scholarly journals Extramedullary Erythropoiesis in Spleen of HIF Prolyl 4-Hydroxylase-2 Deficient Mice Is Mediated By Notch Signaling Downregulation

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2656-2656
Author(s):  
Mikko Myllymäki ◽  
Jenni Määttä ◽  
Elitsa Dimova ◽  
Valerio Izzi ◽  
Timo Väisänen ◽  
...  

Abstract Erythrocytosis, an increase in absolute red cell mass, is mainly driven by erythropoietin, while hypoxia-inducible factor (HIF) regulates the expression of a number of genes involved in it, including erythropoietin. Mutations in HIF prolyl 4-hydroxylase 2 (HIF-P4H-2/PHD2/EGLN1), the major regulator of the stability of HIFα subunits, are found in familiar erythrocytosis, and large-spectrum conditional inactivation of HIF-P4H-2 in mice leads to severe erythrocytosis and premature death. Although bone marrow is the primary site for erythropoiesis, spleen retains a capability for extramedullary erythropoiesis. We studied HIF-P4H-2 hypomorphic mice (Hif-p4h-2gt/gt) which show slightly induced erythropoiesis only upon aging despite no increased erythropoietin levels. Spleen was identified as the site of extramedullary erythropoiesis in these mice. Hematopoietic stem cells (HSCs) from spleens of the Hif-p4h-2gt/gt mice showed increased growth of BFU-Es and the mice were protected against anemia by induced extramedullary erythropoiesis. HIF-1α and HIF-2α were stabilized in the spleens, while the Notch ligands and target Jag1, Jag2, Dll1 and Hes1 became downregulated upon aging dependent on HIF-2α. Inhibition of Notch signaling in wild-type spleen HSCs phenocopied the increased growth of BFU-Es in the Hif-p4h-2gt/gt mice. We conclude that HIFα stabilization can mediate non-erythropoietin-driven extramedullary erythropoiesis in the spleen via altered Notch signaling. Disclosures Myllyharju: FibroGen Inc.: Equity Ownership, Research Funding.

2016 ◽  
Vol 37 (2) ◽  
Author(s):  
Mikko N. M. Myllymäki ◽  
Jenni Määttä ◽  
Elitsa Y. Dimova ◽  
Valerio Izzi ◽  
Timo Väisänen ◽  
...  

ABSTRACT Erythrocytosis is driven mainly by erythropoietin, which is regulated by hypoxia-inducible factor (HIF). Mutations in HIF prolyl 4-hydroxylase 2 (HIF-P4H-2) (PHD2/EGLN1), the major downregulator of HIFα subunits, are found in familiar erythrocytosis, and large-spectrum conditional inactivation of HIF-P4H-2 in mice leads to severe erythrocytosis. Although bone marrow is the primary site for erythropoiesis, spleen remains capable of extramedullary erythropoiesis. We studied HIF-P4H-2-deficient (Hif-p4h-2 gt/gt ) mice, which show slightly induced erythropoiesis upon aging despite nonincreased erythropoietin levels, and identified spleen as the site of extramedullary erythropoiesis. Splenic hematopoietic stem cells (HSCs) of these mice exhibited increased erythroid burst-forming unit (BFU-E) growth, and the mice were protected against anemia. HIF-1α and HIF-2α were stabilized in the spleens, while the Notch ligand genes Jag1, Jag2, and Dll1 and target Hes1 became downregulated upon aging HIF-2α dependently. Inhibition of Notch signaling in wild-type spleen HSCs phenocopied the increased BFU-E growth. HIFα stabilization can thus mediate non-erythropoietin-driven splenic erythropoiesis via altered Notch signaling.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1408-1408
Author(s):  
Yuji Kikuchi ◽  
Akihiro Kume ◽  
Masashi Urabe ◽  
Hiroaki Mizukami ◽  
Takahiro Suzuki ◽  
...  

Abstract Mesenchymal stem cells (MSCs), which are key elements of hematopoietic microenvironment in bone marrow, are known to play a critical role in supporting hematopoiesis. A variety of hematopoietic growth factors are produced from MSCs, and cell-to-cell contact is also believed to be crucial in the interaction between hematopoietic stem cells (HSCs) and MSCs. However, the molecular mechanisms of hematopoiesis-supporting ability of MSCs are still unclear. In particular, there is little information regarding the effects of HSCs on MSC function. In the present study, we investigated the cellular and molecular events in the interactive communication between HSCs and MSCs using a differentiation-inducible MSC model; i.e. parent C3H10T1/2 cells and 10T1/2-derived cell lines, A54 preadipocytes and M1601 myoblasts. These cells were co-cultured with murine HSCs (Lin-Sca1+) isolated from bone marrow. There was 9-fold increase in the number of hematopoietic progenitors after co-culture with A54 preadipocytes, whereas there was no increase when co-cultured with parent 10T1/2 or M1601 cells. More intriguingly, cobblestone areas were observed only when HSCs were co-cultured with A54 cells. Quantitative RT-PCR showed that A54 cells express significantly higher levels of SCF, SDF-1, and angiopoietin-1 (Ang-1) compared with parent 10T1/2 cells and M1601 cells, although these cytokines were not up-regulated when co-cultured with HSCs. To search for the genes involved in the interaction between HSCs and MSCs, we compared gene expression profiles before and after the co-culture by using a microarray analysis. Among the candidate genes with up-regulation after the co-culture, we paid attention to the Notch system, because Notch ligands are considered to play an important role in nurturing HSCs within the hematopoietic microenvironment. As a result, the expression of Notch ligands, Jagged1 and Dll3, increased in A54 cells after the coculture with HSCs. On the other hand, the expression of Notch1 and Hes-1 also increased in HSCs upon co-culture with A54 cells. These data were confirmed by quantitative RT-PCR. Moreover, when HSCs were co-cultured with A54 cells without cell-to-cell contact using Transwell permeable supports, there was neither increase in the number of progenitors in the upper wells, nor the up-regulation of Notch ligands in A54 cells in the lower wells. These findings support the idea that HSCs act on MSCs to induce the expression of Notch ligands via direct cell-to-cell contact and that the Notch ligands derived from MSCs act on HSCs in turn to activate Notch signaling pathway, possibly leading to the cobblestone formation with the maintenance of immature state of HSCs. The Notch system may be one of the critical elements in the interactive communication between HSCs and MSCs.


Blood ◽  
2011 ◽  
Vol 117 (17) ◽  
pp. 4449-4459 ◽  
Author(s):  
Inge Van de Walle ◽  
Greet De Smet ◽  
Martina Gärtner ◽  
Magda De Smedt ◽  
Els Waegemans ◽  
...  

Abstract Notch signaling critically mediates various hematopoietic lineage decisions and is induced in mammals by Notch ligands that are classified into 2 families, Delta-like (Delta-like-1, -3 and -4) and Jagged (Jagged1 and Jagged2), based on structural homology with both Drosophila ligands Delta and Serrate, respectively. Because the functional differences between mammalian Notch ligands were still unclear, we have investigated their influence on early human hematopoiesis and show that Jagged2 affects hematopoietic lineage decisions very similarly as Delta-like-1 and -4, but very different from Jagged1. OP9 coculture experiments revealed that Jagged2, like Delta-like ligands, induces T-lineage differentiation and inhibits B-cell and myeloid development. However, dose-dependent Notch activation studies, gene expression analysis, and promoter activation assays indicated that Jagged2 is a weaker Notch1-activator compared with the Delta-like ligands, revealing a Notch1 specific signal strength hierarchy for mammalian Notch ligands. Strikingly, Lunatic-Fringe– mediated glycosylation of Notch1 potentiated Notch signaling through Delta-like ligands and also Jagged2, in contrast to Jagged1. Thus, our results reveal a unique role for Jagged1 in preventing the induction of T-lineage differentiation in hematopoietic stem cells and show an unexpected functional similarity between Jagged2 and the Delta-like ligands.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 573-573
Author(s):  
Michelle I Lin ◽  
Charles W Carspecken ◽  
Audrey Uong ◽  
Emily Price ◽  
Leonard I. Zon

Abstract Abstract 573 Members of the angiopoietin-like protein (angptl) family have recently been shown to stimulate ex vivo expansion of mouse and human hematopoietic stem cells (HSCs). The requirement of angptls for HSCs during development and their mechanism of action have yet to be determined. To first recapitulate the effects of exogenous angptl on HSC expansion, we made a stable transgenic zebrafish line, Tg(hsp70:angptl2), that overexpresses angptl2 upon heatshock induction. Heatshocked Tg embryos showed a significant increase in cmyb- and runx1-positive HSCs in the aorta-gonad-mesonephros (AGM) region, the site of definitive hematopoiesis at 36 hours post-fertilization (hpf), suggesting that angptl2 can sufficiently expand definitive HSCs during development. We then assessed the effects of angptl1 and/or angptl2 morpholino (MO) knockdown. Single MO knockdown resulted in a decrease in cmyb- and runx1-positive HSCs in the AGM whereas embryos injected with combined angptl1 and 2 MOs (double morphants) exhibited more severe phenotype in which HSCs were completely absent. Angptl regulation of HSC development may occur by stimulating the production or specification of a patent hemogenic endothelium, as the double morphants displayed a disruption of vascular specification at 28 hpf, with decreased expression of the arterial marker ephrinB2 but increased ectopic expression of the venous marker, flt4 in the dorsal aorta. These double morphants also exhibited disrupted intersegmental blood vessel sprouting. Because proper patterning of the developing blood vessels is a prerequisite for subsequent HSC formation, these results strongly suggest an early downstream effect of angptl signaling on hemogenic endothelium specification. To dissect the mechanism of angptl signaling, we asked whether there is a genetic interaction between notch and angptl signaling since the observed phenotype in the angptl double morphants resembled that in the notch mutant mindbomb, mib. We first monitored notch signaling using a transgenic notch reporter zebrafish line that expresses EGFP when the active component of notch signaling, notch intracellular domain (NICD), is expressed. MO knockdown of angptls 1 and 2 resulted in an absence of notch signaling, particularly in the vasculature. Interestingly, overexpressing angptl2 by crossing Tg(hsp70:angptl2) into the mib mutant showed a significant rescue of the HSC phenotype, implying that angptl2 acts downstream of mib signaling. When MOs against angptls 1 and 2 were then injected into the NICD overexpressing Tg embryos, we found that NICD restored HSC formation in the angptl double morphants. Together, these data corroborate the hypothesis that angptls can regulate notch signaling by acting downstream of notch ligand-receptor interaction and upstream of NICD during definitive hematopoiesis. To further probe at the downstream signaling of angptls since their receptor is currently unknown, we found that both angptls can stimulate activation of akt in cultured endothelial cells. To investigate whether akt is involved in angptl signaling during development, we injected the constitutively active myr-akt into angptl double morphants and observed rescue of both HSC and vascular defects. This strongly indicates that akt is the key signaling component downstream of angptl signaling during these biological processes. Injection of myr-akt can also rescue HSC defects in mib, suggesting that notch signaling also requires akt activation for definitive HSC formation. Taken together, our data demonstrate that the angptls are required for HSC development through notch and akt signaling, by coordinating the production of a functional hemogenic endothelium. 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.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 805-805
Author(s):  
Vedran Radojcic ◽  
Ryan P Flynn ◽  
Jooho Chung ◽  
Jing Du ◽  
Eric Perkey ◽  
...  

Abstract Chronic graft-versus-host disease (cGVHD) remains a major cause of morbidity and mortality following allogeneic hematopoietic cell transplantation (allo-HCT) with limited success of current therapeutic options. We have shown that Notch inhibition in donor T cells or systemic neutralization of the Notch ligands Delta-like1 and Delta-like4 (Dll1/4) prevents acute GVHD in multiple mouse allo-HCT models (Zhang et al., Blood 2011; Sandy et al., J Immunol 2013; Tran et al., JCI 2013). However, the role of Notch signaling in cGVHD remains unknown. To address this question, we used genetic and antibody-mediated strategies of Notch inhibition in two established and pathophysiologically distinct mouse models of cGVHD characterized by dominance of sclerodermatous changes (Scl) or non-Scl multi-organ system disease that includes bronchiolitis obliterans (BO), respectively. In the B10.D2→BALB/c MHC-matched, minor antigen mismatched model of Scl-cGVHD, transgenic expression of the pan-Notch inhibitor DNMAML in T cells induced high level protection from cGVHD as assessed using clinical and pathological scoring criteria. Systemic antibody-mediated blockade of Dll1/4 Notch ligands prevented cGVHD when given early after allo-HCT, with dominant effects of Dll4 inhibition alone. However, Dll4 blockade provided no therapeutic benefits if delayed by 48 hours after allo-HCT or when targeting fully established Scl-cGVHD, suggesting that Dll4-mediated Notch signals are critical in this model when delivered to T cells early after allo-HCT. To understand the impact of Notch inhibition during priming of alloantigen-specific T cells, we studied immunodominant Vβ3+CD4+ T cells that expand in response to a host superantigen encoded by Mtv6 in BALB/c recipients. Both control and Notch-inhibited Vβ3+CD4+ T cells upregulated activation markers and expanded to comprise >80% of donor T cells in lymphoid and target organs by day 6. However, Notch inhibition markedly decreased IFN-γ, TNF-α, and IL-17 production in these cells, while expanding Vβ3+CD4+FoxP3+ regulatory T cells. Thus, Notch inhibition preserved in vivo T cell proliferation and expansion, but tilted the balance in favor of alloantigen-specific regulatory T cells over highly inflammatory effector T cells. In the B6→B10.BR MHC-mismatched model of allo-HCT, characterized by prominent germinal center (GC) responses and BO-cGVHD, genetic pan-Notch inhibition in T cells with DNMAML blocked GC formation and provided long-lasting high level protection from BO, as assessed using pulmonary function tests to measure resistance, elastance and compliance, as well as pathological examination (day 56 after allo-HCT). Flow cytometric evaluation showed that follicular helper T cells (CD4+PD-1hiCXCR5+) and cells with a GC B cell phenotype (CD19+GL7+CD95hi) were markedly decreased in numbers. Genetic deletion of Notch1 or Notch2 in donor T cells also prevented the immunological manifestations of cGVHD. Antibody-mediated blockade of the Notch ligands Dll1, Dll4, or both in the peri-transplant period protected recipients from BO-cGVHD. In contrast to our observations in Scl-cGVHD, delayed Dll1/4 blockade starting at day 28 after allo-HCT preserved pulmonary function, decreased GC formation and alloantibody deposition in target tissues, and ameliorated BO-cGVHD. Thus, Dll1/4 inhibition could provide therapeutic benefits even in established BO-cGVHD. Altogether, we identified a key role for early Notch signals in alloantigen-specific T cells that define subsequent cGVHD pathogenesis. Interference with Notch signaling early after allo-HCT provided long-lasting protection from cGVHD. In addition, our observations in the multi-organ system BO-cGVHD model suggest a therapeutic potential for delayed inhibition of Dll1 or Dll4 Notch ligands in an active disease setting driven by alloantibody formation. Our preclinical data suggest that Notch signaling should be explored as a novel druggable target to prevent or treat different forms of cGVHD. Disclosures Yan: Genentech, Inc.: Employment, Equity Ownership. Siebel:Genentech Inc.: Employment, Equity Ownership.


eLife ◽  
2018 ◽  
Vol 7 ◽  
Author(s):  
Shrestha Ghosh ◽  
Sheung Kin Wong ◽  
Zhixin Jiang ◽  
Baohua Liu ◽  
Yi Wang ◽  
...  

Mammalian sirtuin 6 (Sirt6) is a conserved NAD+-dependent deacylase and mono-ADP ribosylase that is known to be involved in DNA damage repair, metabolic homeostasis, inflammation, tumorigenesis, and aging. Loss of Sirt6 in mice results in accelerated aging and premature death within a month. Here, we show that haploinsufficiency (i.e., heterozygous deletion) of Trp53 dramatically extends the lifespan of both female and male Sirt6-deficient mice. Haploinsufficiency of Trp53 in Sirt6-deficient mice rescues several age-related phenotypes of Sirt6-deficient mice, including reduced body size and weight, lordokyphosis, colitis, premature senescence, apoptosis, and bone marrow stem cell decline. Mechanistically, SIRT6 deacetylates p53 at lysine 381 to negatively regulate the stability and activity of p53. These findings establish that elevated p53 activity contributes significantly to accelerated aging in Sirt6-deficient mice. Our study demonstrates that p53 is a substrate of SIRT6, and highlights the importance of SIRT6-p53 axis in the regulation of aging.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2616-2616
Author(s):  
Wilson K. Clements ◽  
Karen G. Ong ◽  
Albert D. Kim ◽  
David Traver

Abstract Abstract 2616 Hematopoietic stem cells (HSCs) are self-renewing progenitor cells that provide all adult blood lineages over the lifetime of an individual. Understanding the signaling events that regulate specification of these cells during embryonic development is a key precondition to recapitulating those processes in vitro for the purposes of regenerative medicine. Here we report that non-canonical Wnt signaling by the conserved vertebrate ligand, Wnt16 is required for specification of HSCs in zebrafish. WNT16 was originally identified as a gene aberrantly expressed in pre-B acute lymphoblastic leukemia (ALL) cells containing the t(1;19) chromosomal translocation, leading to expression of E2A-PBX1. Wnt16 is expressed in mammals at times when hemogenic endothelial cells first appear in the dorsal aorta. In zebrafish, wnt16 is expressed in somites, which lie adjacent to the dorsal aorta. Knock down of Wnt16 function by injection of antisense morpholino oligonucleotides leads to loss of HSCs and definitive blood lineages, such as T-cells, during development. Non-blood tissues including vasculature appear largely unaffected. Thus, wnt16 is required for HSC specification. To better understand the Wnt16 signal transduction pathway, we examined its ability to activate transcription of target genes through β-catenin/Tcf-dependent “canonical” signaling. Although Wnt16 overexpression causes morphological abnormality, it does not yield ectopic expression of endogenous or transgenic canonical reporter genes at time points relevant to blood specification. Thus, Wnt16 signals independently of β-catenin/Tcf through a “non-canonical” Wnt pathway. Notch signaling is required for specification of HSCs across phyla. To determine whether Notch signaling is disrupted in Wnt16 morphants, we examined the expression of all Notch ligands and receptors in these animals and found that expression of two ligands, deltaC (dlc) and deltaD (dld) are decreased. Zebrafish mutants, homozygous for a null allele of dlc, and embryos injected with a dld morpholino each display decreased numbers of HSCs during development, but recover. Dlc mutants injected with dld morpholino show complete loss of HSCs. To determine whether defects in Notch signaling are responsible for loss of HSCs in Wnt16 morphants, we performed a rescue experiment. Transgenic animals carrying an inducible dominant activator of Notch target genes were injected with Wnt16 morpholino, and Notch activity was either induced or not. Wnt16 morphants with enforced Notch activity recovered HSC marker expression. Taken together, our results indicate that Wnt16 regulates expression of the Notch ligands dlc and dld, and these are redundantly required for HSC specification during development. Chimera experiments in mouse using wild type and Notch1-deficient cells indicate that there is a cell-autonomous requirement for Notch signaling in specification of HSCs. To determine when the first Notch-responsive cells that contribute to the adult hematopoietic system appear, we used animals carrying a transgene encoding a photconvertible, green-to-red Kaede protein under the control of a Notch-responsive promoter. By photoconverting all embryonic cells at various times during development, we determined that cells destined to become HSCs first experience a Notch signal at approximately the time when HSCs first appear, just before 24 hours post fertilization (hpf). At earlier time points, Notch responsive cells were present, but did not contribute to blood, although they did contribute to tissues in or near the dorsal aorta, which contains the hemogenic endothelium that gives rise to HSCs. Surprisingly, HSC rescue in Wnt16 morphants by Notch activation could only be achieved earlier than 16 hpf, long before a cell-autonomous Notch signal is received in pre-HSCs. Moreover, loss of the critical ligands, dlc and dld, in Wnt16 morphant animals was confined to somitic tissue prior to 20 hpf, while ligand expression in or near the dorsal aorta at later times was relatively normal. Together, our results strongly suggest a previously unappreciated, non-cell-autonomous requirement for Notch signaling in the somites. We hypothesize that somitic Notch signaling regulates a morphological process or expression of a relay signal required for HSC specification during development. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 652-652
Author(s):  
Lan Zhou ◽  
Weihuan Wang ◽  
Grant Zimmerman ◽  
Jay Myers ◽  
Yuanshuai Huang ◽  
...  

Abstract BACKGROUND: Notch has long been recognized as an important molecule that regulates stem cell self-renewal and differentiation. Despite that Notch is required for the embryonic hematopoietic stem cell (HSC) generation, and that Notch is expressed in the adult HSC, the role of Notch pathway in adult HSC remains unclear. Recently it has been shown that Notch activation and the ability of interacting with Notch ligand Jagged1 is a signature of human primitive HSC and supports HSC regenerative potential. Here we study the physiological significance of Notch in the adult HSC population focusing on how Notch-ligand engagement regulates HSC quiescence and niche retention. METHODS: To better understand the role of Notch in adult HSC homeostasis, we examined HSC frequency, quiescence maintenance, niche occupancy, and HSC mobilization in mice with either conditional lack of RBP-JK, which mediates the canonical Notch signaling activity, or in mice with conditional lack of Pofut1 that catalyzes O-fucosylation of Notch EGF-like repeats and the generation of O-fucose glycans important for the binding of Notch ligands. We also tested Notch ligand neutralizing antibodies and Notch1 and Notch2 inhibitory antibodies to examine the effect of blocking Notch receptor-ligand engagement or the block of Notch signaling activation on HSC homeostasis. RESULTS: We report here that Pofut1-deficient hematopoietic stem and progenitor cells (HSPCs) display enhanced cell cycling and proliferation cell autonomously. These changes are accompanied by G-CSF-independent increased HSPC egress from the marrow to the periphery and other hematopoietic organs, and their enhanced sensitivity to mobilizing stimuli of G-CSF plus the CXCR4 antagonist, AMD3100. This phenotype is caused by reduced adhesion of Pofut1-deficient HSPC to Notch ligand-expressing stromal cells and the osteoblastic lineage cells. Adhesion to these cells by wild type but not Pofut1-deficient HSPCs can be blocked by recombinant Notch ligand Dll1 or Dll4. In addition, adhesion to these cells inhibits wild type but not Pofut1-deficient HSPC cycling, independent of RBP-Jk-mediated Notch signaling. Further, Pofut1-deficient HSPCs exhibit normal expression of key adhesion molecules and normal SDF-1-mediated chemotaxis, but show scattered and distal occupancy in the endosteal or osteoblastic niche. In support of roles for Notch-ligand engagement in facilitating HSPC niche retention, we show that mice receiving 4 doses of neutralizing antibodies to the Notch ligand Dll4 or Jagged1, but not Dll1, display ~2-3-fold increased HSC and progenitor egress when compared to mice receiving isotype control antibody, and further display 60% increased HSC mobilization when compared to mice receiving control antibody and treated similarly with G-CSF and AMD3100. Dll4 blockade also increases the sinusoidal endothelial cell population and HSPC cell cycling. In comparison, only a mild HSPC proliferation and egress is found in RBP-JK-deficient mice, or in mice receiving Notch2 inhibitory antibody. However, Notch2 blockade but not Notch1 blockade induces unique features of HSC and myeloid progenitor mobilization responding to G-CSF plus AMD3100. CONCLUSIONS: Based on these findings, we conclude that HSPC quiescence and retention in the marrow niche is facilitated by the interaction between Notch-expressing HSPCs and Jagged1- or Dll4-expressing niche cells, and is likely also contributed by Notch signaling activation. In addition, Notch receptor-ligand engagement in this process is strengthened by O-fucose modification of Notch receptors. Finally, the observations from our studies may provide a therapeutic indication. Inadequate mobilization in HSPC transplantation remains a clinical problem. Our findings that targeting Notch receptor-ligand interaction and/or inhibiting Notch2 activation increase HSPC emigration suggests a novel approach for enhancing mobilization of stem and progenitor cells for those patients who respond poorly to current mobilizing regimes. Disclosures Shim: Genentech: Employment, Equity Ownership. Yan:Genentech: Employment, Equity Ownership. Lowe:Genentech: Employment, Equity Ownership. Siebel:Genentech: Employment, Equity Ownership.


2008 ◽  
Vol 28 (15) ◽  
pp. 4794-4804 ◽  
Author(s):  
Mi-Jeong Yoon ◽  
Bon-Kyoung Koo ◽  
Ran Song ◽  
Hyun-Woo Jeong ◽  
Juhee Shin ◽  
...  

ABSTRACT Intraembryonic hematopoiesis occurs at two different sites, the floor of the aorta and subaortic patches (SAPs) of the para-aortic splanchnopleura (P-Sp)/aorta-gonad-mesonephros (AGM) region. Notch1 and RBP-jκ are critical for the specification of hematopoietic stem cells (HSCs) in Notch signal-receiving cells. However, the mechanism by which Notch signaling is triggered from the Notch signal-sending cells to support embryonic hematopoiesis remains to be determined. We previously reported that Mind bomb-1 (Mib1) regulates Notch ligands in the Notch signal-sending cells (B. K. Koo, M. J. Yoon, K. J. Yoon, S. K. Im, Y. Y. Kim, C. H. Kim, P. G. Suh, Y. N. Jan, and Y. Y. Kong, PLoS ONE 2:e1221, 2007). Here, we show that intraembryonic hematopoietic progenitors were absent in the P-Sp of Mib1 −/− embryos, whereas they were partly preserved in the Tie2-cre; Mib1 f /f P-Sps, suggesting that Mib1 plays a role in the endothelium and the SAPs. Interestingly, dll1 and dll4/Jag1 are expressed in the SAPs and the endothelium of the AGM, respectively, where mib1 is detected. Indeed, Notch signaling was activated in the nascent HSCs at both sites. In the P-Sp explant culture, the overexpression of Dll1 in OP9 stromal cells rescued the failed production of hematopoietic progenitors in the Mib1 −/− P-Sp, while its activity was abolished by Mib1 knockdown. These results suggest that Mib1 is important for intraembryonic hematopoiesis not only in the aortic endothelium but also in the SAPs.


Sign in / Sign up

Export Citation Format

Share Document