Cord Blood CD34+ Stem Cell Sialyl Lewis X Levels and E-Selectin Binding Are Predictive Of Engraftment In Mice: Functional Separation Of Stemness and Homing To Improve Engraftment

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 893-893 ◽  
Author(s):  
Patrick A Zweidler-McKay ◽  
Simon N Robinson ◽  
Michael W Thomas ◽  
JunJun Lu ◽  
Hong Yang ◽  
...  

Abstract Background Cord blood (CB) is an increasingly used alternative to bone marrow and mobilized peripheral blood as a source of hematopoietic tissue for transplantation. However, the relatively low cell dose and significantly delayed engraftment when compared to BM and mPB remain significant hurdles. A deficit in the homing of CB hematopoietic progenitor and stem cells (HPSC) to the hematopoietic microenvironment due to suboptimal expression and/or activity of homing molecules is thought to be responsible in part for the delayed engraftment seen with CB in patients. Sialyl Lewis X (sLeX) bearing cells can bind E-selectin, and we have previously reported that ex vivo fucosylation of CB HPSCs with fucosyltransferase VI or VII enhance the rapidity and magnitude of engraftment in mice. Here we explore the engraftment potential of endogenous sLeX bearing CD34+ CB HPSCs to determine if physiologic levels of E-selectin binding predicts engraftment in murine xenografts. Approach CB cells were sorted with CD34 and HECA-452 (anti-sLex) antibodies. sLeX-bearing CD34+ HPSCs (CD34+HECA+) and CD34+ HPSCs lacking sLeX (CD34+HECA-) were compared phenotypically for stem cell and differentiation markers, by gene expression profiling, E/P/L-selectin binding, colony-forming assays, and for multi-lineage engraftment into NOD-SCID-IL2Rg immune-deficient mice. Results Cord blood CD34+HECA+ cells represent 10-20% of CB MNCs and show no significant phenotypic differences from the CD34+HECA- cells in stem cell (CD133, CD90 (Thy-1), CD117 (c-kit), CD143/BB9) and differentiation (CD38, CD33, CD14, CD3, CD19) markers. In agreement, similar percentages of CD34+CD38- and CD34+CD38+ CB cells were found to be HECA+ (18% and 15% respectively, p=0.38), showing no significant bias toward the more immature CD34+CD38- phenotype. mRNA-seq expression analysis revealed relatively few differences in gene expression patterns, although CD34+HECA+ cells express higher levels of the gamma globin genes HBG1 and HBG2, the components of fetal hemoglobin. As predicted, CD34+HECA+ cells demonstrated significantly increased ability to bind recombinant E-selectin in vitro, with no differences in P- and L-selectin binding. Importantly, colony forming assays revealed a small (30%) disadvantage to the CD34+HECA+ cells revealing that the CD34+HECA+ CB cells do not have enriched stem cells activity by CFU assay. However, CD34+HECA+ cells demonstrated significantly higher rate and magnitude of engraftment when compared to CD34+HECA- cells in three independent NSG experiments (Figure 1). Indeed bone marrow, peripheral blood and splenic levels of human hematopoietic cells were consistently 3-5-fold higher in CD34+HECA+ injected mice than in CD34+HECA- injected controls (Figure 2). Multi-lineage engraftment data reveals higher levels of myeloid (CD33+/CD14+), B-lymphocytes (CD19+/CD20+) and platelets (6-14-fold, CD41a+/CD61+) in CD34+HECA+ cells, but interestingly lower levels of T-lymphocytes (CD3+). Finally, secondary transplants had equal magnitude of engraftment, indicating no bias in short- versus long-term HSPCs. Conclusions Data presented here supports the hypothesis that endogenous sLex levels on CD34+ cells is associated with enhanced engraftment rapidity and magnitude but that this is not reflective of an enriched stem cell fraction. Rather it appears to be an indicator of homing to the bone marrow through E-selectin binding. Functional separation of stemness and homing supports the approach to improve CB transplantation through decoration of CB cells with sLex via ex vivo fucosylation (see clinical trial abstract by Popat and Shpall) Disclosures: No relevant conflicts of interest to declare.

2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Hao Daniel Lin ◽  
Chui-Yee Fong ◽  
Arijit Biswas ◽  
Ariff Bongso

Abstract Background The transplantation of human umbilical cord blood (UCB) CD34+ cells has been successfully used to treat hematological disorders but one major limitation has been the low cell numbers available. Mesenchymal stem cells (MSCs) lying within the bone marrow in vivo behave like a scaffold on which CD34+ cells interact and proliferate. We therefore evaluated the use of allogeneic MSCs from the human UC Wharton’s jelly (hWJSCs) as stromal support for the ex vivo expansion of CD34+ cells. Methods We performed an in-depth evaluation of the primitiveness, migration, adhesion, maturation, mitochondrial behavior, and pathway mechanisms of this platform using conventional assays followed by the evaluation of engraftment potential of the expanded CD34+ cells in an in vivo murine model. Results We demonstrate that hWJSCs and its conditioned medium (hWJSC-CM) support the production of significantly high fold changes of CD34+, CD34+CD133+, CD34+CD90+, CD34+ALDH+, CD34+CD45+, and CD34+CD49f+ cells after 7 days of interaction when compared to controls. In the presence of hWJSCs or hWJSC-CM, the CD34+ cells produced significantly more primitive CFU-GEMM colonies, HoxB4, and HoxA9 gene expression and lower percentages of CD34+CXCR4+ cells. There were also significantly higher N-cadherin+ cell numbers and increased cell migration in transwell migration assays. The CD34+ cells expanded with hWJSCs had significantly lower mitochondrial mass, mitochondrial membrane potential, and oxidative stress. Green Mitotracker-tagged mitochondria from CD34+ cells were observed lying within red CellTracker-tagged hWJSCs under confocal microscopy indicating mitochondrial transfer via tunneling nanotubes. CD34+ cells expanded with hWJSCs and hWJSC-CM showed significantly reduced oxidative phosphorylation (ATP6VIH and NDUFA10) and increased glycolytic (HIF-1a and HK-1) pathway-related gene expression. CD34+ cells expanded with hWJSCs for 7 days showed significant greater CD45+ cell chimerism in the bone marrow of primary and secondary irradiated mice when transplanted intravenously. Conclusions In this report, we confirmed that allogeneic hWJSCs provide an attractive platform for the ex vivo expansion of high fold numbers of UCB CD34+ cells while keeping them primitive. Allogeneic hWJSCs are readily available in abundance from discarded UCs, can be easily frozen in cord blood banks, thawed, and then used as a platform for UCB-HSC expansion if numbers are inadequate.


2018 ◽  
Vol 70 (3) ◽  
pp. 433-441
Author(s):  
Fatemeh Mohammadali ◽  
Saeid Abroun ◽  
Amir Atashi

Cord blood (CB) is a rich source of hematopoietic stem cells (HSC). It has been used successfully to treat a variety of hematological and non-hematological disorders. Beside the advantages of CB, its main disadvantages are the limited number of stem cells available for transplantation and delayed engraftment. Identifying strategies to enhance expansion and self-renewal of HSCs can improve transplantation efficiency. The aim of this study was to examine different culture conditions on ex vivo expansion and homeobox protein Hox-B4 gene (HOXB4) expression in cord blood CD34+ stem cells. Human cord blood CD34+ HSC were cultured in serum-free medium supplemented with cytokines with and without a feeder layer in normoxia (21% O2) and mild hypoxia (5% O2). At the end of 7 days of culture, the highest number of total nucleated cells (TNC), CD34+ cells, colony forming units (CFUs) and HOXB4 mRNA were observed in a co-culture of HSC with a bone marrow mesenchymal stem cell(MSC) feeder layer at 5% O2.We concluded that the combination of bone marrow (BM)-MSC and mild hypoxia (5% O2) not only improved HSC expansion but also enhanced HOXB4 gene expression as a self-renewal marker of HSC, and better mimicked the niche microenvironment conditions.


Biomedicines ◽  
2020 ◽  
Vol 8 (6) ◽  
pp. 157
Author(s):  
Nicole Zarniko ◽  
Anna Skorska ◽  
Gustav Steinhoff ◽  
Robert David ◽  
Ralf Gaebel

Several cell populations derived from bone marrow (BM) have been shown to possess cardiac regenerative potential. Among these are freshly isolated CD133+ hematopoietic as well as culture-expanded mesenchymal stem cells. Alternatively, by purifying CD271+ cells from BM, mesenchymal progenitors can be enriched without an ex vivo cultivation. With regard to the limited available number of freshly isolated BM-derived stem cells, the effect of the dosage on the therapeutic efficiency is of particular interest. Therefore, in the present pre-clinical study, we investigated human BM-derived CD133+ and CD271+ stem cells for their cardiac regenerative potential three weeks post-myocardial infarction (MI) in a dose-dependent manner. The improvement of the hemodynamic function as well as cardiac remodeling showed no therapeutic difference after the transplantation of both 100,000 and 500,000 stem cells. Therefore, beneficial stem cell transplantation post-MI is widely independent of the cell dose and detrimental stem cell amplification in vitro can likely be avoided.


Blood ◽  
2000 ◽  
Vol 95 (1) ◽  
pp. 102-110 ◽  
Author(s):  
Craig Dorrell ◽  
Olga I. Gan ◽  
Daniel S. Pereira ◽  
Robert G. Hawley ◽  
John E. Dick

Abstract Current procedures for the genetic manipulation of hematopoietic stem cells are relatively inefficient due, in part, to a poor understanding of the conditions for ex vivo maintenance or expansion of stem cells. We report improvements in the retroviral transduction of human stem cells based on the SCID-repopulating cell (SRC) assay and analysis of Lin− CD34+CD38−cells as a surrogate measure of stem cell function. Based on our earlier study of the conditions required for ex vivo expansion of Lin−CD34+ CD38− cells and SRC, CD34+–enriched lineage–depleted umbilical cord blood cells were cultured for 2 to 6 days on fibronectin fragment in MGIN (MSCV-EGFP-Neo) retroviral supernatant (containing 1.5% fetal bovine serum) and IL-6, SCF, Flt-3 ligand, and G-CSF. Both CD34+CD38− cells (20.8%) and CFC (26.3%) were efficiently marked. When the bone marrow of engrafted NOD/SCID mice was examined, 75% (12/16) contained multilineage (myeloid and B lymphoid) EGFP+ human cells composing as much as 59% of the graft. Half of these mice received a limiting dose of SRC, suggesting that the marked cells were derived from a single transduced SRC. Surprisingly, these culture conditions produced a large expansion (166-fold) of cells with the CD34+CD38− phenotype (n = 20). However, there was no increase in SRC numbers, indicating dissociation between the CD34+CD38− phenotype and SRC function. The underlying mechanism involved apparent downregulation of CD38 expression within a population of cultured CD34+CD38+ cells that no longer contained any SRC function. These results suggest that the relationship between stem cell function and cell surface phenotype may not be reliable for cultured cells. (Blood. 2000;95:102-110)


Cancers ◽  
2019 ◽  
Vol 11 (2) ◽  
pp. 181 ◽  
Author(s):  
Maud Plantinga ◽  
Colin G. de Haar ◽  
Ester Dünnebach ◽  
Denise A.M.H. van den Beemt ◽  
Kitty W.M. Bloemenkamp ◽  
...  

Dendritic cells (DCs) are professional antigen-presenting cells which instruct both the innate and adaptive immune systems. Once mature, they have the capacity to activate and prime naïve T cells for recognition and eradication of pathogens and tumor cells. These characteristics make them excellent candidates for vaccination strategies. Most DC vaccines have been generated from ex vivo culture of monocytes (mo). The use of mo-DCs as vaccines to induce adaptive immunity against cancer has resulted in clinical responses but, overall, treatment success is limited. The application of primary DCs or DCs generated from CD34+ stem cells have been suggested to improve clinical efficacy. Cord blood (CB) is a particularly rich source of CD34+ stem cells for the generation of DCs, but the dynamics and plasticity of the specific DC lineage development are poorly understood. Using flow sorting of DC progenitors from CB cultures and subsequent RNA sequencing, we found that CB-derived DCs (CB-DCs) exclusively originate from CD115+-expressing progenitors. Gene set enrichment analysis displayed an enriched conventional DC profile within the CD115-derived DCs compared with CB mo-DCs. Functional assays demonstrated that these DCs matured and migrated upon good manufacturing practice (GMP)-grade stimulation and possessed a high capacity to activate tumor-antigen-specific T cells. In this study, we developed a culture protocol to generate conventional DCs from CB-derived stem cells in sufficient numbers for vaccination strategies. The discovery of a committed DC precursor in CB-derived stem cell cultures further enables utilization of conventional DC-based vaccines to provide powerful antitumor activity and long-term memory immunity.


2006 ◽  
Vol 37 (4) ◽  
pp. 359-366 ◽  
Author(s):  
S N Robinson ◽  
J Ng ◽  
T Niu ◽  
H Yang ◽  
J D McMannis ◽  
...  

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3471-3471
Author(s):  
Sarah Vaiselbuh ◽  
Jeffrey Michael Lipton ◽  
Johnson M. Liu

Abstract CD133 (prominin-1) is the first in a class of novel pentaspan membrane proteins identified in humans and mice, and studies have since confirmed the utility of CD133 as a marker of stem cells with hematopoietic and non-hematopoietic lineage potential. A number of human transplantation studies have documented hematopoietic reconstitution from CD133+ stem cells from mismatched donors, with a suggested advantage over standard grafts in avoidance of graft versus host disease. We have developed a novel hematopoietic culture system (Long-Term Stem Cell Culture or LTSCC) to investigate the potential of human mesenchymal stem cells (MSC) to form stroma that can support short- and long-term hematopoiesis derived from cord blood (CB)-derived CD133+ cells. In addition, we analyzed the effect of stromal derived factor-1 (SDF-1/CXCL12) on survival and short-and long-term colony-forming capacity of CD133+ hematopoiesis. LTSCC induced stroma-like changes in the MSC feeder layer, with adipocyte formation, thought to be needed for formation of stem cell niches, and supported long-term (>9 weeks) survival of CB-CD133+ cells. Cobblestone areas of active CD133-derived hematopoiesis were seen in LTSCC for up to 9 weeks of culture. SDF-1/CXCL12 acted as a survival factor for CB-CD133+ cells and induced a significant ex vivo cell expansion at weeks 3 and 4 of LTSCC (maximal 500-fold increase), while maintaining the capacity for CFU-Mix and BFU-E colony formation up to 7 weeks. Long-term hematopoiesis was assessed by enumeration of long-term culture initiating cells (LTC-IC). When SDF-1/CXCL12 was added to LTSCC, we found a significant increase in LTC-IC: 0.3% (+SDF-1/CXCL12) vs. 0.05% (-SDF-1/CXCL12). Finally, homing capacity, as defined by SDF-1/CXCL12-induced adhesion and migration of CB-CD133+ cells, was maintained and even increased during the first 3 weeks of LTSCC. In summary, MSC can be maintained in LTSCC medium, and this simplified feeder layer is able to provide niches for cobblestone area forming cells derived from CB-CD133+ cells. SDF-1/CXCL12 is critical to support the survival and expansion of CD133+ cells, either directly or indirectly by paracrinesignaled retention of CD133+ cells in contact with specialized MSC niches. We suggest that expansion of CD133+ cells from cord blood may be useful in clinical transplantation limited by insufficient numbers of stem cells.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 92-92
Author(s):  
Klaus Rehe ◽  
Kerrie Wilson ◽  
Simon Bomken ◽  
Hesta McNeill ◽  
Martin Stanulla ◽  
...  

Abstract Abstract 92 Research on cancer stem cells, cells that self-renew and reconstitute the full phenotype of the original malignancy, has yielded controversial results regarding their frequency and identity for many cancers. The hierarchical stem cell model has been well established in some malignancies such as acute myeloid leukemia and states that only rare, immunophenotypically immature blasts harbor stem cell activity, resembling a normal physiological hierarchy. The opposing stochastic model proposes that stemness in cancer cells is supported by extrinsic stimuli and that a substantial fraction of malignant cells have this potential. Continued optimization of in vivo xenotransplantation modeling recently caused a paradigm shift for some cancers, for example in malignant melanoma where stem cell activity was found in as many as 1 in 4 cells. For acute lymphoblastic leukemia (ALL) we and others previously challenged the hierarchical model by demonstrating that both immature and more mature leukemic blasts contain self-renewal properties (Cancer Cell 2008, 14(1), p47-58). In this study we address the frequency of leukemic stem cells in the bulk leukemia and also, more specifically, in subpopulations of different blast maturity by using unsorted and highly purified flow sorted cell fractions. Primary patient material as well as leukemic blasts harvested from engrafted mouse bone marrow (secondary and tertiary material) were sorted for their CD10, CD20 or CD34 expression followed by orthotopic intrafemoral transplantation into severely immunocompromised NOD/scid IL2Rγnull (NSG) mice. Engraftment of transplanted CD19+CD10low and CD19+CD10high, CD19+CD20low and CD19+CD20high and CD19+CD34low and CD19+CD34high blast populations was monitored by 5 color flow cytometry using material from consecutive bone marrow punctures, final bone marrow harvests and/or single cell suspensions from spleens. Primary ALL samples from 15 high risk (BCR/ABL positive (n=8), BCR/ABL like ALL (n=2), high hyperdiploid/MRD positive (n=2), MRD positive (n=1), MLL/AF4 (n=2)), 3 intermediate risk (high WBC/MRD negative (n=2), age >10 years (n=1)) and 3 standard risk (n=3) patients were included. Cells sorted into CD19+CD10low and CD19+CD10high fractions were transplanted from primary patient material (n=4, HR; n=1, SR) and from secondary samples (n=4, HR; n=1; IR) with cells from one HR patient used at limiting dilutions. As few as 100 sorted cells of either fraction were sufficient to repopulate the leukemia. CD19+CD20high and CD19+CD20 low fractions from primary (n=7, HR; n=1, IR), secondary (n=5, HR; n=1, IR) and tertiary material (n=2, HR; n=1, IR) engrafted NSG mice. Limiting dilutions were performed on secondary (n=4, HR) and tertiary material (n=2, HR). Cell numbers required for engraftment varied between leukemias with as few as 100 cells being sufficient to cause engraftment. Limiting dilution experiments using CD19+CD34high and CD19+CD34low fractions from secondary (n=1, HR) and tertiary (n=1, HR) material yielded engraftment with as few as 10 CD19+CD34high and 100 CD19+CD34low cells. Similarly, unsorted primary (n=11, HR; n=2, IR), secondary (n=2, HR) and tertiary material (n=1, HR) required as few as 10 cells for leukemic reconstitution. Taken together, both unsorted and sorted blasts of all immunophenotypes and transplanted with low numbers were able to reconstitute the complete original phenotype of the patient leukemia. All limiting dilutions were transplanted down to 10 cells per mouse and those mice not engrafted yet are still under observation. Furthermore, the ability to self-renew was demonstrated by serial transplantation. Finally, we compared expression of self-renewal associated genes (BMI1, EZH2, HMGA2, MEIS1, TERT) in CD19+CD34low and CD19+CD34high fractions of 5 HR and 1 SR samples with that in cord blood. Interestingly, expression of these genes was not dependent on the CD34 status of the leukemic cells, whereas HMGA2, MEIS1 and TERT were upregulated in CD34+ cord blood cells. In summary we provide strong evidence for the stochastic cancer stem cell model in B precursor ALL by demonstrating that (i) a broad spectrum of blast immunophenotypes exhibit stem cell characteristics and (ii) that this stemness is highly frequent among ALL cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4321-4321
Author(s):  
Miguel Ganuza Fernandez ◽  
Per Holmfeldt ◽  
Himangi Marathe ◽  
Trent Hall ◽  
Jennifer Pardieck ◽  
...  

Abstract Introduction: Hematopoiesis involves the hierarchical generation of the major blood lineages from a common ancestor, the Hematopoietic Stem Cell (HSC). HSC also have the intrinsic ability to repopulate an ablated hematopoietic compartment when introduced into the periphery of a recipient. This has allowed Hematopoietic Stem Cell transplantation (HSCT) to be used as a cell therapy over the last 45 years, benefiting thousands of patients. Unfortunately many patients succumb to disease while waiting for an adequate donor. Others have to undergo unrelated donor transplants, putting themselves at a risk of developing graft-versus-host disease. Improving HSC engraftment could ameliorate transplant morbidity. Thus, understanding mechanisms regulating HSC engraftment is key. Results: We used our recently published gene expression profiles of developing HSC and other public databases to prioritize 58 genes as putative regulators of adult HSC function. We confirmed by qRT-PCR that 51/58 candidates were enriched for gene expression in Lineage-Sca-1+c-Kit+ (LSK) bone marrow cells relative to downstream progeny, suggesting a role in hematopoietic stem and progenitor cells (HSPC). To functionally assay a role for each gene of interest (GOI) in HSC engraftment, we designed and validated ≥2 independent shRNAs/GOI that effected a >75% knockdown in gene expression in LSK cells. LSK cells were lentivirally transduced with control or individual gene-specific shRNAs and transplanted into lethally irradiated recipients along with mock-transduced LSK competitor cells congenic at the CD45 allele. In contrast to previous functional screens, transplant was performed within 24-hours of LSK cell isolation, avoiding extensive ex vivo culture. This minimal manipulation allowed us to detect genes critical for efficient HSC engraftment. Peripheral blood chimerism was analyzed for at least 16 weeks post-transplant. The major bone marrow hematopoietic compartments were also analyzed. 17 of 48 genes tested were identified as necessary for optimal HSPC engraftment (i.e. knockdown induced a significant loss of repopulation) and the knockdown of three genes enhanced HSPC repopulation. shRNAs targeting each “Hit” were interrogated ex vivo for non-specific effects on LSK cell viability and expansion. A 2° screen was performed to validate the results of this primary screen. Here, CD45.2 LSK cells transduced with control or individual gene-specific shRNAs were sorted 48 hours post-transduction for mCherry+ cells and then transplanted into lethally irradiated mice with mock-transduced and mock-sorted CD45.1 congenic LSK cells. 18 “Hits” were confirmed to perturb HSC repopulating potential in this 2° screen, including three whose loss enhanced HSPC repopulation. The transcription factor, Foxa3, is one hit identified here as necessary for HSC repopulation. We further found that that Foxa3-/- bone marrow displays a significant loss of repopulating potential >16 weeks post-transplant, confirming the results of our screen. As Foxa3-/- long-term HSC also display reduced colony forming potential in vitro and fail to contribute to downstream progenitor compartments in transplant recipients, we propose that Foxa3 is a novel regulator of HSC differentiation post-transplant. Foxa3 has never before been implicated in hematopoiesis or HSPC biology. Conclusions: Our novel functional screen has revealed 15 genes required for optimal HSPC engraftment and three genes whose knockdown improved HSPC engraftment. We further validated Foxa3 as a novel regulator of HSC activity by demonstrating that Foxa3-/- HSC are also deficient in repopulating activity. We are currently investigating the molecular mechanism of Foxa3’s role in HSC and, given that Foxa genes are known transcriptional pioneering factors, pursuing the hypothesis that Foxa3 functions as a novel epigenetic regulator of HSC activation and differentiation. Each gene identified in our screen represents a window into the discovery of novel mechanisms regulating HSC biology and engraftment. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document