Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Nature, Springer Science and Business Media LLC, Vol. 523, No. 7561 ( 2015-07-23), p. 468-471
    Type of Medium: Online Resource
    ISSN: 0028-0836 , 1476-4687
    RVK:
    RVK:
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2015
    detail.hit.zdb_id: 120714-3
    detail.hit.zdb_id: 1413423-8
    SSG: 11
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 2344-2344
    Abstract: Abstract 2344 The first hematopoietic stem cells (HSC) that give rise to robust, long-term, multi-lineage reconstitution in irradiated adult recipients arise in the murine embryo at embryonic day 11.5 (E11.5). However, long-term multi-lineage engraftment in neonatal recipients has been observed from E9.0 yolk sac, suggesting that the neonatal hematopoietic microenvironment is more permissive for engraftment of embryonic HSCs. To resolve the apparent discrepancy between the numbers of candidate HSCs detected by direct visualization in the early embryo, relative to the numbers that can be measured by limiting dilution, we sought to characterize engraftment of neonatal recipients versus adult recipients with hematopoietic populations dissected from the aorta-gonad-mesonephros (AGM) region of the early embryo, the first putative site of intraembryonic origin of definitive HSCs. We dissected whole AGM from E11.5 embryos and injected cell dilutions from 2 embryo equivalents (ee) to 0.25 ee into the facial vein of day 1–2 neonatal recipients that had received sublethal conditioning with 350 rad irradiation. In neonatal recipients we detected robust, long-term, multi-lineage hematopoietic engraftment from as little as 0.25 ee. From less than 1 ee of whole AGM, the engraftment chimerism ranged from 5–20%. With 2 ee, chimerism was as high as 70%. Most animals showed balanced donor derived myeloid and lymphoid contribution by 10 weeks post-transplant. Interestingly, some animals had predominantly myeloid reconstitution for as long as 18 weeks, suggesting the presence of a novel long-term, myeloid-restricted, embryonic HSC. We also explored the neonatal engraftment potential of VE-cadherin+ CD45+ and VE-cadherin+ CD45− populations. As expected from the literature, only the VE-cadherin+ CD45+ population engrafted the neonatal recipients. Our data indicate that the neonate harbors a more permissive hematopoietic microenvironment that enables more robust engraftment of early embryonic hematopoietic populations, thereby allowing us to identify potentially novel classes of embryonic hematopoietic progenitors. We are currently exploring the neonatal engraftment potential of E9.5 and E10.5 embryonic populations, additional FACS-purified populations, and hematopoietic populations derived from pluripotent stem cells in vitro. Disclosures: Daley: iPierian, Inc: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Epizyme, Inc: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Verastem, Inc: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Solasia, KK: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; MPM Capital, Inc: Consultancy, Membership on an entity's Board of Directors or advisory committees; Johnson & Johnson: Membership on an entity's Board of Directors or advisory committees.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 860-860
    Abstract: Abstract 860 During bone marrow transplantation, hematopoietic stem/progenitor cells (HSPCs) are exposed to various stress signals, and undergo homing, rapid proliferation and differentiation in order to achieve engraftment. To explore how fate decisions are made under such stress conditions, we developed a novel imaging-based competitive marrow transplantation in zebrafish. The feasibility of handling hundreds of zebrafish for transplantation per day allowed us to screen a library of 480 small molecules with known bioactivity, aimed at identifying new drugs and pathways regulating HSPC engraftment. Two structurally related eicosanoids, 11,12-epoxyeicosatrienoic acid (EET) and 14,15-EET, were able to enhance GFP+ marrow engraftment compared to DsRed2+ engraftment in zebrafish. This remarkable effect of EETs on adult marrow prompted us to study the effect of EETs in embryonic hematopoiesis. Treating zebrafish embryos with 11,12-EET during definitive hematopoiesis increased the HSPC marker Runx1 expression in the AGM (Aorta-Gonad-Mesonephros), resulting in a significant increase of HSPC in the next hematopoietic site, caudal hematopoietic tissue, the equivalent of fetal liver/placenta in mammals. The same treatment condition also induced ectopic Runx1 expression in the tail mesenchyme, a non-hematopoietic tissue. Microarray analysis on EET-treated zebrafish embryos revealed an upregulation of genes involved in stress response, especially Activator Protein 1 (AP-1) family members. Genetic knockdown experiments confirmed AP-1 members, especially JunB and its binding partners, cFos and Fosl2, are required for Runx1 induction. Motif analysis also predicted several conserved AP-1 binding sites in the Runx1 enhancer regions. To understand how EETs induced AP-1 expression, a suppressor screen was performed in zebrafish embryos. The screen revealed that activation of both PI3K/Akt and Stat3 are required for induced AP-1 expression, and therefore Runx1 upregulation. Similarly, ex vivo treatment of mouse whole bone marrow with 11,12-EET resulted in a 2-fold increase of long-term repopulating units. Microarray data had previously shown that Cyp2j6, one of the cytochrome P450 enzymes involved in EET biosynthesis from arachidonic acid, is enriched in quiescent mouse long-term HSCs. To further increase the EET levels in HSPCs, human CYP2C8 enzyme was over-expressed in transgenic mice using the Tie2 promoter. These transgenic mice have a 4-fold increase of long-term multi-lineage repopulating unit compared to their wild-type siblings. In purified mouse HSPCs, EETs directly and cell-autonomously activate PI3K/AKT pathway. Co-treatment of mouse bone marrow with EET and a PI3K inhibitor, LY294,002, completely blocked EET-induced enhancement of mouse bone marrow engraftment. In conclusion, we performed the first competitive marrow transplantation-based chemical screen, leading to the discovery of arachidonic acid-cytochrome P450-EETs as a novel modulator of HSC cell fate decision. PI3K/Akt and Stat3 pathways activated by EETs are required for adult HSPC engraftment and/or embryonic HSC specification, partially through transcriptional regulation of AP-1. We also demonstrated the requirement of AP-1 family members for Runx1 expression during embryonic development. This discovery may have clinical application in marrow or cord blood transplantation. Disclosures: Daley: iPierian, Inc: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Epizyme, Inc: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Verastem, Inc: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Solasia, KK: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; MPM Capital, Inc: Consultancy, Membership on an entity's Board of Directors or advisory committees; Johnson & Johnson: Membership on an entity's Board of Directors or advisory committees. Zon:Fate Therapeutics: Founder; Stemgent: Consultancy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Nature, Springer Science and Business Media LLC, Vol. 573, No. 7772 ( 2019-09-05), p. E1-E1
    Type of Medium: Online Resource
    ISSN: 0028-0836 , 1476-4687
    RVK:
    RVK:
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2019
    detail.hit.zdb_id: 120714-3
    detail.hit.zdb_id: 1413423-8
    SSG: 11
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    Public Library of Science (PLoS) ; 2005
    In:  PLoS Genetics Vol. 1, No. 3 ( 2005-9-2), p. e28-
  • 6
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 1283-1283
    Abstract: Abstract 1283 Adult hematopoietic cells transition through a hemogenic endothelial (HE) intermediate during development, but the signaling pathways modulating this transition are incompletely characterized. Although the Hedgehog (Hh) pathway is hypothesized to play a role in blood and endothelial cell formation, early embryonic lethality of mice lacking Hedgehog signaling precludes such analysis. To determine a role for Hh signaling in HE patterning, we assessed the effect of altered Hh signaling in differentiating mouse embryonic stem cells (mESCs), cultured embryonic day 9.5 mouse embryos, and developing zebrafish embryos. In differentiating mESCs, addition of Indian Hh ligand (IHH) increased the number of CD41+c-Kit+ hematopoietic progenitors, whereas chemical inhibition of Hh signaling led to a decrease without affecting primitive-streak mesoderm gene expression. In the setting of Hh inhibition, Notch induction rescued hemogenic VE-cadherin+ cells, demonstrating that Notch expands HE. Scl/Tal1 (stem cell leukemia/T-cell associated leukemia 1) induction rescued VE-cadherin+CD41+ cells, demonstrating that Scl/Tal1 converts endothelial cells to hematopoietic tissue. Similar experiments using cultured mouse yolk sacs demonstrated that signaling pathways are conserved in vivo. Moreover, VE-cadherin+ cells isolated from the mouse yolk sac or paraaortic splanchnopleura, when virally transduced with Notch signaling or Scl, had increased hematopoietic colony-forming activity. Finally, ectopic Notch or Scl induction in zebrafish embryos rescued the expression of the prototypical hemogenic endothelium marker Runx1 in the absence of Hh signalling. Together, our results reveal that the Hh-Notch-Scl axis promotes embryonic hematopoiesis through endothelial-to-hematopoietic transition. Disclosures: Zon: Fate Therapeutics: Consultancy; Stemgent: Consultancy. Daley:iPierian, Inc: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Epizyme, Inc: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Verastem, Inc: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Solasia, KK: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; MPM Capital, Inc: Consultancy, Membership on an entity's Board of Directors or advisory committees; Johnson & Johnson: Membership on an entity's Board of Directors or advisory committees.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Proceedings of the National Academy of Sciences, Proceedings of the National Academy of Sciences, Vol. 110, No. 2 ( 2013-01-08)
    Abstract: The function of the Hedgehog-Notch-Scl axis in endothelial-to-hematopoietic conversion provides useful insight into the origins of vertebrate hematopoiesis; however, many aspects of the endothelial-to-hematopoietic transition are still unknown. One broad question is what combination of transcription factors is required to induce hematopoiesis from endothelial cells. A recent study noted that VE-cadherin endothelial cells are heterogeneous in that distinct populations give rise to erythroid/myeloid progenitors and hematopoietic stem cells ( 5 ). Future studies will likely focus on this heterogeneity in an attempt to fine-tune the subpopulation of VE-cadherin needed for the generation of hematopoietic stem cells for therapeutic approaches. The long-term goal would be to derive hematopoietic stem cells using patient-specific pluripotent stem cells for bone marrow transplantation in individuals without a matched donor. These approaches revealed that Hedgehog signaling is required for the generation of VE-cadherin–positive endothelial cells that can give rise to hematopoietic cells ( Fig. P1 ), thus providing the link between endothelial and hematopoietic development. In this regard, Hedgehog plays a functionally redundant role with the Notch signaling pathway, which has previously been shown to promote arterial specification in zebrafish embryos ( 4 ). Moreover, the transcription factor stem cell leukemia (Scl) can compensate for the loss of Hedgehog or Notch signaling during the endothelial-to-hematopoietic transition. The redundancy of Hedgehog and Notch and the potent effect of Scl in converting endothelial cells into hematopoietic cells were confirmed using zebrafish embryos in which the ectopic expression of Scl overcame dual inhibition of Hedgehog and Notch and rescued the expression of the prototypical hemogenic endothelial marker Runx1. In this scenario, endothelial cells did not need to form functional vessels for Scl to exert its effect. To study the role of Hedgehog signaling in hematopoiesis, we first induced mouse ES cells to become hematopoietic cells while altering components of the Hedgehog pathway to measure the effect on hematopoietic output and quantity of VE-cadherin–positive endothelial cells. We next dissected the hematopoietic compartments in the mouse embryo, such as the yolk sac and the aortic trunk, and assessed the effect of altered Hedgehog signaling on hematopoietic output. We also altered Hedgehog signaling in developing zebrafish embryos and assessed the changes in the level of gene expression associated with hematopoiesis. We took these three approaches, because each model system has its advantages and disadvantages. We used developing mouse ES cells to dissect signaling pathway components in large quantities and avoided confounders, such as early embryo lethality and cardiovascular deformities. Using this information, we were able to dissect the same pathways in mouse and zebrafish embryos. Zebrafish embryos finally allowed us to visualize the anatomical relationship between angioblasts and their subsequent differentiation into hematopoietic cells. Here, we studied the Hedgehog signaling pathway, which plays a role in vessel remodeling and is implicated in hematopoiesis. Research in this area is hampered, because transgenic mice defective in Hedgehog signaling die before birth. For example, mouse embryos that lack Hedgehog signaling fail to form a functioning aorta and show arrest around embryonic day 10 ( 2 ). Interestingly, Hedgehog signaling is not required in hematopoietic cells in adults ( 3 ). These prior observations led us to hypothesize that endothelial cells respond to Hedgehog signaling, and perhaps through this process, the Hedgehog pathway contributes to the endothelial-to-hematopoietic transition. Hematopoietic cells arise in many compartments in the developing embryo before they migrate to the bone marrow in the newborn, where they will continue to regenerate blood cells throughout their lifespan. In the developing mouse embryo, these hematopoietic compartments include extraembryonic tissues, such as the yolk sac and placenta, and embryonic compartments, such the dorsal aorta and fetal liver. Although it is unclear exactly how hematopoietic cells eventually populate the bone marrow, evidence suggests that adult hematopoietic cells transition through an endothelial intermediate, and the origins of adult hematopoiesis are in these endothelial cells ( 1 ). These endothelial cells express vascular endothelial cadherin (VE-cadherin), which is important for vascular integrity at cell–cell junctions, particularly in newly formed vessels. When Chen et al. ( 1 ) labeled cells in the developing mouse embryo, which were positive for VE-cadherin, they found that virtually all hematopoietic cells in the adult retained this label. The molecular events that modulate the transition of endothelial cells to hematopoietic cells are poorly understood. Understanding how adult hematopoietic cells arise from endothelial cells in the embryo may facilitate the generation of hematopoietic progenitors in vitro. These cells might serve as alternatives to donor-derived bone marrow transplantation to treat hematopoietic diseases, such as refractory leukemias.
    Type of Medium: Online Resource
    ISSN: 0027-8424 , 1091-6490
    RVK:
    RVK:
    Language: English
    Publisher: Proceedings of the National Academy of Sciences
    Publication Date: 2013
    detail.hit.zdb_id: 209104-5
    detail.hit.zdb_id: 1461794-8
    SSG: 11
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Cell, Elsevier BV, Vol. 147, No. 3 ( 2011-10), p. 577-589
    Type of Medium: Online Resource
    ISSN: 0092-8674
    RVK:
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2011
    detail.hit.zdb_id: 187009-9
    detail.hit.zdb_id: 2001951-8
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 3387-3387
    Abstract: Abstract 3387 BMP and Wnt signaling pathways control essential cellular responses through activation of the transcription factors SMAD (BMP) and TCF (Wnt). Here, we have evaluated their function during hematopoietic regeneration after irradiation. Using heat-shock inducible transgenic zebrafish lines that overexpress BMP2 or Wnt8, we demonstrated accelerated marrow recovery following irradiation. Heat-shock induced overexpression of the respective inhibitors Chordin and DKK1 blunted the recovery. Surprisingly, gene expression profiling after induction of BMP or Wnt signaling in zebrafish marrow cells post-irradiation revealed increased expression of the key hematopoietic genes scl, runx1, and gata2. To determine if the effect of BMP and Wnt signaling on hematopoietic genes during regeneration was direct, we performed ChIP-PCR for Smad1 and the hematopoietic regulator Gata2 in murine lineage-negative progenitors seven days after a sublethal irradiation. We found that Smad1 and Gata2 co-occupy hematopoietic genes including Cd9, Il13, Mapk6, and Meis1. To examine the binding of SMAD1 and TCF7L2 throughout the genome of hematopoietic cells, we employed ChIP-seq in human erythroid and myeloid leukemia cell lines, K562 and U937, respectively. More than 70% of the genes bound by SMAD1 and TCF7L2 were co-occupied with the lineage transcription factors GATA1 and GATA2 in erythroid cells, and with C/EBPα in myeloid cells. This finding suggests that signaling transcription factors control hematopoietic gene programs by binding DNA adjacent to lineage-specific transcription factors. The transcriptional output of BMP and Wnt activity was tested on an LMO2 enhancer reporter construct. Expression of SMAD1 or TCF7L2 alone had little effect, but markedly increased reporter activity in conjunction with GATA2, indicating that BMP and Wnt signaling cooperate with lineage regulators to enhance transcription of cell-type specific target genes. To establish the order of transcription factor occupancy, we utilized estrogen-inducible C/EBPα-ER in K562 cells or GATA1 induction in murine G1ER cell lines, and assessed SMAD1 occupancy before and after induction of each respective lineage regulator. Induction of the myeloid lineage regulator C/EBPα in K562 cells shifted binding of SMAD1, such that SMAD1 co-occupancy with C/EBPα changed from 6% to 15% of C/EBPα targets. In contrast, expression of the erythroid regulator GATA1 promoted loss of SMAD1 on 82% of its targets, and restricted more than 98% of the remaining SMAD1 sites to erythroid targets adjacent to GATA1. Co-occupancy of signaling factors and lineage regulators was further tested in primary human CD34+ multipotent hematopoietic progenitors and CD34+ cells directed to the erythroid lineage. Both SMAD1 and TCF7L2 co-localized with GATA2 on greater than 75% of bound genes in multipotent CD34+ progenitor cells. Similar to our results following GATA1 induction in G1ER cells, SMAD1 occupancy shifted to 65% erythroid targets upon differentiation of progenitors to the erythroid lineage. These data provide strong evidence that the binding of signaling factors follows the genomic occupancy of the dominant lineage regulator during differentiation. Together, our findings demonstrate that hematopoietic regeneration is driven by collaboration of master regulators and signaling transcription factors to control the entire hematopoietic program. Disclosures: Daley: Verastem, Inc: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; iPierian, Inc: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Epizyme, Inc: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Solasia, KK: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; MPM Capital, Inc: Consultancy, Membership on an entity's Board of Directors or advisory committees; Johnson & Johnson: Membership on an entity's Board of Directors or advisory committees. Zon:Fate Therapeutics:; Stemgent: Consultancy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Society of Hematology ; 2009
    In:  Blood Vol. 113, No. 22 ( 2009-05-28), p. 5476-5479
    In: Blood, American Society of Hematology, Vol. 113, No. 22 ( 2009-05-28), p. 5476-5479
    Abstract: Human dermal fibroblasts obtained by skin biopsy can be reprogrammed directly to pluripotency by the ectopic expression of defined transcription factors. Here, we describe the derivation of induced pluripotent stem cells from CD34+ mobilized human peripheral blood cells using retroviral transduction of OCT4/SOX2/KLF4/MYC. Blood-derived human induced pluripotent stem cells are indistinguishable from human embryonic stem cells with respect to morphology, expression of surface antigens, and pluripotency-associated transcription factors, DNA methylation status at pluripotent cell-specific genes, and the capacity to differentiate in vitro and in teratomas. The ability to reprogram cells from human blood will allow the generation of patient-specific stem cells for diseases in which the disease-causing somatic mutations are restricted to cells of the hematopoietic lineage.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages