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
Material
Language
Subjects(RVK)
  • 1
    In: American Journal of Respiratory Cell and Molecular Biology, American Thoracic Society, Vol. 58, No. 5 ( 2018-05), p. 636-647
    Type of Medium: Online Resource
    ISSN: 1044-1549 , 1535-4989
    RVK:
    Language: English
    Publisher: American Thoracic Society
    Publication Date: 2018
    detail.hit.zdb_id: 1473629-9
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal of Clinical Investigation, American Society for Clinical Investigation, Vol. 125, No. 12 ( 2015-11-9), p. 4638-4654
    Type of Medium: Online Resource
    ISSN: 0021-9738 , 1558-8238
    Language: English
    Publisher: American Society for Clinical Investigation
    Publication Date: 2015
    detail.hit.zdb_id: 2018375-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Society of Hematology ; 2018
    In:  Blood Vol. 132, No. Supplement 1 ( 2018-11-29), p. 1060-1060
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 1060-1060
    Abstract: Sickle cell anemia results from a point mutation in both alleles of the β-globin gene. This homozygous mutation ultimately leads to a structural alteration of the hemoglobin protein that promotes polymerization of the mutant sickle hemoglobin tetramer (HbS) upon deoxygenation. HbS polymerization results in rigid, sickle-shaped RBCs with increased cell-to-cell adhesion properties. These sticky and rigid RBCs are prone to become trapped in small capillary networks leading to ischemia-reperfusion injury, endothelial damage and the hallmark pain crisis of sickle cell anemia. Neonates are protected from deoxy-HbS polymerization by high levels of fetal hemoglobin (HbF). HbF is composed of two α-globin and two γ-globin subunits(α2γ2), The γ-globin molecule cannot interact with deoxy-βS-globin polymers, which makes HbF an effective inhibitor of deoxy-HbS polymerization. The level of HbF required to reduce the symptoms of sickle cell anemia is 20-25%, but levels as low as 9% can prolong red cell survival. Treatment with hydroxyurea (HU) induces HbF and reduces the hematologic and clinical consequences of sickle cell anemia. Basal and inducible HbF levels are important in predicting the severity of sickle cell anemia and are highly phenotypically variable among patients, leading to varied responses to treatment. Patients are also variably susceptible to HU-induced cytopenias, which limits the use of HU in certain patients. HU is currently the only FDA-approved HbF-inducer for the treatment of sickle cell anemia and there is clearly a need for alternative HbF-inducers. Understanding the signaling pathways that regulate HbF induction will lead to novel therapeutic targets for sickle cell anemia. The soluble guanylate cyclase/cyclic guanosine monophosphate-dependent protein kinase (sGC/PKG) signaling pathway potentially links HU to the induction of HbF expression. In this study we investigated the direct role of sGC in HbF induction using novel pharmacologic modulators of sGC. Nitric oxide (NO) activates sGC by binding to the ferrous iron (Fe2+) in the active site heme moiety. Once activated, sGC converts GTP to cGMP, which in-turn activates PKG. Reactive oxygen species (ROS) oxidize the active site heme of sGC leading to NO-insensitivity. We tested the ability of a novel sGC activator, BAY 58-2667, to induce γ-globinin primary and immortalized (HUDEP-2) human erythroid progenitor cells. BAY 54-6544 binds to heme-free inactivated sGC to restore its guanylyl cyclase activity independent of NO. We also tested the ability of the sGC stimulator, BAY 41-2272, to induce γ-globinin primary and HUDEP-2 human erythroid progenitor cells. BAY 41-2272, binds to the ferrous iron at the active site of non-oxidized sGC to stimulate guanylyl cyclase activity in a synergistic manner with NO. We compared g-globin mRNA and protein expressionin the primary and immortalized human erythroid progenitors after treatment with different concentrations and combinations of BAY 54-6544, BAY 41-2667 and HU. We also evaluated g-globin induction in cellstreated with the pan-phosphodiesterase inhibitor IBMX and a synthetic cGMP analog. Although we see robust induction of cGMP and activation of PKG with all treatments, we only see significant induction of g-globin expression in the HU treated cells. This data suggests that the induction of HbF occurs through a non-sGC/PKG-dependent signaling pathway. These data demonstrate a very limited induction of γ-globin by BAY 54-6544 and BAY 41-2667 that appears to be disproportionate to, and independent of, cGMP/PKG signaling. These data also demonstrate, for the first time, that HU treatment of the immortalized HUDEP-2 cell line induces γ-globin expression more consistently than in primary erythroid progenitors. 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: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Society of Hematology ; 2007
    In:  Blood Vol. 110, No. 11 ( 2007-11-16), p. 707-707
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 707-707
    Abstract: Erythroid iron deficiency, whether due to dimished body stores or histiocytic retention, diminishes marrow responsiveness to erythropoietin (Epo). Conversely, intravenous iron infusion augments marrow responsiveness to Epo, even in anemia patients with adequate pre-existing iron stores. Iron regulation of Epo-driven erythropoiesis affects proliferation and differentiation of early progenitors, prior to the commitment to heme production. Thus, while iron is essential for all cells, erythroid precursors manifest an exquisite sensitivity to iron deficiency, most likely as a rationing mechanism to protect other, more vital iron-dependent functions. Using primary human hematopoietic cultures with defined levels of transferrin saturation, we have confirmed the existence of a critical threshold of iron deprivation, at which erythroid progenitors display proliferative and maturation blockade while granulopoiesis and megakaryopoiesis remain unaffected. Extensive pharmacologic and genetic screening for components of this erythroid iron response pathway have identified the iron-sulfur cluster containing aconitase enzymes as a critical signaling node. Mitochondrial and cytosolic aconitase (mAcon & cAcon) interconvert citrate and isocitrate as a key step in the Krebs cycle. Firstly, treatment of iron deprived erythroid cultures with exogenous isocitrate, but not citrate, completely restored differentiation, as judged by glycophorin A (GPA) expression and hemoglobinization. By contrast, both citrate and isocitrate enhanced erythroid differentiation under iron replete conditions. Secondly, treatment of iron replete erythroid cultures with a specific aconitase inhibitor, fluorocitrate, induced a lineage-selective maturation blockade identical to that seen with iron deprivation. Thirdly, enzymography showed erythroid-lineage specific inactivation of both cAcon and mAcon in response to iron deprivation; immunoblotting showed no change mAcon protein levels as a function of either lineage or iron status. Fourthly, a retroviral genetic screen identified HBLD2, an iron-sulfur cluster assembly factor, as a protein whose overexpression reversed the erythroid maturation blockade associated with iron deprivation. Enzymography confirmed that overexpression of HBLD2 enhanced both cAcon and mAcon activities. Fifthly, administration to wild type, iron replete C57BL/6 mice of isocitrate at 200 mg/kg/day for 5 days caused a significant increase in peripheral red cell hemoglobinization, reflected by MCHC and Hb levels. Taken together, these results identify isocitrate as a positive regulator of Epo-mediated erythroid differentiation. In Epo-independent CD34+ cell culture systems, isocitrate did not enhance erythropoiesis. Therefore aconitase enzymes serve as a critical nexus in iron and Epo regulation of erythropoiesis, integrating cellular metabolism with developmental programming. The unique sensitivity of the erythroid lineage to iron deprivation appears to derive from a cellular milieu effect on the aconitase enzymes, promoting their inactivation under iron deprivation. Notably, IRP repression of mAcon translation does not contribute to the erythroid iron deprivation response pathway. This pathway may have relevance for future clinical approaches to Epo-refractory chronic anemias and polycythemia vera.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Society of Hematology ; 2012
    In:  Blood Vol. 120, No. 20 ( 2012-11-15), p. 4219-4228
    In: Blood, American Society of Hematology, Vol. 120, No. 20 ( 2012-11-15), p. 4219-4228
    Abstract: In red cell development, the differentiation program directed by the transcriptional regulator GATA1 requires signaling by the cytokine erythropoietin, but the mechanistic basis for this signaling requirement has remained unknown. Here we show that erythropoietin regulates GATA1 through protein kinase D activation, promoting histone deacetylase 5 (HDAC5) dissociation from GATA1, and subsequent GATA1 acetylation. Mice deficient for HDAC5 show resistance to anemic challenge and altered marrow responsiveness to erythropoietin injections. In ex vivo studies, HDAC5−/− progenitors display enhanced entry into and passage through the erythroid lineage, as well as evidence of erythropoietin–independent differentiation. These results reveal a molecular pathway that contributes to cytokine regulation of hematopoietic differentiation and offer a potential mechanism for fine tuning of lineage-restricted transcription factors by lineage-specific cytokines.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Society of Hematology ; 2009
    In:  Blood Vol. 114, No. 22 ( 2009-11-20), p. 75-75
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 75-75
    Abstract: Abstract 75 Erythropoiesis is a complex developmental program regulated in part by erythropoietin (EPO) and iron levels. We have previously shown that erythroid iron deprivation causes diminished enzymatic activity of the multifunctional aconitase proteins (Bullock et al., ASH 2007 #707). In addition, pharmacological inhibition of aconitase impairs erythroid growth and maturation in a lineage-selective manner without compromising ATP levels, suggesting a novel, non-metabolic regulatory role for aconitase in erythropoiesis (Talbot et al., ASH 2008 #3865). The nature of this regulatory role was addressed in the present study by examining the connection between aconitase activity and erythroid signal transduction. The impact of aconitase inhibition by fluoroacetate (FA) on several candidate pathways was assessed in primary human erythroid progenitors by standard immunoblotting. This approach consistently implicated the mitogen activated protein kinases (MAPKs) ERK1/2, whose phosphorylation was strongly induced both at early (30 minutes) and late (4 days) time points. In addition, phosphorylation of the other MAPKs p38 and JNK was unchanged, ruling out a non-specific stress response. To determine why aconitase inhibition augmented ERK1/2 phosphorylation, the effects of FA were determined on the upstream kinases MEK1/2. Surprisingly, phosphorylation of MEK1/2 showed no changes at earlier time points (up to 24 hours) and decreases at later time points (2–4 days). Activation of ERK/1/2 in the absence of MEK1/2 activation suggests an unconventional pathway in which aconitase regulates dephosphorylation, scaffolding, or subcellular localization. The inhibition of erythropoiesis by FA could result from increased ERK1/2 activity or diminished MEK1/2 activity. The first mechanism predicts phenotypic rescue by addition of MEK inhibitor (U0126), whereas the second mechanism predicts increased inhibition. In fact, U0126 alone recapitulated the anti-proliferative and anti-differentiative effects of FA on primary erythroid cultures, and the combination of U0126 and FA caused increased inhibition of growth and differentiation, arguing in favor of MEK1/2 repression as a mechanism for FA. Our findings establish that aconitase modulates signaling through the MEK-ERK pathway, perturbation of which may block erythropoiesis. To expand our studies on the in vivo role of aconitase in erythropoiesis, the impact of aconitase blockade was assessed in two additional murine models: phenylhydrazine (PHZ)-induced stress erythropoiesis and Polycythemia vera (PV). In both systems, erythropoiesis was found to be extremely sensitive to aconitase blockade. In the first system, adult C57Bl/6 female mice were treated with continuous FA infusion plus bolus intraperitoneal PHZ. FA-treated mice developed a much more severe anemia in response to PHZ than did non-FA-treated mice despite receiving 50% less PHZ. Their anemia showed a lower nadir (red cell count of 3.56 M/ul vs. 4.75 M/ul, p=0.0013, n=6) and a more prolonged trough. Their reticulocytosis was delayed and diminished. In the second system, PV female mice carrying one copy of the JAK2 V617F allele were treated with continuous FA infusion. Decreased red indices were observed by day 11 of treatment, with a decline in hematocrit from 51.3% to 42.5% in PV mice (p=0.0002, n=4), compared with a decline from 44.6% to 37.9% in wild type mice (p=0.04, n=4). No effects were observed on platelet and leukocyte counts. Interestingly, the rate of decrease of the red cell parameters was greater in the PV mice, suggesting that erythropoiesis driven by JAK2 V617F is highly responsive to changes in aconitase activity levels. We conclude that aconitase functions as a modifier of erythroid signaling to regulate proliferation and differentiation. Indeed, our results indicate that aconitase activity regulates steady state, stress, and JAK2 V617F-driven erythropoiesis, most likely through its novel modulation of MEK-ERK signaling. This novel function may provide an alternate targeting strategy in the treatment of Polycythemia vera. 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: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Society of Hematology ; 2014
    In:  Blood Vol. 124, No. 21 ( 2014-12-06), p. 216-216
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 216-216
    Abstract: Iron and erythropoietin (Epo) are intimately linked regulators of erythropoiesis. Moderate iron restriction suppresses erythropoiesis at the Epo-dependent, CFU-E stage, without induction of apoptosis and without suppression of other hematopoietic cell lineages. Iron modulates Epo bioactivity in patients with iron deficiency anemia (IDA) and patients with anemia of chronic disease and inflammation (ACDI). To conserve iron when supplies are low, this erythroid iron restriction response reduces iron consumption by suppressing erythropoiesis. The erythroid iron sensor is unknown. Aconitases are multifunctional iron-sulfur cluster proteins localized in the cytosol (Aco1) and mitochondria (Aco2) that convert citrate into isocitrate. We have shown that iron restriction inhibits Aco2 enzymatic activity leading to suppression of erythropoiesis in vitro, and these effects are reversed by isocitrate. Isocitrate corrects IDA in mice and ACDI in rats (Bullock GC, et al. Blood. 2010;116:97-108; Richardson CL, et al. J Clin Invest. 2013 Aug 1;123(8):3614-3623). Iron restriction also alters the cross-talk between transferrin receptor and Epo receptor signaling pathways. These results suggest that Aco2 is an iron-responsive regulator of erythropoiesis. We are investigating the downstream molecular signaling mechanisms by which iron restriction induced-inactivation of Aco2 suppresses erythropoiesis. Our novel preliminary data show that mitochondrial oxidative metabolism rates change over time during erythropoiesis and that iron restriction reduces erythroid mitochondrial metabolism 4 to 7-fold compared to iron replete controls. This iron restriction induced change in respiration is associated with a significant, 1.5 to 3-fold, increase in mitochondrial superoxide production without a corresponding increase in hydrogen peroxide. Importantly, these mitochondrial alterations are reproduced by direct inhibition of aconitase with fluoroacetate (FA) and are not due to changes in mitochondrial number. Further, isocitrate reverses the effects of iron restriction or aconitase inhibition on mitochondrial metabolism and attenuates superoxide production. Based on these data and the known role of reactive oxygen species (superoxide/hydrogen peroxide) in Epo signaling, we propose the overarching hypothesis that iron restriction inhibits mitochondrial aconitase which, in turn, alters erythroid mitochondrial metabolism and ROS signaling resulting in suppression of erythropoiesis (Figure 1). We show for the first time bioenergetics profiles from iron restricted and iron replete primary human erythroid progenitor cells undergoing erythropoiesis. We also show that moderate levels of iron restriction cause mitochondrial dysfunction and alterations in mitochondrial ROS in differentiating erythroid progenitors. The clinical relevance of this project lies in its potential for the development of new iron-free agonists and antagonists of red blood cell production. Agonists may benefit patients with anemia due to iron deficiency or chronic inflammation and antagonists may benefit patients with myeloproliferative neoplasms. Figure 1: Proposed mechanism of iron-dependent regulation of erythropoiesis Figure 1 Figure 1. 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 ...
  • 8
    Online Resource
    Online Resource
    American Society of Hematology ; 2010
    In:  Blood Vol. 116, No. 21 ( 2010-11-19), p. 166-166
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 166-166
    Abstract: Abstract 166 The erythroid iron deprivation response results from lineage-selective inactivation of aconitase enzymes, causing diminished erythropoietin (Epo) responsiveness in erythroid progenitors. Provision of exogenous isocitrate in either cell culture or murine models of iron deficiency restores Epo responsiveness and abrogates the erythropoietic block characteristic of the iron deprivation response. Although isocitrate administration can restore erythropoiesis in iron deficient mice, the response is transient, and iron administration is required for sustained correction. In anemias other than those due to iron deficiency, inappropriate activation of this iron deprivation pathway might also contribute to suppression of erythropoiesis. Such anemias are predicted to respond to isocitrate administration. A major area of clinical controversy is the degree to which iron restriction plays a role anemia of chronic inflammation (ACI). In support of such a role, inflammatory stimuli promote increased hepcidin production by the liver, and diminished serum iron represents a consistent finding in patients with ACI. Challenging such a role, a recently published series of elderly patients with ACI showed no evidence of increased hepcidin production (Ferrucci et al., Blood 115:3810, 2010). Furthermore, ACI usually manifests as a normochromic, normocytic anemia in contrast to the microcytic, hypochromic anemia of iron deficiency. Finally, administration of anti-TNF antibody to patients with rheumatoid arthritis corrected their associated anemia, suggesting a role for direct cytokine repression of erythropoiesis (Papadaki et al., Blood 100:474, 2002). To experimentally examine the role of the erythroid iron deprivation response in ACI, we determined the effects of isocitrate administration in a classic rat arthritis model of ACI. 6 week-old female Lewis rats received a single IP injection (15 μ g rhamnose/g) of Streptococcal cell wall peptidoglycan-polysaccharide (PG-PS). Normochromic, normocytic anemia developed at 2 weeks post injection. Specifically, the mean red cell count (RBC) in rats receiving PG-PS was 7.18 ± 0.22 × 106 cells/μ l vs 8.65 ± 0.39 × 106 cells/μ l in rats not receiving PG-PS (P = 0.014). At this time, the anemic rats underwent daily IP injections with either trisodium isocitrate (200 mg/kg/day) or with equivalent volumes of saline. Rats receiving isocitrate showed correction of anemia after 3 days of treatment, with a mean RBC of 8.14 ± 0.06 × 106 cells/μ l as opposed to a mean RBC in the control group of 6.42 ± 0.45 × 106 cells/μ l (P = 0.018). This correction was maintained after 5 additional days of treatment: RBC in isocitrate-treated group of 8.36 ± 0.24 × 106 cells/μ l versus RBC in saline-treated group of 7.01 ± 0.19 × 106 cells/μ l (P = 0.004). No differences in neutrophil or platelet counts were observed at any point in rats receiving isocitrate vs saline. These results support a role for the erythroid iron deprivation response in the impaired erythropoiesis associated with ACI. These results also support our previous in vitro findings that iron deprivation sensitizes erythroid progenitors to inhibition by inflammatory cytokines (i.e. IFNγ or TNFα) (Richardson et al., ASH 2009 #159). More recent in vitro studies on the mechanisms underlying this sensitization have addressed whether iron restriction synergizes with inflammatory cytokines in promoting aconitase inactivation. Using a gel-based enzymatic assay, we confirmed the inhibitory effect of iron deprivation on both cytosolic and mitochondrial aconitase isoforms, but could find no inhibitory effects associated with either IFNγ or TNFα treatment. In subsequent experiments, a functional screen for elements integrating the erythroid iron deprivation response with inflammatory signaling implicated a calmodulin-regulated factor. Specifically, the calmodulin inhibitor, KN62 reversed the cell death observed with the combination of iron deprivation plus inflammatory cytokines but had minimal effects on viability of cells subjected to either iron deprivation or inflammatory cytokines separately (n = 4). These data thus delineate an iron-regulated signaling element downstream of aconitase which employs calmodulin to modulate erythroid responsiveness to inflammatory cytokines. Pharmacologic targeting of this element, as with isocitrate administration, may provide a new avenue for clinical management of ACI. 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: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 116, No. 1 ( 2010-07-08), p. 97-108
    Abstract: Human red cell differentiation requires the action of erythropoietin on committed progenitor cells. In iron deficiency, committed erythroid progenitors lose responsiveness to erythropoietin, resulting in hypoplastic anemia. To address the basis for iron regulation of erythropoiesis, we established primary hematopoietic cultures with transferrin saturation levels that restricted erythropoiesis but permitted granulopoiesis and megakaryopoiesis. Experiments in this system identified as a critical regulatory element the aconitases, multifunctional iron-sulfur cluster proteins that metabolize citrate to isocitrate. Iron restriction suppressed mitochondrial and cytosolic aconitase activity in erythroid but not granulocytic or megakaryocytic progenitors. An active site aconitase inhibitor, fluorocitrate, blocked erythroid differentiation in a manner similar to iron deprivation. Exogenous isocitrate abrogated the erythroid iron restriction response in vitro and reversed anemia progression in iron-deprived mice. The mechanism for aconitase regulation of erythropoiesis most probably involves both production of metabolic intermediates and modulation of erythropoietin signaling. One relevant signaling pathway appeared to involve protein kinase Cα/β, or possibly protein kinase Cδ, whose activities were regulated by iron, isocitrate, and erythropoietin.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: British Journal of Haematology, Wiley, Vol. 187, No. 5 ( 2019-12), p. 666-675
    Abstract: Haemolysis is a major feature of sickle cell disease (SCD) that contributes to organ damage. It is well established that haem, a product of haemolysis, induces expression of the enzyme that degrades it, haem oxygenase‐1 (HMOX1). We have also shown that haem induces expression of placental growth factor (PGF), but the organ specificity of these responses has not been well‐defined. As expected, we found high level expression of Hmox1 and Pgf transcripts in the reticuloendothelial system organs of transgenic sickle cell mice, but surprisingly strong expression in the heart ( P   〈  0·0001). This pattern was largely replicated in wild type mice by intravenous injection of exogenous haem. In the heart, haem induced unexpectedly strong mRNA responses for Hmox1 (18‐fold), Pgf (4‐fold), and the haem transporter Slc48a1 (also termed Hrg1 ; 2·4‐fold). This was comparable to the liver, the principal known haem‐detoxifying organ. The NFE2L2 (also termed NRF2 ) transcription factor mediated much of the haem induction of Hmox1 and Hrg1 in all organs, but less so for Pgf . Our results indicate that the heart expresses haem response pathway genes at surprisingly high basal levels and shares with the liver a similar transcriptional response to circulating haem. The role of the heart in haem response should be investigated further.
    Type of Medium: Online Resource
    ISSN: 0007-1048 , 1365-2141
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2019
    detail.hit.zdb_id: 1475751-5
    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...