Delayed Platelet Recovery Following Cord Blood Transplantation: Insights from a Mouse Model.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1727-1727
Author(s):  
Tao Du ◽  
Camelia Iancu-Rubin ◽  
George F. Atweh ◽  
Rona Singer Weinberg

Abstract Umbilical cord blood (CB) is an important source of hematopoietic stem cells for stem cell transplantation and is being used with increasing frequency. A major concern related to clinical CB transplantation is the long delay in platelet recovery. Megakaryopoiesis is characterized by the acquisition of lineage-specific markers (e.g. CD41) during the early stages that is followed by polyploidization (DNA content > 4N) during the later stages of megakaryopoiesis. Using a newborn blood (NB) model of CB transplantation that was previously developed in our laboratory, we asked whether the delay in platelet recovery is a result of a decrease in the rate of megakaryocyte production or a delay in their maturation. C57BL/6 mice were transplanted with either murine adult bone marrow (BM) cells or murine new-born blood (NB) cells following lethal irradiation. We had previously shown that the concentration of Lin-Sca-1+c-Kit+ stem cells in murine adult BM was approximately 3 times higher than that in NB. To correct for this difference in stem cell concentration, irradiated mice were transplanted with either 0.5 ×106 BM cells or 2×106 NB cells. Platelet counts at 2 and 4 weeks were lower in mice transplanted with NB cells than in mice transplanted with BM cells (Table 1). Interestingly, the platelet counts became comparable in NB and BM recipients at 8 weeks post-transplantation. We compared the ability of BM cells from both NB and BM recipients to form CFU-Meg colonies in methylcellulose. At 2 and 4 weeks post-transplantation, BM cultures derived from NB recipients generated fewer CFU-Meg colonies than cultures from BM recipients (Table 1). Interestingly, after 8 weeks, the numbers of CFU-Meg from both stem cell sources were similar. We also compared the ability of BM cells from NB and BM recipients to differentiate into megakaryocytes in liquid culture, using CD41 expression and polyploidy as markers of megakaryocytic maturation. At 2 weeks post-transplantation, cultures from NB recipients generated 13% CD41+ cells whereas cultures from BM recipients generated 22% CD41+ cells. However, by 4 weeks post-transplantation, the numbers of CD41+ cells were similar in cultures derived from NB recipients and BM recipients (Table 1). Moreover, at 2 and 4 weeks post-transplantation, there were fewer polyploid cells in liquid cultures from NB recipients compared to BM recipients (Table 1). The lower number of polyploid cells was commensurate with the lower number of CD41+ cells. This suggests that the rate of maturation of megakaryocyte is similar in NB and BM. In conclusion, our studies show that CB stem cells generate megakaryocytic progenitors at a slower rate than BM stem cells and that the delay in platelet recovery is not a result of a delay in the maturation of megakaryocytic progenitors. Thus, in order to increase the rate of platelet recovery following CB transplantation, the focus should be on enhancing the rate of production of megakaryocytic progenitors from hematopoietic stem cells. Table 1. Platelet(×103/μl) CFU-Meg(Colonies/1 × 105 Cells) CD41(%) Ploidy(% >4N DNAContent) normal BM(n=5) 1200±120 35.5±5 55±5 27±3 2 weeks NBT-2M(n=5) 190± 38 10±2 13±2 7.5±1 BMT-0.5M(n=5) 400±50 17±4 22±1 11±2 4 weeks NBT-2M(n=5) 550±59 18±2 26±2 14.5±2 BMT-0.5M(n=5) 730±110 23±3 27±1 18±3 8 weeks NBT-2M(n=5) 930±115 39±7 N/A N/A BMT-0.5M(n=5) 970±130 40±4 N/A N/A

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 35-35 ◽  
Author(s):  
Ewa K. Zuba-Surma ◽  
Izabela Klich ◽  
Marcin Wysoczynski ◽  
Nicholas J Greco ◽  
Mary J. Laughlin ◽  
...  

Abstract Abstract 35 Recently, we identified in umbilical cord blood (UCB) a population of very small embryonic/epiblast-like (VSEL) stem cells (Leukemia 2007;21:297–303) that are i) smaller than erythrocytes, ii) SSEA-4+/Oct-4+/CD133+/CXCR4+/Lin−/CD45−, iii) respond to SDF-1 gradient and iv) possess large nuclei containing primitive euchromatin. We have demonstrated in vitro that UCB-derived VSELs did not reveal hematopoietic activity freshly after isolation, but grow hematopoietic colonies following co-culture/activation over OP-9 cells. To investigate the hierarchy of UCB-derived, CD45 negative VSELs, we employed staining with Aldefluor - detecting aldehyde dehydrogenase (ALDH), the enzyme expressed in primitive hematopoietic cells. Subsequently, we sorted CD45−/CD133+/ALDHhigh and CD45−/CD133+/ALDHlow sub-fractions of VSELs from UCB samples and established that freshly sorted from UCB VSELs in contrast to sorted CD45+/ CD133+/ALDHhigh and CD45+/CD133+/ALDHlow hematopoietic stem cells (HSC) did not grow colonies in vitro. However, when CD45− VSELs were activated/expanded over OP-9 stroma cells, they exhibit hematopoietic potential and grew in routine methylcellulose cultures hematopoietic colonies composed of CD45+ cells. Interestingly, while CD45−/CD133+/ALDHhigh VSELs gave raise to hematopoietic colonies after the first replating, the formation of colonies by CD45−/CD133+/ALDHlow VSELs was somehow delayed, what suggest that they needed more time to acquire hematopoietic commitment. Thus our in vitro data indicate that both populations of CD45− cells may acquire hematopoietic potential; however hematopoietic specification is delayed for CD45−/CD133+/ALDHlow cells, suggesting their more primitive nature. In parallel, real time PCR analysis confirmed that while freshly isolated CD45−/CD133+/ALDHhigh VSELs express more hematopoietic transcripts (e.g., c-myb, 80.2±27.4 fold difference), CD45−/CD133+/ALDHlow exhibit higher levels of pluripotent stem cell markers (e.g., Oct-4, 119.5±15.5 fold difference as compared to total UCB mononuclear cells) (Figure 1 panel A). Next hematopoietic potential of UCB-derived VSELs was tested in vivo after transplantation into NOD/SCID mice (Figure 1 panel B and C). We noticed that both CD45−/CD133+/ALDHhigh and CD45−/CD133+/ALDHlow VSELs, give rise to human lympho-hematopoietic chimerism in lethally irradiated NOD/SCID mice as assayed 4–6 weeks after transplantation. The level of human hematopoietic CD45+ cells in murine peripheral blood (PB), bone marrow (BM) and spleen (SP) were comparable for both transplanted UCB-VSELs fractions - 7.1±2.9% (PB), 23.2±0.2% (SP) and 25.2±1.0% (BM). In conclusion, our data suggest that freshly isolated very small CD45 negative UCB-VSELs are depleted from clonogeneic progenitors, however they are highly enriched for primitive HSC. Based on our in vitro and in vivo data we postulate following hierarchy of hematopoietic stem cells in UCB (from most primitive to more differentiated) i) CD45−/CD133+/ALDHlow, ii) CD45−/CD133+/ALDHhigh , iii) CD45+/CD133+/ALDHlow and iv) CD45−/CD133+/ALDHhigh. We also postulate that as we have already shown for murine BM-derived VSELs, human UCB-derived CD45 negative VSELs correspond to a population of most primitive long term repopulating HSC (LT-HSC). Of note, we also found that currently employed, routine UCB processing strategies may lead up to ∼50% unwanted loss of these small cells that are endowed with such remarkable hematopoietic activity! Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4054-4054
Author(s):  
Aaron Victor ◽  
Mary J. Laughlin ◽  
Marcie R. Finney ◽  
Nicholas J. Greco

Abstract There is a significant unmet need for novel therapeutic treatments for patients presenting with chronic ischemic conditions such as coronary artery disease and diabetes. Revascularization measures, such as infusions with endothelial progenitor cells (EPC) characterized by the expression of early hematopoietic stem cell markers, hold significant potential in treating these patients. Pre-clinical and clinical studies using transplanted EPC to restore blood flow and improve cardiac function in animal models of ischemia have proven effective. Recent studies have used bone marrow mononuclear cells while some more recent studies have focused on enriched stem cell treatments, such as purified bone marrow hematopoietic stem cell (HSC) CD34+/133+ cell populations, in patients with coronary artery ischemia. In this study, the hypothesis to be tested was that umbilical cord blood-derived hematopoietic stem cells (CD34+/CD133+) cells may augment the formation and stability of angiogenic networks of cord-like structures derived from umbilical vein endothelial cells (HUVEC) cultured in growth factor-reduced Matrigel (GFR MG) assays. Umbilical cord blood MNC were isolated with ficoll and separated into HSC CD34+/133+ and CD34−/133− fractions. Positive fractions were flow cytometry, sorted for HSC, and stained with the lipophilic fluorescent red dye CM-DiI and the HUVEC were stained with the lipophilic fluorescent green dye Oregon Green. HUVEC alone or HSC and HUVEC were then co-cultured under hypoxic conditions (1% O2) on the GFR MG in 96 well plates. Cells were photographed with a fluorescent microscope at 16, 48, and 72 hours. Transwell experiments (0.4μm pores) were also performed with HSC CD34+/133+ and CD34−/133− fractions prepared and suspended in transwells above HUVEC plated on GFR MG on bottom wells. The presence of both HSC CD34+/133+ and CD34−/133− fractions increased the numbers of nodes (branch points of structures) and allowed the structures to persist when observed over three days (a representative experiment of N =3) (Table): Day 1 Day 1 Day 2 Day 2 Day 3 Day 3 Node # % Total Node # % Total Node # % Total HUVEC 11.6 ± 4.9 100 1.3 ± 1.2 9.2 0.33 ± 0.58 2.2 HUVEC + HSC CD34+/133+ 17.3 ± 9.2 100 6.3 ± 4.5 35.3 4.7 ± 5.5 21.4 HUVEC + HSC CD34−/133− 34 ± 13.2 100 19.7 ± 2.5 61.6 10 ± 3.6 29.8 The HSC CD34−/133− fraction resulted in a greater increase in node formation than the HSC CD34+/133+ and both fractions stimulated significant persistence in formed structures. In addition, CM-Dil labeled cells were localized at nodes points. Results with the transwell assay demonstrated that when either HSC CD34+/133+ or CD34−/133− fractions were suspended above HUVEC, augmentation of the formation of cord-like structures was not observed. In summary, both umbilical cord blood-derived HSC CD34+/133+ and CD34−/133− fractions possess properties that augment the formation of angiogenic structures. We observed that the number of nodes are greater in the presence of both HSC CD34+/133+ and CD34−/133− fractions than with HUVEC alone. The transwell experiment suggested that cell-to-cell interactions are necessary for augmentation of the cord structures. In future studies, we will address the mechanism of intercellular interactions that result in the augmentation of cord-like structures and which particular subpopulations within cord blood, both from HSC CD34+/133+ and CD34−/133− fractions are required for augmentation of structure formation.


Blood ◽  
1996 ◽  
Vol 87 (12) ◽  
pp. 5370-5378 ◽  
Author(s):  
DA Breems ◽  
PB van Hennik ◽  
N Kusadasi ◽  
A Boudewijn ◽  
JJ Cornelissen ◽  
...  

Peripheral blood stem cells (PBSC) are used for stem cell support in patients after intensive chemotherapy and generally permit faster hematopoietic recovery than bone marrow. The development of different protocols for chemotherapy conditioning, mobilization, and ex vivo manipulation of PBSC may potentially lead to loss of primitive hematopoietic stem cells or reduction of their quality. To characterize the frequency of different stem cell subsets and their quality per mobilized PBSC, we have studied 47 leukapheresis products (LPs) of 21 cancer patients using stroma-dependent long-term culture (LTC) and limiting dilution-type cobblestone area forming cell (CAFC) assays. A large variation in CAFC week-type frequencies between the LPs was observed. The frequencies of CAFC week 2 as a tentative indicator of progenitor cells and transiently repopulating hematopoietic stem cells ranged from 0.89 to 205 per 10(5) mobilized nucleated cells and the frequencies of more primitive CAFC week 6 varied between 0.37 and 48. The average total colony-forming cell (CFC) production per CAFC at week 6 varied between 1.2 and 730, as determined in parallel LTC. In contrast to LPs, bone marrow samples generated 4.2 to 48 CFC per CAFC at week 6. Notably, a poor stem cell quality was consistently found in LPs that contained less than 5,000 CAFC week 6 per kilogram of body weight. Frequency analyses of CFCs, CAFC subtypes, and immunophenotypic subsets showed a good level of mutual correlation, suggesting identical mobilization kinetics of different stem cell subsets. The premobilization chemotherapy intensity was directly correlated with both decreasing frequency and quality of the CAFC week 6 in LPs. The frequency of CFCs, immunophenotypic subsets, and CAFC subsets transplanted and the transplant quality as determined in LTC assays was related to the neutrophil and platelet recovery time after PBSC transplantation. Although the number of progenitor cells transplanted and the in vitro transplant quality showed the best correlation with early hematopoietic recovery, the data did not permit determination of which stem cell subsets are predominantly responsible for early posttransplantation recovery. As a result, frequency and quality analysis of stem cell subsets may be a useful tool to monitor and calibrate the efficacy of novel mobilization regimens and ex vivo manipulation of PBSC.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1332-1332
Author(s):  
Takahiro Suzuki ◽  
Yasuhisa Yokoyama ◽  
Keiki Kumano ◽  
Minoko Takanashi ◽  
Shiro Kozuma ◽  
...  

Abstract [Background and purposes] Ex vivo expansion of hematopoietic stem cells (HSCs) has been explored in the fields of stem cell biology, gene therapy and clinical transplantation. The use of Notch ligands or soluble IL-6 receptor combined with IL-6 has been a major technique that revealed several fold expansion of human cord blood SCID repopulating cells (SRCs). These studies, however, have been conducted in an independent manner, which hampered direct comparison and evaluation of the effect of combination of these methods. Our purpose of this study is to clarify these issues in a chemically defined serum-free medium that allows us to develop clinical usage of the culture condition. We also compared the efficiencies of SRC isolation with magnetic beads targeting CD34 and CD133. [Methods] Human cord blood CD133-sorted cells were cultured on immobilized Delta1 supplemented with stem cell factor, thrombopoietin, flt-3 ligand, IL-3 and IL-6/soluble IL-6 receptor chimeric protein (FP6) for three weeks, and cultured cells were transplanted into NOD/SCID mice after limiting dilution to calculate the number of SRCs. To confirm whether full multipotency and self-renewal capacity of SRCs were maintained during the culture, cells were transplanted serially into NOD/SCID/γcnull (NOG) mice, and hematopoietic reconstitution was examined. To compare the efficiencies of CD34- and CD133-sorting, we divided each sample into two aliquots and separated CD34+ and CD133+ cells, and calculated the SRC numbers recovered by both separation methods. [Results and discussion] The frequencies of SRCs in the culture-initiating CD133-sorted cells and cultured progeny were calculated as one out of 1,020 and one out of 175 (adjusted to culture-initiating cells), respectively, indicating 6-fold expansion of SRCs that was statistically significant. Delta1 significantly enhanced the expansion rate of SRCs, and addition of IL-3 to this condition further promoted the expansion. In the serial transplantation assays, we found human myeloid and lymphoid reconstitution both in the primary and secondary NOG recipients, verifying the SRC capacity in the cultured cells. Notably, the CD133-sorting was approximately 4.5 times more efficient in collecting SRCs than the CD34-sorting from the same number of mononuclear cells (MNCs) (308 and 254 SRCs by CD133-sorting vs. 67 and 59 SRCs by CD34-sorting from 108 MNCs). Our study provides a promising method to expand HSCs and encourages future trials on clinical transplantation.


2016 ◽  
Vol 157 (46) ◽  
pp. 1819-1829
Author(s):  
Zsolt Matula ◽  
Gyöngyi Kudlik ◽  
Veronika Urbán S. ◽  
Ferenc Uher

For decades, developing hematopoietic cells have been strictly compartmentalized into a small population of multipotent self-renewing hematopoietic stem cells, multipotent hematopoietic progenitor cells that are undergoing commitment to myeloid or lymphoid fates, and unipotent precursor cells that mature towards peripheral blood and immune cells. Recent studies, however, have provided a battery of findings that cannot be explained by this “classical” hierarchical model for the architecture of hematopoiesis. It is emerging that heterogeneous hematopoietic stem cell populations in the bone marrow coexist, each with distinct, preprogrammed differentiation and proliferation behaviors. Three subsets can be distinguished among them: myeloid-biased (α), balanced (β), and lymphoid-biased (γ/δ) hematopoietic stem cells. The ratio of these hematopoietic stem cell subsets is developmentally regulated in the foetal liver and hematopoietic stem cells adult bone marrow, and coordinately gives rise to hematopoiesis. Beta- and γ/δ-hematopoietic stem cells are found predominantly early in the life of an organism, whereas α-hematopoietic stem cells accumulate in aged mice and humans. In addition, new sophisticated genetic experiments in mice have identified a major role of long-lived, committed progenitor cells downstream from hematopoietic stem cells as drivers of normal adult hematopoiesis, and revealed that post-transplantation hematopoiesis differs qualitatively and quantitatively from normal steady-state hematopoiesis. These findings have important implications for understanding in situ the regulation of haematopoiesis in health and disease. Orv. Hetil., 2016, 157(46), 1819–1829.


Author(s):  
Valentina Orticelli ◽  
Andrea Papait ◽  
Elsa Vertua ◽  
Patrizia Bonassi Signoroni ◽  
Pietro Romele ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document