当前位置: 首页 > 期刊 > 《干细胞学杂志》 > 2004年第1期 > 正文
编号:11342468
Characterization of a Lineage-Negative Stem-Progenitor Cell Population Optimized for Ex Vivo Expansion and Enriched for LTC-IC
http://www.100md.com 《干细胞学杂志》
     a King-George Laboratory, St. George’s Hospital Medical School and Kingston University, London, UK;

    b School of Life Sciences, Kingston University, Kingston upon Thames, UK;

    c Department of Haematology, St. George’s Hospital Medical School, London, UK

    Key Words. Negative selection ? Stem cells ? LTC-IC ? CD34 ? Ex vivo expansion

    Colin P. McGuckin, Ph.D., Reader, School of Life Sciences, Faculty of Science, Kingston University, Penrhyn Road, Kingston upon Thames, Surrey KT1 2EE, United Kingdom. Telephone: 44-797-126-6764; Fax: 44-208-725-0245; e-mail: c.mcguckin@kingston.ac.uk

    ABSTRACT

    Since the 1980s, hematopoietic stem cell transplantation and clinical research have mainly revolved around the cell surface protein, CD34, used as a marker for positive selection of heterogeneous hematopoietic stem and progenitor cells (HSPC) . Although the true role of the CD34 molecule continues to be debated, CD34+ HSPC have been functionally defined as capable of generating progenitor-derived clones in vitro and by their potential in reconstituting the lymphomyelopoietic system in myelocompromised hosts . CD133 was reported as an alternative marker for positive selection methods targeting more primitive HSPC enriched with CD34bright cells . However, we have recently reported that although CD133+ cells had interesting ex vivo expansion potential, they still encompassed cells at various stages of differentiation .

    Several studies have demonstrated that cells that lack CD34 and hematopoietic lineage markers (Lin-/CD34-) could engraft immunocompromised animal hosts and sustain long-term in vivo hematopoiesis . Controversies then rapidly appeared by other groups suggesting limited hematopoietic engraftment potential to Lin-CD34- cells when compared with Lin-CD34+ HSPC . These investigators then hypothesized that small numbers of contaminating CD34+ HSPC could account for CD34- cell engraftment to the marrow . This controversy has been partly reconciled due to papers indicating the reversibility of CD34 expression, which may vary in vivo according to variable engraftment requirements .

    Much of the problem with Lin- cell populations described so far is that they are poorly characterized. Modifications in previously described isolation protocols may also account for the variability of engraftment in immunocompromised animal models . Due to the need to find an optimized population, we have developed a reproducible strategy for primitive cell harvest.

    MATERIALS AND METHODS

    Cell Purification and Phenotypic Characterization

    The mean purity of CD133+ immunomagnetically selected cells was 94.3% ± 0.9% and represented 0.4% ± 0.04% of the original UCB MNC (n = 19). CD133+ cells coexpressed other surface markers as follows: CD34 (93.8% ± 0.9%), CD38 (62.7% ± 3.7%), CD164 (63.8% ± 6.0%), CD162 (69.3% ± 2.6%), and CXCR4 (67.7% ± 1.4%).

    Lin- HSPC were characterized as a discrete cell population representing 0.1% ± 0.02% (n = 11) of original UCB MNC. The Lin- cell subset was negative for a wide range of hematopoietic lineage markers, including CD45, glycophorin-A, CD38, CD7, CD33, CD56, CD16, CD3, and CD2. A proportion of Lin- HSPC nonetheless expressed markers: A) reflecting their immaturity status , such as CD133 (7.0% ± 0.8%), CD34 (14.4% ± 3.6%), intracellular CD34 (16.2% ± 0.6%), and CD164 (16.0% ± 4.1%), or B) that were involved in HSPC migration, adhesion, and homing to the bone marrow (BM) , CXCR4 (48.6% ± 4.0%), and CD162 (96.7% ± 2.0%) (Fig. 1). Our negative isolation protocol appeared to be highly reproducible and isolated a rare primitive Lin- cell population.

    Figure 1. Phenotypic characterization of Lin- cells after isolation from UCB. Lin- cells expressed higher levels of immaturity markers, such as CD133, CD34 (n = 7), intracellular CD34 (n = 2), and CD164 (n = 5), than low-density UCB MNC. Lin- cells also expressed CXCR4 (n = 3) and CD162 (n = 3), both involved in hematopoietic progenitors homing to the BM. Results are expressed as percentage of the Lin- cell population ± SE.

    CD133+ Cells Demonstrated a High Proliferation Potential in Cytokine-Stimulated Liquid Culture

    Over 8 weeks in liquid culture, CD133+ cells proliferated more rapidly and yielded a significantly higher viable total cell number-fold increase (FI) than Lin- HSPC under both K36EG (p < 0.01) and TPOFLK (p < 0.05) stimulations (Fig. 2). In both culture systems, MNC were baseline and exhausted by week 7 of culture. Interestingly, TPOFLK cytokine mix induced a significantly higher viable cell FI when growing CD133+ cells for 8 weeks in culture (TPOFLK FI: 77.10 ± 0.27 x 107 versus K36EG FI: 1.53 ± 0.60 x 107, p < 0.05, n = 4). A similar pattern was observed for Lin- HSPC with a higher proliferation potential under TPOFLK synergism (week 8 FI: 33.20 ± 1.75) when compared with K36EG-stimulated liquid culture in which they stopped growing by week 6 (p < 0.001, n = 4). At first analysis, when compared with Lin- cells, CD133+ had a better ability to produce large cell numbers in TPOFLK-stimulated liquid culture.

    Figure 2. CD133+ cells demonstrated a high proliferation potential in cytokine-stimulated liquid culture. Ex vivo expansion of Lin- cells, CD133+ cells, and MNC over 8 weeks in liquid culture stimulated either by TPOFLK or K36EG. Over 8 weeks, CD133+ cells proliferated more rapidly and yielded significantly higher cumulative CFC counts than Lin- cells under both K36EG (p < 0.01) and TPOFLK (p < 0.05) growth factor stimulations. When compared with K36EG stimulation, TPOFLK synergism induced significantly cumulative CFC counts for both CD133+ cells and Lin- cells. MNC did not expand CFC and exhausted by week 8. Results are expressed as mean ± SE of four separate experiments.

    TPOFLK-Stimulated Liquid Culture of Lin- Cells Maintains a More Primitive Population of HSPC than CD133+ Cells

    The TPOFLK-stimulated liquid culture expansion system consistently maintained a higher proportion of CD133+ and Lin- cell-derived primitive colony-forming cells (CFC) when compared with the effect K36EG cytokine mix on these cells (Fig. 3) (p < 0.05, n = 4). This confirmed the value of TPOFLK synergism when expanding primitive HSPC in liquid culture.

    Figure 3. TPOFLK-stimulated liquid culture of Lin- cells maintains a more primitive population of HSPC than CD133+ cells. Lin-, CD133+, and MNC were respectively grown over 8 weeks in liquid culture stimulated either by TPOFLK or K36EG. At weeks 2, 4, 6, and 8, 104 cells from each cell type (Lin-, CD133+, and MNC), in both TPOFLK and K36EG conditions, were seeded in a clonogenic assay to enumerate CFC capacity. TPOFLK synergism consistently maintained a higher proportion of CD133+ and Lin- cell-derived CFC when compared with K36EG cytokine mix stimulation (p < 0.05). In TPOFLK-stimulated expansion system, Lin- cells produced more CFC/104 seeded cells than CD133+ cells (*p < 0.05). Results are expressed as mean ± SE of four separate experiments.

    After immediate selection, CD133+ cells were enriched for more CFC/104-seeded cells (485 ± 111) when compared with Lin- cells (251 ± 41) CFC and MNC (12 ± 2) (p < 0.05, n = 5). However, in TPOFLK-stimulated liquid culture, Lin- cells consistently maintained a higher proportion of CFC at weeks 4, 6, and 8 than CD133+ cells and MNC (Fig. 3). This difference was particularly significant at 6 and 8 weeks (p < 0.05). Interestingly, in TPOFLK-stimulated liquid culture, the peak in CFC maintenance/expansion from Lin- cells was observed at 6 weeks of expansion (34 ± 8 CFC/104 cells), correlating with their slow proliferation pattern. Under the same conditions, the highest CFC frequency from CD133+ cells appeared earlier at week 4 and gradually faded by week 8 (Fig. 3).

    After selection, both Lin- and CD133+ cells were predominantly enriched with CFC from the erythroid line (56% and 72% of total CFC, respectively). Over 8 weeks, TPOFLK-stimulated liquid cultures gradually favored CFU-granulocyte macrophage (GM) expansion/maintenance, with CFC-erythroid exhausting from week 4 for Lin- cells and week 6 for CD133+ cells. However, from week 2 to week 8 of TPOFLK-supplemented liquid culture, Lin- HSPC yielded a higher ratio of CFC-GM than CD133+ cells (74% versus 58%, respectively, of total scored CFC) (Fig. 4).

    Figure 4. Lin- cells preferentially expand CFC-GM in liquid culture stimulated by TPOFLK. After isolation, both Lin- and CD133+ cells were predominantly enriched with CFC-erythroid. However, when growing both cell subsets in TPOFLK-stimulated culture systems over 8 weeks, a higher proportion of CFC-GM was progressively expanded. Results are expressed as mean percentage of total CFC scored at each time point of four separate experiments.

    Lin- Cells Contain More LTC-IC than CD133+ Cells and MNC

    After 6 weeks in stroma-supported LTC assay, Lin- cells produced significantly more CFC than CD133+ cells (fivefold) and MNC (3,346-fold) (p < 0.05, n = 3) as shown by Figure 5. Taken together, these data showed that Lin- cells were enriched for more LTC-IC than CD133+ cells and MNC.

    Figure 5. Lin- cells produced significantly more CFC in an LTC assay than MNC and CD133+ cells. After 6 weeks in an LTC system, Lin- cells scored a significantly higher cumulative CFC count than MNC and CD133+ cells (p < 0.05). When compared with respective CFC baseline seeded in the LTC assay, Lin- was the only cell population to expand CFC. Results are expressed as mean cumulative CFC count ± SE of three separate experiments.

    DISCUSSION

    Civin CI, Strauss LC, Brovall C et al. Antigenic analysis of hematopoiesis. III. A hematopoietic progenitor cell surface antigen defined by a monoclonal antibody raised against KG-1a cells. J Immunol 1984;133:157–165.

    Tindle RW, Nichols RA, Chan L et al. A novel monoclonal antibody BI-3C5 recognises myeloblasts and non-B non-T lymphoblasts in acute leukaemias and CGL blast crises, and reacts with immature cells in normal bone marrow. Leuk Res 1985;9:1–9.

    Stella CC, Cazzola M, De Fabritiis P et al. CD34-positive cells: biology and clinical relevance. Haematologica 1995;80:367–387.

    Kvalheim G. Clinical use of enriched CD34+ cells. Blood Ther Med 2002;2:57–64.

    Ogawa M. Differentiation and proliferation of hematopoietic stem cells. Blood 1993;81:2844–2853.

    Pasino M, Lanza T, Marotta F et al. Flow cytometric and functional characterization of AC133(+) cells from human umbilical cord blood. Br J Haematol 2000;108:793–800.

    Yin AH, Miraglia S, Zanjani ED et al. AC133, a novel marker for human hematopoietic stem and progenitor cells. Blood 1997;90:5002–5012.

    Forraz N, Pettengell R, Deglesne PA et al. AC133 (+) umbilical cord blood progenitors demonstrate rapid self-renewal and low apoptosis. Br J Haematol 2002;119:516–524.

    Osawa M, Hanada K, Hamada H et al. Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science 1996;273:242–245.

    Bhatia M, Bonnet D, Murdoch B et al. A newly discovered class of human hematopoietic cells with SCID-repopulating activity. Nat Med 1998;4:1038–1045.

    Zanjani ED, Almeida-Porada G, Livingston AG et al. Human bone marrow CD34(-) cells engraft in vivo and undergo multilineage expression that includes giving rise to CD34(+) cells. Exp Hematol 1998;26:353–360.

    Uchida N, Fujisaki T, Eaves AC et al. Transplantable hematopoietic stem cells in human fetal liver have a CD34(+) side population (SP) phenotype. J Clin Invest 2001;108:1071–1077.

    Gao ZG, Fackler MJ, Leung W et al. Human CD34(+) cell preparations contain over 100-fold greater NOD/SCID mouse engrafting capacity than do CD34(-) cell preparations. Exp Hematol 2001;29:910–921.

    Sato T, Laver JH, Ogawa M. Reversible expression of CD34 by murine hematopoietic stem cells. Blood 1999;94:2548–2552.

    Knaan-Shanzer S. No hierarchy within the hemopoietic stem cell compartment. Abstract presented at The New Stem Cells. September 18–19, 2000. Leiden, The Netherlands. In: van Bekkum DW. New stem cell workshop. STEM CELLS 2001;19:260–262.

    Dao MA, Arevalo J, Nolta JA. Reversibility of CD34 expression on human hematopoietic stem cells that retain the capacity for secondary reconstitution. Blood 2003;101:112–118.

    Engelhardt M, Lubbert M, Guo Y. CD34(+) or CD34(-): which is the more primitive? Leukemia 2002;16:1603–1608.

    McGuckin CP, Pearce D, Forraz N et al. Multiparametric analysis of immature cell populations in umbilical cord blood and bone marrow. Eur J Haematol 2003;71:341–350.

    Issaad C, Croisille L, Katz A et al. A murine stromal cell line allows the proliferation of very primitive human CD34++/CD38- progenitor cells in long-term cultures and semisolid assays. Blood 1993;81:2916–2924.

    Bennaceur-Griscelli A, Tourino C, Izac B et al. Murine stromal cells counteract the loss of long-term culture-initiating cell potential induced by cytokines in CD34(+)CD38(low/neg) human bone marrow cells. Blood 1999;94:529–538.

    Pearce D, Pettengell R, Gordon-Smith EC et al. Intracellular analysis of AC133+ cell subsets. Blood 1999;94:132a.

    McGuckin CP, Forraz N, Baradez MO et al. Colocalization analysis of sialomucins CD34 and CD164. STEM CELLS 2003;21:162–170.

    Zannettino AC, Buhring HJ, Niutta S et al. The sialomucin CD164 (MGC-24v) is an adhesive glycoprotein expressed by human hematopoietic progenitors and bone marrow stromal cells that serves as a potent negative regulator of hematopoiesis. Blood 1998;92:2613–2628.

    Mohle R, Bautz F, Rafii S et al. The chemokine receptor CXCR-4 is expressed on CD34(+) hematopoietic progenitors and leukemic cells and mediates transendothelial migration induced by stromal cell-derived factor-1. Blood 1998;91:4523–4530.

    Voermans C, Rood PML, Hordijk PL et al. Adhesion molecules involved in transendothelial migration of human hematopoietic progenitor cells. STEM CELLS 2000;18:435–443.

    Voermans C, Anthony EC, Mul E et al. SDF-1-induced actin polymerization and migration in human hematopoietic progenitor cells. Exp Hematol 2001;29:1456–1464.

    Greenberg AW, Kerr WG, Hammer DA et al. Relationship between selectin-mediated rolling of hematopoietic stem and progenitor cells and progression in hematopoietic development. Blood 2000;95:478–486.

    Sutherland HJ, Lansdorp PM, Henkelman DH et al. Functional characterization of individual human hematopoietic stem cells cultured at limiting dilution on supportive marrow stromal layers. Proc Natl Acad Sci USA 1990;87:3584–3588.

    Hogge DE, Lansdorp PM, Reid D et al. Enhanced detection, maintenance, and differentiation of primitive human hematopoietic cells in cultures containing murine fibroblasts engineered to produce human steel factor, interleukin-3, and granulocyte colony-stimulating factor. Blood 1996;88:3765–3773.

    Pettengell RP, Luft T, Henschler R et al. Direct comparison by limiting dilution analysis of long-term culture initiating cells in human bone marrow, umbilical cord blood and blood stem cells. Blood 1994;84:3653–3659.

    Falzetti F, Ballanti S, De Ioanni M et al. Selection and autologous transplantation of rh-GCSF mobilized CD133+ cells from peripheral blood in patients with indolent Non Hodgkins’ lymphoma under high-dose chemotherapy. Blood 2002;100:831a.

    Tabilio A. Autologous hematopoeitic stem cell transplantation after rhG-CSF mobilization and CD133 selection in patients with multiple Myeloma. Proceeding from Cellular Therapy: New Insights and Novel Strategies. 15–16. Corporate Friday Symposium. 44th Annual Meeting of the American Society of Hematology, 2002.

    Servida F, Soligo D, Caneva L et al. Functional and morphological characterization of immunomagnetically selected CD34(+) hematopoietic progenitor cells. STEM CELLS 1996;14:430–438.

    Majdic O, St?ckl J, Pickl WF et al. Signaling and induction of enhanced cytoadhesiveness via the hematopoietic progenitor cell surface molecule CD34. Blood 1994;83:1226–1234.

    Tada J, Omine M, Suda T et al. A common signaling pathway via syk and lyn tyrosine kinases generated from capping of the sialomucins CD34 and CD43 in immature hematopoietic cells. Blood 1999;93:3723–3735.

    Fujisaki T, Berger MG, Rose-John S et al. Rapid differentiation of a rare subset of adult human Lin(-)CD34(-)CD38(-) cells stimulated by multiple growth factors in vitro. Blood 1999;94:1926–1932.

    Preffer FI, Dombkowski D, Sykes M et al. Lineage-negative side population (SP) cells with restricted hematopoietic capacity circulate in normal human adult blood: immunophenotypic and functional characterization. STEM CELLS 2002;20:417–427.

    Lewis DL, Verfaillie CM. Multi-lineage expansion potential of primitive hematopoietic progenitors: superiority of umbilical cord blood compared to mobilized peripheral blood. Exp Hematol 2000;28:1087–1095.

    Terstappen LW, Safford M, Konemann S et al. Flow cytometric characterization of acute myeloid leukemia: phenotypic heterogeneity at diagnosis. Leukemia 1992;6:70–80.

    Ferrero D, Cherasco C, Ortolano B et al. In vitro growth and quantification of early (CD33-/CD38-) myeloid progenitor cells: stem cell factor requirement and effects of previous chemotherapy. Haematologica 1999;84:390–396.

    Fernandez M, Simon V, Minguell JJ. Production of soluble CD34 by human myeloid cells. Br J Haematol 2000;111:426–431.

    Lapidot T, Kollet O. The essential roles of the chemokine SDF-1 and its receptor CXCR4 in human stem cell homing and repopulation of transplanted immune-deficient NOD/SCID and NOD/SCID/B2m(null) mice. Leukemia 2002;16:1992–2003.

    Levesque JP, Zannettino ACW, Pudney M et al. PSGL-1-mediated adhesion of human hematopoietic progenitors to P-selectin results in suppression of hematopoiesis. Immunity 1999;11:369–378.

    Simmons PJ, Levesque JP, Zannettino AC. Adhesion molecules in haemopoiesis. Baillieres Clin Haematol 1997;10:485–505.

    Chan JY, Watt SM. Adhesion receptors on haematopoietic progenitor cells. Br J Haematol 2001;112:541–557.

    Fackler MJ, Civin CI, May WS. Up-regulation of surface CD34 is associated with protein kinase C-mediated hyperphosphorylation of CD34. J Biol Chem 1992;267:17540–17546.

    Ridings J, Weedon H, Ioannou C et al. Purification of cord blood lymphocytes. J Immunol Methods 1996;195:43–48.

    Forraz N, Pettengell R, McGuckin CP. Haemopoietic and neuroglial progenitors are promoted during cord blood ex-vivo expansion. Br J Haematol 2002;119:888.

    Forraz N, Pettengell R, McGuckin CP. In vitro production of haemopoietic and neuroglial progenitors from umbilical cord blood lineage negative stem cells. Cell Prolif 2002;35:290.

    Sieburg HB, Cho RH, Müller-Sieburg CE. Limiting dilution analysis for estimating the frequency of hematopoietic stem cells: uncertainty and significance. Exp Hematol 2002;30:1436–1443.

    Lemieux ME, Eaves CJ. Identification of properties that can distinguish primitive populations of stromal cell-responsive lympho-myeloid cells from cells that are stromal cell-responsive but lymphoid-restricted and cells that have lympho-myeloid potential but are also capable of competitively repopulating myeloablated recipients. Blood 1996;88:1639–1648.

    Petzer AL, Hogge DE, Lansdorp PM et al. Self-renewal of primitive human hematopoietic cells (long-term-culture-initiating cells) in vitro and their expansion in defined medium. Proc Natl Acad Sci USA 1996;93:1470–1474.

    Brummendorf TH, Dragowska W, Zijlmans JMJM et al. Asymmetric cell divisions sustain long-term hematopoiesis from single-sorted human fetal liver cells. J Exp Med 1998;188:1117–1124.

    Ku?i S, Wessels JT, Bühring HJ et al. Identification of a novel class of human adherent CD34Neg stem cells that give rise to SCID-repopulating cells. Blood 2003;101:869–876.(Nicolas Forraza,b,c, Ruth)