1
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Toward a Novel Therapeutic Option for Polycythemia

      review-article
      HemaSphere
      Wolters Kluwer Health

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Red blood cell (RBC) production is a finely tuned process that requires coordinated oxygen- and iron-dependent regulation of cell differentiation and iron homeostasis. Excess production of erythrocytes, referred to as erythrocytosis or polycythemia, occurs physiologically, as an adjustment to high altitude, or pathologically, because of intrinsic abnormalities in erythroid precursors or inappropriately high level of erythropoietin (EPO). In response to hypoxia, RBC number is increased as a compensatory mechanism in an attempt to enhance oxygen availability and respiratory capacity. Hypoxia promotes EPO release by the kidney, which in turn stimulates RBC production. This process is orchestrated by key transcription factors, called hypoxia-inducible factors (HIFs). HIFs are increased in response to anemia as well as low oxygen and iron levels. Exposure to low oxygen levels causes the stabilization of the α subunits of HIFs (HIF1α, HIF2α), thus leading to the transcription of HIF targets, including EPO, and therefore erythrocytosis. HIF2α is regulated at the posttranslational level by prolyl hydroxylases (PHDs) that use oxygen and iron as substrates to hydroxylate HIF2α. Following hydroxylation, HIF2α undergoes ubiquitination by the von Hippel–Lindau (VHL)-E3 ubiquitin ligase and degradation through the proteasomal pathway. Hence, HIF2α can sense hypoxia and iron deficiency, and then increases EPO expression and drives RBC production. Missense mutations in VHL cause Chuvash polycythemia, an autosomal recessive disorder hallmarked by congenital erythrocytosis, due to excessive HIF2α stabilization (Fig. 1). Figure 1 Tempol prevents hypoxia-induced erythrocytosis by inhibiting HIF2α-mediated EPO production. In iron-replete conditions, IRP1 exerts cytosolic aconitase activity dependent on iron–sulfur cluster; in iron-deplete conditions, IRP1 loses the iron–sulfur cluster group and binds to the IRE sequence of target mRNAs, thus regulating the expression of iron-related genes. In renal fibroblasts, by binding HIF2α IRE, IRP1 regulates translationally the expression of HIF2α according to iron and oxygen status. In hypoxia or iron deficiency conditions, HIF2α protein is stabilized due to the inactivation of PHD-VHL degradation pathway and translocates to the nucleus, where it transcriptionally activates EPO expression. EPO in turn acts on erythroblasts to stimulate RBC production. This process occurs under hypoxic conditions associated with high altitude and with pathological elevation of EPO (e.g., Chuvash mutation). Tempol, by activating the IRE binding activity of IRP1, mediates HIF2α translational repression, which leads to reduced EPO production. This effect prevents hypoxia-induced erythrocytosis. Tempol can be applied as therapeutic strategy to counteract high altitude-triggered eryhtrocytosis as well as high HIF2α-mediated polytcythemia. The intracellular level of HIFα subunits are also regulated by iron regulatory proteins (IRP1, IRP2). IRPs act as cytosolic iron sensors and control the fate of messenger RNAs (mRNA) encoding proteins involved in iron metabolism. This role is dependent on their ability to bind specific RNA stem-loop structures, termed iron-responsive elements (IREs), on specific target mRNAs and control the translation of the encoded protein. IRP1 is a bifunctional protein that binds to IREs as an apo-protein in iron-deficient conditions (apo-IRP1), and it converts to cytosolic aconitase through the acquisition of an iron–sulfur [4Fe–4S] cluster in iron-replete conditions (holo-IRP1). IRP targets include transferrin receptor 1 (TfR1), divalent metal ion transporter 1 (DMT1), the iron exporter ferroportin (FPN) and the iron storage protein ferritin (Ft). The RNA binding activity of the IRPs is therefore regulated by cellular iron, being decreased in iron-replete cells and increased in iron-deplete ones. Hence, by regulating IRP activity, intracellular iron levels control the expression of IRP targets, adjusting iron handling (uptake, storage, and export) accordingly. As for iron, hypoxic conditions inactivate IRP1 by favoring holo-IRP1 formation and inhibiting IRE-binding activity, and relieve IRE-bearing transcripts from IRP1-mediated repression. HIF2α has been described as one of the targets of IRP1. In 2013, different groups showed that mice lacking IRP1 are hallmarked by elevated HIF2α levels, increased EPO production and polycythemia. 1–3 These findings supported the concept that IRP1 has a crucial role in repressing HIF2α translation, thus keeping in balance RBC production. Conversely, translational derepression of HIF2α triggers hypoxia-like erythrocytosis. IRP1-deficient mice develop severe iron deficiency, likely caused by increased erythropoiesis that consumes large amounts of iron for RBC production, and consequently depletes circulating transferrin-bound iron as well as tissue iron stores. IRP1-mediated regulation of HIF2α translation acts as a protective mechanism that prevents erythropoiesis from consuming too much iron, thus depleting systemic iron. While HIF2α senses hypoxia and stimulates EPO expression and RBC production, IRP1 fine-tunes HIF2α expression to ensure that there is enough iron available for iron–sulfur cluster synthesis. When too much iron is consumed, and systemic iron levels are low, iron–sulfur cluster synthesis is impaired and IRP1 is converted to the IRE-binding form, which represses HIF2α translation, and thereby restricts RBC production. Through this feedback mechanism, IRP1 controls the balance between systemic iron homeostasis and erythropoiesis. 3 Recently Ghosh et al 4 showed that mice bearing the human missense mutation VHLR200W develop Chuvash polycythemia, which closely reflects erythrocytosis observed in IRP1-deficient animals. VHLR200W-mutant mice feature increased RBC counts, elevated hemoglobin levels, splenomegaly, and skin erythema. The authors used a stable nitroxide radical, Tempol (4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl), as a therapeutic approach to correct polycythemia in these animals. 4 Tempol is a membrane-permeable free radical scavenger with antioxidant properties. It can degrade superoxide radicals in a superoxide dismutase (SOD) mimetic manner and suppress the formation of hydroxyl radicals by inhibiting Fenton's reaction. Importantly, Tempol has the ability to activate the IRE-binding activity of IRP1. Therefore the rational for its administration in Chuvash polycythemia is based on the attempt to inhibit HIF2α translation through enhanced HIF2α IRE binding of IRP1. In the mouse model of Chuvash polycythemia, Tempol ameliorates erythrocytosis by lowering HIF2α expression and EPO levels (Fig. 1). 4 The lack of hematocrit improvement in IRP1-deficient animals confirmed that Tempol-mediated amelioration of erythrocytosis requires IRP1 activation. Consistently, the authors elegantly showed that the therapeutic action of Tempol on Chuvash polycythemia is abrogated by the deletion of IRP1 in VHLR200W-mutant mice. These findings open a new perspective in the treatment of Chuvash polycythemia, for which phlebotomy is the primary standard therapy to date. Recurrent phlebotomies might result in a contradictory effect: phlebotomy-induced iron deficiency, by directly stabilizing HIF2α, can further stimulate erythropoiesis, thus only temporarily and partially restoring hematocrit levels. The use of Tempol would overcome the limitation of the current therapeutic approach, by reducing HIF2α levels. Interestingly, Tempol shows beneficial effects in other diseases hallmarked by elevated HIF2α and/or VHL mutations, including VHL-deficient clear cell renal cell carcinoma (CCRCC), or neurodegeneration associated with IRP2 loss and altered cell iron homeostasis. 5 Importantly, Ghosh et al demonstrated the even broader relevance of this therapeutic strategy for individuals exposed to hypoxic conditions in high altitude, who commonly develop erythrocytosis. Wild-type mice exposed to prolonged hypoxia are in part protected from polycythemia development and show a longer life expectancy when treated with Tempol (Fig. 1). 4 Thus Tempol represents a potentially valuable therapy to limit polycythemia triggered by high altitude and its associated side effects.

          Related collections

          Most cited references5

          • Record: found
          • Abstract: found
          • Article: not found

          Deletion of iron regulatory protein 1 causes polycythemia and pulmonary hypertension in mice through translational derepression of HIF2α.

          Iron regulatory proteins (Irps) 1 and 2 posttranscriptionally control the expression of transcripts that contain iron-responsive element (IRE) sequences, including ferritin, ferroportin, transferrin receptor, and hypoxia-inducible factor 2α (HIF2α). We report here that mice with targeted deletion of Irp1 developed pulmonary hypertension and polycythemia that was exacerbated by a low-iron diet. Hematocrits increased to 65% in iron-starved mice, and many polycythemic mice died of abdominal hemorrhages. Irp1 deletion enhanced HIF2α protein expression in kidneys of Irp1(-/-) mice, which led to increased erythropoietin (EPO) expression, polycythemia, and concomitant tissue iron deficiency. Increased HIF2α expression in pulmonary endothelial cells induced high expression of endothelin-1, likely contributing to the pulmonary hypertension of Irp1(-/-) mice. Our results reveal why anemia is an early physiological consequence of iron deficiency, highlight the physiological significance of Irp1 in regulating erythropoiesis and iron distribution, and provide important insights into the molecular pathogenesis of pulmonary hypertension. Copyright © 2013 Elsevier Inc. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The IRP1-HIF-2α axis coordinates iron and oxygen sensing with erythropoiesis and iron absorption.

            Red blood cell production is a finely tuned process that requires coordinated oxygen- and iron-dependent regulation of cell differentiation and iron metabolism. Here, we show that translational regulation of hypoxia-inducible factor 2α (HIF-2α) synthesis by iron regulatory protein 1 (IRP1) is critical for controlling erythrocyte number. IRP1-null (Irp1(-/-)) mice display a marked transient polycythemia. HIF-2α messenger RNA (mRNA) is derepressed in kidneys of Irp1(-/-) mice but not in kidneys of Irp2(-/-) mice, leading to increased renal erythropoietin (Epo) mRNA and inappropriately elevated serum Epo levels. Expression of the iron transport genes DCytb, Dmt1, and ferroportin, as well as other HIF-2α targets, is enhanced in Irp1(-/-) duodenum. Analysis of mRNA translation state in the liver revealed IRP1-dependent dysregulation of HIF-2α mRNA translation, whereas IRP2 deficiency derepressed translation of all other known 5' iron response element (IRE)-containing mRNAs expressed in the liver. These results uncover separable physiological roles of each IRP and identify IRP1 as a therapeutic target for manipulating HIF-2α action in hematologic, oncologic, and other disorders. Copyright © 2013 Elsevier Inc. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              IRP1 regulates erythropoiesis and systemic iron homeostasis by controlling HIF2α mRNA translation.

              Hypoxia inducible factor 2α (HIF2α) transcriptionally activates several genes in response to hypoxia. Under normoxic conditions, it undergoes oxygen-dependent degradation by the prolyl hydroxylase (PHD)/von Hippel-Lindau (VHL) system. The presence of an iron-responsive element (IRE) within the 5' untranslated region of HIF2α mRNA suggests a further iron- and oxygen-dependent mechanism for translational regulation of its expression via iron regulatory proteins 1 and 2 (IRP1 and IRP2, respectively). We show here that the disruption of mouse IRP1, but not IRP2, leads to profound HIF2α-dependent abnormalities in erythropoiesis and systemic iron metabolism. Thus, 4- to 6-week-old IRP1(-/-) mice exhibit splenomegaly and extramedullary hematopoiesis, which is corrected in older animals. These erythropoietic abnormalities are caused by translational de-repression of HIF2α mRNA and subsequent accumulation of HIF2α, which induces expression of erythropoietin (Epo). Increased levels of circulating Epo lead to reticulocytosis, polycythemia, and suppression of hepatic hepcidin mRNA. This in turn promotes hyperferremia and iron depletion in splenic macrophages due to unrestricted expression of ferroportin. Our data demonstrate that IRP1 is the principal regulator of HIF2α mRNA translation in vivo and provide evidence that translational control of HIF2α expression dominates over PHD/VHL-mediated regulation of HIF2α stability in juvenile IRP1(-/-) mice.
                Bookmark

                Author and article information

                Journal
                Hemasphere
                Hemasphere
                HS9
                HemaSphere
                Wolters Kluwer Health
                2572-9241
                27 July 2018
                August 2018
                : 2
                : 4
                : e139
                Affiliations
                Lindsley F. Kimball Research Institute (LFKRI), New York Blood Center, New York, NY, USA
                Author notes
                Correspondence: Francesca Vinchi (e-mail: FVinchi@ 123456nybc.org ).
                Article
                HemaSphere-2018-0143 003
                10.1097/HS9.0000000000000139
                6746006
                8bdeb5a6-f46c-4f98-bda6-58615e1f50fc
                Copyright © 2018 the Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the European Hematology Association.

                This is an open access article distributed under the terms of the Creative Commons Attribution-Non Commercial-No Derivatives License 4.0 (CCBY-NC-ND), where it is permissible to download and share the work provided it is properly cited. The work cannot be changed in any way or used commercially without permission from the journal. http://creativecommons.org/licenses/by-nc-nd/4.0

                History
                Categories
                HemaTopics
                Custom metadata
                TRUE

                Comments

                Comment on this article