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
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Elsevier BV ; 2011
    In:  Biophysical Journal Vol. 100, No. 3 ( 2011-02), p. 233a-
    In: Biophysical Journal, Elsevier BV, Vol. 100, No. 3 ( 2011-02), p. 233a-
    Type of Medium: Online Resource
    ISSN: 0006-3495
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2011
    detail.hit.zdb_id: 1477214-0
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Society of Hematology ; 2014
    In:  Blood Vol. 124, No. 21 ( 2014-12-06), p. 4068-4068
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 4068-4068
    Abstract: Thrombospondin-1 (TSP1) is a secreted matricellular protein found preformed in platelet α-granules and upregulated in the plasma of patients with sickle cell disease (SCD). In preclinical models TSP1 enhances sickle red blood cell (RBC) binding to endothelial cells, while in murine sickle mice TSP1 promotes RBC echinocytosis and vaso-occlusion. Recently we reported elevated plasma TSP1 was associated with vaso-occlusive complications in SCD patients. Herein we tested the hypothesis that TSP1 compromises RBC membrane integrity via enhanced calcium influx leading to morphological changes, such as echinocytosis. RBC were collected via centrifugation of heparinized blood from human HbAA donors (n=3), SCD patients with homozygous HbSS SCD (n=4), and wild type C57BL/6J mice (n=2). Cells were then re-suspended in glucose and calcium-supplemented PBS, and treated with several concentrations of exogenous TSP1 (2.75 nM, 11 nM, 22 nM and 44 nM) for 2 hours at 37°C. RBC morphology was assessed by both light and electron microscopy. TSP1 modulation of RBC calcium influx was studied using the calcium fluorophore Fluo-3, AM. TSP1 treatment, in a dose-dependent manner, stimulated echinocytosis in human SCD and non-SCD RBC and in murine cells. In some instances electron microscopy showed echinocyte membrane projections with a string-of-beads appearance, suggestive of imminent microparticle shedding. TSP1 treatment, again in a dose-dependent manner, also significantly increased calcium influx in SCD (Figure, right panel) and non SCD RBC (Figure, left panel) and murine RBC (not shown). Interestingly, SCD RBC displayed more sensitivity to TSP1-mediated increases in cellular calcium as compared to normal cells. Our results show for the first time that TSP1, at doses found in patient plasma, stimulates significant echinocytosis and calcium influx in both normal and SCD RBC. These data identify a novel role for plasma TSP1 in promoting RBC pathology. Future studies are underway to elucidate this mechanism and its pathogenicity in humans with and without SCD. Figure 1 Figure 1. Disclosures Isenberg: Vasculox: Membership on an entity's Board of Directors or advisory committees; Radiation Control Technologies: 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: 2014
    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
    Online Resource
    Online Resource
    SAGE Publications ; 2015
    In:  Journal of Investigative Medicine Vol. 63, No. 6 ( 2015-08), p. 806-810
    In: Journal of Investigative Medicine, SAGE Publications, Vol. 63, No. 6 ( 2015-08), p. 806-810
    Abstract: Interleukin-33 (IL-33) is a member of the IL-1 cytokine superfamily that potently drives production of a variety of cytokines and contributes to the pathogenesis of inflammatory diseases. The IL-33 is a nuclear protein and is released from apoptotic or necrotic cells. Serum IL-33 levels are increased in various diseases, such as atopic dermatitis, chronic hepatitis C infection, and asthma. Here, we show that red blood cells (RBCs) are one of the major sources of plasma IL-33. The IL-33 levels are significantly increased in supernatants from lysed RBCs. Plasma IL-33 levels are increased in patients during hemolysis, and plasma IL-33 levels show a positive correlation with degree of hemolysis. The IL-33 protein and messenger RNA levels were detected in the late stages of differentiation in ex vivo primary human erythroid progenitor cell cultures, suggesting that IL-33 is expressed during maturation of RBCs. Furthermore, hemoglobin depleted red cell lysates induced IL-8 expression in human epithelial cells. This effect was attenuated in IL-33 decoy receptor expressing cells and was enhanced in IL-33 receptor expressing cells. These results suggest that erythroid progenitor cells produce IL-33 and circulating RBCs represent a major source of IL-33 that is released upon hemolysis.
    Type of Medium: Online Resource
    ISSN: 1081-5589 , 1708-8267
    Language: English
    Publisher: SAGE Publications
    Publication Date: 2015
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    SAGE Publications ; 2018
    In:  Journal of Investigative Medicine Vol. 66, No. 1 ( 2018-01), p. 1-6
    In: Journal of Investigative Medicine, SAGE Publications, Vol. 66, No. 1 ( 2018-01), p. 1-6
    Abstract: Anemia is a very common blood disorder that affects the lives of billions of people worldwide. Anemia is caused by the loss of blood, increased destruction of red blood cells (RBCs), or reduced production of RBCs. Erythropoiesis is the complex process of RBC differentiation and maturation, in which protein degradation plays a crucial role. Protein ubiquitination regulates programmed protein degradation, which can be reversed by deubiquitinating enzymes (DUBs); however, the role of DUBs in erythropoiesis has not been well studied. We examined the expression of DUBs during erythropoiesis using an ex vivo human CD34+ hematopoietic progenitor cell culture system. Here we show that ubiquitin-specific protease 50 (USP50) levels are increased during erythropoiesis. USP50 mRNA levels are significantly increased on day 3 and protein levels are elevated on day 9 of erythroid differentiation. Coimmunoprecipitation and proteomics analyses reveal that Ku70, a DNA-binding protein, is associated with USP50. Overexpression of USP50 has no effect on Ku70 mRNA levels, while it reduces Ku70 protein levels by promoting Ku70 degradation, suggesting that USP50 may indirectly regulate Ku70 protein stability. USP50 protein is also not stable. USP50 protein degradation is independent of the proteasomal and the lysosomal degradation systems. This study suggests that DUBs like USP50 may regulate protein stability during erythropoiesis; however, more investigation is warranted.
    Type of Medium: Online Resource
    ISSN: 1081-5589 , 1708-8267
    Language: English
    Publisher: SAGE Publications
    Publication Date: 2018
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Society of Hematology ; 2013
    In:  Blood Vol. 122, No. 21 ( 2013-11-15), p. 3443-3443
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 3443-3443
    Abstract: Several clinical observations illustrate the link between iron and erythropoietin (Epo)-mediated signaling in early erythroid progenitor cells. In iron deficiency anemia (IDA) erythropoiesis is blocked at an early stage despite increased serum Epo concentrations. Intravenous iron improves the effectiveness of exogenous Epo in patients with Epo-refractory anemia of chronic disease. These clinical observations suggest that iron dominantly regulates Epo-receptor (EpoR) signaling. However, the mechanism of this iron-mediated signaling remains unclear. We recently demonstrated that 1) the aconitases, multifunctional iron-sulfur cluster proteins that convert citrate into isocitrate are essential in the iron- Epo-signaling pathway in early erythroid progenitors, and that 2) isocitrate, the downstream product of aconitase, can enhance the effectiveness of Epo during iron deficiency in vitro and in vivo in mice with IDA and in rats with the anemia of chronic inflammation. These observations suggest that isocitrate or derivatives of isocitrate that synergize with erythropoiesis stimulating agents have important therapeutic application in the treatment of anemia. New data from my lab also shows that cellular iron restriction regulates mitochondrial oxygen consumption rates differentially over time during red blood cell differentiation, suggesting a novel link between mitochondrial function and erythropoeisis. We also see an increase in mitochondrial superoxide anion production in iron deprived erythroid progenitors. Based on these data, we hypothesize that mitochondrial aconitase is an iron sensor that integrates mitochondrial redox signaling to EpoR signaling and subsequent red blood cell production. To test this hypothesis we are investigating the iron dependent mechanisms by which aconitase, isocitrate, reactive oxygen species and mitochondrial metabolic pathways alter EpoR signaling. 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. 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: 2013
    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. 126, No. 23 ( 2015-12-03), p. 2145-2145
    Abstract: Several clinical observations illustrate the link between iron and erythropoietin (EPO)-mediated signaling in committed erythroid progenitor cells. In iron deficiency anemia (IDA), erythropoiesis is blocked despite increased serum EPO concentrations. Intravenous iron improves the effectiveness of exogenous EPO in patients with EPO-refractory anemia of chronic disease. These clinical observations suggest that iron dominantly regulates EPO-receptor signaling. However, the mechanism of this iron-mediated signaling remains unclear. We recently demonstrated that 1) the aconitases, multifunctional iron-sulfur cluster proteins that convert citrate into isocitrate are essential in the iron- EPO-signaling pathway in erythroid progenitor cells, and that 2) isocitrate, the product of aconitase, can enhance the effectiveness of EPO during iron deficiency in vitro and in mice with IDA and in rats with the anemia of chronic inflammation. These observations suggest that isocitrate, or its derivatives that synergize with erythropoiesis stimulating agents, have important therapeutic application in the treatment of anemia. New data shows that cellular iron restriction regulates mitochondrial oxygen consumption rates (OCR) differentially over time during erythropoiesis, suggesting a novel link between mitochondrial function and erythropoeisis. It is unknown how iron deficiency induced inhibition of mitochondrial aconitase (ACO2) regulates mitochondrial metabolism during RBC production. Pilot data show that ACO2 inhibition by cellular iron deprivation or pharmacological inhibition of ACO2 decreases mitochondrial respiratory rates (RRs) and alters reactive oxygen species (ROS) production. Further, isocitrate normalizes mitochondrial RRs and ROS and restores RBC production. Importantly, disruption of mitochondrial ROS generation with a mitochondrial-specific anti-oxidant blocks RBC production and a subset of oxidant generators promote erythropoiesis. Targeted reduction of ACO2 protein expression and enzyme activity in iron replete stably transduced K562 cells decreases OCRs. This confirms the link between ACO2 and mitochondrial metabolism in human erythroid cells. These data inform our overarching hypothesis that iron-restriction inhibits ACO2, thereby inhibiting mitochondrial metabolism, resulting in the loss of a mitochondrial ROS signal that is required for erythropoiesis. The loss of this critical mitochondrial ROS signal inhibits the EPO signaling that is required for RBC production. These data also suggest that ACO2 is an iron-sensing regulator of mitochondrial metabolism and ROS signaling. 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: 2015
    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: 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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    Elsevier BV ; 2013
    In:  Biophysical Journal Vol. 104, No. 2 ( 2013-01), p. 264a-
    In: Biophysical Journal, Elsevier BV, Vol. 104, No. 2 ( 2013-01), p. 264a-
    Type of Medium: Online Resource
    ISSN: 0006-3495
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2013
    detail.hit.zdb_id: 1477214-0
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 74-74
    Abstract: Anemia is a global health problem that decreases quality of life for billions of people. Previous studies have concluded that there is an iron-regulated checkpoint in erythropoiesis that suppresses red blood cell production. This results in anemia and the conservation of iron for use in other vital processes. We have shown that the aconitase enzymes are key to this pathway and that inhibition of aconitase enzyme activity by iron restriction or pharmacologic inhibitors blocks erythropoiesis in primary human hematopoietic progenitor cells (HPCs). Mitochondrial aconitase (ACO2) functions as an isomerase within the tricarboxylic acid (TCA) cycle to convert citrate into isocitrate, which contributes to ATP and heme synthesis. During iron restriction, there is a significant decrease of intracellular isocitrate with only a slight increase in intracellular citrate. A corresponding increase in ATP-citrate lyase activity suggests that excess citrate is shunted into acetyl-CoA production. The addition of exogenous isocitrate to iron-deprived HPCs abrogates the block in erythropoiesis and protects iron-deprived mice and chronically-inflamed rats from anemia. These results suggest that ACO2 regulates mitochondrial metabolism and erythropoiesis. Recent unpublished data shows that ACO2 inhibition by iron deprivation or by treatment with an ACO2 inhibitor decreases mitochondrial respiratory rates (RR) and increases mitochondrial reactive oxygen species (mito-ROS). Isocitrate normalizes RR and mito-ROS and restores erythropoiesis. Importantly, disruption of ROS generation with a variety of anti-oxidants blocks erythropoiesis, while surprisingly, treatment of iron restricted HPCs with oxidant generators or ROS promotes erythropoiesis. These data inform our overarchinghypothesis that iron-restriction inhibits ACO2, thereby inhibiting mitochondrial metabolism, resulting in the loss of a mitochondrial ROS signal that is required for erythropoiesis. We have recently extended these studies to ACO2 knock down (ACO2-KD) K562 cell lines which provide more material for biochemical assessment of mitochondrial function. New pilot data shows that a 70% decrease in ACO2 expression significantly reduces the induction of erythroid specific genes during hydroxyurea or hemin treatment. This confirms that ACO2 plays a direct role in erythropoiesis. Extracellular flux (XF, Seahorse Bioscience) experiments show a decreased RR in ACO2-KD cells. Mitochondrial complex activity assays show no differences in complex IV or citrate synthase activity between control and ACO2-KD cell lines. These studies also demonstrate that the shRNA-ACO2 lentiviral constructs are effectively targeting ACO2 and can be used in our HPC model system. We now have evidence in two different human cell culture models of erythropoiesis that mitochondrial aconitase is an iron-sensing regulator of both mitochondrial respiration and erythropoiesis. Our long term goals are to identify novel therapeutic targets in this iron dependent metabolic regulatory pathway that enhance or suppress erythropoiesis and have potential clinical application in the treatment of anemia or polycythemia. We are also investigating the role of the mitochondrion in the differentiation of other hematopoietic cell lineages. 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: 2016
    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