751
|
Wallace DF, Subramaniam VN. Analysis of IL-22 contribution to hepcidin induction and hypoferremia during the response to LPS in vivo. Int Immunol 2015; 27:281-7. [PMID: 25568302 DOI: 10.1093/intimm/dxu144] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 12/23/2014] [Indexed: 12/16/2023] Open
Abstract
The anaemia of chronic disease (ACD) results from inflammation-mediated up-regulation of the iron regulatory hormone hepcidin, with the consequent sequestration of iron limiting its availability for erythropoiesis. The inflammatory cytokine IL-6, a regulator of hepcidin, has been implicated in this process. Recent in vivo and in vitro studies indicate that IL-22 is also able to stimulate hepcidin expression. We aimed to determine if IL-22 had a role in causing the hypoferremia associated with the inflammatory response. Wild-type and Il22-knockout mice were subjected to an acute inflammatory stimulus via administration of LPS and the response of hepcidin and iron homeostasis was analysed. In the absence of IL-22, there was a response of hepcidin, resulting in a reduction in serum iron levels. However, the hypoferremic response to LPS was slightly blunted in mice lacking IL-22, suggesting that, during LPS-mediated inflammation, IL-22 may play a minor role in mediating the hypoferremic response. These results may have implications for the treatment and management of the ACD.
Collapse
Affiliation(s)
- Daniel F Wallace
- Membrane Transport Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia and School of Medicine, University of Queensland, Brisbane, Queensland 4006, Australia
| | - V Nathan Subramaniam
- Membrane Transport Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia and School of Medicine, University of Queensland, Brisbane, Queensland 4006, Australia
| |
Collapse
|
752
|
Abstract
Macrophages play a critical role in iron homeostasis via their intimate association with developing and dying red cells. Central nurse macrophages promote erythropoiesis in the erythroblastic island niche. These macrophages make physical contact with erythroblasts, enabling signaling and the transfer of growth factors and possibly nutrients to the cells in their care. Human mature red cells have a lifespan of 120 days before they become senescent and again come into contact with macrophages. Phagocytosis of red blood cells is the main source of iron flux in the body, because heme must be recycled from approximately 270 billion hemoglobin molecules in each red cell, and roughly 2 million senescent red cells are recycled each second. Here we will review pathways for iron trafficking found at the macrophage-erythroid axis, with a focus on possible roles for the transport of heme in toto.
Collapse
|
753
|
|
754
|
Sankaran VG, Weiss MJ. Anemia: progress in molecular mechanisms and therapies. Nat Med 2015; 21:221-30. [PMID: 25742458 DOI: 10.1038/nm.3814] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 02/04/2015] [Indexed: 12/12/2022]
Abstract
Anemia is a major source of morbidity and mortality worldwide. Here we review recent insights into how red blood cells (RBCs) are produced, the pathogenic mechanisms underlying various forms of anemia, and novel therapies derived from these findings. It is likely that these new insights, mainly arising from basic scientific studies, will contribute immensely to both the understanding of frequently debilitating forms of anemia and the ability to treat affected patients. Major worldwide diseases that are likely to benefit from new advances include the hemoglobinopathies (β-thalassemia and sickle cell disease); rare genetic disorders of RBC production; and anemias associated with chronic kidney disease, inflammation, and cancer. Promising new approaches to treatment include drugs that target recently defined pathways in RBC production, iron metabolism, and fetal globin-family gene expression, as well as gene therapies that use improved viral vectors and newly developed genome editing technologies.
Collapse
Affiliation(s)
- Vijay G Sankaran
- 1] Division of Hematology and Oncology, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA. [2] Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA. [3] Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
755
|
|
756
|
Emerging EPO and EPO receptor regulators and signal transducers. Blood 2015; 125:3536-41. [PMID: 25887776 DOI: 10.1182/blood-2014-11-575357] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 04/13/2015] [Indexed: 12/13/2022] Open
Abstract
As essential mediators of red cell production, erythropoietin (EPO) and its cell surface receptor (EPO receptor [EPOR]) have been intensely studied. Early investigations defined basic mechanisms for hypoxia-inducible factor induction of EPO expression, and within erythroid progenitors EPOR engagement of canonical Janus kinase 2/signal transducer and activator of transcription 5 (JAK2/STAT5), rat sarcoma/mitogen-activated protein kinase/extracellular signal-regulated kinase (RAS/MEK/ERK), and phosphatidylinositol 3-kinase (PI3K) pathways. Contemporary genetic, bioinformatic, and proteomic approaches continue to uncover new clinically relevant modulators of EPO and EPOR expression, and EPO's biological effects. This Spotlight review highlights such factors and their emerging roles during erythropoiesis and anemia.
Collapse
|
757
|
Shovlin CL. Circulatory contributors to the phenotype in hereditary hemorrhagic telangiectasia. Front Genet 2015; 6:101. [PMID: 25914716 PMCID: PMC4391027 DOI: 10.3389/fgene.2015.00101] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/25/2015] [Indexed: 12/11/2022] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is mechanistically and therapeutically challenging, not only because of the molecular and cellular perturbations that generate vascular abnormalities, but also the modifications to circulatory physiology that result, and are likely to exacerbate vascular injury. First, most HHT patients have visceral arteriovenous malformations (AVMs). Significant visceral AVMs reduce the systemic vascular resistance: supra-normal cardiac outputs are required to maintain arterial blood pressure, and may result in significant pulmonary venous hypertension. Secondly, bleeding from nasal and gastrointestinal telangiectasia leads to iron losses of such magnitude that in most cases, diet is insufficient to meet the ‘hemorrhage adjusted iron requirement.’ Resultant iron deficiency restricts erythropoiesis, leading to anemia and further increases in cardiac output. Low iron levels are also associated with venous and arterial thromboses, elevated Factor VIII, and increased platelet aggregation to circulating 5HT (serotonin). Third, recent data highlight that reduced oxygenation of blood due to pulmonary AVMs results in a graded erythrocytotic response to maintain arterial oxygen content, and higher stroke volumes and/or heart rates to maintain oxygen delivery. Finally, HHT-independent factors such as diet, pregnancy, sepsis, and other intercurrent illnesses also influence vascular structures, hemorrhage, and iron handling in HHT patients. These considerations emphasize the complexity of mechanisms that impact on vascular structures in HHT, and also offer opportunities for targeted therapeutic approaches.
Collapse
Affiliation(s)
- Claire L Shovlin
- NHLI Cardiovascular Sciences, Imperial Centre for Translational and Experimental Medicine, Imperial College London London, UK ; Respiratory Medicine, Hammersmith Hospital, Imperial College Healthcare NHS Trust London, UK
| |
Collapse
|
758
|
Silva B, Faustino P. An overview of molecular basis of iron metabolism regulation and the associated pathologies. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1347-59. [PMID: 25843914 DOI: 10.1016/j.bbadis.2015.03.011] [Citation(s) in RCA: 189] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 03/05/2015] [Accepted: 03/27/2015] [Indexed: 12/18/2022]
Abstract
Iron is essential for several vital biological processes. Its deficiency or overload drives to the development of several pathologies. To maintain iron homeostasis, the organism controls the dietary iron absorption by enterocytes, its recycling by macrophages and storage in hepatocytes. These processes are mainly controlled by hepcidin, a liver-derived hormone which synthesis is regulated by iron levels, inflammation, infection, anemia and erythropoiesis. Besides the systemic regulation of iron metabolism mediated by hepcidin, cellular regulatory processes also occur. Cells are able to regulate themselves the expression of the iron metabolism-related genes through different post-transcriptional mechanisms, such as the alternative splicing, microRNAs, the IRP/IRE system and the proteolytic cleavage. Whenever those mechanisms are disturbed, due to genetic or environmental factors, iron homeostasis is disrupted and iron related pathologies may arise.
Collapse
Affiliation(s)
- Bruno Silva
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisboa, Portugal
| | - Paula Faustino
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisboa, Portugal.
| |
Collapse
|
759
|
Affiliation(s)
- Tomas Ganz
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
760
|
Abstract
Iron, an essential nutrient, is required for many diverse biological processes. The absence of a defined pathway to excrete excess iron makes it essential for the body to regulate the amount of iron absorbed; a deficiency could lead to iron deficiency and an excess to iron overload and associated disorders such as anaemia and haemochromatosis respectively. This regulation is mediated by the iron-regulatory hormone hepcidin. Hepcidin binds to the only known iron export protein, ferroportin (FPN), inducing its internalization and degradation, thus limiting the amount of iron released into the blood. The major factors that are implicated in hepcidin regulation include iron stores, hypoxia, inflammation and erythropoiesis. The present review summarizes our present knowledge about the molecular mechanisms and signalling pathways contributing to hepcidin regulation by these factors.
Collapse
|
761
|
Aboul-Enein A, El-Beshlawy A, Hamdy M, Shaheen I, El-Saadany Z, Samir A, El-Samie HA. Peripheral expression of hepcidin gene in Egyptian β-thalassemia major. Gene 2015; 564:206-9. [PMID: 25816754 DOI: 10.1016/j.gene.2015.03.048] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 03/23/2015] [Accepted: 03/24/2015] [Indexed: 01/06/2023]
Abstract
Iron overload is the major cause of morbidity and mortality in transfusion dependent β-thalassemia major patients. There is a sophisticated balance of body iron metabolism of storage and transport which is regulated by several factors including the peptide hepcidin. Hepcidin is the main iron regulatory molecule; it is secreted mainly by the liver and other tissues including monocytes and lymphocytes. Expression of hepcidin in such cells is unclear and has been studied in few reports with controverted result. Peripheral expression of hepcidin was measured using quantitative real time PCR (qRT-PCR) in 50 β-thalassemia major patients, in addition to 20 healthy volunteers as a control group. Hepcidin levels in β-thalassemia major patients showed statistically significant decrease in comparison to the control group, and was correlated to cardiac iron stores (T2*). However, hepcidin level was not different among the patients according to the HCV status or whether splenectomized or not. In conclusion; peripheral expression of hepcidin, in iron overloaded β-thalassemia major patients, is a reflection of hepatic expression. It can be used as a molecular predictor for the severity of cardiac iron overload and can be used as a future target for therapy in β-thalassemia major patients.
Collapse
Affiliation(s)
| | | | - Mona Hamdy
- Pediatric Department, Cairo University, Cairo, Egypt
| | - Iman Shaheen
- Clinical Pathology Department, Cairo University, Cairo, Egypt
| | | | - Ahmed Samir
- Radiology Department, Ain Shams University, Cairo, Egypt
| | | |
Collapse
|
762
|
Macrophages and iron trafficking at the birth and death of red cells. Blood 2015; 125:2893-7. [PMID: 25778532 DOI: 10.1182/blood-2014-12-567776] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/05/2015] [Indexed: 01/25/2023] Open
Abstract
Macrophages play a critical role in iron homeostasis via their intimate association with developing and dying red cells. Central nurse macrophages promote erythropoiesis in the erythroblastic island niche. These macrophages make physical contact with erythroblasts, enabling signaling and the transfer of growth factors and possibly nutrients to the cells in their care. Human mature red cells have a lifespan of 120 days before they become senescent and again come into contact with macrophages. Phagocytosis of red blood cells is the main source of iron flux in the body, because heme must be recycled from approximately 270 billion hemoglobin molecules in each red cell, and roughly 2 million senescent red cells are recycled each second. Here we will review pathways for iron trafficking found at the macrophage-erythroid axis, with a focus on possible roles for the transport of heme in toto.
Collapse
|
763
|
Evidence that the expression of transferrin receptor 1 on erythroid marrow cells mediates hepcidin suppression in the liver. Exp Hematol 2015; 43:469-78.e6. [PMID: 25782630 DOI: 10.1016/j.exphem.2015.03.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 03/05/2015] [Indexed: 01/19/2023]
Abstract
Hepcidin is the key regulator of iron absorption and recycling, and its expression is suppressed by red blood cell production. When erythropoiesis is expanded, hepcidin expression decreases. To gain insight into the stage of erythroid differentiation at which the regulation might originate, we measured serum hepcidin levels in archived pure red cell aplasia samples from patients whose block in erythroid differentiation was well defined by hematopoietic colony assays and marrow morphologic review. Hepcidin values are high or high normal in pure red cell aplasia patients in whom erythropoiesis is inhibited prior to the proerythroblast stage, but are suppressed in patients with excess proerythroblasts and few later erythroid cells. These data suggest that the suppressive effect of erythropoietic activity on hepcidin expression can arise from proerythroblasts, the stage at which transferrin receptor 1 expression peaks, prompting the hypothesis that transferrin receptor 1 expression on erythroid precursors is a proximal mediator of the erythroid regulator of hepcidin expression. Our characterization of erythropoiesis, iron status, and hepcidin expression in mice with global or hematopoietic cell-specific haploinsufficiency of transferrin receptor 1 provides initial supporting data for this model. The regulation appears independent of erythroferrone and growth differentiation factor 15, supporting the concept that several mechanisms signal iron need in response to an expanded erythron.
Collapse
|
764
|
Abstract
Iron overload is becoming an increasing problem as haemoglobinopathy patients gain greater access to good medical care and as therapies for myelodysplastic syndromes improve. Therapeutic options for iron chelation therapy have increased and many patients now receive combination therapies. However, optimal utilization of iron chelation therapy requires knowledge not only of the total body iron burden but the relative iron distribution among the different organs. The physiological basis for extrahepatic iron deposition is presented in order to help identify patients at highest risk for cardiac and endocrine complications. This manuscript reviews the current state of the art for monitoring global iron overload status as well as its compartmentalization. Plasma markers, computerized tomography, liver biopsy, magnetic susceptibility devices and magnetic resonance imaging (MRI) techniques are all discussed but MRI has come to dominate clinical practice. The potential impact of recent pancreatic and pituitary MRI studies on clinical practice are discussed as well as other works-in-progress. Clinical protocols are derived from experience in haemoglobinopathies but may provide useful guiding principles for other iron overload disorders, such as myelodysplastic syndromes.
Collapse
Affiliation(s)
- John C Wood
- Division of Cardiology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
765
|
An increase in hemoglobin, platelets and white blood cells levels by iron chelation as single treatment in multitransfused patients with myelodysplastic syndromes: clinical evidences and possible biological mechanisms. Ann Hematol 2015; 94:771-7. [DOI: 10.1007/s00277-015-2341-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 02/16/2015] [Indexed: 12/16/2022]
|
766
|
Sankaran VG, Ulirsch JC, Tchaikovskii V, Ludwig LS, Wakabayashi A, Kadirvel S, Lindsley RC, Bejar R, Shi J, Lovitch SB, Bishop DF, Steensma DP. X-linked macrocytic dyserythropoietic anemia in females with an ALAS2 mutation. J Clin Invest 2015; 125:1665-9. [PMID: 25705881 DOI: 10.1172/jci78619] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 01/08/2015] [Indexed: 01/19/2023] Open
Abstract
Macrocytic anemia with abnormal erythropoiesis is a common feature of megaloblastic anemias, congenital dyserythropoietic anemias, and myelodysplastic syndromes. Here, we characterized a family with multiple female individuals who have macrocytic anemia. The proband was noted to have dyserythropoiesis and iron overload. After an extensive diagnostic evaluation that did not provide insight into the cause of the disease, whole-exome sequencing of multiple family members revealed the presence of a mutation in the X chromosomal gene ALAS2, which encodes 5'-aminolevulinate synthase 2, in the affected females. We determined that this mutation (Y365C) impairs binding of the essential cofactor pyridoxal 5'-phosphate to ALAS2, resulting in destabilization of the enzyme and consequent loss of function. X inactivation was not highly skewed in wbc from the affected individuals. In contrast, and consistent with the severity of the ALAS2 mutation, there was a complete skewing toward expression of the WT allele in mRNA from reticulocytes that could be recapitulated in primary erythroid cultures. Together, the results of the X inactivation and mRNA studies illustrate how this X-linked dominant mutation in ALAS2 can perturb normal erythropoiesis through cell-nonautonomous effects. Moreover, our findings highlight the value of whole-exome sequencing in diagnostically challenging cases for the identification of disease etiology and extension of the known phenotypic spectrum of disease.
Collapse
MESH Headings
- 5-Aminolevulinate Synthetase/genetics
- 5-Aminolevulinate Synthetase/metabolism
- Adult
- Anemia, Dyserythropoietic, Congenital/genetics
- Anemia, Macrocytic/genetics
- Cells, Cultured
- Erythropoiesis/genetics
- Exome/genetics
- Female
- Genes, Dominant
- Genes, X-Linked
- Genetic Diseases, X-Linked/blood
- Genetic Diseases, X-Linked/genetics
- Hemorrhage/etiology
- Humans
- Iron Overload/etiology
- Male
- Models, Molecular
- Molecular Sequence Data
- Mutation, Missense
- Point Mutation
- Pregnancy
- Pregnancy Complications, Hematologic/genetics
- Protein Binding
- Protein Conformation
- Puerperal Disorders/etiology
- Pyridoxal Phosphate/metabolism
- RNA, Messenger/genetics
- Reticulocytes/metabolism
- X Chromosome Inactivation
Collapse
|
767
|
Yun S, Vincelette ND. Update on iron metabolism and molecular perspective of common genetic and acquired disorder, hemochromatosis. Crit Rev Oncol Hematol 2015; 95:12-25. [PMID: 25737209 DOI: 10.1016/j.critrevonc.2015.02.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 01/29/2015] [Accepted: 02/11/2015] [Indexed: 12/14/2022] Open
Abstract
Iron is an essential component of erythropoiesis and its metabolism is tightly regulated by a variety of internal and external cues including iron storage, tissue hypoxia, inflammation and degree of erythropoiesis. There has been remarkable improvement in our understanding of the molecular mechanisms of iron metabolism past decades. The classical model of iron metabolism with iron response element/iron response protein (IRE/IRP) is now extended to include hepcidin model. Endogenous and exogenous signals funnel down to hepcidin via wide range of signaling pathways including Janus Kinase/Signal Transducer and Activator of Transcription 3 (JAK/STAT3), Bone Morphogenetic Protein/Hemojuvelin/Mothers Against Decapentaplegic Homolog (BMP/HJV/SMAD), and Von Hippel Lindau/Hypoxia-inducible factor/Erythropoietin (VHL/HIF/EPO), then relay to ferroportin, which directly regulates intra- and extracellular iron levels. The successful molecular delineation of iron metabolism further enhanced our understanding of common genetic and acquired disorder, hemochromatosis. The majority of the hereditary hemochromatosis (HH) patients are now shown to have mutations in the genes coding either upstream or downstream proteins of hepcidin, resulting in iron overload. The update on hepcidin centered mechanisms of iron metabolism and their clinical perspective in hemochromatosis will be discussed in this review.
Collapse
Affiliation(s)
- Seongseok Yun
- Department of Medicine, University of Arizona, Tucson, AZ 85721, USA.
| | - Nicole D Vincelette
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55902, USA
| |
Collapse
|
768
|
Bu JT, Bartnikas TB. The use of hypotransferrinemic mice in studies of iron biology. Biometals 2015; 28:473-80. [PMID: 25663418 DOI: 10.1007/s10534-015-9833-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/04/2015] [Indexed: 12/15/2022]
Abstract
The hypotransferrinemic (hpx) mouse is a model of inherited transferrin deficiency that originated several decades ago in the BALB/cJ mouse strain. Also known as the hpx mouse, this line is almost completely devoid of transferrin, an abundant serum iron-binding protein. Two of the most prominent phenotypes of the hpx mouse are severe anemia and tissue iron overload. These phenotypes reflect the essential role of transferrin in iron delivery to bone marrow and regulation of iron homeostasis. Over the years, the hpx mouse has been utilized in studies on the role of transferrin, iron and other metals in a variety of organ systems and biological processes. This review summarizes the lessons learned from these studies and suggests possible areas of future exploration using this versatile yet complex mouse model.
Collapse
Affiliation(s)
- Julia T Bu
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship Street, Providence, RI, 02912, USA
| | | |
Collapse
|
769
|
Puliyel M, Mainous AG, Berdoukas V, Coates TD. Iron toxicity and its possible association with treatment of Cancer: lessons from hemoglobinopathies and rare, transfusion-dependent anemias. Free Radic Biol Med 2015; 79:343-51. [PMID: 25463277 DOI: 10.1016/j.freeradbiomed.2014.10.861] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/22/2014] [Accepted: 10/30/2014] [Indexed: 01/19/2023]
Abstract
Exposure to elevated levels of iron causes tissue damage and organ failure, and increases the risk of cancer. The toxicity of iron is mediated through generation of oxidants. There is also solid evidence indicating that oxidant stress plays a significant role in a variety of human disease states, including malignant transformation. Iron toxicity is the main focus when managing thalassemia. However, the short- and long-term toxicities of iron have not been extensively considered in children and adults treated for malignancy, and only recently have begun to draw oncologists' attention. The treatment of malignancy can markedly increase exposure of patients to elevated toxic iron species without the need for excess iron input from transfusion. This under-recognized exposure likely enhances organ toxicity and may contribute to long-term development of secondary malignancy and organ failure. This review discusses the current understanding of iron metabolism, the mechanisms of production of toxic free iron species in humans, and the relation of the clinical marker, transferrin saturation (TS), to the presence of toxic free iron. We will present epidemiological data showing that high TS is associated with poor outcomes and development of cancer, and that lowering free iron may improve outcomes. Finally, we will discuss the possible relation between some late complications seen in survivors of cancer and those due to iron toxicity.
Collapse
Affiliation(s)
- Mammen Puliyel
- Section of Hematology, Childrens Center for Cancer, Blood Disease and Bone Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles California, USA
| | - Arch G Mainous
- Department of Health Services Research, Management and Policy, University of Florida, Gainesville, Fla. USA
| | - Vasilios Berdoukas
- Section of Hematology, Childrens Center for Cancer, Blood Disease and Bone Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles California, USA
| | - Thomas D Coates
- Section of Hematology, Childrens Center for Cancer, Blood Disease and Bone Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles California, USA.
| |
Collapse
|
770
|
Kawabata H, Uchiyama T, Sakamoto S, Kanda J, Oishi S, Fujii N, Tomosugi N, Kadowaki N, Takaori-Kondo A. A HAMP promoter bioassay system for identifying chemical compounds that modulate hepcidin expression. Exp Hematol 2015; 43:404-413.e5. [PMID: 25633564 DOI: 10.1016/j.exphem.2015.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 11/28/2014] [Accepted: 01/16/2015] [Indexed: 12/11/2022]
Abstract
Hepcidin is the central regulator of systemic iron homeostasis; dysregulation of hepcidin expression causes various iron metabolic disorders, including hereditary hemochromatosis and anemia of inflammation. To identify molecules that modulate hepcidin expression, we developed a bioassay system for hepcidin gene (HAMP) promoter activity by stable transfection of Hep3B hepatoma cells with an expression plasmid in which EGFP was linked to a 2.5-kb human HAMP promoter. Interleukin 6, bone morphogenetic protein 6 (BMP-6), and oncostatin M, well-characterized stimulators of the HAMP promoter, strongly enhanced the green fluorescence intensity of these cells. Dorsomorphin, heparin, and cobalt chloride, known inhibitors of hepcidin expression, significantly suppressed green fluorescence intensity, and these inhibitory effects were more prominent when the cells were stimulated with BMP-6. Employing this system, we screened 1,280 biologically active small molecules and found several candidate inhibitors of hepcidin expression. Apomorphine, benzamil, etoposide, CGS-15943, kenpaullone, and rutaecarpine (all at 10 μmol/L) significantly inhibited hepcidin mRNA expression by Hep3B cells without affecting cell viability. CGS-15943 was the strongest suppressor of BMP-6-induced hepcidin-25 secretion in these cells. We conclude that our newly developed hepcidin promoter bioassay system is useful for identifying and evaluating compounds that modulate hepcidin expression.
Collapse
Affiliation(s)
- Hiroshi Kawabata
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Tatsuki Uchiyama
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Soichiro Sakamoto
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junya Kanda
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Shinya Oishi
- Laboratory of Bioorganic Medical Chemistry and Chemogenomics, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Nobutaka Fujii
- Laboratory of Bioorganic Medical Chemistry and Chemogenomics, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Naohisa Tomosugi
- Division of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Japan
| | - Norimitsu Kadowaki
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
771
|
Gelderman MP, Baek JH, Yalamanoglu A, Puglia M, Vallelian F, Burla B, Vostal J, Schaer DJ, Buehler PW. Reversal of hemochromatosis by apotransferrin in non-transfused and transfused Hbbth3/+ (heterozygous B1/B2 globin gene deletion) mice. Haematologica 2015; 100:611-22. [PMID: 25616571 DOI: 10.3324/haematol.2014.117325] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 01/20/2015] [Indexed: 01/17/2023] Open
Abstract
Intermediate beta-thalassemia has a broad spectrum of sequelae and affected subjects may require occasional blood transfusions over their lifetime to correct anemia. Iron overload in intermediate beta-thalassemia results from a paradoxical intestinal absorption, iron release from macrophages and hepatocytes, and sporadic transfusions. Pathological iron accumulation in parenchyma is caused by chronic exposure to non-transferrin bound iron in plasma. The iron scavenger and transport protein transferrin is a potential treatment being studied for correction of anemia. However, transferrin may also function to prevent or reduce iron loading of tissues when exposure to non-transferrin bound iron increases. Here we evaluate the effects of apotransferrin administration on tissue iron loading and early tissue pathology in non-transfused and transfused Hbb(th3/+) mice. Mice with the Hbb(th3/+) phenotype have mild to moderate anemia and consistent tissue iron accumulation in the spleen, liver, kidneys and myocardium. Chronic apotransferrin administration resulted in normalization of the anemia. Furthermore, it normalized tissue iron content in the liver, kidney and heart and attenuated early tissue changes in non-transfused Hbb(th3/+) mice. Apotransferrin treatment was also found to attenuate transfusion-mediated increases in plasma non-transferrin bound iron and associated excess tissue iron loading. These therapeutic effects were associated with normalization of transferrin saturation and suppressed plasma non-transferrin bound iron. Apotransferrin treatment modulated a fundamental iron regulatory pathway, as evidenced by decreased erythroid Fam132b gene (erythroferrone) expression, increased liver hepcidin gene expression and plasma hepcidin-25 levels and consequently reduced intestinal ferroportin-1 in apotransferrin-treated thalassemic mice.
Collapse
Affiliation(s)
- Monique P Gelderman
- Laboratory of Biochemistry and Vascular Biology, Center of Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Jin Hyen Baek
- Laboratory of Cellular Hematology, Center of Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Ayla Yalamanoglu
- Laboratory of Cellular Hematology, Center of Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA Division of Internal Medicine, University of Zurich, Switzerland
| | - Michele Puglia
- Functional Genomics Center Zurich, Swiss Federal Institute of Technology Zurich / University of Zurich, Switzerland Division of Internal Medicine, University of Zurich, Switzerland
| | | | - Bo Burla
- Division of Internal Medicine, University of Zurich, Switzerland
| | - Jaroslav Vostal
- Laboratory of Biochemistry and Vascular Biology, Center of Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Dominik J Schaer
- Division of Internal Medicine, University of Zurich, Switzerland Center of Evolutionary Medicine, University of Zurich, Switzerland
| | - Paul W Buehler
- Laboratory of Cellular Hematology, Center of Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| |
Collapse
|
772
|
Vallelian F, Gelderman-Fuhrmann MP, Schaer CA, Puglia M, Opitz L, Baek JH, Vostal J, Buehler PW, Schaer DJ. Integrative proteome and transcriptome analysis of extramedullary erythropoiesis and its reversal by transferrin treatment in a mouse model of beta-thalassemia. J Proteome Res 2015; 14:1089-100. [PMID: 25566950 DOI: 10.1021/pr5010778] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Beta-thalassemia results from mutations of the β-hemoglobin (Hbb) gene and reduced functional Hbb synthesis. Excess α-Hb causes globin chain aggregation, oxidation, cytoskeletal damage, and increased red blood cell clearance. These events result in anemia, altered iron homeostasis, and expansion of extramedullary erythropoiesis. Serum transferrin (Tf) is suggested to be an important regulator of erythropoiesis in murine models of thalassemia. The present study was conducted to establish a quantitative proteomic and transcriptomic analysis of transferrin-modulated extramedullary erythropoiesis in the spleen of wild type and thalassemic Hbb(th3/+) mice. Our LC-MS/MS protein analysis and mRNA sequencing data provide quantitative expression estimates of 1590 proteins and 24,581 transcripts of the murine spleen and characterize key processes of erythropoiesis and RBC homeostasis such as the whole heme synthesis pathway as well as critical components of the red blood cell antioxidant systems and the proliferative cell cycling pathway. The data confirm that Tf treatment of nontransfused Hbb(th3/+) mice induces a systematic correction of these processes at a molecular level. Tf treatment of Hbb(th3/+) mice for 60 days leads to a complete molecular restoration of the normal murine spleen phenotype. These findings support further investigation of plasma-derived Tf as a treatment for thalassemia.
Collapse
Affiliation(s)
- Florence Vallelian
- Division of Internal Medicine, University of Zurich , CH-8091 Zurich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
773
|
Origa R, Cazzola M, Mereu E, Danjou F, Barella S, Giagu N, Galanello R, Swinkels DW. Differences in the erythropoiesis-hepcidin-iron store axis between hemoglobin H disease and β-thalassemia intermedia. Haematologica 2015; 100:e169-71. [PMID: 25596269 DOI: 10.3324/haematol.2014.115733] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Raffaella Origa
- Ospedale Microcitemico-Department of Public Health, Clinical and Molecular Medicine, University of Cagliari, Italy
| | - Mario Cazzola
- Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, and Department of Molecular Medicine, University of Pavia, Italy
| | - Elisabetta Mereu
- Ospedale Microcitemico-Department of Public Health, Clinical and Molecular Medicine, University of Cagliari, Italy
| | - Fabrice Danjou
- Ospedale Microcitemico-Department of Public Health, Clinical and Molecular Medicine, University of Cagliari, Italy
| | | | | | - Renzo Galanello
- Ospedale Microcitemico-Department of Public Health, Clinical and Molecular Medicine, University of Cagliari, Italy
| | - Dorine W Swinkels
- Department of Laboratory Medicine, Radboud University medical center, Nijmegen, The Netherlands
| |
Collapse
|
774
|
Abstract
Iron and oxygen metabolism are intimately linked with one another.
Collapse
Affiliation(s)
- Robert J. Simpson
- Diabetes and Nutritional Sciences
- School of Medicine
- Kings College London
- , UK
| | - Andrew T. McKie
- Diabetes and Nutritional Sciences
- School of Medicine
- Kings College London
- , UK
| |
Collapse
|
775
|
Camaschella C. Iron deficiency: new insights into diagnosis and treatment. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2015; 2015:8-13. [PMID: 26637694 DOI: 10.1182/asheducation-2015.1.8] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Iron deficiency and iron deficiency anemia are common conditions worldwide affecting especially children and young women. In developing countries, iron deficiency is caused by poor iron intake and/or parasitic infection, whereas vegetarian dietary choices, poor iron absorption, and chronic blood loss are common causes in high-income countries. Erythropoiesis stimulating agents can result in functional iron deficiency for erythropoiesis even when stores are iron-replete. Diagnosis of iron deficiency is straightforward, except when it occurs in the context of inflammatory disorders. Oral iron salts correct absolute iron deficiency in most patients, because low hepcidin levels facilitate iron absorption. Unfortunately frequent side effects limit oral iron efficacy. Intravenous iron is increasingly utilized, because currently available preparations allow rapid normalization of total body iron even with a single infusion and are effective also in functional iron deficiency and in iron deficiency associated with inflammatory disorders. The evidence is accumulating that these preparations are safe and effective. However, long-term safety issues of high doses of iron need to be further explored.
Collapse
Affiliation(s)
- Clara Camaschella
- Vita Salute University, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
776
|
Langdon JM, Barkataki S, Berger AE, Cheadle C, Xue QL, Sung V, Roy CN. RAP-011, an activin receptor ligand trap, increases hemoglobin concentration in hepcidin transgenic mice. Am J Hematol 2015; 90:8-14. [PMID: 25236856 DOI: 10.1002/ajh.23856] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 09/10/2014] [Accepted: 09/16/2014] [Indexed: 12/21/2022]
Abstract
Over expression of hepcidin antimicrobial peptide is a common feature of iron-restricted anemia in humans. We investigated the erythroid response to either erythropoietin or RAP-011, a "murinized" ortholog of sotatercept, in C57BL/6 mice and in hepcidin antimicrobial peptide 1 over expressing mice. Sotatercept, a soluble, activin receptor type IIA ligand trap, is currently being evaluated for the treatment of anemias associated with chronic renal disease, myelodysplastic syndrome, β-thalassemia, and Diamond Blackfan anemia and acts by inhibiting signaling downstream of activin and other Transforming Growth Factor-β superfamily members. We found that erythropoietin and RAP-011 increased hemoglobin concentration in C57BL/6 mice and in hepcidin antimicrobial peptide 1 over expressing mice. While erythropoietin treatment depleted splenic iron stores in C57BL/6 mice, RAP-011 treatment did not deplete splenic iron stores in mice of either genotype. Bone marrow erythroid progenitors from erythropoietin-treated mice exhibited iron-restricted erythropoiesis, as indicated by increased median fluorescence intensity of transferrin receptor immunostaining by flow cytometry. In contrast, RAP-011-treated mice did not exhibit the same degree of iron-restricted erythropoiesis. In conclusion, we have demonstrated that RAP-011 can improve hemoglobin concentration in hepcidin antimicrobial peptide 1 transgenic mice. Our data support the hypothesis that RAP-011 has unique biologic effects which prevent or circumvent depletion of mouse splenic iron stores. RAP-011 may, therefore, be an appropriate therapeutic for trials in human anemias characterized by increased expression of hepcidin antimicrobial peptide and iron-restricted erythropoiesis.
Collapse
Affiliation(s)
- Jacqueline M. Langdon
- Division of Geriatric Medicine and Gerontology; Johns Hopkins University School of Medicine; Baltimore Maryland
| | - Sangjucta Barkataki
- Lowe Family Genomics Core; Johns Hopkins University School of Medicine; Baltimore Maryland
| | - Alan E. Berger
- Lowe Family Genomics Core; Johns Hopkins University School of Medicine; Baltimore Maryland
| | - Chris Cheadle
- Lowe Family Genomics Core; Johns Hopkins University School of Medicine; Baltimore Maryland
| | - Qian-Li Xue
- Division of Geriatric Medicine and Gerontology; Johns Hopkins University School of Medicine; Baltimore Maryland
| | - Victoria Sung
- Translational Development; Celgene Corporation; San Francisco CA
| | - Cindy N. Roy
- Division of Geriatric Medicine and Gerontology; Johns Hopkins University School of Medicine; Baltimore Maryland
- Division of Hematology; Johns Hopkins School of Medicine; Baltimore Maryland
| |
Collapse
|
777
|
Gammella E, Diaz V, Recalcati S, Buratti P, Samaja M, Dey S, Noguchi CT, Gassmann M, Cairo G. Erythropoietin's inhibiting impact on hepcidin expression occurs indirectly. Am J Physiol Regul Integr Comp Physiol 2014; 308:R330-5. [PMID: 25519735 DOI: 10.1152/ajpregu.00410.2014] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Under conditions of accelerated erythropoiesis, elevated erythropoietin (Epo) levels are associated with inhibition of hepcidin synthesis, a response that ultimately increases iron availability to meet the enhanced iron needs of erythropoietic cells. In the search for erythroid regulators of hepcidin, many candidates have been proposed, including Epo itself. We aimed to test whether direct interaction between Epo and the liver is required to regulate hepcidin. We found that prolonged administration of high doses of Epo in mice leads to great inhibition of liver hepcidin mRNA levels, and concomitant induction of the hepcidin inhibitor erythroferrone (ERFE). Epo treatment also resulted in liver iron mobilization, mediated by increased ferroportin activity and accompanied by reduced ferritin levels and increased TfR1 expression. The same inhibitory effect was observed in mice that do not express the homodimeric Epo receptor (EpoR) in liver cells because EpoR expression is restricted to erythroid cells. Similarly, liver signaling pathways involved in hepcidin regulation were not influenced by the presence or absence of hepatic EpoR. Moreover, Epo analogs, possibly interacting with the postulated heterodimeric β common EpoR, did not affect hepcidin expression. These findings were supported by the lack of inhibition on hepcidin found in hepatoma cells exposed to various concentrations of Epo for different periods of times. Our results demonstrate that hepcidin suppression does not require the direct binding of Epo to its liver receptors and rather suggest that the role of Epo is to stimulate the synthesis of the erythroid regulator ERFE in erythroblasts, which ultimately downregulates hepcidin.
Collapse
Affiliation(s)
- Elena Gammella
- Department of Biomedical Sciences for Health, University of Milano, Milan, Italy
| | - Victor Diaz
- Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Stefania Recalcati
- Department of Biomedical Sciences for Health, University of Milano, Milan, Italy
| | - Paolo Buratti
- Department of Biomedical Sciences for Health, University of Milano, Milan, Italy
| | - Michele Samaja
- Department of Health Science, University of Milano, Milan, Italy
| | - Soumyadeep Dey
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland; and
| | - Constance Tom Noguchi
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland; and
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland; Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Gaetano Cairo
- Department of Biomedical Sciences for Health, University of Milano, Milan, Italy;
| |
Collapse
|
778
|
Hepcidin is suppressed by erythropoiesis in hemoglobin E β-thalassemia and β-thalassemia trait. Blood 2014; 125:873-80. [PMID: 25519750 DOI: 10.1182/blood-2014-10-606491] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Hemoglobin E (HbE) β-thalassemia is the most common severe thalassemia syndrome across Asia, and millions of people are carriers. Clinical heterogeneity in HbE β-thalassemia is incompletely explained by genotype, and the interaction of phenotypic variation with hepcidin is unknown. The effect of thalassemia carriage on hepcidin is also unknown, but it could be relevant for iron supplementation programs aimed at combating anemia. In 62 of 69 Sri Lankan patients with HbE β-thalassemia with moderate or severe phenotype, hepcidin was suppressed, and overall hepcidin inversely correlated with iron accumulation. On segregating by phenotype, there were no differences in hepcidin, erythropoiesis, or hemoglobin between severe or moderate disease, but multiple linear regression showed that erythropoiesis inversely correlated with hepcidin only in severe phenotypes. In moderate disease, no independent predictors of hepcidin were identifiable; nevertheless, the low hepcidin levels indicate a significant risk for iron overload. In a population survey of Sri Lankan schoolchildren, β-thalassemia (but not HbE) trait was associated with increased erythropoiesis and mildly suppressed hepcidin, suggesting an enhanced propensity to accumulate iron. In summary, the influence of erythropoiesis on hepcidin suppression associates with phenotypic disease variation and pathogenesis in HbE β-thalassemia and indicates that the epidemiology of β-thalassemia trait requires consideration when planning public health iron interventions.
Collapse
|
779
|
Abstract
Transferrin receptor 2 (TFR2) contributes to hepcidin regulation in the liver and associates with erythropoietin receptor in erythroid cells. Nevertheless, TFR2 mutations cause iron overload (hemochromatosis type 3) without overt erythroid abnormalities. To clarify TFR2 erythroid function, we generated a mouse lacking Tfr2 exclusively in the bone marrow (Tfr2(BMKO)). Tfr2(BMKO) mice have normal iron parameters, reduced hepcidin levels, higher hemoglobin and red blood cell counts, and lower mean corpuscular volume than normal control mice, a phenotype that becomes more evident in iron deficiency. In Tfr2(BMKO) mice, the proportion of nucleated erythroid cells in the bone marrow is higher and the apoptosis lower than in controls, irrespective of comparable erythropoietin levels. Induction of moderate iron deficiency increases erythroblasts number, reduces apoptosis, and enhances erythropoietin (Epo) levels in controls, but not in Tfr2(BMKO) mice. Epo-target genes such as Bcl-xL and Epor are highly expressed in the spleen and in isolated erythroblasts from Tfr2(BMKO) mice. Low hepcidin expression in Tfr2(BMKO) is accounted for by erythroid expansion and production of the erythroid regulator erythroferrone. We suggest that Tfr2 is a component of a novel iron-sensing mechanism that adjusts erythrocyte production according to iron availability, likely by modulating the erythroblast Epo sensitivity.
Collapse
|
780
|
Heeney MM. Iron clad: iron homeostasis and the diagnosis of hereditary iron overload. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2014; 2014:202-209. [PMID: 25696856 DOI: 10.1182/asheducation-2014.1.202] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Iron is an ubiquitous metal of vital importance to the normal physiologic processes of many organisms. Over the last 2 decades, the discovery of mutations in genes leading to hereditary disorders of iron overload, iron deficiency, and iron maldistribution have accelerated our understanding of human iron homeostasis. This chapter provides an updated overview of the human iron cycle, regulation of iron homeostasis, and how perturbations in these homeostatic mechanisms lead to iron overload disease and provides strategies for the diagnosis of hereditary iron overload.
Collapse
Affiliation(s)
- Matthew M Heeney
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
| |
Collapse
|
781
|
Casu C, Rivella S. Iron age: novel targets for iron overload. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2014; 2014:216-221. [PMID: 25696858 PMCID: PMC5292264 DOI: 10.1182/asheducation-2014.1.216] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Excess iron deposition in vital organs is the main cause of morbidity and mortality in patients affected by β-thalassemia and hereditary hemochromatosis. In both disorders, inappropriately low levels of the liver hormone hepcidin are responsible for the increased iron absorption, leading to toxic iron accumulation in many organs. Several studies have shown that targeting iron absorption could be beneficial in reducing or preventing iron overload in these 2 disorders, with promising preclinical data. New approaches target Tmprss6, the main suppressor of hepcidin expression, or use minihepcidins, small peptide hepcidin agonists. Additional strategies in β-thalassemia are showing beneficial effects in ameliorating ineffective erythropoiesis and anemia. Due to the suppressive nature of the erythropoiesis on hepcidin expression, these approaches are also showing beneficial effects on iron metabolism. The goal of this review is to discuss the major factors controlling iron metabolism and erythropoiesis and to discuss potential novel therapeutic approaches to reduce or prevent iron overload in these 2 disorders and ameliorate anemia in β-thalassemia.
Collapse
Affiliation(s)
- Carla Casu
- Department of Pediatrics, Division of Hematology-Oncology, Weill Medical College, Cornell University, New York, NY
| | - Stefano Rivella
- Department of Pediatrics, Division of Hematology-Oncology, Weill Medical College, Cornell University, New York, NY
- Department of Cell and Developmental Biology, Weill Medical College, Cornell University, New York, NY
| |
Collapse
|
782
|
Archer NM, Shmukler BE, Andolfo I, Vandorpe DH, Gnanasambandam R, Higgins JM, Rivera A, Fleming MD, Sachs F, Gottlieb PA, Iolascon A, Brugnara C, Alper SL, Nathan DG. Hereditary xerocytosis revisited. Am J Hematol 2014; 89:1142-6. [PMID: 25044010 DOI: 10.1002/ajh.23799] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 06/30/2014] [Indexed: 01/16/2023]
Affiliation(s)
- Natasha M. Archer
- Division of Hematology and Oncology; Boston Children's Hospital; Boston Massachusetts
- Dana-Farber Cancer Center; Boston Massachusetts
| | - Boris E. Shmukler
- Renal Division; Beth Israel Deaconess Medical Center; Boston Massachusetts
- Molecular and Vascular Medicine Division; Beth Israel Deaconess Medical Center; Boston Massachusetts
| | - Immacolata Andolfo
- Department of Molecular Medicine and Medical Biotechnologies; Federico II University of Naples; Naples Italy
- CEINGE, Advanced Biotechnologies; Naples Italy
| | - David H. Vandorpe
- Renal Division; Beth Israel Deaconess Medical Center; Boston Massachusetts
- Molecular and Vascular Medicine Division; Beth Israel Deaconess Medical Center; Boston Massachusetts
| | | | - John M. Higgins
- Department of Systems Biology; Harvard Medical School; Boston Massachusetts
- Center for Systems Biology and Department of Pathology; Massachusetts General Hospital; Boston Massachusetts
| | - Alicia Rivera
- Department of Laboratory Medicine; Boston Children's Hospital; Boston Massachusetts
- Department of Pathology; Harvard Medical School; Boston Massachusetts
| | - Mark D. Fleming
- Department of Pathology; Harvard Medical School; Boston Massachusetts
| | - Frederick Sachs
- Department of Physiology and Biophysics; University of Buffalo; Buffalo New York
| | - Philip A. Gottlieb
- Department of Physiology and Biophysics; University of Buffalo; Buffalo New York
| | - Achille Iolascon
- Department of Molecular Medicine and Medical Biotechnologies; Federico II University of Naples; Naples Italy
- CEINGE, Advanced Biotechnologies; Naples Italy
| | - Carlo Brugnara
- Department of Laboratory Medicine; Boston Children's Hospital; Boston Massachusetts
- Department of Pathology; Harvard Medical School; Boston Massachusetts
| | - Seth L. Alper
- Renal Division; Beth Israel Deaconess Medical Center; Boston Massachusetts
- Molecular and Vascular Medicine Division; Beth Israel Deaconess Medical Center; Boston Massachusetts
- Department of Medicine; Harvard Medical School; Boston Massachusetts
| | - David G. Nathan
- Division of Hematology and Oncology; Boston Children's Hospital; Boston Massachusetts
- Dana-Farber Cancer Center; Boston Massachusetts
- Department of Pediatrics; Harvard Medical School; Boston Massachusetts
| |
Collapse
|
783
|
Shovlin CL. Pulmonary arteriovenous malformations. Am J Respir Crit Care Med 2014; 190:1217-28. [PMID: 25420112 PMCID: PMC4315816 DOI: 10.1164/rccm.201407-1254ci] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 10/20/2014] [Indexed: 11/16/2022] Open
Abstract
Within the past decade, pulmonary arteriovenous malformations (PAVMs) have evolved from rare curiosities to not uncommon clinical states, with the latest estimates suggesting a prevalence of ~1 in 2,600. PAVMs provide anatomic right-to-left shunts, allowing systemic venous blood to bypass gas exchange and pulmonary capillary bed processing. Hypoxemia and enhanced ventilatory demands result, although both are usually asymptomatic. Paradoxical emboli lead to strokes and cerebral abscesses, and these commonly occur in individuals with previously undiagnosed PAVMs. PAVM hemorrhage is rare but is the main cause of maternal death in pregnancy. PAVM occlusion by embolization is the standard of care to reduce these risks. However, recent data demonstrate that currently recommended management protocols can result in levels of radiation exposure that would be classified as harmful. Recent publications also provide a better appreciation of the hematologic and cardiovascular demands required to maintain arterial oxygen content and oxygen consumption in hypoxemic patients, identify patient subgroups at higher risk of complications, and emphasize the proportion of radiologically visible PAVMs too small to treat by embolization. This review, therefore, outlines medical states that exacerbate the consequences of PAVMs. Chief among these is iron deficiency, which is commonly present due to concurrent hereditary hemorrhagic telangiectasia: iron deficiency impairs hypoxemia compensations by restricting erythropoiesis and increases the risk of ischemic strokes. Management of periodontal disease, dental interventions, pulmonary hypertension, and pregnancy also requires specific consideration in the setting of PAVMs. The review concludes by discussing to what extent previously recommended protocols may benefit from modification or revision.
Collapse
Affiliation(s)
- Claire L. Shovlin
- NHLI Cardiovascular Sciences, Imperial College, London; and
- Respiratory Medicine, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
784
|
Wallace DF, Secondes ES, Rishi G, Ostini L, McDonald CJ, Lane SW, Vu T, Hooper JD, Velasco G, Ramsay AJ, Lopez-Otin C, Subramaniam VN. A critical role for murine transferrin receptor 2 in erythropoiesis during iron restriction. Br J Haematol 2014; 168:891-901. [PMID: 25403101 DOI: 10.1111/bjh.13225] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/22/2014] [Indexed: 12/29/2022]
Abstract
Effective erythropoiesis requires an appropriate supply of iron and mechanisms regulating iron homeostasis and erythropoiesis are intrinsically linked. Iron dysregulation, typified by iron-deficiency anaemia and iron overload, is common in many clinical conditions and impacts the health of up to 30% of the world's population. The proteins transmembrane protease, serine 6 (TMPRSS6; also termed matriptase-2), HFE and transferrin receptor 2 (TFR2) play important and opposing roles in systemic iron homeostasis, by regulating expression of the iron regulatory hormone hepcidin. We have performed a systematic analysis of mice deficient in these three proteins and show that TMPRSS6 predominates over HFE and TFR2 in hepcidin regulation. The phenotype of mice lacking TMPRSS6 and TFR2 is characterized by severe anaemia and extramedullary haematopoiesis in the spleen. Stress erythropoiesis in these mice results in increased expression of the newly identified erythroid iron regulator erythroferrone, which does not appear to overcome the hepcidin overproduction mediated by loss of TMPRSS6. Extended analysis reveals that TFR2 plays an important role in erythroid cells, where it is involved in terminal erythroblast differentiation and the regulation of erythropoietin. In conclusion, we have identified an essential role for TFR2 in erythropoiesis that may provide new targets for the treatment of anaemia.
Collapse
Affiliation(s)
- Daniel F Wallace
- Membrane Transport Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Qld, Australia; School of Medicine, The University of Queensland, Brisbane, Qld, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
785
|
|
786
|
Prise en charge actuelle des thalassémies intermédiaires. Transfus Clin Biol 2014; 21:143-9. [DOI: 10.1016/j.tracli.2014.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 07/22/2014] [Indexed: 12/19/2022]
|
787
|
Affiliation(s)
- Léon Kautz
- Department of Medicine ; David Geffen School of Medicine, University of California, Los Angeles, UCLA, department of Medicine, 10833 LeConte avenue, CHS37-131, CA90095 Los Angeles, CA, États-Unis
| |
Collapse
|
788
|
Porter JB, Walter PB, Neumayr LD, Evans P, Bansal S, Garbowski M, Weyhmiller MG, Harmatz PR, Wood JC, Miller JL, Byrnes C, Weiss G, Seifert M, Grosse R, Grabowski D, Schmidt A, Fischer R, Nielsen P, Niemeyer C, Vichinsky E. Mechanisms of plasma non-transferrin bound iron generation: insights from comparing transfused diamond blackfan anaemia with sickle cell and thalassaemia patients. Br J Haematol 2014; 167:692-6. [PMID: 25209728 DOI: 10.1111/bjh.13081] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 04/23/2014] [Indexed: 01/17/2023]
Abstract
In transfusional iron overload, extra-hepatic iron distribution differs, depending on the underlying condition. Relative mechanisms of plasma non-transferrin bound iron (NTBI) generation may account for these differences. Markers of iron metabolism (plasma NTBI, labile iron, hepcidin, transferrin, monocyte SLC40A1 [ferroportin]), erythropoiesis (growth differentiation factor 15, soluble transferrin receptor) and tissue hypoxia (erythropoietin) were compared in patients with Thalassaemia Major (TM), Sickle Cell Disease and Diamond-Blackfan Anaemia (DBA), with matched transfusion histories. The most striking differences between these conditions were relationships of NTBI to erythropoietic markers, leading us to propose three mechanisms of NTBI generation: iron overload (all), ineffective erythropoiesis (predominantly TM) and low transferrin-iron utilization (DBA).
Collapse
Affiliation(s)
- John B Porter
- Department of Haematology, University College London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
789
|
Abstract
Erythroferrone (ERFE) is an erythropoiesis-driven regulator of iron homeostasis. ERFE mediates the suppression of the iron-regulatory hormone hepcidin to increase iron absorption and mobilization of iron from stores. We examined the role of ERFE in the recovery from anemia of inflammation (AI) induced by injection of heat-killed Brucella abortus. B abortus-treated wild-type mice developed a moderate anemia and reached nadir hemoglobin 14 days after injection and partially recovered by 28 days. We observed that Erfe expression in the bone marrow and the spleen was greatly increased during anemia and peaked at 14 days after injection, a time course similar to serum erythropoietin. To determine whether ERFE facilitates the recovery from anemia, we analyzed Erfe-deficient mice injected with B abortus. Compared with wild-type mice, Erfe-deficient mice exhibited a more severe anemia, had higher hepcidin levels and consequently lower serum iron concentration on days 14 and 21, and manifested impaired mobilization of iron from stores (liver and spleen). Erfe(-/-) mice eventually compensated by further stimulating erythropoiesis and reticulocyte production. Thus, ERFE contributes to the recovery from AI by suppressing hepcidin and increasing iron availability.
Collapse
|
790
|
Ravasi G, Pelucchi S, Greni F, Mariani R, Giuliano A, Parati G, Silvestri L, Piperno A. Circulating factors are involved in hypoxia-induced hepcidin suppression. Blood Cells Mol Dis 2014; 53:204-10. [PMID: 25065484 DOI: 10.1016/j.bcmd.2014.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 06/30/2014] [Indexed: 12/21/2022]
Abstract
Hepcidin transcription is strongly down-regulated under hypoxic conditions, however whether hypoxia inhibits hepcidin directly or indirectly is still unknown. We investigated the time course of hypoxia-mediated hepcidin down-regulation in vivo in healthy volunteers exposed to hypobaric hypoxia at high altitude and, based on the hypothesis that circulating factors are implicated in hepcidin inhibition, we analyzed the effect of sera of these volunteers exposed to normoxia and hypoxia on hepcidin expression in Huh-7 cell lines. Hypoxia led to a significant hepcidin down-regulation in vivo that was almost complete within 72h of exposure and followed erythropoietin induction. This delay in hepcidin down-regulation suggests the existence of soluble factor/s regulating hepcidin production. We then stimulated HuH-7 cells with normoxic and hypoxic sera to analyze the effects of sera on hepcidin regulation. Hypoxic sera had a significant inhibitory effect on hepcidin promoter activity assessed by a luciferase assay, although the amount of such decrease was not as relevant as that observed in vivo. Cellular mRNA analysis showed that a number of volunteers' sera inhibited hepcidin expression, concurrently with ID1 inhibition, suggesting that inhibitory factor(s) may act through the SMAD-pathway.
Collapse
Affiliation(s)
- Giulia Ravasi
- Department of Health Science, University Milano-Bicocca, Monza, Italy
| | - Sara Pelucchi
- Department of Health Science, University Milano-Bicocca, Monza, Italy
| | - Federico Greni
- Department of Health Science, University Milano-Bicocca, Monza, Italy
| | | | - Andrea Giuliano
- Department of Health Science, University Milano-Bicocca, Monza, Italy; Department of Cardiology, Italian Institute for Auxology, Milan, Italy
| | - Gianfranco Parati
- Department of Health Science, University Milano-Bicocca, Monza, Italy; San Gerardo Hospital, Monza, Italy; Department of Cardiology, Italian Institute for Auxology, Milan, Italy
| | - Laura Silvestri
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute and Vita Salute University, Milan, Italy
| | - Alberto Piperno
- Department of Health Science, University Milano-Bicocca, Monza, Italy; San Gerardo Hospital, Monza, Italy; Consortium of Human Molecular Genetics, Monza, Italy.
| |
Collapse
|
791
|
Anemia: Giving iron a boost. Nat Med 2014. [DOI: 10.1038/nm.3635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
792
|
Impact of iron overload and potential benefit from iron chelation in low-risk myelodysplastic syndrome. Blood 2014; 124:873-81. [PMID: 24923296 DOI: 10.1182/blood-2014-03-563221] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Myelodysplastic syndromes (MDSs) are a group of heterogeneous clonal bone marrow disorders characterized by ineffective hematopoiesis, peripheral blood cytopenias, and potential for malignant transformation. Lower/intermediate-risk MDSs are associated with longer survival and high red blood cell (RBC) transfusion requirements resulting in secondary iron overload. Recent data suggest that markers of iron overload portend a relatively poor prognosis, and retrospective analysis demonstrates that iron chelation therapy is associated with prolonged survival in transfusion-dependent MDS patients. New data provide concrete evidence of iron's adverse effects on erythroid precursors in vitro and in vivo. Renewed interest in the iron field was heralded by the discovery of hepcidin, the main serum peptide hormone negative regulator of body iron. Evidence from β-thalassemia suggests that regulation of hepcidin by erythropoiesis dominates regulation by iron. Because iron overload develops in some MDS patients who do not require RBC transfusions, the suppressive effect of ineffective erythropoiesis on hepcidin may also play a role in iron overload. We anticipate that additional novel tools for measuring iron overload and a molecular-mechanism-driven description of MDS subtypes will provide a deeper understanding of how iron metabolism and erythropoiesis intersect in MDSs and improve clinical management of this patient population.
Collapse
|
793
|
Abstract
Although most circulating iron in blood plasma is destined for erythropoiesis, the mechanisms by which erythropoietic demand modulates the iron supply ("erythroid regulators") remain largely unknown. Iron absorption, plasma iron concentrations, and tissue iron distribution are tightly controlled by the liver-produced hormone hepcidin. During the last decade, much progress has been made in elucidating hepcidin regulation by iron and inflammation. This review discusses the less understood mechanisms and mediators of hepcidin suppression in physiologically and pathologically stimulated erythropoiesis.
Collapse
|