Faculty Opinions recommendation of Bone marrow origin of hematopoietic progenitors and stem cells in murine muscle.

Author(s):  
Gerald Spangrude
Blood ◽  
2001 ◽  
Vol 98 (7) ◽  
pp. 2008-2013 ◽  
Author(s):  
Hiroshi Kawada ◽  
Makio Ogawa

It has been reported that mononuclear cells harvested from murine skeletal muscle are capable of hematopoietic reconstitution of lethally irradiated mice. First, the nature of the hematopoietic progenitors in the muscle of C57BL/6–Ly-5.1 mice was examined by means of methylcellulose culture. The types and incidences of colonies grown from muscle mononuclear cells were different from those cultured from bone marrow (BM) or peripheral blood mononuclear cells. The next step was to examine the origin of the hematopoietic progenitors and stem cells in the muscle with the use of Ly-5.2 mice that had been made chimeric by transplantation of Ly-5.1 BM cells. The percentages of Ly-5.1 cells cultured from the muscle of the chimeric mice correlated with those cultured from BM, indicating BM origin of hematopoietic progenitors in the muscle. Long-term hematopoietic engrafting cells in the muscle of the chimeric mice were also derived from BM. However, mobilization of progenitors into circulation by granulocyte colony-stimulating factor did not change the population of hematopoietic progenitors in the muscle. It is proposed that hematopoietic progenitors and stem cells in the muscle tissue are of BM origin but their transition from BM to muscle may be a slow process.


Blood ◽  
1993 ◽  
Vol 81 (6) ◽  
pp. 1497-1504 ◽  
Author(s):  
VF Quesniaux ◽  
GJ Graham ◽  
I Pragnell ◽  
D Donaldson ◽  
SD Wolpe ◽  
...  

Abstract A macrophage-derived inhibitor of early hematopoietic progenitors (colony-forming unit-spleen, CFU-A) called stem cell inhibitor was found to be identical to macrophage inflammatory protein-1 alpha (MIP-1 alpha). We investigated the effect of MIP-1 alpha on the earliest stem cells that sustain long-term hematopoiesis in vivo in a competitive bone marrow repopulation assay. Because long-term reconstituting (LTR) stem cells are normally quiescent, an in vivo model was first developed in which they are triggered to cycle. A first 5-fluorouracil (5-FU) injection was used to eliminate later progenitors, causing the LTR stem cells, which are normally resistant to 5-FU, to enter the cell cycle and become sensitive to a second 5-FU injection administered 5 days later. Human MIP-1 alpha administered from day 0 to 7 was unable to prevent the depletion of the LTR stem cells by the second 5-FU treatment, as observed on day 7 in this model, suggesting that the LTR stem cells were not prevented from being triggered into cycle despite the MIP-1 alpha treatment. However, the MIP-1 alpha protocol used here did substantially decrease the number of more mature hematopoietic progenitors (granulocyte-macrophage colony-forming cells [CFC], burst- forming unit-erythroid, CFCmulti, and preCFCmulti) recovered in the bone marrow shortly after a single 5-FU injection. In vitro, MIP-1 alpha had no inhibitory effect on the ability of these progenitors to form colonies. This study confirms the in vivo inhibitory effect of MIP- 1 alpha on subpopulations of hematopoietic progenitors that are activated in myelodepressed animals. However, MIP-1 alpha had no effect on the long-term reconstituting stem cells in vivo under conditions in which it effectively reduced all later progenitors.


Blood ◽  
1995 ◽  
Vol 85 (10) ◽  
pp. 2742-2746 ◽  
Author(s):  
RJ Jones ◽  
JP Barber ◽  
MS Vala ◽  
MI Collector ◽  
SH Kaufmann ◽  
...  

Cytosolic aldehyde dehydrogenase (ALDH), an enzyme responsible for oxidizing intracellular aldehydes, has an important role in ethanol, vitamin A, and cyclophosphamide metabolism. High expression of this enzyme in primitive stem cells from multiple tissues, including bone marrow and intestine, appears to be an important mechanism by which these cells are resistant to cyclophosphamide. However, although hematopoietic stem cells (HSC) express high levels of cytosolic ALDH, isolating viable HSC by their ALDH expression has not been possible because ALDH is an intracellular protein. We found that a fluorescent aldehyde, dansyl aminoacetaldehyde (DAAA), could be used in flow cytometry experiments to isolate viable mouse and human cells based on their ALDH content. The level of dansyl fluorescence exhibited by cells after incubation with DAAA paralleled cytosolic ALDH levels determined by Western blotting and the sensitivity of the cells to cyclophosphamide. Moreover, DAAA appeared to be a more sensitive means of assessing cytosolic ALDH levels than Western blotting. Bone marrow progenitors treated with DAAA proliferated normally. Furthermore, marrow cells expressing high levels of dansyl fluorescence after incubation with DAAA were enriched for hematopoietic progenitors. The ability to isolate viable cells that express high levels of cytosolic ALDH could be an important component of methodology for identifying and purifying HSC and for studying cyclophosphamide-resistant tumor cell populations.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3767-3767
Author(s):  
Kran Suknuntha ◽  
Yuki Ishii ◽  
Kejin Hu ◽  
Jean YJ Wang ◽  
Igor Slukvin

Abstract Abstract 3767 Reprogramming of neoplastic cells to pluripotency provides a unique tool to personalize the exploration of tumor pathogenic mechanisms and drug resistance using iPSCs with patient-specific chromosomal abnormalities. We have developed a technology to generate transgene-free iPSCs from bone marrow and cord blood cells employing episomal vectors. Using this approach we created transgene-free iPSCs from a patient with CML in the chronic phase. CMLiPSCs showed a unique complex chromosomal translocation identified in the patinet's marrow sample while displaying typical embryonic stem cell phenotype and pluripotent differentiation potential. Importantly, these CMLiPSCs are devoid of genomic integration and expression of reprogramming factors, which are incompatible for modeling tumor development and drug response (Hu et al. Blood 117:e109). We have also shown that these CMLiPSCs contain the BCR-ABL oncogene without any detectable mutations in its kinase domain. By coculture with OP9, we generated APLNR+ mesodermal cells, MSCs, and lin-CD34+CD45+ hematopoietic progenitors from CMLiPSCs, and control BMiPSCs from a normal subject and analyzed the levels of BCR-ABL protein and tyrosine-phosphorylated (pTyr) cellular proteins in the different cell populations. The highest level of BCR-ABL protein expression was found in the in undifferentiated iPSCs, however, the overall cellular pTyr levels was lower than the control BMiPSCs, suggesting that BCR-ABL kinase activity was suppressed in the CMLiPScs. Consistent with these findings, imatinib does not inhibit the growth and survival of these CMLiPSCs. The levels of BCR-ABL protein decreased upon differentiation with a major reduction observed when cells became mesoderm. Following differentiation of CMLiPSC-derived mesoderm into the MSCs and lin-CD34+CD45+ hematopoietic progenitors, the levels of BCR-ABL protein did not change significantly, indicating that the major epigenetic regulation of BCR-ABL expression occurs during the transition to mesoderm. In spite of the decrease in BCR-ABL expression, the total pTyr levels significantly increased following transition of CMLiPSCs to mesoderm and blood cells, suggesting recovery of BCR-ABL kinase activity during differentiation. Interestingly, we found that imatinib had no effect on CFC potential of the most primitive lin-CD34+CD45+ hematopoietic progenitors derived from CMLiPSCs, while significant inhibition in hematopoietic CFC potential was observed when we used the patient's bone marrow cells. Following expansion of lin-CD34+CD45+ progenitors in serum-free medium with cytokines, we found that more differentiated hematopoietic cells became imatinib sensitive. The differential response of progenitors versus more differentiated cells to imatinib recapitulate the clinical observation that CML stem cells display innate resistance to imatinib but their differentiated progenies become sensitive to this BCR-ABL kinase inhibitor. The iPSC-based models provide several advantages for the study of CML pathogenesis. iPSCs can provide an unlimited supply of hematopoietic cells carrying patient-specific genetic abnormalities. Using well-defined temporal windows and surface markers, distinct cell subsets with tumor-initiating/tumor-propagating potential after transplantation in immunodeficient mice could be identified and used for drug screening. iPSC models make it possible to address CML stem-cell potential at various stages of differentiation for which it may be difficult to obtain samples from the patient, for example, at the hemangioblast stage. They also provide a unique opportunity to explore the interplays between epigenetics and oncogene function, as we have demonstrated using the CMLiPSCs. The major unsolved question is why CML stem cells are naturally resistant to imatinib, and this question can be addressed using the iPS system. Disclosures: Slukvin: CDI: Consultancy, Equity Ownership.


Blood ◽  
2005 ◽  
Vol 105 (11) ◽  
pp. 4170-4178 ◽  
Author(s):  
Regis Doyonnas ◽  
Julie S. Nielsen ◽  
Shierley Chelliah ◽  
Erin Drew ◽  
Takahiko Hara ◽  
...  

Abstract Podocalyxin/podocalyxin-like protein 1 [PCLP1]/thrombomucin/MEP21 is a CD34-related sialomucin. We have performed a detailed analysis of its expression during murine development and assessed its utility as a marker of hematopoietic stem cells (HSCs) and their more differentiated progeny. We find that podocalyxin is highly expressed by the first primitive hematopoietic progenitors and nucleated red blood cells to form in the embryonic yolk sac. Likewise, podocalyxin is expressed by definitive multilineage hematopoietic progenitors and erythroid precursors in fetal liver. The level of podocalyxin expression gradually declines with further embryo maturation and reaches near-background levels at birth. This is followed by a postnatal burst of expression that correlates with the seeding of new hematopoietic progenitors to the spleen and bone marrow. Shortly thereafter, podocalyxin expression gradually declines, and by 4 weeks postpartum it is restricted to a rare population of Sca-1+, c-kit+, lineage marker- (Lin-) cells in the bone marrow. These rare podocalyxin-expressing cells are capable of serially reconstituting myeloid and lymphoid lineages in lethally irradiated recipients, suggesting they have HSC activity. In summary, we find that podocalyxin is a marker of embryonic HSCs and erythroid cells and of adult HSCs and that it may be a valuable marker for the purification of these cells for transplantation.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1196-1196 ◽  
Author(s):  
Bijender Kumar ◽  
Mayra Garcia ◽  
Guido Marcucci ◽  
Ching-Cheng Chen

Abstract MicroRNAs (miRNAs) regulate hematopoietic cell fate and their global down-regulation by Dicer1 deletion promotes tumorigenesis in a cancer-cell-autonomous manner (Kumar M.S. et al, 2007). Raajimakers MH et al. (2010) using neonatal Osterix specific dicer deletion showed altered hematopoiesis and developed myelodysplasia. However, there is no study illustrating the role of the ablation of bone marrow (BM) niche specific miRNA processing machinery in the adult mice. Since expression and functions of different mesenchymal and osteoprogenitors vary from embryonic development to adulthood, studying the dicer ablation in adult mice may provide more insight about the role of miRNA processing in adult mice niche. Here we investigate whether adult Osterix expressing cells play a similar role in the HSC niche compared to fetal Osterix expressing cells. We crossed Osx-GFP-tTA-Cre recombinase mice with mice with floxed Dicer1 allele. Crossing generated Osx- GFP-tTA-Cre+Dicerfl/+ (OCDfl/+control) and Osx-GFP- tTA-Cre+ Dicerfl/fl (OCDfl/fl mutant) mice. Osx-GFP-tTA-Cre expression was either activated during embryonic development (young dicer KO) or suppressed using tetracycline until mice were 6 weeks of age (adult dicer KO). We found young dicer KO mice had reduced weight (p=0.0031), leukopenia, anemia, reduced mature CD19+B220- B lymphocytes (p=0.0034) and increased CD11b+Gr- monocytes and CD11b+Gr+ neutrophils (p=0.02 and p=0.04 respectively) in peripheral blood compared to OCDfl/+ control aged littler mates. The leucocytes and platelets showed dysplastic changes suggestive of myelodysplasia and had extra-medullary hematopoiesis. Adult dice KO, on the other hand, show no leukemia development 6 months after Cre activation. The number of BM hematopoietic progenitors (Lin-Sca1+ c-Kit+ cells, LSK) and long term hematopoietic stem cells (LT-HSCs, LSK CD150+CD48+ cells) in young dicer KO mice were significantly reduced compared to age matched control (OCDfl/+ control) mice. We observed increased Annexin V positive LSK, LT-HSCs and megakaryocytes erythroid progenitors (MEP) in the young dicer KO mice indicating increased apoptosis. Adult dicer KO mice didn't have significant changes in apoptosis in different hematopoietic progenitors. In young dicer KO mice, BM derived LSK and LT-HSCs showed increased cycling (SG2M phase, p=0.0133) and less quiescenece (Go phase, p=0.013). However LT-HSC from adult dicer KO didn't show any difference in cell cycling (p=0.18 and 0.09 respectively). Together these results indicate that while Osterix expressing cells in fetal and young mice give rise to a variety of HSC niche supporting cells the adult expression is limited to more mature osteoblast that are not absolutely essential for HSC maintenance. Our study provides the rationale for further exploration of the complexity in hierarchy of activity within niche constituting mesenchymal stroma progenitors and their role in different developmental stages to maintain hematopoiesis. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1996 ◽  
Vol 88 (8) ◽  
pp. 2927-2938 ◽  
Author(s):  
NJ Drize ◽  
JR Keller ◽  
JL Chertkov

We describe here a technique to study the clonal contribution of primitive stem cells that account for long-term hematopoiesis in the same mouse over a 14-month period. Specifically, irradiated recipient female mice were transplanted with retrovirally marked male hematopoietic progenitors. Bone marrow was then collected repeatedly from local sites from the same mice throughout a 14-month period and injected into secondary irradiated recipients for analysis of donor retrovirally marked day-11 colony-forming unit-spleen (CFU-S-11). We have tracked the temporal in vivo fate of 194 individual CFU-S-derived cell clones in 38 mice reconstituted with such retrovirally marked bone marrow cells. Our data show that long-term hematopoiesis is maintained by a large number of simultaneously functioning small, shortlived (1 to 3 months) clones that usually grow locally with little or no dispersion between different regions of the hematopoietic system. Furthermore, the clones that disappeared were never detected again. The data suggest that normal hematopoiesis is supported by the sequential recruitment of marrow repopulating cells into a differentiation mode.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2303-2303
Author(s):  
M. Kozik ◽  
J. J. Banks ◽  
L. R. Fanning ◽  
M. R. Finney ◽  
Y. Huang ◽  
...  

Abstract Background. Umbilical cord blood (UCB), a source of hematopoeitic stem cells (HSC), is marked by delayed engraftment attributed to the limited cellular content of a single UCB unit. Cytokine-based ex vivo expansion of UCB is a way of increasing the number of cells available for allogeneic transplants, however, this strategy has not demonstrated improved engraftment in human clinical trials. Further studies have incorporated human mesenchymal stem cells (huMSC) which may provide signals that control the proliferation, survival, and differentiation of HSC. In attempt to reduce the occurrence and severity of GVHD following allogeneic transplants, strategies such as utilizing T-cell depleted grafts have been pursued, however, clinical trials using these grafts have shown decreased rates of donor engraftment, suggesting the requirement of accessory cells as well as HSC to achieve engraftment. Methods. UCB mononuclear cells (MNC) were cultured using cytokines (IL-3, IL-6, G-CSF, SCF, Flt-3L, EPO) with or without a feeder-layer of huMSC for 12 days. On day 12, viability, 4-color flow cytometry, and human engraftment potential were measured. Human engraftment potential was determined by injecting cells (without CD34+ selection) from each culture condition and non-cultured UCB MNC, via tail vein, into sublethal irradiated NOD/SCID mice. Mice were injected with unexpanded UCB MNC (n=23), UCB expanded in huMSC+cytokines (n=21) and UCB expanded in cytokines alone (n=10). 7–9 weeks following injection of human cells, bone marrow was harvested and analyzed for human content. Positive human engraftment was determined by a human %CD45+ of ≥ 0.4%. Results. An 8.77 fold expansion of UCB cultured in cytokines alone compared to a 7.14 fold expansion of UCB cultured in huMSC + cytokines was observed. Surface phenotyping of expanded UCB, and human cells emerging in the bone marrow of NOD/SCID mice following injection of cultured and non-cultured UCB are in Table 1. Unexpanded huMSC+cytokines Cytokines % CD3 44.0 (4.40M) 2.04 (10.1M) 1.52 (4.35M) % CD56 17.0 (1.70M) 7.07 (36.1M) 3.69 (13.8M) % CD34 3.41 (.341M) 2.25 (10.9M) 3.52 (12.3M) Bone marrow of NOD/SCID mice % CD45+ 2.57 3.58 2.38 % of CD45+ co-expressing CD3 11.0 9.25 18.7 % of CD45+ co-expressing CD19 33.5 16.6 19.4 % of CD45+ co-expressing CD56 10.1 8.04 1.60 Human engraftment was seen in 13 mice which received unexpanded UCB, 10 mice which received UCB expanded in huMSC+cytokines and only 3 mice which received UCB expanded in cytokines alone. Statistical analysis, using multivariable logistic regression to determine the factors that predict engraftment, revealed that the proportions of T and NK cells present in expanded UCB correlated with engraftment. A 10% increase in the proportion of CD45+ co-expressing CD3 was associated with a 1.79 fold increase in engraftment (p=0.016), whereas each 10% increase in the proportion of CD45+ co-expressing CD56 increased the odds of engrafting by 104% (p= 0.003). Conclusions. We observed an expansion of CD34 hematopoietic progenitors as well as a greater proportion of CD3+ cells, in expansion conditions incorporating huMSC. Additionally, we observed improved rates of engraftment in this expansion condition. Therefore, although the mechanism by which accessory cells including T and NK cells facilitate HSC engraftment is not known, we observed that the presence of accessory cells in addition to CD34 hematopoietic progenitors facilitated engraftment in NOD/SCID mice.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1599-1599
Author(s):  
Kathleen E McGrath ◽  
Jenna M Frame ◽  
Anne Koniski ◽  
Paul D Kingsley ◽  
James Palis

Abstract Abstract 1599 The ontogeny of hematopoiesis in mammalian embryos is complicated by the requirement for functional blood cells prior to the emergence of hematopoietic stem cells or the bone marrow microenvironment. In the murine embryo, transplantable HSC are first evident at embryonic day (E) 10.5 and the first few HSC are found in the fetal liver hematopoietic environment by E12.5. However, two overlapping waves of hematopoietic potential arise in the yolk sac before E10.5. The first “primitive” wave produces progenitors from E7.25 to E8.5 with primitive erythroid, megakaryocyte and macrophage potentials. The resulting primitive erythroid cells mature within the circulation and support embryonic growth past E9.5. At E8.5, a second wave of hematopoiesis begins in the yolk sac and generates definitive erythroid and multiple myeloid progenitors that are the proposed source of the hematopoietic progenitors seeding the fetal liver before HSC colonization. We have identified a cell population displaying a unique cell surface immunophenotype in the E9.5 yolk sac that contains the potential to form definitive erythroid cells, megakaryocytes, macrophages and all forms of granulocytes within days of in vitro culture. Furthermore, all definitive hematopoietic colony-forming cells (BFU-E, CFC-myeloid and HPP-CFC) in the E9.5 yolk sac have this immunophenotype. These erythro-myeloid progenitors (EMP) are lineage-negative and co-express ckit, CD41, CD16/32 and Endoglin. Interestingly, this is not an immunophenotype evident in the adult bone marrow. Other markers that have been associated with HSC formation (AA4.1, ScaI) or with lymphoid potential (IL7R, Flt3) are not present on these cells at E9.5. Consistent with the lack of lymphoid markers, we also do not observe short-term development of B-cells (CD19+B220+ expressing Rag2 RNA) in cultures of the E9.5 sorted EMP, while bone marrow Lin-/ckit+/ScaI- cells do form B-cells under the same conditions. Clonal analysis of sorted EMP cells revealed single cells with both erythroid and granulocyte potential, similar to the common myeloid progenitors in adult bone marrow. Though these EMP are enriched at E9.5 in the yolk sac, they are also found at low levels in the fetal blood, embryo proper and placenta, consistent with their entrance into the circulation. By E10.5, EMP were most highly enriched in the newly formed fetal liver. Additionally by E12.5, a time when the first few HSCs are detected in the fetal liver, we find active erythropoiesis and granulopoiesis in the liver and the first definitive red blood cells and neutrophils in the bloodstream. Therefore, we believe the yolk sac definitive progenitors' fate is to populate the fetal liver and thus provide the first definitive erythrocytes and granulocytes for the embryo. The differentiation of embryonic stem cells (ES) and induced pluripotent stem cells (iPS) cells into mature cells types offers the hope of cell-based therapies. Analysis of differentiating murine ES cells reveals overlapping waves of primitive and definitive hematopoietic colony forming potential. We demonstrate the appearance of an EMP-like (ckit+/CD41+/FcGR+) population coincident with the emergence of definitive hematopoietic progenitors during murine ES cell differentiation as embryoid bodies. We have confirmed with colony forming assays that definitive hematopoietic potential is associated with this immunophenotypic group. Our studies support the concept that blood cell emergence during ES cell differentiation closely mimics pre-HSC hematopoiesis in the yolk sac. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 466-466
Author(s):  
Jennifer L Gori ◽  
Jason M Butler ◽  
Devikha Chandrasekaran ◽  
Brian C Beard ◽  
Daniel J Nolan ◽  
...  

Clinical use of human pluripotent stem cell (PSC)-hematopoietic stem cells (HSCs) is impeded by low engraftment potential. This block suggests that additional vascular derived angiocrine signals and hematopoietic cues must be provided to produce authentic HSCs. In addition, gene modification of induced (i)PSCs with a chemotherapy resistance transgene would provide a selective mechanism to stabilize or increase engraftment of HSCs. We therefore hypothesized that modifying iPSCs to express the O6-benzylguanine (O6BG)-resistant P140K variant of methylguanine methyltransferase (MGMT), would support in vivo selection of early-engrafted iPSC-HSCs. We further postulated that Akt-activated human endothelial cells afforded by transduction of the E4ORF1 gene (E4ORF1+ECs) through angiocrine upregulation of Notch and IGF ligands would provide the necessary signals under xenobiotic-free conditions to promote definitive hematopoiesis. This vascular induction platform could drive the emergence of true HSCs. We focused on pigtail macaque (Mn)iPSCs, as a scalable, clinically relevant nonhuman primate model. MniPSCs modified to express P140K had 15-fold higher MGMT levels compared to levels in human peripheral blood mononuclear cells. P140K-MniPSCs differentiated into chemoresistant CD34+ hematopoietic progenitors (50% CD34+) with a predominant long-term (LT)-HSC-like phenotype (CD34+CD38-Thy1+CD45RA-CD49f+). Hematopoietic progenitors maintained colony forming potential after O6BG and bis-chloroethylnitrosourea (BCNU) treatment. HSCs expanded on E4ORF1+ECs maintained colony forming potential, in contrast to cells cultured with cytokines alone, with a 22-fold increase in CD34+ cell content and 10-fold increase in LT-HSC-like cells. Importantly, MniPSC-HSCs expanded with the E4ORF1+ECs had long-term engraftment in NSG mice at levels comparable to Mn bone marrow HSC engrafted mice. O6BG/BCNU treatment increased engraftment to 35% CD45+ cells the blood of mice transplanted with E4ORF1+EC expanded P140K-MniPSC-HSCs, which was maintained 16 weeks post transplantation. Primate CD45+ cell levels in the blood after selection were significantly higher for this cohort compared to mice transplanted with P140K-MniPSC-HSCs expanded in the “cytokines alone” condition (18% vs. 3% CD45+, P<0.05). On average, 15% CD34+ and 37% CD45+ cells were detected in the bone marrow of mice transplanted with E4ORF1+EC-expanded P140K-MniPSC HSCs, which is significantly higher than levels detected in the other cohorts (Table 1). CD45+ cells in the marrow were predominantly myeloid but lymphoid subsets were also present (10-25% CD3+ cells). Remarkably, the level of gene marking in CFCs and number of gene marked CFCs from mouse bone marrow was substantially higher for mice transplanted with E4ORF1+EC expanded compared to cytokine expanded P140K-MniPSC-HSCs (Table 1). Finally, to confirm engraftment of authentic HSCs, secondary transplants were established. Although engraftment was achieved in all secondary transplanted cohorts, the level of nonhuman primate cells detected was significantly higher in animals transplanted with E4ORF1+EC expanded P140K-MniPSC-HSCs. Significantly more lymphocytes (CD45+CD3+ and CD45+CD56+) and monocytes (CD45+CD14+) were detected in the blood of these secondary transplant recipients. These findings confirm generation of bona fide HSCs derived from nonhuman primate iPSCs and demonstrate that O6BG/BCNU chemotherapy supports in vivo selection of P140K-MniPSC-HSCs generated by co-culture with the E4ORF1+EC vascular platform. Our studies mark a significant advance toward clinical translation of PSC-based blood therapeutics and the development of a nonhuman primate preclinical model. Table 1 CD34+ and CD45+ engraftment and gene marking in the bone marrow of mice transplanted with nonhuman primate HPSCs from MniPSCs and bone marrow. HSCs E4ORF1+ECs O6BG/BCNU Mean %CD34+ Mean %CD45+ % gene marking in CFCs (lentivirus+) total lentivirus+ CFCs per 105 cells GFP-MniPSC + - 3 16 9 ± 2 13 ± 2 P140K-MniPSC + - 4 19 12 ± 5 17 ± 7 P140K-MniPSC - + 0.4 24 3 ± 2 2 ± 1 P140K-MniPSC + + 15 37 27 ± 24 111 ± 96 Mn BM CD34+ - - 2 21 0 0 Disclosures: Nolan: Angiocrine Bioscience: Employment. Ginsberg:Angiocrine Bioscience: Employment. Rafii:Angiocrine Bioscience: Founder Other.


Sign in / Sign up

Export Citation Format

Share Document