Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    Online Resource
    Online Resource
    Ovid Technologies (Wolters Kluwer Health) ; 2008
    In:  Critical Care Medicine Vol. 36, No. 10 ( 2008-10), p. 2734-2739
    In: Critical Care Medicine, Ovid Technologies (Wolters Kluwer Health), Vol. 36, No. 10 ( 2008-10), p. 2734-2739
    Type of Medium: Online Resource
    ISSN: 0090-3493
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2008
    detail.hit.zdb_id: 2034247-0
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 3870-3870
    Abstract: Abstract 3870 The BMP and WNT signaling pathways are two highly conserved signaling pathways that cooperate in many developmental processes, ultimately through alteration of transcription via SMAD and TCF transcription factors. These pathways elicit pleiotropic outcomes across cell types, yet only a few cell-specific direct target genes are known for the signaling transcription factors that mitigate these effects. We took a genome-wide approach to define the binding sites of BMP and WNT-directed transcription factors in different hematopoietic lineages. Using heat-shock inducible transgenic fish lines that overexpress BMP2 or WNT8, we demonstrated accelerated marrow recovery following irradiation. Irradiation recovery was blunted by heat shock induced overexpression of the respective inhibitors Chordin and DKK1. Similar to the zebrafish regeneration results, competitive transplants with mouse bone marrow treated with the WNT agonist BIO led to enhanced chimerism. Inhibition of BMP diminished peripheral blood contribution even in the presence of WNT stimulation, suggesting a conserved and cell intrinsic interaction for these signaling pathways in adult stress hematopoiesis. To examine potential target genes that could account for the synergy, we performed chromatin immunoprecipitation with WNT- and BMP-activated transcription factors followed by sequencing (ChIP-seq) in K562 cells. ChIP-seq was performed with TCF7L2/TCF4, a mediator of the WNT pathway, and SMAD1, a mediator of the BMP signaling pathway, and 〉 2000 binding sites were identified for each factor. Motif discovery revealed that the DNA sequences bound by TCF7L2 and SMAD1 were not only enriched for TCF and SMAD binding elements, respectively, but were also enriched for a GATA motif. Comparison of the TCF7L2 and SMAD1 bound genes with published ChIP-Seq data for GATA1 and GATA2 in K562 cells revealed that both signaling factors bind more than 40% of GATA1 bound genes and greater than 70% of GATA2 bound genes. Ingenuity and GSEA analysis revealed that genes important for erythropoiesis were among the genes co-bound by these factors. To evaluate the effect of cell lineage on signaling factor binding, ChIP-seq of TCF7L2 and SMAD1 in U937, a monocytic leukemia cell line, was performed. Motif discovery of sequences bound in U937 found enrichment for an ETS motif, which is bound by the key myeloid transcription factor Pu.1. In addition, TCF7L2 and SMAD1 bound genes in U937 overlapped genes bound by C/EBPalpha in U937 by greater than 70%. These genes are implicated in monocytic development. The overlap of binding between TCF7L2 in K562 and U937 was less than 15% and the overlap of SMAD1 binding sites between the cell lines was less than 10%, indicating a substantial influence of cell lineage on transcription factor binding. Confirmation of cell type selective binding of TCF7L2 and SMAD1 in vivo was accomplished by ChIP of the transcription factors in zebrafish nucleated erythrocytes. Binding of TCF7L2 and SMAD1 in these cells showed that these factors co-bind with GATA1 in many genes with established roles in erythropoiesis. Together our data suggest the co-binding of WNT- and BMP-specific transcription factors with master regulators of each hematopoietic cell type results in regulation of distinct blood genes based on lineage. (First two authors contributed equally to this work) Disclosures: Zon: FATE, Inc.: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties; Stemgent: Consultancy, Equity Ownership, 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: 2010
    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. 114, No. 22 ( 2009-11-20), p. 2430-2430
    Abstract: Abstract 2430 Poster Board II-407 After transplantation, hematopoietic stem/progenitor cells (HSPCs) home to the marrow, where they engraft and self-renew. To explore the mechanism of this multi-step and dynamic repopulation process, we performed the first in vivo adult vertebrate chemical screen aimed at identifying novel chemical modulators of HSPC repopulation using a novel competitive marrow transplantation assay in zebrafish. To distinguish between the donors, we used ubiquitous GFP or DsRed2 transgenic fish, Tg(β-actin:GFP) and Red GloFish®, for marrow cell isolation. 20,000 GFP+ cells were treated with a chemical and mixed with 80,000 untreated DsRed2+ marrows. This pool of cells was injected retro-orbitally into a transparent adult zebrafish. After a recovery period, the fish was then anesthetized and the region of the kidney (the adult site of hematopoiesis) was examined by fluorescence microscopy. The competition between the two donors was determined by analyzing the ratios of GFP and DsRed2 fluorescence intensity with ImageJ software. Using this assay, we demonstrated that dmPGE2 and/or GSK-3β inhibitor treatment of GFP+ marrows for 3 hrs could dramatically increase repopulation in fish. A chemical library of 480 chemicals with known bioactivities was screened using this in vivo assay. GFP+ marrows were incubated with different chemicals for 3 hrs and ten recipient fish were transplanted for each chemical. By examining engraftment at 4 weeks, we found 10 chemicals that improved HSPC repopulation. Based on the known bioactivity, these chemicals were categorized into several signaling pathways, including prostaglandin metabolism and retinoic acid pathways. Several of the compounds also increased HSC formation in zebrafish embryos, indicating that some pathways might be shared by different developmental stages. To examine whether the bioactivities of these hits are conserved in mammals, CD45.1 mouse whole bone marrow cells were treated with hit compounds for 3 hrs and competitively transplanted into CD45.2 recipients. Peripheral blood was sampled at 3, 6, 12, and 20 week post transplant. Several hits were confirmed to increase long-term chimerism in mice. The retinoic acid pathway has been shown to play an important role in hematopoiesis. Among the six retinoic acid receptor (RAR) agonists in the chemical library, which includes all-trans retinoic acid (ATRA), only two structurally highly related compounds, AM-580 and TTNPB scored positive in the screen. These two compounds have distinctive chemical moieties from ATRA. This structural difference likely leads to stronger agonistic effects on RAR than ATRA and resists degradation. In conclusion, the in vivo chemical screening using zebrafish competitive marrow transplantation provides a successful example of phenotypic screening in whole adult vertebrates. The discovery of novel repopulation modulators should provide a better understanding of signaling events that regulate homing and self-renewal, and may have clinical application in marrow or cord blood transplantation. Disclosures: Zon: FATE Inc: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Stemgent: Consultancy.
    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 ...
  • 4
    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 ...
  • 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: 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 ...
  • 8
    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 ...
  • 9
    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 ...
  • 10
    Online Resource
    Online Resource
    Elsevier BV ; 2018
    In:  Molecular Genetics and Metabolism Vol. 123, No. 2 ( 2018-02), p. S55-S56
    In: Molecular Genetics and Metabolism, Elsevier BV, Vol. 123, No. 2 ( 2018-02), p. S55-S56
    Type of Medium: Online Resource
    ISSN: 1096-7192
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2018
    detail.hit.zdb_id: 1471393-7
    SSG: 12
    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