GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • American Society of Hematology  (4)
  • 1
    In: Blood, American Society of Hematology, Vol. 128, No. 20 ( 2016-11-17), p. 2457-2461
    Abstract: CXCR4 and α4β1/7 inhibition by AMD3100 and firategrast mobilizes fetal liver HSCs with α4β1/7 inhibition having a stronger effect. Fetal HSC mobilization followed by IUHCT results in increased donor HSC homing to the FL and enhanced long-term allogeneic engraftment.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 35-35
    Abstract: Introduction In utero hematopoietic cell transplantation (IUHCT) is a nonmyeloablative, nonimmunosuppressive allogeneic transplant approach that has the potential to treat a number of congenital disorders, including hemoglobinopathies and immunodeficiencies. Donor cell engraftment at levels high enough to induce donor specific immune tolerance or to treat a target disease has been elusive and remains a major limitation to the clinical application of IUHCT. One of the most significant barriers to high levels of donor cell engraftment is competition with endogenous fetal hematopoietic stem cells (HSCs) for limited hematopoietic niches following transplant. 16,16-dimethyl-prostaglandin E2 (PGE2) and the epoxyeicosatrienoic acids (EETs) 11,12-EET and 14,15-EET have been shown to enhance donor HSC homing, survival, and cell cycling in postnatal murine and zebrafish models of HSC transplantation. We hypothesized that a single ex vivo treatment of donor cells with these eicosanoids would improve donor cell homing and survival following IUHCT,resulting in higher levels of postnatal donor engraftment. Methods Ten million bone marrow (BM) mononuclear cells from C57Bl/6-GFP mice (H2Kb) were injected intravenously into embryonic day (E14) Balb/c fetuses (H2Kd) via the vitelline vein. Donor cells were treated with vehicle, PGE2, 11,12-EET, or 14,15-EET immediately prior to IUHCT. Early homing to and engraftment of the fetal liver (FL) and spleen (FS) of PGE2 and vehicle treated BM cells at 4, 24 and 72 hours after IUHCT was assessed by flow cytometry. Donor cell survival and apoptosis was also assessed in the FL 96 hours post-IUHCT in these two treatment groups by flow cytometric analysis of intracellular expression of survivin (anti-apoptotic) and anti-caspase 3 (pro-apoptotic). Long-term peripheral blood donor cell engraftment was assessed monthly up to 6 months of age and multilineage engraftment (donor T cells, B cells, granulocytes, and macrophages) was determined at 6 months of age in recipients of all donor cell treatment groups. Statistical analysis was performed using ANOVA with BonferroniÕs multiple comparison test or Kruskal-Wallis with DunnÕs multiple comparison test for normal and non-normal data, respectively. Data reported as mean +/- SEM. Results PGE2 pre-treatment produced a significant increase in FL and FS engraftment at 72 hours post-IUHCT compared to vehicle treated donor cells (FL: 37.5 +/- 3.1% vs 21.6 +/- 1.3%; FS: 52.2 +/- 3.7% vs 39.2 +/- 1.8%; p 〈 0.05). There was no significant increase in donor cell engraftment in the FL at earlier time points associated with PGE2 treatment. PGE2 treatment was also associated with increased survival of donor cells compared to vehicle treated cells as indicated by increased donor cell expression of survivin (19.8±5.6% vs. 4.6±1.3%; p 〈 0.05) and decreased donor cell anti-caspase 3 staining (9.3±1.2% vs. 64.1±10.6%; p 〈 0.05) at 96 hours post-IUHCT. These findings of increased FL/FS engraftment and increased survival at 72 to 96 hours post IUHCT following donor cell treatment with PGE2 translated into significantly increased long-term donor cell engraftment up to 6 months of age. Similar to PGE2, ex vivo treatment of donor cells with 11,12-EET or 14,15-EET resulted in enhanced long-term allogeneic donor cell engraftment (Figure 1). Balanced multilineage engraftment was seen for all treatments suggesting enhanced engraftment at the level of the HSC. No deleterious effects were observed on murine growth or overall health. Conclusions The ex vivo treatment of donor cells with eicosanoids including PGE2, 11,12-EET, and 14,15 EET prior to IUHCT results in enhanced long-term multilineage allogeneic donor cell engraftment. Detailed studies of early homing to fetal hematopoietic organs and survival at 96 hours following IUHCT of PGE2 treated donor cells suggest the mechanism of enhanced engraftment is mainly do to a pro-survival effect of PGE2 and related eicosanoids. This work represents a novel application of these eicosanoids in an allogeneic model of IUHCT and highlights their potential to assist in future clinical applications of IUHCT. Disclosures Zon: FATE Therapeutics: Employment, Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Other: Founder; Scholar Rock: Employment, Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Other: Founder.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 2414-2414
    Abstract: Introduction In utero hematopoietic cell transplantation (IUHCT) has tremendous potential for prenatal treatment of congenital hematological disorders, but clinical application has been limited by low engraftment levels. Host cell competition is one of the most formidable barriers to successful engraftment post IUHCT, as fetal stem cells have a proliferative advantage over their postnatal equivalents. Glycogen synthase kinase-3 beta (GSK3β) inhibition has been shown to augment expansion kinetics of bone marrow-derived hematopoietic stem cells (HSC), resulting in enhanced repopulating capacity in vivo. We have developed a strategy that allows targeted, sustained delivery of a GSK3β inhibitor to donor cells by conjugating biodegradable nanocarriers to the cell surface. The aim of the present study was to determine whether donor cell engineering with GSK3β inhibitor-loaded nanoparticles enhances hematopoietic engraftment following IUHCT. Methods Bone marrow mononuclear cells (MNC) were isolated from 6-week old C57BL/6TgN(act-EGFP)OsbY01 mice. GSK3β inhibitor (CHIR99021) loaded multilamelar lipid vehicles (MLV) were synthesized from simple liposomes with the addition of divalent cations to induce liposome fusion. The presence of thiol-reactive maleimide groups in MLV lipid bilayers allowed stable conjugation to the cell membrane of donor MNC. IUHCT was performed in Balb/c mice at E14 and 107 MNC were injected intravenously into each fetus. Donor cell engraftment was assessed in peripheral blood at 4 and 12 weeks of age by flow cytometry (% GFP+ cells within CD45+ population). Comparisons were made between animals that received untreated MNC (control), MNC conjugated with inhibitor-loaded MLV (CHIR99021-MLV), and MNC co-injected with a bolus dose of the inhibitor equivalent to that loaded in MLV (CHIR99021-bolus). Lineage characterization of donor cells was performed using antibodies against lymphoid (CD3, B220), and myeloid (CD11b, Gr-1) cell surface markers. Results are expressed as mean±SEM, and statistical analysis was performed using 1- or 2-way ANOVA with Bonferroni post-hoc tests. Experimental protocols were approved by the Institutional Animal Care and Use Committee at The Children’s Hospital of Philadelphia. Results MLV had a diameter of 489±8nm, and contained 21.7±2.1μg CHIR99021 per 1010 nanoparticles. Inhibitor release was gradual, with 92.7±0.2% of the encapsulated mass released within 7 days. 64±9 nanoparticles were conjugated on each donor cell, resulting in inhibitor dose of 1.3*10-7μg per cell (1.3μg or 4mg/kg per fetus). MLV conjugation did not affect the ability of MNC to migrate to fetal hematopoietic organs post IUHCT. Fetal survival was comparable in control (20/27; 74.1%) and CHIR99021-MLV (10/12; 83.3%) groups, but was reduced in the CHIR99021-bolus group (7/15; 46.7%; p 〈 0.05). Sustained in vivo release of the inhibitor in CHIR99021-MLV animals resulted in increased donor cell engraftment at 4 weeks of age (52.9±2.8%) that was 3 times greater to that observed in control animals (15.4±1.4%; p 〈 0.0001). Bolus CHIR99021 administration had no effect on engraftment (10.5±2.0%; p 〈 0.0001 vs. CHIR99021-MLV, p=0.6 vs. control). Engraftment levels were lower at 12 weeks in control and bolus-CHIR99021 groups (control: 5.9±0.8%, bolus-CHIR99021: 4.5±1.7%; p 〈 0.05 vs. 4 weeks), but were maintained in CHIR99021-MLV offspring (48.3±2.2%; p 〈 0.0001 vs. control and bolus-CHIR99021). Donor cell characterization showed multi-lineage hematopoietic differentiation, with distribution similar to that of host cells and no difference between experimental groups (p=0.6). In vitro pre-treatment for 7 days with the same amount of CHIR99021 per cell resulted in enhanced HSC (lineage-/c-Kit+/Sca-1+) proliferation (p 〈 0.01 vs. control) and augmented their ability to generate hematopoietic progenitors (p 〈 0.05 vs. control), by modulating the Wnt, Notch and Hedgehog pathways. Conclusion Cell engineering with GSK3β inhibitor-loaded synthetic nanoparticles enhances hematopoietic engraftment of BM-MNC following IUHCT. Prolonged retention of the biodegradable nanocarriers on donor cell surfaces enables sustained CHIR99021 release, and allows pseudo-autocrine bioactivity. Conjugation of drug-loaded particles directly to donor cells allows targeted augmentation of HSC function, and could markedly increase the therapeutic potential of IUHCT. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 5800-5800
    Abstract: Introduction In Utero Hematopoietic Cell Transplantation (IUHCT) is a promising therapeutic strategy for congenital hematopoietic disorders. While mixed allogeneic hematopoietic chimerism with associated donor specific tolerance is routinely achieved by a predominant mechanism of central deletion, the critical events of donor and host thymocyte development have not been determined. In this study, we utilized the murine model of allogeneic IUHCT to analyze donor and host thymocyte development in the context of normal immune ontogeny. Methods Bone marrow (BM) cells (10x106) from C57/BL6 (B6, H2kb) mice were injected intravenously into Balb/c (H2kd) fetuses at embryonic day 14 (E14). E14 B6 fetuses injected with GFP B6 BM were used as congeneic controls. At indicated postnatal ages the thymocytes were delineated by multi-color flow cytometry. Cell apoptosis and proliferation were determined by Annexin V staining and in vivo BrdU incorporation, respectively. T cell alloreactivity was assessed by in vitro and in vivo MLR. Results Our findings demonstrate that the thymic processing of donor BM-derived thymocytes differs significantly from host thymocyte processing and from thymocyte development in normal control mice. While the phenotypic development of host thymocytes remained comparable with that of normal control Balb/c mice, the four major subsets of donor thymocytes showed altered distribution, with significantly higher proportions of single positive (SP) cells, and a dramatically lower proportion of CD4+CD8+ double positive (DP) cells, compared to their host-derived counterparts and B6 controls. The extent of the alteration is directly related to both BM chimerism levels and age. Higher levels of chimerism and/or older age are associated with more profound alterations in donor thymocyte distribution. Moreover, DP cells of donor origin show higher apoptosis and lower proliferation than those of the host. Donor TCR gamma/delta cells in DN cells which do not require positive selection based on MHC recognition are relatively increased compared to the host. Moreover, compared with the naive mice and congeneic chimeric mice, the donor BM-derived thymocytes in the allogeneic chimeric mice show increased proportion of DN3 and decreased proportion of DN4, but increased TCRβ+ proportions in both DN3 and DN4 cells, indicating that donor BM-derived thymocyte development is impeded during DN to DP transition, resulting from a MHC-restriction associated mechanism. In addition, in allogeneic chimeric mice, both host and donor BM-derived T cells are tolerant to allogeneic antigens in in vivo and in vitro MLR. Conclusion Our data suggests that in an allogeneic IUHCT system the immune reconstitution of the donor bone marrow-derived thymocytes differs from that of the host cells and that of normal mice. The data supports a mechanism of impaired MHC based positive selection of donor cells by the predominantly host thymic stroma resulting in lack of progression of a higher proportion of donor cells from the DN to DP stage of thymocyte development. Taken together, although donor BM-derived T cells undergo differential thymic development, permanent host-donor two-way tolerance could be achieved in the allogeneic IUHCT mouse model. These findings add to our understanding of the requirements for tolerance induction after IUHCT and have important clinical implications in choosing an optimal donor for IUHCT. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...