation and engraftment ability enhanced by stem cell niche replacing components. Current usage of SR1 and Notch ligand supplemented HSC ex vivo expansion could benefit the HSC engraftment ability and epigenetic regulation.

  • Fig. 2 Schematic view of HSC expansion, non-hematological origin HSC generation, and application. Considering the current shortage of HLA-matched HSCs for patients who need allogeneic HSCT, ex vivo expanded and HLA-matched iPSC-derived HSCs could be the next generation solution. Humanized large animals could be used for the blood product generation applicable to human and for the long-term engraftment studies, because they mimic human immune complexity and might replace the artificial large scale HSC expansion.


Reference

1. Meuwissen HJ, Gatti RA, Terasaki PI, Hong R, Good RA. Treatment of lymphopenic hypogammaglobulinemia and bone-marrow aplasia by transplantation of allogeneic marrow. Crucial role of histocompatiility matching. N Engl J Med. 1969; 281:691–697. PMID: 4186068.
2. Oran B, Shpall E. Umbilical cord blood transplantation: a maturing technology. Hematology Am Soc Hematol Educ Program. 2012; 2012:215–222. PMID: 23233584.
Article
3. Pineault N, Abu-Khader A. Advances in umbilical cord blood stem cell expansion and clinical translation. Exp Hematol. 2015; 43:498–513. PMID: 25970610.
Article
4. Scaradavou A, Brunstein CG, Eapen M, et al. Double unit grafts successfully extend the application of umbilical cord blood transplantation in adults with acute leukemia. Blood. 2013; 121:752–758. PMID: 23223509.
Article
5. Barker JN, Weisdorf DJ, DeFor TE, et al. Transplantation of 2 partially HLA-matched umbilical cord blood units to enhance engraftment in adults with hematologic malignancy. Blood. 2005; 105:1343–1347. PMID: 15466923.
Article
6. Pasquini MC, Zhu X. Current uses and outcomes of hematopoietic stem cell transplantation: 2014 CIBMTR summary slides. Minneapolis, MN: Center for International Blood and Marrow Transplantation Research (CIBMTR);2015. Accessed October 11, 2015. https://www.cibmtr.org/ReferenceCenter/SlidesReports/SummarySlides/pages/index.aspx.
7. de Lima M, McNiece I, Robinson SN, et al. Cord-blood engraftment with ex vivo mesenchymal-cell coculture. N Engl J Med. 2012; 367:2305–2315. PMID: 23234514.
8. Horwitz ME, Chao NJ, Rizzieri DA, et al. Umbilical cord blood expansion with nicotinamide provides long-term multilineage engraftment. J Clin Invest. 2014; 124:3121–3128. PMID: 24911148.
Article
9. Delaney C, Heimfeld S, Brashem-Stein C, Voorhies H, Manger RL, Bernstein ID. Notch-mediated expansion of human cord blood progenitor cells capable of rapid myeloid reconstitution. Nat Med. 2010; 16:232–236. PMID: 20081862.
Article
10. Wagner JE, Brunstein C, McKenna D, et al. Acceleration of umbilical cord blood (UCB) stem engraftment: Results of a phase I clinical trial with stemregenin-1 (SR1) expansion culture. Biol Blood Marrow Transplant. 2015; 21:S48–S49.
Article
11. Silva LC, Ortigosa LC, Benard G. Anti-TNF-α agents in the treatment of immune-mediated inflammatory diseases: mechanisms of action and pitfalls. Immunotherapy. 2010; 2:817–833. PMID: 21091114.
Article
12. Elman JS, Li M, Wang F, Gimble JM, Parekkadan B. A comparison of adipose and bone marrow-derived mesenchymal stromal cell secreted factors in the treatment of systemic inflammation. J Inflamm (Lond). 2014; 11:1. PMID: 24397734.
Article
13. Reardon S, Cyranoski D. Japan stem-cell trial stirs envy. Nature. 2014; 513:287–288. PMID: 25230622.
Article
14. Petersdorf EW, Gooley TA, Anasetti C, et al. Optimizing outcome after unrelated marrow transplantation by comprehensive matching of HLA class I and II alleles in the donor and recipient. Blood. 1998; 92:3515–3520. PMID: 9808542.
Article
15. Hansen JA, Gooley TA, Martin PJ, et al. Bone marrow transplants from unrelated donors for patients with chronic myeloid leukemia. N Engl J Med. 1998; 338:962–968. PMID: 9521984.
Article
16. de Lima M, McMannis J, Gee A, et al. Transplantation of ex vivo expanded cord blood cells using the copper chelator tetraethylenepentamine: a phase I/II clinical trial. Bone Marrow Transplant. 2008; 41:771–778. PMID: 18209724.
Article
17. Taylor CJ, Peacock S, Chaudhry AN, Bradley JA, Bolton EM. Generating an iPSC bank for HLA-matched tissue transplantation based on known donor and recipient HLA types. Cell Stem Cell. 2012; 11:147–152. PMID: 22862941.
Article
18. Wagner JE Jr, Eapen M, Carter S, Wang Y, et al. One-unit versus two-unit cord-blood transplantation for hematologic cancers. N Engl J Med. 2014; 371:1685–1694. PMID: 25354103.
Article
19. Weidner CI, Walenda T, Lin Q, et al. Hematopoietic stem and progenitor cells acquire distinct DNA-hypermethylation during in vitro culture. Sci Rep. 2013; 3:3372. PMID: 24284763.
Article
20. Xie J, Zhang C. Ex vivo expansion of hematopoietic stem cells. Sci China Life Sci. 2015; 58:839–853. PMID: 26246379.
Article
21. Chaurasia P, Gajzer DC, Schaniel C, D'Souza S, Hoffman R. Epigenetic reprogramming induces the expansion of cord blood stem cells. J Clin Invest. 2014; 124:2378–2395. PMID: 24762436.
Article
22. Young JC, Wu S, Hansteen G, et al. Inhibitors of histone deacetylases promote hematopoietic stem cell self-renewal. Cytotherapy. 2004; 6:328–336. PMID: 16146885.
Article
23. Araki H, Yoshinaga K, Boccuni P, Zhao Y, Hoffman R, Mahmud N. Chromatin-modifying agents permit human hematopoietic stem cells to undergo multiple cell divisions while retaining their repopulating potential. Blood. 2007; 109:3570–3578. PMID: 17185465.
Article
24. Peled T, Shoham H, Aschengrau D, et al. Nicotinamide, a SIRT1 inhibitor, inhibits differentiation and facilitates expansion of hematopoietic progenitor cells with enhanced bone marrow homing and engraftment. Exp Hematol. 2012; 40:342–355.e1. PMID: 22198152.
25. Lang J, Weiss N, Freed BM, Torres RM, Pelanda R. Generation of hematopoietic humanized mice in the newborn BALB/c-Rag2null Il2rγnull mouse model: a multivariable optimization approach. Clin Immunol. 2011; 140:102–116. PMID: 21536497.
26. Berges BK, Wheat WH, Palmer BE, Connick E, Akkina R. HIV-1 infection and CD4 T cell depletion in the humanized Rag2-/-gamma c-/- (RAG-hu) mouse model. Retrovirology. 2006; 3:76. PMID: 17078891.
Article
27. Matsuoka Y, Sumide K, Kawamura H, et al. Human cord blood-derived primitive CD34-negative hematopoietic stem cells (HSCs) are myeloid-biased long-term repopulating HSCs. Blood Cancer J. 2015; 5:e290. PMID: 25768404.
Article
28. Tanner A, Taylor SE, Decottignies W, Berges BK. Humanized mice as a model to study human hematopoietic stem cell transplantation. Stem Cells Dev. 2014; 23:76–82. PMID: 23962058.
Article
29. Brehm MA, Shultz LD, Luban J, Greiner DL. Overcoming current limitations in humanized mouse research. J Infect Dis. 2013; 208(Suppl 2):S125–S130. PMID: 24151318.
Article
30. Lee K, Kwon DN, Ezashi T, et al. Engraftment of human iPS cells and allogeneic porcine cells into pigs with inactivated RAG2 and accompanying severe combined immunodeficiency. Proc Natl Acad Sci U S A. 2014; 111:7260–7265. PMID: 24799706.
Article
31. Larochelle A, Vormoor J, Hanenberg H, et al. Identification of primitive human hematopoietic cells capable of repopulating NOD/SCID mouse bone marrow: implications for gene therapy. Nat Med. 1996; 2:1329–1337. PMID: 8946831.
Article
32. Ishii M, Matsuoka Y, Sasaki Y, et al. Development of a high-resolution purification method for precise functional characterization of primitive human cord blood-derived CD34-negative SCID-repopulating cells. Exp Hematol. 2011; 39:203–213.e1. PMID: 21112372.
Article
33. Lu R, Neff NF, Quake SR, Weissman IL. Tracking single hematopoietic stem cells in vivo using high-throughput sequencing in conjunction with viral genetic barcoding. Nat Biotechnol. 2011; 29:928–933. PMID: 21964413.
Article
34. Kim C. iPSC technology-Powerful hand for disease modeling and therapeutic screen. BMB Rep. 2015; 48:256–265. PMID: 25104399.
Article
35. Kim H, Kim JS. A guide to genome engineering with programmable nucleases. Nat Rev Genet. 2014; 15:321–334. PMID: 24690881.
Article
36. Maeder ML, Linder SJ, Cascio VM, Fu Y, Ho QH, Joung JK. CRISPR RNA-guided activation of endogenous human genes. Nat Methods. 2013; 10:977–979. PMID: 23892898.
Article
37. Kim C. Disease modeling and cell based therapy with iPSC: future therapeutic option with fast and safe application. Blood Res. 2014; 49:7–14. PMID: 24724061.
Article
38. Ding Q, Lee YK, Schaefer EA, et al. A TALEN genome-editing system for generating human stem cell-based disease models. Cell Stem Cell. 2013; 12:238–251. PMID: 23246482.
Article
39. Gaj T, Gersbach CA, Barbas CF 3rd. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol. 2013; 31:397–405. PMID: 23664777.
Article
40. Joung JK, Sander JD. TALENs: a widely applicable technology for targeted genome editing. Nat Rev Mol Cell Biol. 2013; 14:49–55. PMID: 23169466.
Article
41. Schwartz SD, Hubschman JP, Heilwell G, et al. Embryonic stem cell trials for macular degeneration: a preliminary report. Lancet. 2012; 379:713–720. PMID: 22281388.
Article
42. Song WK, Park KM, Kim HJ, et al. Treatment of macular degeneration using embryonic stem cell-derived retinal pigment epithelium: preliminary results in Asian patients. Stem Cell Reports. 2015; 4:860–872. PMID: 25937371.
Article
43. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006; 126:663–676. PMID: 16904174.
Article
44. Kanemura H, Go MJ, Shikamura M, et al. Tumorigenicity studies of induced pluripotent stem cell (iPSC)-derived retinal pigment epithelium (RPE) for the treatment of age-related macular degeneration. PLoS One. 2014; 9:e85336. PMID: 24454843.
Article
45. Schlaeger TM, Daheron L, Brickler TR, et al. A comparison of non-integrating reprogramming methods. Nat Biotechnol. 2015; 33:58–63. PMID: 25437882.
Article
46. Geron Corporation. World's first clinical trial of human embryonic stem cell therapy cleared. Regen Med. 2009; 4:161. PMID: 19322956.
47. Hanna J, Wernig M, Markoulaki S, et al. Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin. Science. 2007; 318:1920–1923. PMID: 18063756.
Article
48. Wang L, Li L, Menendez P, Cerdan C, Bhatia M. Human embryonic stem cells maintained in the absence of mouse embryonic fibroblasts or conditioned media are capable of hematopoietic development. Blood. 2005; 105:4598–4603. PMID: 15718421.
Article
49. Woll PS, Grzywacz B, Tian X, et al. Human embryonic stem cells differentiate into a homogeneous population of natural killer cells with potent in vivo antitumor activity. Blood. 2009; 113:6094–6101. PMID: 19365083.
Article
50. Galic Z, Kitchen SG, Kacena A, et al. T lineage differentiation from human embryonic stem cells. Proc Natl Acad Sci U S A. 2006; 103:11742–11747. PMID: 16844782.
Article
51. Carpenter L, Malladi R, Yang CT, et al. Human induced pluripotent stem cells are capable of B-cell lymphopoiesis. Blood. 2011; 117:4008–4011. PMID: 21343609.
Article
52. Knorr DA, Ni Z, Hermanson D, et al. Clinical-scale derivation of natural killer cells from human pluripotent stem cells for cancer therapy. Stem Cells Transl Med. 2013; 2:274–283. PMID: 23515118.
Article
53. Ledran MH, Krassowska A, Armstrong L, et al. Efficient hematopoietic differentiation of human embryonic stem cells on stromal cells derived from hematopoietic niches. Cell Stem Cell. 2008; 3:85–98. PMID: 18593561.
Article
54. Gori JL, Butler JM, Chan YY, et al. Vascular niche promotes hematopoietic multipotent progenitor formation from pluripotent stem cells. J Clin Invest. 2015; 125:1243–1254. PMID: 25664855.
Article
55. Ramos-Mejía V, Navarro-Montero O, Ayllón V, et al. HOXA9 promotes hematopoietic commitment of human embryonic stem cells. Blood. 2014; 124:3065–3075. PMID: 25185710.
Article
56. Real PJ, Ligero G, Ayllon V, et al. SCL/TAL1 regulates hematopoietic specification from human embryonic stem cells. Mol Ther. 2012; 20:1443–1453. PMID: 22491213.
Article
57. Doulatov S, Vo LT, Chou SS, et al. Induction of multipotential hematopoietic progenitors from human pluripotent stem cells via respecification of lineage-restricted precursors. Cell Stem Cell. 2013; 13:459–470. PMID: 24094326.
Article
58. Ran D, Shia WJ, Lo MC, et al. RUNX1a enhances hematopoietic lineage commitment from human embryonic stem cells and inducible pluripotent stem cells. Blood. 2013; 121:2882–2890. PMID: 23372166.
Article
59. Amabile G, Welner RS, Nombela-Arrieta C, et al. In vivo generation of transplantable human hematopoietic cells from induced pluripotent stem cells. Blood. 2013; 121:1255–1264. PMID: 23212524.
Article
60. Suzuki N, Yamazaki S, Yamaguchi T, et al. Generation of engraftable hematopoietic stem cells from induced pluripotent stem cells by way of teratoma formation. Mol Ther. 2013; 21:1424–1431. PMID: 23670574.
Article
61. Szabo E, Rampalli S, Risueño RM, et al. Direct conversion of human fibroblasts to multilineage blood progenitors. Nature. 2010; 468:521–526. PMID: 21057492.
Article
62. Sandler VM, Lis R, Liu Y, et al. Reprogramming human endothelial cells to haematopoietic cells requires vascular induction. Nature. 2014; 511:312–318. PMID: 25030167.
Article
63. Riddell J, Gazit R, Garrison BS, et al. Reprogramming committed murine blood cells to induced hematopoietic stem cells with defined factors. Cell. 2014; 157:549–564. PMID: 24766805.
Article
64. Batta K, Florkowska M, Kouskoff V, Lacaud G. Direct reprogramming of murine fibroblasts to hematopoietic progenitor cells. Cell Rep. 2014; 9:1871–1884. PMID: 25466247.
Article
65. Lu SJ, Li F, Yin H, et al. Platelets generated from human embryonic stem cells are functional in vitro and in the microcirculation of living mice. Cell Res. 2011; 21:530–545. PMID: 21221130.
Article
66. Nakamura S, Takayama N, Hirata S, et al. Expandable megakaryocyte cell lines enable clinically applicable generation of platelets from human induced pluripotent stem cells. Cell Stem Cell. 2014; 14:535–548. PMID: 24529595.
Article
67. Kennedy M, Awong G, Sturgeon CM, et al. T lymphocyte potential marks the emergence of definitive hematopoietic progenitors in human pluripotent stem cell differentiation cultures. Cell Rep. 2012; 2:1722–1735. PMID: 23219550.
Article
68. Liu Z, Lu SJ, Lu Y, et al. Transdifferentiation of Human Hair Follicle Mesenchymal Stem Cells into Red Blood Cells by OCT4. Stem Cells Int. 2015; 2015:389628. PMID: 25755671.
Article
69. Vodyanik MA, Bork JA, Thomson JA, Slukvin II. Human embryonic stem cell-derived CD34+ cells: efficient production in the coculture with OP9 stromal cells and analysis of lymphohematopoietic potential. Blood. 2005; 105:617–626. PMID: 15374881.
Article
70. Wada H, Kojo S, Kusama C, et al. Successful differentiation to T cells, but unsuccessful B-cell generation, from B-cell-derived induced pluripotent stem cells. Int Immunol. 2011; 23:65–74. PMID: 21135032.
Article
71. Sturgeon CM, Ditadi A, Awong G, Kennedy M, Keller G. Wnt signaling controls the specification of definitive and primitive hematopoiesis from human pluripotent stem cells. Nat Biotechnol. 2014; 32:554–561. PMID: 24837661.
Article
72. Chen U. Do we have a workable clinical protocol for differentiating lympho-hematopoietic stem cells from the source of embryonic stem cells and induced pluripotent stem cells in culture. Scand J Immunol. 2014; 80:247–249. PMID: 25041639.
Article
73. Gori JL, Chandrasekaran D, Kowalski JP, et al. Efficient generation, purification, and expansion of CD34(+) hematopoietic progenitor cells from nonhuman primate-induced pluripotent stem cells. Blood. 2012; 120:e35–e44. PMID: 22898598.
Article
Full Text Links
  • BR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr