Endothelial Cells Promote Expansion of Long-Term Engrafting Marrow Hematopoietic Stem and Progenitor Cells in Primates

Jennifer L Gori, Jason M Butler, Balvir Kunar, Michael G Poulos, Michael Ginsberg, Daniel J Nolan, Zachary K Norgaard, Jennifer E Adair, Shahin Rafii, Hans-Peter Kiem, Jennifer L Gori, Jason M Butler, Balvir Kunar, Michael G Poulos, Michael Ginsberg, Daniel J Nolan, Zachary K Norgaard, Jennifer E Adair, Shahin Rafii, Hans-Peter Kiem

Abstract

Successful expansion of bone marrow (BM) hematopoietic stem and progenitor cells (HSPCs) would benefit many HSPC transplantation and gene therapy/editing applications. However, current expansion technologies have been limited by a loss of multipotency and self-renewal properties ex vivo. We hypothesized that an ex vivo vascular niche would provide prohematopoietic signals to expand HSPCs while maintaining multipotency and self-renewal. To test this hypothesis, BM autologous CD34+ cells were expanded in endothelial cell (EC) coculture and transplanted in nonhuman primates. CD34+ C38- HSPCs cocultured with ECs expanded up to 17-fold, with a significant increase in hematopoietic colony-forming activity compared with cells cultured with cytokines alone (colony-forming unit-granulocyte-erythroid-macrophage-monocyte; p < .005). BM CD34+ cells that were transduced with green fluorescent protein lentivirus vector and expanded on ECs engrafted long term with multilineage polyclonal reconstitution. Gene marking was observed in granulocytes, lymphocytes, platelets, and erythrocytes. Whole transcriptome analysis indicated that EC coculture altered the expression profile of 75 genes in the BM CD34+ cells without impeding the long-term engraftment potential. These findings show that an ex vivo vascular niche is an effective platform for expansion of adult BM HSPCs. Stem Cells Translational Medicine 2017;6:864-876.

Keywords: Bone marrow transplantation; CD34+ cells; Endothelial cells; Gene therapy; Hematopoietic stem progenitor cell.

© 2016 The Authors Stem Cells Translational Medicine published by Wiley Periodicals, Inc. on behalf of AlphaMed Press.

Figures

Figure 1
Figure 1
Human ECs support robust expansion of hematopoietic stem/progenitor cells. (A): Kinetics of expansion of SS and granulocyte colony‐stimulating factor/stem cell factor‐primed BM‐derived CD34+CD38− cells after coculture with cytokines with or without human ECs (n = 3 naïve donors per condition). (B): Analysis of hematopoietic colony‐forming cell (CFC) potential of BM‐derived CD34+ cells after 7‐day expansion with cytokines with or without EC. Right: Images of CFU‐M (top) and BFU‐E (bottom) colonies. (C): Phase contrast (left) and fluorescent (middle) images of P140K‐MGMT‐GFP transduced macaque SS BM CD34+ cells and EC after 7 days of coculture. Right: Flow cytometry analysis of GFP and CD34 coexpression in gene‐modified CD34+ cells after EC expansion. (D): Summary of P140K‐MGMT‐GFP lentivirus‐transduced SS BM CD34+ cell expansion by flow cytometry analysis for detection of CD34+CD38− and LT‐HSPC phenotype (CD34+CD49f+Thy1+CD38−CD45RA−; n = 3 naïve donors). (E): Microscopy showing morphology of Wright‐stained cytospin samples of gene‐modified CD34+ hematopoietic cells after coculture with cytokines with or without EC. (F): CFC analysis showing frequency and morphology of CFUs generated from gene‐modified CD34+ cells after expansion with cytokines with or without EC. Data are shown as the mean from the three experiments (donors) ± SD. CFC assays were conducted with three macaque donors and three biologic replicates per donor. Total CFUs are expressed per 105 cells plated in MethoCult (StemCell Technologies). The colony types included BFU‐E, CFU‐M, CFU‐GM, and CFU‐GEMM. Statistical analysis used the Student t test: ∗, p < .05; ∗∗, p < .005. Abbreviations: BM, bone marrow; BFU‐E, burst‐forming unit‐erythroid; CFU, colony‐forming unit; CFU‐E, colony‐forming unit‐erythroid burst; CFU‐GEMM, colony‐forming unit‐granulocyte‐erythroid‐monocyte‐macrophage; CFU‐GM, colony‐forming unit‐granulocyte‐macrophage; CFU‐M, colony‐forming unit‐macrophage; EC, endothelial cell; GFP = green fluorescent protein; LT‐HSPC, long‐term hematopoietic and progenitor stem cell; SS, steady‐state.
Figure 2
Figure 2
Hematopoietic reconstitution after transplantation with endothelial cell (EC) expanded CD34+ cells. (A): Cell doses used in autologous hematopoietic and progenitor stem cell transplantation in three nonhuman primates (animal identification nos.: A11224, Z13018, A11208). For each naïve animal, CD34+ cells were collected from bone marrow (BM), prestimulated with cytokines for 2 days, transduced with P140K‐green fluorescent protein lentivirus vector, cryopreserved, thawed, and then expanded in endothelial cell coculture for 7 days. Each animal received myeloablative conditioning (1,020 cGy) followed by intravenous infusion with a heterogeneous mixture of the transduced CD34+ cells and endothelial cell coculture. Cell doses are indicated per kilogram of body weight of BM‐derived transduced CD34+ cells before (pre‐expansion) and after (postexpansion infusion product) coculture with endothelial cells. The infusion products contained both the autologous CD34+ cells and the xenogeneic human EC dose. (B): Human ECs detected in peripheral blood up to 1 week after cell infusion. Data shown represent mean ± SD for n = 3 monkeys (A11224, Z13018, A11208). Representative flow cytometry analysis of peripheral blood sample 4 days after transplantation of CD34+/EC cocultures for detection of the human‐specific cell surface marker CD147 (Tra‐1‐85 antibody) and EC‐specific cell surface markers CD31 and KDR. (C): Hematopoietic recovery (over the first 30 days for each of the three indicated animals) as indicated by complete blood count analysis. Horizontal shaded lines represent minimal threshold of recover for (from top to bottom): platelets, neutrophils, lymphocytes. Abbreviation: EC, endothelial cell.
Figure 3
Figure 3
Long‐term engraftment and in vivo multilineage contribution to hematopoiesis by endothelial cell‐expanded gene‐modified autologous CD34+ cells. Detection of gene‐modified cells in animals A11224 (A), Z13018 (B), and A11208 (C). Left: Percentages of GFP+ cells (gene marking by flow cytometry) in peripheral blood lymphocytes and granulocytes. Middle: Absolute cell numbers of GFP+ cells in peripheral blood lymphocytes and neutrophils. Right: Gene marking as determined by the mean VCN per circulating leukocyte genome equivalent (normalized to β‐globin copy number) by Taqman qPCR to detect the lentiviral LTR and GFP transgene. Abbreviations: GFP, green fluorescent protein; LTR, long terminal repeat; VCN, vector copy number.
Figure 4
Figure 4
Detection of gene‐modified platelets, RBCs, and lymphoid subsets in vivo. (A): Detection of GFP+ platelets and RBCs in animal A11224 (top) and animal Z13018 (bottom). (B): Detection of GFP+ CD3+ T lymphocytes and CD20+ B lymphocytes in animal A11224 (top) and animal Z13018 (bottom). (C): Detection of GFP+ CD4 and CD8 T‐lymphocyte subsets in animal A11224 (left) and animal Z13018 (right). For determination of gene marking in lymphoid subsets, cells in the lymphoid subgate within the forward scatter by side scatter (FSC × SSC) gate were costained with fluorophore‐conjugated CD3, CD4, CD8, and CD20 antibodies. The percentages of GFP+ cells were then evaluated within the CD3, CD4, CD8, and CD20 subset gates. Abbreviations: GFP, green fluorescent protein; RBCs, red blood cells.
Figure 5
Figure 5
Gene‐modified peripheral blood leukocytes demonstrate polyclonal engraftment after endothelial cell (EC) expansion and myeloablative transplantation. Each graph represents the clonal diversity observed at each time point and/or in each blood cell subpopulation examined as a function of the frequency of genomic sequences identified in the pool (primary y‐axis) for each animal: A11224 (A, B), Z13018 (C), and A11208 (D). Each bar designates the relative frequency of each clonal integration sequence identified in the sample from the most abundant (bottom of bar) to the least abundant (top of bar). The total number of unique sites of integration identified in each sample is listed at the top of each bar. Unique integration sites (clones) sequenced at a frequency >1% of the pool are designated by open boxes. All clones contributing <1% sequence frequency to the pool are grouped in a single gray box. Colored boxes designate clones tracked in multiple samples over time in the same animal. White boxes indicate the clones that were not observed in any other sample from the same animal. Clones tracked across multiple samples that did not contribute >1% frequency in a time point are designated by colored text citing the percentage of sequences identified within or next to the corresponding gray box. Overlay lines designate the percentage of GFP+ cells in the sample analyzed as determined by flow cytometry (secondary y‐axis). For (B), the sorted lineage is indicated and the time point (days after transplantation) are indicated in parentheses. Abbreviation: GFP, green fluorescent protein.
Figure 6
Figure 6
Differential gene expression in CD34+ cells unexpanded or expanded with cytokines or expanded with cytokines and ECs. Hierarchical clustering and transcriptional analysis of EC‐expanded CD34+ cells and cytokine‐expanded CD34+ cells from three donors (A11224, R11145, Z13018) and from unexpanded CD34+ cells (two donors). Heat maps display the relative abundance of genes that were differentially expressed across the three cell populations (unexpanded CD34+ cells, cytokine‐expanded CD34+ cells, and EC‐expanded CD34+ cells). EC‐expanded CD34+ cells were EC depleted and enriched for CD34+ cells before analysis. All other subsets were sorted according to expression of CD34 before analysis. Scaling is relative to the maximum FPKM for each gene across all samples. (For example, if the unexpanded CD34+ cells had the greatest mRNA abundance for a particular gene, all other values would be relative to the level in unexpanded CD34+ cells.) The numbers within parentheses next to each gene name provide that value of the maximum FPKM from all samples. All genes shown in this heat map are differentially expressed. The color scale to the left of the heat map is scaled to the FPKM values to the maximum. The genes are ordered according to the p value (from top to bottom, smallest to largest). For all genes shown, the following criterion was applied: FPKM cutoff of 20 (p ≤ .05, log twofold change). Abbreviations: ECs, endothelial cells; FPKM, fragments per kilobase of exon per million reads mapped; Max, maximum.

References

    1. Anguita‐Compagnon AT, Dibarrart MT, Palma J et al. Mobilization and collection of peripheral blood stem cells: Guidelines for blood volume to process, based on CD34‐positive blood cell count in adults and children. Transplant Proc 2010;42:339–344.
    1. Petrecca K, Atanasiu R, Akhavan A et al. N‐linked glycosylation sites determine HERG channel surface membrane expression. J Physiol 1999;515:41–48.
    1. Perseghin P, Terruzzi E, Dassi M et al. Management of poor peripheral blood stem cell mobilization: Incidence, predictive factors, alternative strategies and outcome. A retrospective analysis on 2177 patients from three major Italian institutions. Transfus Apheresis Sci 2009;41:33–37.
    1. Sun CL, Francisco L, Kawashima T et al. Prevalence and predictors of chronic health conditions after hematopoietic cell transplantation: A report from the Bone Marrow Transplant Survivor Study. Blood 2010;116:3129–3139; quiz 3377.
    1. Delaney C, Heimfeld S, Brashem‐Stein C et al. Notch‐mediated expansion of human cord blood progenitor cells capable of rapid myeloid reconstitution. Nat Med 2010;16:232–236.
    1. Dahlberg A, Brashem‐Stein C, Delaney C et al. Enhanced generation of cord blood hematopoietic stem and progenitor cells by culture with StemRegenin1 and Delta1(Ext‐IgG.). Leukemia 2014;28:2097–2101.
    1. Hoggatt J, Mohammad KS, Singh P et al. Prostaglandin E2 enhances long‐term repopulation but does not permanently alter inherent stem cell competitiveness. Blood 2013;122:2997–3000.
    1. Cutler C, Multani P, Robbins D et al. Prostaglandin‐modulated umbilical cord blood hematopoietic stem cell transplantation. Blood 2013;122:3074–3081.
    1. Goessling W, Allen RS, Guan X et al. Prostaglandin E2 enhances human cord blood stem cell xenotransplants and shows long‐term safety in preclinical nonhuman primate transplant models. Cell Stem Cell 2011;8:445–458.
    1. Fan X, Gay FP, Lim FW et al. Low‐dose insulin‐like growth factor binding proteins 1 and 2 and angiopoietin‐like protein 3 coordinately stimulate ex vivo expansion of human umbilical cord blood hematopoietic stem cells as assayed in NOD/SCID gamma null mice. Stem Cell Res Ther 2014;5:71.
    1. Blank U, Ehrnström B, Heinz N et al. Angptl4 maintains in vivo repopulation capacity of CD34+ human cord blood cells. Eur J Haematol 2012;89:198–205.
    1. Lam AC, Li K, Zhang XB et al. Preclinical ex vivo expansion of cord blood hematopoietic stem and progenitor cells: Duration of culture; the media, serum supplements, and growth factors used and engraftment in NOD/SCID mice. Transfusion 2001;41:1567–1576.
    1. Boitano AE, Wang J, Romeo R et al. Aryl hydrocarbon receptor antagonists promote the expansion of human hematopoietic stem cells. Science 2010;329:1345–1348.
    1. Fares I, Chagraoui J, Gareau Y et al. Cord blood expansion: Pyrimidoindole derivatives are agonists of human hematopoietic stem cell self‐renewal. Science 2014;345:1509–1512.
    1. Kobayashi H, Butler JM, O’Donnell R et al. Angiocrine factors from Akt‐activated endothelial cells balance self‐renewal and differentiation of haematopoietic stem cells. Nat Cell Biol 2010;12:1046–1056.
    1. Butler JM, Gars EJ, James DJ et al. Development of a vascular niche platform for expansion of repopulating human cord blood stem and progenitor cells. Blood 2012;120:1344–1347.
    1. Beard BC, Trobridge GD, Ironside C et al. Efficient and stable MGMT‐mediated selection of long‐term repopulating stem cells in nonhuman primates. J Clin Invest 2010;120:2345–2354.
    1. Trobridge GD, Beard BC, Gooch C et al. Efficient transduction of pigtailed macaque hematopoietic repopulating cells with HIV‐based lentiviral vectors. Blood 2008;111:5537–5543.
    1. Watts KL, Delaney C, Humphries RK et al. Combination of HOXB4 and Delta‐1 ligand improves expansion of cord blood cells. Blood 2010;116:5859–5866.
    1. Butler JM, Nolan DJ, Vertes EL et al. Endothelial cells are essential for the self‐renewal and repopulation of Notch‐dependent hematopoietic stem cells. Cell Stem Cell 2010;6:251–264.
    1. Bernstein SH, Nademanee AP, Vose JM et al. A multicenter study of platelet recovery and utilization in patients after myeloablative therapy and hematopoietic stem cell transplantation. Blood 1998;91:3509–3517.
    1. 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.
    1. Beard BC, Adair JE, Trobridge GD et al. High‐throughput genomic mapping of vector integration sites in gene therapy studies. Methods Mol Biol 2014;1185:321–344.
    1. Ginsberg M, James D, Ding BS et al. Efficient direct reprogramming of mature amniotic cells into endothelial cells by ETS factors and TGFβ suppression. Cell 2012;151:559–575.
    1. Yoo KH, Lee SH, Kim HJ et al. The impact of post‐thaw colony‐forming units‐granulocyte/macrophage on engraftment following unrelated cord blood transplantation in pediatric recipients. Bone Marrow Transplant 2007;39:515–521.
    1. Page KM, Zhang L, Mendizabal A et al. Total colony‐forming units are a strong, independent predictor of neutrophil and platelet engraftment after unrelated umbilical cord blood transplantation: A single‐center analysis of 435 cord blood transplants. Biol Blood Marrow Transplant 2011;17:1362–1374.
    1. Bedi R, Kumar L, Kochupillai V. Autologous peripheral blood stem cell transplantation: Predictors for haematopoietic reconstitution. Natl Med J India 2003;16:255–259.
    1. Schmitz N, Linch DC, Dreger P et al. Randomised trial of filgrastim‐mobilised peripheral blood progenitor cell transplantation versus autologous bone‐marrow transplantation in lymphoma patients. Lancet 1996;347:353–357.
    1. Gonçalves TL, Benvegnú DM, Bonfanti G. Specific factors influence the success of autologous and allogeneic hematopoietic stem cell transplantation. Oxid Med Cell Longev 2009;2:82–87.
    1. Notta F, Doulatov S, Laurenti E et al. Isolation of single human hematopoietic stem cells capable of long‐term multilineage engraftment. Science 2011;333:218–221.
    1. Okada S, Fukuda T, Inada K et al. Prolonged expression of c‐fos suppresses cell cycle entry of dormant hematopoietic stem cells. Blood 1999;93:816–825.
    1. McKinney‐Freeman S, Cahan P, Li H et al. The transcriptional landscape of hematopoietic stem cell ontogeny. Cell Stem Cell 2012;11:701–714.
    1. Shimazu T, Iida R, Zhang Q et al. CD86 is expressed on murine hematopoietic stem cells and denotes lymphopoietic potential. Blood 2012;119:4889–4897.
    1. Chute JP, Saini AA, Chute DJ et al. Ex vivo culture with human brain endothelial cells increases the SCID‐repopulating capacity of adult human bone marrow. Blood 2002;100:4433–4439.
    1. Chute JP, Muramoto G, Fung J et al. Quantitative analysis demonstrates expansion of SCID‐repopulating cells and increased engraftment capacity in human cord blood following ex vivo culture with human brain endothelial cells. Stem Cells 2004;22:202–215.
    1. Chute JP, Muramoto GG, Fung J et al. Soluble factors elaborated by human brain endothelial cells induce the concomitant expansion of purified human BM CD34+CD38− cells and SCID‐repopulating cells. Blood 2005;105:576–583.
    1. Brandt JE, Bartholomew AM, Fortman JD et al. Ex vivo expansion of autologous bone marrow CD34(+) cells with porcine microvascular endothelial cells results in a graft capable of rescuing lethally irradiated baboons. Blood 1999;94:106–113.
    1. Araki H, Chute JP, Petro B et al. Bone marrow CD34+ cells expanded on human brain endothelial cells reconstitute lethally irradiated baboons in a variable manner. Leuk Lymphoma 2010;51:1121–1127.
    1. Chute JP, Muramoto GG, Salter AB et al. Transplantation of vascular endothelial cells mediates the hematopoietic recovery and survival of lethally irradiated mice. Blood 2007;109:2365–2372.
    1. Muramoto GG, Chen B, Cui X et al. Vascular endothelial cells produce soluble factors that mediate the recovery of human hematopoietic stem cells after radiation injury. Biol Blood Marrow Transplant 2006;12:530–540.
    1. Hooper AT, Butler JM, Nolan DJ et al. Engraftment and reconstitution of hematopoiesis is dependent on VEGFR2‐mediated regeneration of sinusoidal endothelial cells. Cell Stem Cell 2009;4:263–274.
    1. Ding L, Saunders TL, Enikolopov G et al. Endothelial and perivascular cells maintain haematopoietic stem cells. Nature 2012;481:457–462.
    1. Ding L, Morrison SJ. Haematopoietic stem cells and early lymphoid progenitors occupy distinct bone marrow niches. Nature 2013;495:231–235.
    1. Poulos MG, Crowley MJ, Gutkin MC et al. Vascular platform to define hematopoietic stem cell factors and enhance regenerative hematopoiesis. Stem Cell Rep 2015;5:881–894.
    1. Himburg HA, Harris JR, Ito T et al. Pleiotrophin regulates the retention and self‐renewal of hematopoietic stem cells in the bone marrow vascular niche. Cell Reports 2012;2:964–975.
    1. Parekh C, Crooks GM. Critical differences in hematopoiesis and lymphoid development between humans and mice. J Clin Immunol 2013;33:711–715.
    1. Csaszar E, Wang W, Usenko T et al. Blood stem cell fate regulation by delta‐1‐mediated rewiring of IL‐6 paracrine signaling. Blood 2014;123:650–658.
    1. Watts KL, Nelson V, Wood BL et al. Hematopoietic stem cell expansion facilitates multilineage engraftment in a nonhuman primate cord blood transplantation model. Exp Hematol 2012;40:187–196.
    1. Watts KL, Delaney C, Nelson V et al. CD34(+) expansion with delta‐1 and HOXB4 promotes rapid engraftment and transfusion independence in a Macaca nemestrina cord blood transplant model. Mol Ther 2013;21:1270–1278.
    1. Trobridge GD, Wu RA, Beard BC et al. Protection of stem cell‐derived lymphocytes in a primate AIDS gene therapy model after in vivo selection. PLoS One 2009;4:e7693.
    1. Kiem HP, Arumugam PI, Burtner CR et al. Pigtailed macaques as a model to study long‐term safety of lentivirus vector‐mediated gene therapy for hemoglobinopathies. Mol Ther Methods Clin Dev 2014;1:14055.
    1. Hayakawa J, Ueda T, Lisowski L et al. Transient in vivo beta‐globin production after lentiviral gene transfer to hematopoietic stem cells in the nonhuman primate. Hum Gene Ther 2009;20:563–572.

Source: PubMed

3
S'abonner