scholarly journals 207. In Vivo Assessment of the Efficacy of Gene Therapy Targeted to Diseased Human Hematopoietic Progenitor Cells in a Xenotransplantable Murine Model of a Lysosomal Storage Disorder

2003 ◽  
Vol 7 (5) ◽  
pp. S81-S82 ◽  
Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1463-1463
Author(s):  
Filippo Milano ◽  
Ian Nicoud ◽  
Daniel Weber ◽  
Shelly Heimfeld ◽  
Irwin D. Bernstein ◽  
...  

Abstract Abstract 1463 Introduction: Practical clinical countermeasures that could enhance survival after accidental or deliberate radiation exposure are lacking. We have previously demonstrated that culture of murine hematopoietic stem/progenitor cells in the presence of immobilized Notch ligand Delta1 results in a multi-log increase in the number of lin-/sca-1+/c-kit+ (LSK) cells with short term lymphoid and myeloid repopulating ability. Here we show that Delta1 cultured LSK cells, cryopreserved after expansion, can be an effective therapy in a murine model of the hematopoietic acute radiation syndrome (hARS). Methods: Whole bone marrow (BM) was obtained from Ly5a mice (H-2b, CD45.1). LSK cells were isolated by flow cytometry and placed in culture in the presence of either immobilized Delta1 or control human IgG. Serum free conditions were used, consisting of Iscove's medium supplemented with cytokines mSCF, hFlt-3 ligand, hIL-6, and hIL-11. After 14 days, expanded cells were harvested and cryopreserved in 90% FBS + 10% DMSO. On the day of transplant, frozen cells were thawed, washed, and resuspended in PBS + 1% FBS; manual viable cell counts and LSK phenotyping were performed prior to tail vein infusion with escalating doses of Delta1 or IgG cultured cells at doses of 1, 3, 5, or 10 × 106 (Delta-1 group only at 10×106) into supralethally irradiated (8.5 Gy) MHC-mismatched Balb/c mice (H-2d, CD45.2). Peripheral blood (PB) and BM were collected from mice at 1, 2, 3 and 4 weeks after transplantation for chimerism determination by cytofluorometry. Results: Culture with Delta1 resulted in significantly greater increases in absolute numbers of LSK cells (7.2 × 104-fold expansion) as compared to growth with control IgG (0.8 × 104-fold expansion). Approximately 90% of viable LSK cells were recovered post thaw. PB samples from recipient Balb/c mice receiving Delta1 cells demonstrated significantly higher Ly5a+ donor cell engraftment as compared to recipients receiving IgG-cultured LSK cells (p=0.0001). Donor IgG cultured cells were detectable only at day 7, whereas cells grown in the presence of Delta1 persisted through day 14 and 21. Only mice transplanted with Delta1-expanded cells showed engraftment in marrow, although by 2 weeks donor cells had decreased substantially. No signs of graft versus host disease (GVHD) were observed. Survival was significantly prolonged among mice that received Delta1-cultured cells, whereas all mice that received IgG cultured cells died within the first 3 weeks after irradiation (p=0.0001). Overall survival at day 30 was 11, 20, 26 and 63 percent after receiving 1, 3, 5 and 10 × 106 Delta1-cultured cells, respectively. Mice that received 10 × 106 cultured cells showed a statistically significant better survival (p=0.02), demonstrating a dose response relationship with the highest survival observed in mice that received the highest dose of expanded cells. Conclusions: Using the Notch ligand Delta1 for the ex vivo expansion of murine hematopoietic progenitor cells, we have demonstrated that the cultured cells can be efficiently cryopreserved without loss of in vivo function. Infusion of Notch-expanded and cryopreserved cells into lethally irradiated mismatched recipients demonstrated that short-term engraftment without manifestations of GVHD can be achieved across major H-2 barriers and resulted in. significantly enhanced survival in a dose dependent manner. We have previously demonstrated that culture of human cord blood CD34+ cells in the presence of Delta1 also results in a significant increase in the absolute number of hematopoietic progenitor cells that are capable of rapid myeloid reconstitution in vivo. The findings presented herein thus support further development of a parallel human ex vivo expanded and cryopreserved product for clinical application in a non-HLA matched setting. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1998 ◽  
Vol 91 (8) ◽  
pp. 2664-2671 ◽  
Author(s):  
Punam Malik ◽  
Timothy C. Fisher ◽  
Lora L.W. Barsky ◽  
Licheng Zeng ◽  
Parvin Izadi ◽  
...  

Hemoglobinopathies, such as β-thalassemias and sickle cell anemia (SCA), are among the most common inherited gene defects. Novel models of human erythropoiesis that result in terminally differentiated red blood cells (RBCs) would be able to address the pathophysiological abnormalities in erythrocytes in congenital RBC disorders and to test the potential of reversing these problems by gene therapy. We have developed an in vitro model of production of human RBCs from normal CD34+ hematopoietic progenitor cells, using recombinant growth factors to promote terminal RBC differentiation. Enucleated RBCs were then isolated to a pure population by flow cytometry in sufficient numbers for physiological studies. Morphologically, the RBCs derived in vitro ranged from early polylobulated forms, resembling normal reticulocytes to smooth biconcave discocytes. The hemoglobin pattern in the in vitro-derived RBCs mimicked the in vivo adult or postnatal pattern of β-globin production, with negligible γ-globin synthesis. To test the gene therapy potential using this model, CD34+ cells were genetically marked with a retroviral vector carrying a cell-surface reporter. Gene transfer into CD34+ cells followed by erythroid differentiation resulted in expression of the marker gene on the surface of the enucleated RBC progeny. This model of human erythropoiesis will allow studies on pathophysiology of congenital RBC disorders and test effective therapeutic strategies.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2674-2674
Author(s):  
Seiji Fukuda ◽  
Hal E. Broxmeyer ◽  
Louis M. Pelus

Abstract The Flt3 receptor tyrosine kinase (Flt3) is expressed on primitive normal and transformed hematopoietic cells and Flt3 ligand (FL) facilitates hematopoietic stem cell mobilization in vivo. The CXC chemokine SDF-1α(CXCL12) attracts primitive hematopoietic cells to the bone marrow microenvironment while disruption of interaction between SDF-1α and its receptor CXCR4 within bone marrow may facilitate their mobilization to the peripheral circulation. We have previously shown that Flt3 ligand has chemokinetic activity and synergistically increases migration of CD34+ cells and Ba/F3-Flt3 cells to SDF-1α in short-term migration assays; this was associated with synergistic phosphorylation of MAPKp42/p44, CREB and Akt. Consistent with these findings, over-expression of constitutively active ITD (internal tandem duplication) Flt3 found in patients with AML dramatically increased migration to SDF-1α in Ba/F3 cells. Since FL can induce mobilization of hematopoietic stem cells, we examined if FL could antagonize SDF-1α/CXCR4 function and evaluated the effect of FL on in vivo homing of normal hematopoietic progenitor cells. FL synergistically increased migration of human RS4;11 acute leukemia cells, which co-express wild-type Flt3 and CXCR4, to SDF-1α in short term migration assay. Exogenous FL had no effect on SDF-1α induced migration of MV4-11 cells that express ITD-Flt3 and CXCR4 however migration to SDF-1α was partially blocked by treatment with the tyrosine kinase inhibitor AG1296, which inhibits Flt3 kinase activity. These results suggest that FL/Flt3 signaling positively regulates SDF-1α mediated chemotaxis of human acute leukemia cells in short-term assays in vitro, similar to that seen with normal CD34+ cells. In contrast to the enhancing effect of FL on SDF-1α, prolonged incubation of RS4;11 and THP-1 acute myeloid leukemia cells, which also express Flt3 and CXCR4, with FL for 48hr, significantly inhibited migration to SDF-1α, coincident with reduction of cell surface CXCR4. Similarly, prolonged exposure of CD34+ or Ba/F3-Flt3 cells to FL down-regulates CXCR4 expression, inhibits SDF-1α-mediated phosphorylation of MAPKp42/p44, CREB and Akt and impairs migration to SDF-1α. Despite reduction of surface CXCR4, CXCR4 mRNA and intracellular CXCR4 in Ba/F3-Flt3 cells were equivalent in cells incubated with or without FL, determined by RT-PCR and flow cytometry after cell permeabilization, suggesting that the reduction of cell surface CXCR4 expression is due to accelerated internalization of CXCR4. Furthermore, incubation of Ba/F3-Flt3 cells with FL for 48hr or over-expression of ITD-Flt3 in Ba/F3 cells significantly reduced adhesion to VCAM1. Consistent with the negative effect of FL on in vitro migration and adhesion to VCAM1, pretreatment of mouse bone marrow cells with 100ng/ml of FL decreased in vivo homing of CFU-GM to recipient marrow by 36±7% (P<0.01), indicating that FL can negatively regulate in vivo homing of hematopoietic progenitor cells. These findings indicate that short term effect of FL can provide stimulatory signals whereas prolonged exposure has negative effects on SDF-1α/CXCR4-mediated signaling and migration and suggest that the FL/Flt3 axis regulates hematopoietic cell trafficking in vivo. Manipulation of SDF-1α/CXCR4 and FL/Flt3 interaction could be clinically useful for hematopoietic cell transplantation and for treatment of hematopoietic malignancies in which both Flt3 and CXCR4 are expressed.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1206-1206 ◽  
Author(s):  
Heather A. O'Leary ◽  
Charlie Mantel ◽  
Xianyin Lai ◽  
Scott Cooper ◽  
Giao Hangoc ◽  
...  

Abstract DPP4 (CD26) is a dipeptidyl peptidase that functions by enzymatically cleaving the penultimate proline, alanine or select other amino acids such as serine of proteins, resulting in functional alterations of the protein. We recently published that many cytokines, chemokines and growth factors have putative DPP4 truncations sites and that DPP4 specifically was able to truncate some colony stimulating factors such as GM-CSF and IL-3 with resultant blunting of their activity. However, the mechanism of action of the truncated factors is still unknown and requires further investigation. The expression, and activity, of DPP4 is relevant in normal and malignant hematopoiesis as we have data showing that CD34+ umbilical cord blood cells (UCB) as well as Acute Myelogenous Leukemia (AML) patient samples express active DPP4. Further, specific inhibition of DPP4 increases homing and engraftment of both human UCB and mouse bone marrow cells after transplantation in mice indicating the therapeutic potential of DPP4 activity altering compounds. Due to its potential importance in disease states, and their subsequent treatment, it is relevant to study how the activity of DPP4 alters the functions of the molecules it cleaves, and subsequently their interactions with each other. DPP4 can cleave the penultimate proline of GM-CSF and IL-3 resulting in truncated forms which have blunted colony stimulating factor activity for hematopoietic progenitor cells (HPC). Since GM-CSF and IL-3 receptors share a common receptor beta chain, we investigated if DPP4 truncation of GM-CSF (TGM) or IL-3 (T3) could inhibit the receptor binding and functional activity of the full length (FL) alternate compound (i.e TGM inhibition of FL3 activity or T3 inhibition of FLGM activity) in the factor dependent TF-1 cell line, UCB cells and in in vivo mouse studies. We determined using TF-1 and UCB that both T3 and TGM bound to the receptors with higher affinity than their FL forms and could blunt the receptor binding of the FLGM and FL3. Additionally, TGM and T3 decreased colony formation induced by either FLGM or FL3 in both TF-1, UCB, and primary AML patient cell samples. Strikingly, this inhibition of colony formation did not require a 1:1 ratio of the full length to truncated forms of these cytokines. Rather, approximately 4-10 fold less truncated molecules could be used to efficiently inhibit the colony formation activity of the full length form, even across molecules. In vivo injection of FL, T, or a mixture of FL/T or T/T factors into DPP4 activity knockout mice followed by colony assays showed the TGM and T3 suppresed the effect of FLGM or FL3 on progenitor cell numbers per femur and diminished cycling of hematopoietic progenitor cells as detected by high specificity tritiated thymidine kill assay. Proteomic analysis of the effects of full length and truncated factors (FLGM, FL3, TGM, T3) were performed with TF-1 cells where we detected differential protein regulation by the full length vs truncated factors. After 24 hour treatment with 10ng/ml of FLGM or TGM, TF-1 cells displayed statistically significant (p < .05) differences in 26 proteins of which 17 were higher in the FL vs the T, and 9 higher in the T vs FL treated groups. These proteins included, but were not limited to, cell cycle proteins such as CDK6, HDAC6, as well as signal transduction proteins and redox control proteins such as STAM1 and Glutaredoxin. Additionally, alterations in protein phosphphorylation were detected for TF-1 cells treated for 15 or 30 min with the full length vs truncated IL-3 and GM-CSF proteins. Interestingly, the protein expression or phosphorylation levels were not always decreased by the truncated protein compared to the full length. In some cases, the truncated molecules induced an increase in the protein expression or phosphorylation. These data suggest interesting roles for full length and truncated GM-CSF and IL-3 in both normal and malignant hematopoiesis. Further investigation into the regulation of DPP4, and the roles that full length and truncated factors play during normal and malignant hematopoiesis, are important and will allow for a better understanding of the signficance of DPP4 activity during steady state, stressed, and disease hematopoiesis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4718-4718
Author(s):  
Mayte Medrano ◽  
María Victoria Barbado-Gonzalez ◽  
Nuria Campillo ◽  
Francisco Hidalgo ◽  
Teresa Caballero-Velazquez ◽  
...  

Abstract Endocannabinoid system is a set of ligands, receptors and endogenous enzymes which modulate a variety of physiological effects. There are two well-characterized cannabinoid receptors, CB1 (mainly expressed in Central Nervous System) and CB2 (mainly in hematopoietic cells). Here, we tested the effect of the cannabinoid WIN-55 212-2 in acute myeloid leukemia (AML) in vitro, ex vivo and in vivo and studied the molecular signaling pathways involved in this effect. Moreover, we synthesized a new family of twelve cannabinoids that are specific to CB2 receptor. For their design and synthesis, computational techniques of docking, analytical and spectroscopic techniques such as mass spectrometry (MS) were used. To assess the anti-leukemia effect of the different cannabinoids, we analyzed cell viability by MTT and flow cytometry using six human AML cell lines, primary cells from healthy donors (hematopoietic progenitor cells (HPC) and lymphocytes) and blasts from AML patients. Cannabinoids induced a potent proapoptotic effect on AML cell lines and on primary leukemic cells, which was not observed in normal HPC and lymphocytes from healthy donors. Fragmentation of PARP and activation of caspases 2, 3, 8 and 9 were confirmed by western-blot. Other proteins involved in the effect of cannabinoids were p-AKT, p-ERK 1/2, p-38 and p- JNK. Also studies on p-PERK, p-IRE1 and CHOP confirmed an increased endoplasmic reticulum stress upon exposure to cannabinoids. Mitochondrial damage was analyzed by flow cytometry using TMRE and by MitoSOXTMRed. These assays confirmed a very early mitochondrial damage in leukemic cells which was not observed in normal hematopoietic progenitor cells. Moreover, we analyzed the ceramide levels, a membrane lipid associated with death/survival cell processes by HPLC and immunohistochemistry. Remarkably, we observed significant differences in the amounts of certain subtypes of ceramides in untreated versus treated leukemic cells. The proapoptotic effect of cannabinoids on AML cells was abolished upon co-culture with either CB2 receptor antagonists or with pancaspase inhibitors. Finally, NOD/scid/IL-2R gammae null (NSG) mice were xenotransplanted with HL60 cell line. We confirmed disease infiltration in bone marrow (BM) by BM aspirates and flow cytometry assays. Once the presence of leukemic cells was confirmed, treatment with vehicle, WIN-55 cannabinoid at a dose of 5 mg/kg/day or citarabine (ARA-C) at 50 mg/kg during 5 days was administered. We observed a significantly increased survival among mice treated with WIN-55 cannabinoid as compared to both the control group and the group treated with ARA-C. In addition, we tested in vivo the effect of these compounds on normal hematopoiesis by treating healthy BALB-C mice. We confirmed that cannabinoids did not affect the viability of the different populations of hematopoietic progenitors (LK, GMP, CMP) and, moreover, an increased platelet count was observed in treated mice. Our findings indicate that cannabinoids display a highly selective proapoptotic effect against leukemic cells. Several pathways are involved in this effect, the modification in the ceramide pattern playing a main role. Figure 1 Figure 1. Figure 2 Figure 2. Figure 3 Figure 3. Disclosures No relevant conflicts of interest to declare.


Haematologica ◽  
2017 ◽  
Vol 102 (9) ◽  
pp. 1558-1566 ◽  
Author(s):  
Jana Reimer ◽  
Sabine Knöß ◽  
Maurice Labuhn ◽  
Emmanuelle M. Charpentier ◽  
Gudrun Göhring ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document