1
|
Kowdley KV, Modi NB, Peltekian K, Vierling JM, Ferris C, Valone FH, Gupta S. Rusfertide for the treatment of iron overload in HFE-related haemochromatosis: an open-label, multicentre, proof-of-concept phase 2 trial. Lancet Gastroenterol Hepatol 2023; 8:1118-1128. [PMID: 37863080 DOI: 10.1016/s2468-1253(23)00250-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/23/2023] [Accepted: 07/27/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND Hereditary haemochromatosis protein (HFE)-related haemochromatosis, an inherited iron overload disorder caused by insufficient hepcidin production, results in excessive iron absorption and tissue and organ injury, and is treated with first-line therapeutic phlebotomy. We aimed to investigate the efficacy and safety of rusfertide, a peptidic mimetic of hepcidin, in patients with HFE-related haemochromatosis. METHODS This open-label, multicentre, proof-of-concept phase 2 trial was done across nine academic and community centres in the USA and Canada. Adults (aged ≥18 years) with HFE-related haemochromatosis on a stable therapeutic phlebotomy regimen (maintenance phase) for at least 6 months before screening and who had a phlebotomy frequency of at least 0·25 per month (eg, at least three phlebotomies in 12 months or at least four phlebotomies in 15 months) and less than one phlebotomy per month, with serum ferritin of less than 300 ng/mL and haemoglobin of more than 11·5 g/dL, were eligible. Patients initiated 24 weeks of subcutaneous rusfertide treatment within 7 days of a scheduled phlebotomy at 10 mg once weekly. Rusfertide doses and dosing schedules could be adjusted to maintain serum transferrin iron saturation (TSAT) at less than 40%. During rusfertide treatment, investigators were to consider the need for phlebotomy when the serum ferritin and TSAT values exceeded the patient's individual pre-phlebotomy serum ferritin and TSAT values. No primary endpoint or testing hierarchy was prespecified. Prespecified efficacy endpoints included the change in the frequency of phlebotomies; the proportion of patients achieving phlebotomy independence; change in serum iron, TSAT, serum transferrin, serum ferritin, and liver iron concentration (LIC) as measured by MRI; and treatment-emergent adverse events (TEAEs). The key efficacy analyses for phlebotomy rate and LIC were conducted by use of paired t tests in the intention-to-treat population, defined as all patients who received any study drug and who had pretreatment and at least one post-dose measurement. We included all participants who received at least one dose of rusfertide in the safety analyses. This trial is closed and completed and is registered with ClinicalTrials.gov, NCT04202965. FINDINGS Between March 11, 2020, and April 23, 2021, 28 patients were screened and 16 (ten [63%] men and six [38%] women) were enrolled. 16 were included in analyses of phlebotomy endpoints and 14 for the LIC endpoint. 12 (75%) patients completed 24 weeks of treatment. The mean number of phlebotomies was significantly reduced during the 24-week rusfertide treatment (0·06 phlebotomies [95% CI -0·07 to 0·20]) compared with 24 weeks pre-study (2·31 phlebotomies [95% CI 1·77 to 2·85]; p<0·0001). 15 (94%) of 16 patients were phlebotomy-free during the treatment period. Mean LIC in the 14 patients in the intention-to-treat population was 1·4 mg iron per g dry liver weight (95% CI 1·0 to 1·8) at screening and 1·1 mg iron per g dry liver weight (95% CI 0·9 to 1·3) at the end of treatment (p=0·068). Mean TSAT was 45·3% (95% CI 33·2 to 57·3) at screening, 36·7% (24·2 to 49·2) after the pretreatment phlebotomy, 21·8% (15·8 to 27·9) 24 h after the first dose of rusfertide, 40·4% (27·1 to 53·8) at the end of treatment, and 32·6% (25·0 to 40·1) over the treatment duration. Mean serum iron was 24·6 μmol/L (95% CI 18·6 to 30·6), 20·1 μmol/L (14·8 to 25·3), 11·9 μmol/L (9·2 to 14·7), 22·5 μmol/L (15·9 to 29·1), and 19·0 μmol/L (15·3 to 22·6) at these same timepoints, respectively. Mean serum ferritin was 83·3 μg/L (52·2 to 114.4), 65·5 μg/L (32·1 to 98·9), 62·8 μg/L (33·8 to 91·9), 150·0 μg/L (86·6 to 213.3), and 94·3 μg/L (54·9 to 133.6) at these same timepoints, respectively. There were only minor changes in serum transferrin concentration. 12 (75%) patients had at least one TEAE, the most common of which was injection site pain (five [31%] patients). All TEAEs were mild or moderate in severity, except for a serious adverse event of pancreatic adenocarcinoma, which was considered severe and unrelated to treatment and was pre-existing and diagnosed 21 days after starting rusfertide treatment. INTERPRETATION Rusfertide prevents iron re-accumulation in the absence of phlebotomies and could be a viable therapeutic option for selected patients with haemochromatosis. FUNDING Protagonist Therapeutics.
Collapse
Affiliation(s)
- Kris V Kowdley
- Liver Institute Northwest, Seattle, WA, USA; Elson S Floyd College of Medicine, Washington State University, Seattle, WA, USA.
| | | | - Kevork Peltekian
- Division of Digestive Care and Endoscopy, Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - John M Vierling
- Section of Gastroenterology and Hepatology and Division of Abdominal Transplantation, Baylor College of Medicine, Houston, TX, USA
| | | | | | | |
Collapse
|
2
|
Ginzburg YZ. Hepcidin and its multiple partners: Complex regulation of iron metabolism in health and disease. VITAMINS AND HORMONES 2023; 123:249-284. [PMID: 37717987 DOI: 10.1016/bs.vh.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
The peptide hormone hepcidin is central to the regulation of iron metabolism, influencing the movement of iron into the circulation and determining total body iron stores. Its effect on a cellular level involves binding ferroportin, the main iron export protein, preventing iron egress and leading to iron sequestration within ferroportin-expressing cells. Hepcidin expression is enhanced by iron loading and inflammation and suppressed by erythropoietic stimulation. Aberrantly increased hepcidin leads to systemic iron deficiency and/or iron restricted erythropoiesis as occurs in anemia of chronic inflammation. Furthermore, insufficiently elevated hepcidin occurs in multiple diseases associated with iron overload such as hereditary hemochromatosis and iron loading anemias. Abnormal iron metabolism as a consequence of hepcidin dysregulation is an underlying factor resulting in pathophysiology of multiple diseases and several agents aimed at manipulating this pathway have been designed, with some already in clinical trials. In this chapter, we assess the complex regulation of hepcidin, delineate the many binding partners involved in its regulation, and present an update on the development of hepcidin agonists and antagonists in various clinical scenarios.
Collapse
Affiliation(s)
- Yelena Z Ginzburg
- Tisch Cancer Institute, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, United Sates.
| |
Collapse
|
3
|
Fisher AL, Babitt JL. Coordination of iron homeostasis by bone morphogenetic proteins: Current understanding and unanswered questions. Dev Dyn 2022; 251:26-46. [PMID: 33993583 PMCID: PMC8594283 DOI: 10.1002/dvdy.372] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/15/2021] [Accepted: 05/07/2021] [Indexed: 01/19/2023] Open
Abstract
Iron homeostasis is tightly regulated to balance the iron requirement for erythropoiesis and other vital cellular functions, while preventing cellular injury from iron excess. The liver hormone hepcidin is the master regulator of systemic iron balance by controlling the degradation and function of the sole known mammalian iron exporter ferroportin. Liver hepcidin expression is coordinately regulated by several signals that indicate the need for more or less iron, including plasma and tissue iron levels, inflammation, and erythropoietic drive. Most of these signals regulate hepcidin expression by modulating the activity of the bone morphogenetic protein (BMP)-SMAD pathway, which controls hepcidin transcription. Genetic disorders of iron overload and iron deficiency have identified several hepatocyte membrane proteins that play a critical role in mediating the BMP-SMAD and hepcidin regulatory response to iron. However, the precise molecular mechanisms by which serum and tissue iron levels are sensed to regulate BMP ligand production and promote the physical and/or functional interaction of these proteins to modulate SMAD signaling and hepcidin expression remain uncertain. This critical commentary will focus on the current understanding and key unanswered questions regarding how the liver senses iron levels to regulate BMP-SMAD signaling and thereby hepcidin expression to control systemic iron homeostasis.
Collapse
Affiliation(s)
| | - Jodie L Babitt
- Corresponding author: Jodie L Babitt, Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA. Mailing address: 185 Cambridge St., CPZN-8208, Boston, MA 02114. Telephone: +1 (617) 643-3181.
| |
Collapse
|
4
|
Vaulont S. Et le fer dans tout çà ? Med Sci (Paris) 2021; 37 Hors série n° 2:38-40. [PMID: 34895462 DOI: 10.1051/medsci/2021228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
5
|
Duarte TL, Talbot NP, Drakesmith H. NRF2 and Hypoxia-Inducible Factors: Key Players in the Redox Control of Systemic Iron Homeostasis. Antioxid Redox Signal 2021; 35:433-452. [PMID: 32791852 DOI: 10.1089/ars.2020.8148] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Significance: Oxygen metabolism and iron homeostasis are closely linked. Iron facilitates the oxygen-carrying capacity of blood, and its deficiency causes anemia. Conversely, excess free iron is detrimental for stimulating the formation of reactive oxygen species, causing tissue damage. The amount and distribution of iron thus need to be tightly regulated by the liver-expressed hormone hepcidin. This review analyzes the roles of key oxygen-sensing pathways in cellular and systemic regulation of iron homeostasis; specifically, the prolyl hydroxylase domain (PHD)/hypoxia-inducible factor (HIF) and the Kelch-like ECH-associated protein 1/NF-E2 p45-related factor 2 (KEAP1/NRF2) pathways, which mediate tissue adaptation to low and high oxygen, respectively. Recent Advances: In macrophages, NRF2 regulates genes involved in hemoglobin catabolism, iron storage, and iron export. NRF2 was recently identified as the molecular sensor of iron-induced oxidative stress and is responsible for BMP6 expression by liver sinusoidal endothelial cells, which in turn activates hepcidin synthesis by hepatocytes to restore systemic iron levels. Moreover, NRF2 orchestrates the activation of antioxidant defenses that are crucial to protect against iron toxicity. On the contrary, low iron/hypoxia stabilizes renal HIF2a via inactivation of iron-dependent PHD dioxygenases, causing an erythropoietic stimulus that represses hepcidin via an inhibitory effect of erythroferrone on bone morphogenetic proteins. Intestinal HIF2a is also stabilized, increasing the expression of genes involved in dietary iron absorption. Critical Issues: An intimate crosstalk between oxygen-sensing pathways and iron regulatory mechanisms ensures that fluctuations in systemic iron levels are promptly detected and restored. Future Directions: The realization that redox-sensitive transcription factors regulate systemic iron levels suggests novel therapeutic approaches. Antioxid. Redox Signal. 35, 433-452.
Collapse
Affiliation(s)
- Tiago L Duarte
- Instituto de Biologia Molecular e Celular, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Nick P Talbot
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, United Kingdom
| | - Hal Drakesmith
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- Haematology Theme, Oxford Biomedical Research Centre, Oxford, United Kingdom
| |
Collapse
|
6
|
Traeger L, Wiegand SB, Sauer AJ, Corman BHP, Peneyra KM, Wunderer F, Fischbach A, Bagchi A, Malhotra R, Zapol WM, Bloch DB. UBA6 and NDFIP1 regulate the degradation of ferroportin. Haematologica 2021; 107:478-488. [PMID: 34320783 PMCID: PMC8804582 DOI: 10.3324/haematol.2021.278530] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Indexed: 11/17/2022] Open
Abstract
Hepcidin regulates iron homeostasis by controlling the level of ferroportin, the only membrane channel that facilitates export of iron from within cells. Binding of hepcidin to ferroportin induces the ubiquitination of ferroportin at multiple lysine residues and subsequently causes the internalization and degradation of the ligand-channel complex within lysosomes. The objective of this study was to identify components of the ubiquitin system that are involved in ferroportin degradation. A HepG2 cell line, which inducibly expresses ferroportingreen fluorescent protein (FPN-GFP), was established to test the ability of small interfering (siRNA) directed against components of the ubiquitin system to prevent BMP6- and exogenous hepcidin-induced ferroportin degradation. Of the 88 siRNA directed against components of the ubiquitin pathway that were tested, siRNA-mediated depletion of the alternative E1 enzyme UBA6 as well as the adaptor protein NDFIP1 prevented BMP6- and hepcidin-induced degradation of ferroportin in vitro. A third component of the ubiquitin pathway, ARIH1, indirectly inhibited ferroportin degradation by impairing BMP6-mediated induction of hepcidin. In mice, the AAV-mediated silencing of Ndfip1 in the murine liver increased the level of hepatic ferroportin and increased circulating iron. The results suggest that the E1 enzyme UBA6 and the adaptor protein NDFIP1 are involved in iron homeostasis by regulating the degradation of ferroportin. These specific components of the ubiquitin system may be promising targets for the treatment of iron-related diseases, including iron overload and anemia of inflammation.
Collapse
Affiliation(s)
- Lisa Traeger
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston.
| | - Steffen B Wiegand
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Andrew J Sauer
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Benjamin H P Corman
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Kathryn M Peneyra
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Florian Wunderer
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, United States; Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, Frankfurt
| | - Anna Fischbach
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Aranya Bagchi
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Rajeev Malhotra
- Cardiovascular Research Center and the Cardiology Division of the Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Warren M Zapol
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Donald B Bloch
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, United States; Division of Rheumatology, Allergy and Immunology of the Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston.
| |
Collapse
|
7
|
Renassia C, Peyssonnaux C. [Hepcidin: An iron hand in mucosal healing in inflammatory bowel diseases]. Med Sci (Paris) 2021; 37:581-583. [PMID: 34180813 DOI: 10.1051/medsci/2021074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Cyril Renassia
- Institut Cochin, Inserm, CNRS, Université de Paris, Laboratoire d'excellence GR-Ex, 24 rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Carole Peyssonnaux
- Institut Cochin, Inserm, CNRS, Université de Paris, Laboratoire d'excellence GR-Ex, 24 rue du Faubourg Saint-Jacques, 75014 Paris, France
| |
Collapse
|
8
|
Bloomer SA, Brown KE. Hepcidin and Iron Metabolism in Experimental Liver Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1165-1179. [PMID: 33891874 DOI: 10.1016/j.ajpath.2021.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/25/2021] [Accepted: 04/06/2021] [Indexed: 11/18/2022]
Abstract
The liver plays a pivotal role in the regulation of iron metabolism through its ability to sense and respond to iron stores by release of the hormone hepcidin. Under physiologic conditions, regulation of hepcidin expression in response to iron status maintains iron homeostasis. In response to tissue injury, hepcidin expression can be modulated by other factors, such as inflammation and oxidative stress. The resulting dysregulation of hepcidin is proposed to account for alterations in iron homeostasis that are sometimes observed in patients with liver disease. This review describes the effects of experimental forms of liver injury on iron metabolism and hepcidin expression. In general, models of acute liver injury demonstrate increases in hepcidin mRNA and hypoferremia, consistent with hepcidin's role as an acute-phase reactant. Conversely, diverse models of chronic liver injury are associated with decreased hepcidin mRNA but with variable effects on iron status. Elucidating the reasons for the disparate impact of different chronic injuries on iron metabolism is an important research priority, as is a deeper understanding of the interplay among various stimuli, both positive and negative, on hepcidin regulation. Future studies should provide a clearer picture of how dysregulation of hepcidin expression and altered iron homeostasis impact the progression of liver diseases and whether they are a cause or consequence of these pathologies.
Collapse
Affiliation(s)
- Steven A Bloomer
- Division of Science and Engineering, Penn State Abington, Abington, Pennsylvania
| | - Kyle E Brown
- Iowa City Veterans Administration Medical Center, Iowa City, Iowa; Division of Gastroenterology-Hepatology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa; Program in Free Radical and Radiation Biology, Department of Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, Iowa.
| |
Collapse
|
9
|
Neutrophils from hereditary hemochromatosis patients are protected from iron excess and are primed. Blood Adv 2020; 4:3853-3863. [PMID: 32810223 DOI: 10.1182/bloodadvances.2020002198] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023] Open
Abstract
Iron is required for the oxidative response of neutrophils to allow the production of reactive oxygen species (ROS). However, neutrophil function may be severely altered in conditions of iron overload, as observed in chronically transfused patients. Therefore, a tight regulation of neutrophil iron homeostasis seems to be critical for avoiding iron toxicity. Hepcidin is the key iron regulator in organisms; however, no studies have investigated its role in maintaining neutrophil iron homeostasis or characterized neutrophil function in patients with hereditary hemochromatosis (HH), a common iron overload genetic disorder that results from a defect in hepcidin production. To explore these issues, we studied 2 mouse models of iron overload: an experimentally induced iron overload model (EIO), in which hepcidin is increased, and a genetic HH model of iron overload with a deletion of hepatic hepcidin. We found that iron-dependent increase of hepatic hepcidin results in neutrophil intracellular iron trapping and consecutive defects in oxidative burst activity. In contrast, in both HH mouse models and HH patients, the lack of hepcidin expression protects neutrophils from toxic iron accumulation. Moreover, systemic iron overload correlated with a surprising neutrophil priming and resulted in a more powerful oxidative burst. Indeed, important factors in neutrophil priming and activation, such as tumor necrosis factor α (TNF-α), VCAM-1, and ICAM-1 are increased in the plasma of HH patients and are associated with an increase in HH neutrophil phagocytosis capacity and a decrease in L-selectin surface expression. This is the first study to characterize neutrophil iron homeostasis and associated functions in patients with HH.
Collapse
|
10
|
Bondu S, Alary AS, Lefèvre C, Houy A, Jung G, Lefebvre T, Rombaut D, Boussaid I, Bousta A, Guillonneau F, Perrier P, Alsafadi S, Wassef M, Margueron R, Rousseau A, Droin N, Cagnard N, Kaltenbach S, Winter S, Kubasch AS, Bouscary D, Santini V, Toma A, Hunault M, Stamatoullas A, Gyan E, Cluzeau T, Platzbecker U, Adès L, Puy H, Stern MH, Karim Z, Mayeux P, Nemeth E, Park S, Ganz T, Kautz L, Kosmider O, Fontenay M. A variant erythroferrone disrupts iron homeostasis in SF3B1-mutated myelodysplastic syndrome. Sci Transl Med 2020; 11:11/500/eaav5467. [PMID: 31292266 DOI: 10.1126/scitranslmed.aav5467] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 03/19/2019] [Accepted: 06/12/2019] [Indexed: 12/21/2022]
Abstract
Myelodysplastic syndromes (MDS) with ring sideroblasts are hematopoietic stem cell disorders with erythroid dysplasia and mutations in the SF3B1 splicing factor gene. Patients with MDS with SF3B1 mutations often accumulate excessive tissue iron, even in the absence of transfusions, but the mechanisms that are responsible for their parenchymal iron overload are unknown. Body iron content, tissue distribution, and the supply of iron for erythropoiesis are controlled by the hormone hepcidin, which is regulated by erythroblasts through secretion of the erythroid hormone erythroferrone (ERFE). Here, we identified an alternative ERFE transcript in patients with MDS with the SF3B1 mutation. Induction of this ERFE transcript in primary SF3B1-mutated bone marrow erythroblasts generated a variant protein that maintained the capacity to suppress hepcidin transcription. Plasma concentrations of ERFE were higher in patients with MDS with an SF3B1 gene mutation than in patients with SF3B1 wild-type MDS. Thus, hepcidin suppression by a variant ERFE is likely responsible for the increased iron loading in patients with SF3B1-mutated MDS, suggesting that ERFE could be targeted to prevent iron-mediated toxicity. The expression of the variant ERFE transcript that was restricted to SF3B1-mutated erythroblasts decreased in lenalidomide-responsive anemic patients, identifying variant ERFE as a specific biomarker of clonal erythropoiesis.
Collapse
Affiliation(s)
- Sabrina Bondu
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France
| | - Anne-Sophie Alary
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France.,Service d'hématologie biologique, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Paris Centre-Cochin, Paris 75014, France
| | - Carine Lefèvre
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France.,Laboratoire d'excellence du Globule Rouge GR-Ex, Paris 75015, France
| | - Alexandre Houy
- Institut Curie, PSL Research University, Human Genetics and Oncogenesis, Paris 75005, France
| | - Grace Jung
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Thibaud Lefebvre
- Université de Paris, Paris 75006, France.,Laboratoire d'excellence du Globule Rouge GR-Ex, Paris 75015, France.,INSERM, UMR 1149/ERL CNRS 8252, Centre de Recherches sur l'inflammation, Université de Paris, Paris 75018, France
| | - David Rombaut
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France
| | - Ismael Boussaid
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France
| | - Abderrahmane Bousta
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France
| | - François Guillonneau
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France.,Proteomic platform 3P5, Université de Paris, Paris 75014, France
| | - Prunelle Perrier
- Institut de Recherche en Santé Digestive (IRSD), Université de Toulouse, INSERM U1220, Institut National de la Recherche Agronomique U1416, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse 31024, France
| | - Samar Alsafadi
- Institut Curie, PSL Research University, Department of Translational Research, Paris 75005, France
| | - Michel Wassef
- Institut Curie, PSL Research University, INSERM 934/UMR 3215, Genetics and biology of Development, Paris 75005 France
| | - Raphaël Margueron
- Institut Curie, PSL Research University, INSERM 934/UMR 3215, Genetics and biology of Development, Paris 75005 France
| | - Alice Rousseau
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France
| | - Nathalie Droin
- Institut Gustave Roussy, Genomic platform, Villejuif 94805, France
| | - Nicolas Cagnard
- Université de Paris, Paris 75006, France.,Platform Bioinformatics, Université de Paris, Paris 75015, France
| | - Sophie Kaltenbach
- Université de Paris, Paris 75006, France.,Laboratoire de Génétique, AP-HP, Hôpital Necker, Paris 75015, France
| | - Susann Winter
- Medical Clinic und Policlinic 1, Technische Universität Dresden, Dresden 01307, Germany
| | - Anne-Sophie Kubasch
- Medical Clinic und Policlinic 1, Hematology and Cellular Therapy, University Hospital, Leipzig 04103, Germany
| | - Didier Bouscary
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France.,Service d'Hématologie clinique, AP-HP, Hôpitaux Universitaires Paris Centre-Cochin, Paris 75014, France
| | - Valeria Santini
- MDS unit, Hematology, AOU Careggi, University of Florence, Florence 50134, Italy
| | - Andrea Toma
- Département d'Hématologie, AP-HP, Hôpital Henri-Mondor, Université Paris 12, Créteil 94000, France
| | - Mathilde Hunault
- Service des Maladies du Sang, Centre hospitalo-universitaire, Angers 49100, France
| | | | - Emmanuel Gyan
- Service d'hématologie et thérapie cellulaire, Centre hospitalo-universitaire, CNRS ERL 7001 LNOx, Université de Tours, Tours 37044, France
| | - Thomas Cluzeau
- Côte d'Azur University, CHU of Nice, Hematology department and INSERM U1065, Mediterranean Center of Molecular Medecine, Nice 06204, France
| | - Uwe Platzbecker
- Medical Clinic und Policlinic 1, Hematology and Cellular Therapy, University Hospital, Leipzig 04103, Germany
| | - Lionel Adès
- Université de Paris, Paris 75006, France.,Service d'Hématologie Senior, AP-HP, Hôpital Saint-Louis, Paris 75010, France
| | - Hervé Puy
- Université de Paris, Paris 75006, France.,Laboratoire d'excellence du Globule Rouge GR-Ex, Paris 75015, France.,INSERM, UMR 1149/ERL CNRS 8252, Centre de Recherches sur l'inflammation, Université de Paris, Paris 75018, France
| | - Marc-Henri Stern
- Institut Curie, PSL Research University, INSERM U830, Genetics and biology of cancers, DNA repair and uveal melanoma (D.R.U.M.), Équipe labellisée par la Ligue nationale contre le cancer, Paris 75005, France
| | - Zoubida Karim
- Université de Paris, Paris 75006, France.,Laboratoire d'excellence du Globule Rouge GR-Ex, Paris 75015, France.,INSERM, UMR 1149/ERL CNRS 8252, Centre de Recherches sur l'inflammation, Université de Paris, Paris 75018, France
| | - Patrick Mayeux
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France.,Laboratoire d'excellence du Globule Rouge GR-Ex, Paris 75015, France.,Proteomic platform 3P5, Université de Paris, Paris 75014, France
| | - Elizabeta Nemeth
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Sophie Park
- Département d'Hématologie, Centre Hospitalier Universitaire, Université de Grenoble Alpes, La Tronche 38700, France
| | - Tomas Ganz
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Léon Kautz
- Institut de Recherche en Santé Digestive (IRSD), Université de Toulouse, INSERM U1220, Institut National de la Recherche Agronomique U1416, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse 31024, France
| | - Olivier Kosmider
- Université de Paris, Paris 75006, France. .,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France.,Service d'hématologie biologique, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Paris Centre-Cochin, Paris 75014, France.,Laboratoire d'excellence du Globule Rouge GR-Ex, Paris 75015, France
| | - Michaëla Fontenay
- Université de Paris, Paris 75006, France. .,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France.,Service d'hématologie biologique, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Paris Centre-Cochin, Paris 75014, France.,Laboratoire d'excellence du Globule Rouge GR-Ex, Paris 75015, France
| |
Collapse
|
11
|
Baek JH, Shin HKH, Gao Y, Buehler PW. Ferroportin inhibition attenuates plasma iron, oxidant stress, and renal injury following red blood cell transfusion in guinea pigs. Transfusion 2020; 60:513-523. [PMID: 32064619 DOI: 10.1111/trf.15720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/03/2019] [Accepted: 12/18/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Red blood cell (RBC) transfusions result in the sequestration and metabolism of storage-damaged RBCs within the spleen and liver. These events are followed by increased plasma iron concentrations that can contribute to oxidant stress and cellular injury. We hypothesized that administration of a ferroportin inhibitor (FPN-INH) immediately after acute RBC exchange transfusion could attenuate posttransfusion circulatory compartment iron exposure, by retaining iron in spleen and hepatic macrophages. STUDY DESIGN AND METHODS Donor guinea pig blood was leukoreduced, and RBCs were preserved at 4°C. Recipient guinea pigs (n = 5/group) were exchange transfused with donor RBCs after refrigerator preservation and dosed intravenously with a small-molecule FPN-INH. Groups included transfusion with vehicle (saline), 5 mg/kg or 25 mg/kg FPN-INH. A time course of RBC morphology, plasma non-transferrin-bound iron (NTBI) and plasma hemoglobin (Hb) were evaluated. End-study spleen, liver, and kidney organ iron levels, as well as renal tissue oxidation and injury, were measured acutely (24-hr after transfusion). RESULTS RBC transfusion increased plasma NTBI, with maximal concentrations occurring 8 hours after transfusion. Posttransfusion iron accumulation resulted in tubule oxidation and acute kidney injury. FPN inhibition increased spleen and liver parenchymal/macrophage iron accumulation, but attenuated plasma NTBI, and subsequent renal tissue oxidation/injury. CONCLUSION In situations of acute RBC transfusion, minimizing circulatory NTBI exposure by FPN inhibition may attenuate organ-specific adverse consequences of iron exposure.
Collapse
Affiliation(s)
- Jin Hyen Baek
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices, Center of Biologics Evaluation and Research (CBER), FDA, Silver Spring, Maryland, USA
| | - Hye Kyung H Shin
- Laboratory of Biochemistry and Vascular Biology, Division of Blood Components and Devices, Center of Biologics Evaluation and Research (CBER), FDA, Silver Spring, Maryland, USA
| | - Yamei Gao
- Division of Viral Products, Center of Biologics Evaluation and Research (CBER), FDA, Silver Spring, Maryland, USA
| | - Paul W Buehler
- Department of Pathology, Center for Blood Oxygen Transport, Baltimore, Maryland, USA.,Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, University of Maryland Baltimore School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
12
|
Wang X, Garrick MD, Collins JF. Animal Models of Normal and Disturbed Iron and Copper Metabolism. J Nutr 2019; 149:2085-2100. [PMID: 31504675 PMCID: PMC6887953 DOI: 10.1093/jn/nxz172] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/04/2019] [Accepted: 06/28/2019] [Indexed: 01/19/2023] Open
Abstract
Research on the interplay between iron and copper metabolism in humans began to flourish in the mid-20th century, and diseases associated with dysregulated homeostasis of these essential trace minerals are common even today. Iron deficiency is the most frequent cause of anemia worldwide, leading to significant morbidity, particularly in developing countries. Iron overload is also quite common, usually being the result of genetic mutations which lead to inappropriate expression of the iron-regulatory hormone hepcidin. Perturbations of copper homeostasis in humans have also been described, including rare genetic conditions which lead to severe copper deficiency (Menkes disease) or copper overload (Wilson disease). Historically, the common laboratory rat (Rattus norvegicus) was the most frequently utilized species to model human physiology and pathophysiology. Recently, however, the development of genetic-engineering technology combined with the worldwide availability of numerous genetically homogenous (i.e., inbred) mouse strains shifted most research on iron and copper metabolism to laboratory mice. This created new opportunities to understand the function of individual genes in the context of a living animal, but thoughtful consideration of whether mice are the most appropriate models of human pathophysiology was not necessarily involved. Given this background, this review is intended to provide a guide for future research on iron- and copper-related disorders in humans. Generation of complementary experimental models in rats, swine, and other mammals is now facile given the advent of newer genetic technologies, thus providing the opportunity to accelerate the identification of pathogenic mechanisms and expedite the development of new treatments to mitigate these important human disorders.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | - Michael D Garrick
- Department of Biochemistry, University at Buffalo–The State University of New York, Buffalo, NY, USA
| | - James F Collins
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, USA,Address correspondence to JFC (e-mail: )
| |
Collapse
|
13
|
Liu J, Liu W, Liu Y, Miao Y, Guo Y, Song H, Wang F, Zhou H, Ganz T, Yan B, Liu S. New thiazolidinones reduce iron overload in mouse models of hereditary hemochromatosis and β-thalassemia. Haematologica 2019; 104:1768-1781. [PMID: 30792208 PMCID: PMC6717595 DOI: 10.3324/haematol.2018.209874] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 02/15/2019] [Indexed: 02/06/2023] Open
Abstract
Genetic iron-overload disorders, mainly hereditary hemochromatosis and untransfused β-thalassemia, affect a large population worldwide. The primary etiology of iron overload in these diseases is insufficient production of hepcidin by the liver, leading to excessive intestinal iron absorption and iron efflux from macrophages. Hepcidin agonists would therefore be expected to ameliorate iron overload in hereditary hemochromatosis and β-thalassemia. In the current study, we screened our synthetic library of 210 thiazolidinone compounds and identified three thiazolidinone compounds, 93, 156 and 165, which stimulated hepatic hepcidin production. In a hemochromatosis mouse model with hemochromatosis deficiency, the three compounds prevented the development of iron overload and elicited iron redistribution from the liver to the spleen. Moreover, these compounds also greatly ameliorated iron overload and mitigated ineffective erythropoiesis in β-thalassemic mice. Compounds 93, 156 and 165 acted by promoting SMAD1/5/8 signaling through differentially repressing ERK1/2 phosphorylation and decreasing transmembrane protease serine 6 activity. Additionally, compounds 93, 156 and 165 targeted erythroid regulators to strengthen hepcidin expression. Therefore, our hepcidin agonists induced hepcidin expression synergistically through a direct action on hepatocytes via SMAD1/5/8 signaling and an indirect action via eythroid cells. By increasing hepcidin production, thiazolidinone compounds may provide a useful alternative for the treatment of iron-overload disorders.
Collapse
Affiliation(s)
- Jing Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wei Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- School of Environmental Science and Engineering, Shandong University, Shandong, China
| | - Yang Miao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
| | - Yifan Guo
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
| | - Haoyang Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
| | - Fudi Wang
- Department of Nutrition, Nutrition Discovery Innovation Center, Institute of Nutrition and Food Safety, School of Public Health, School of Medicine, Zhejiang University, Zhejiang, China
| | - Hongyu Zhou
- Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Institute of Environmental Research at Greater Bay, Guangzhou University, Guangzhou, China
| | - Tomas Ganz
- Department of Medicine and Department of Pathology, David Geffen School of Medicine at University of California, California, Los Angeles, CA, USA
| | - Bing Yan
- School of Environmental Science and Engineering, Shandong University, Shandong, China
- Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Institute of Environmental Research at Greater Bay, Guangzhou University, Guangzhou, China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
14
|
Qian ZM, Ke Y. Hepcidin and its therapeutic potential in neurodegenerative disorders. Med Res Rev 2019; 40:633-653. [PMID: 31471929 DOI: 10.1002/med.21631] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/18/2019] [Accepted: 08/05/2019] [Indexed: 12/12/2022]
Abstract
Abnormally high brain iron, resulting from the disrupted expression or function of proteins involved in iron metabolism in the brain, is an initial cause of neuronal death in neuroferritinopathy and aceruloplasminemia, and also plays a causative role in at least some of the other neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, and Friedreich's ataxia. As such, iron is believed to be a novel target for pharmacological intervention in these disorders. Reducing iron toward normal levels or hampering the increases in iron associated with age in the brain is a promising therapeutic strategy for all iron-related neurodegenerative disorders. Hepcidin is a crucial regulator of iron homeostasis in the brain. Recent studies have suggested that upregulating brain hepcidin levels can significantly reduce brain iron content through the regulation of iron transport protein expression in the blood-brain barrier and in neurons and astrocytes. In this review, we focus on the discussion of the therapeutic potential of hepcidin in iron-associated neurodegenerative diseases and also provide a systematic overview of recent research progress on how misregulated brain iron metabolism is involved in the development of multiple neurodegenerative disorders.
Collapse
Affiliation(s)
- Zhong-Ming Qian
- Institute of Translational & Precision Medicine, Nantong University, Nantong, Jiangsu, China.,Laboratory of Neuropharmacology, School of Pharmacy & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ya Ke
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| |
Collapse
|
15
|
Iron Pathophysiology in Alzheimer’s Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1173:67-104. [DOI: 10.1007/978-981-13-9589-5_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
16
|
Asperti M, Denardo A, Gryzik M, Arosio P, Poli M. The role of heparin, heparanase and heparan sulfates in hepcidin regulation. VITAMINS AND HORMONES 2019; 110:157-188. [PMID: 30798810 DOI: 10.1016/bs.vh.2019.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hepcidin is considered the major regulator of systemic iron homeostasis in human and mice, and its expression in the liver is mainly regulated at a transcriptional level. Central to its regulation are the bone morphogenetic proteins, particularly BMP6, that are heparin binding proteins. Heparin was found to inhibit hepcidin expression and BMP6 activity in hepatic cell lines and in mice, suggesting that endogenous heparan sulfates are involved in the pathway of hepcidin expression. This was confirmed by the study of cells and mice overexpressing heparanase, the enzyme that hydrolyzes heparan sulfates, and by cellular models with altered heparan sulfates. The evidences supporting the role of heparan sulfate in hepcidin expression are summarized in this chapter and open the way for new understanding in hepcidin expression and its control in pathological condition.
Collapse
Affiliation(s)
- Michela Asperti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Andrea Denardo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Magdalena Gryzik
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Paolo Arosio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Maura Poli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
17
|
Abstract
Hepcidin is central to regulation of iron metabolism. Its effect on a cellular level involves binding ferroportin, the main iron export protein, resulting in its internalization and degradation and leading to iron sequestration within ferroportin-expressing cells. Aberrantly increased hepcidin leads to systemic iron deficiency and/or iron restricted erythropoiesis. Furthermore, insufficiently elevated hepcidin occurs in multiple diseases associated with iron overload. Abnormal iron metabolism as a consequence of hepcidin dysregulation is an underlying factor resulting in pathophysiology of multiple diseases and several agents aimed at manipulating this pathway have been designed, with some already in clinical trials. In this chapter, we present an overview of and rationale for exploring the development of hepcidin agonists and antagonists in various clinical scenarios.
Collapse
Affiliation(s)
- Yelena Z Ginzburg
- Tisch Cancer Institute, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
18
|
Jiang Y, Chen B, Yan Y, Zhu GX. Hepcidin protects against iron overload-induced inhibition of bone formation in zebrafish. FISH PHYSIOLOGY AND BIOCHEMISTRY 2019; 45:365-374. [PMID: 30361820 DOI: 10.1007/s10695-018-0568-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/29/2018] [Indexed: 06/08/2023]
Abstract
Iron overload increases the risk of osteoporosis, which leads to an increase in the incidences of bone fracture after menopause. In vitro studies have demonstrated that excess iron can inhibit osteoblast activity. Hepcidin, a central regulator of iron homeostasis, prevents iron overload, and thus, it is considered to have anti-osteoporosis effects. In this study, a zebrafish model was employed to investigate the therapeutic role of hepcidin in iron overload-induced inhibition of bone formation. Our results show that ferric ammonium citrate (FAC) treatment decreased osteoblast-specific gene expression (runx2a, runx2b, and bglap) and bone mineralization in the zebrafish embryo, accompanied with increased whole-body iron levels and oxidative stress. Bone mineralization and osteoblast-specific gene expression increased with the microinjection of hepcidin-flag Capped-mRNA into zebrafish embryos. Moreover, the whole-body iron content and oxidative stress in the iron-overloaded zebrafish embryos decreased when microinjection of hepcidin preceded the FAC treatment. Therefore, our study suggests that hepcidin could prevent and rescue reduced bone formation caused by FAC treatment by preventing iron absorption.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Orthopedics, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi, 214000, China
| | - Bin Chen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, 215004, China
| | - Yilin Yan
- Institute of Neuroscience, University of Oregon, Eugene, OR, USA
| | - Guo-Xing Zhu
- Department of Orthopedics, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi, 214000, China.
| |
Collapse
|
19
|
Almutairi MMA, Xu G, Shi H. Iron Pathophysiology in Stroke. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1173:105-123. [PMID: 31456207 DOI: 10.1007/978-981-13-9589-5_6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ischemic and hemorrhagic stroke are the common types of stroke that lead to brain injury neurological deficits and mortality. All forms of stroke remain a serious health issue, and there is little successful development of drugs for treating stroke. Incomplete understanding of stroke pathophysiology is considered the main barrier that limits this research progress. Besides mitochondria and free radical-producing enzymes, labile iron is an important contributor to oxidative stress. Although iron regulation and metabolism in cerebral stroke are not fully understood, much progress has been achieved in recent years. For example, hepcidin has recently been recognized as the principal regulator of systemic iron homeostasis and a bridge between inflammation and iron regulation. This review discusses recent research progress in iron pathophysiology following cerebral stroke, focusing molecular regulation of iron metabolism and potential treatment targets.
Collapse
Affiliation(s)
- Mohammed M A Almutairi
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, 66045, USA.,Department of Pharmacology and Toxicology, School of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Grace Xu
- Department of Anesthesiology, School of Medicine, University of Kansas, Kansas City, KS, 66160, USA
| | - Honglian Shi
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, 66045, USA.
| |
Collapse
|
20
|
Sukhbaatar N, Weichhart T. Iron Regulation: Macrophages in Control. Pharmaceuticals (Basel) 2018; 11:ph11040137. [PMID: 30558109 PMCID: PMC6316009 DOI: 10.3390/ph11040137] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 12/21/2022] Open
Abstract
Macrophages are sentinel cells of the innate immune system and have important functions in development, tissue homeostasis, and immunity. These phylogenetically ancient cells also developed a variety of mechanisms to control erythropoiesis and the handling of iron. Red pulp macrophages in the spleen, Kupffer cells in the liver, and central nurse macrophages in the bone marrow ensure a coordinated metabolism of iron to support erythropoiesis. Phagocytosis of senescent red blood cells by macrophages in the spleen and the liver provide a continuous delivery of recycled iron under steady-state conditions and during anemic stress. Central nurse macrophages in the bone marrow utilize this iron and provide a cellular scaffold and niche to promote differentiation of erythroblasts. This review focuses on the role of the distinct macrophage populations that contribute to efficient iron metabolism and highlight important cellular and systemic mechanisms involved in iron-regulating processes.
Collapse
Affiliation(s)
- Nyamdelger Sukhbaatar
- Medical University of Vienna, Center for Pathobiochemistry and Genetics, Vienna 1090, Austria.
| | - Thomas Weichhart
- Medical University of Vienna, Center for Pathobiochemistry and Genetics, Vienna 1090, Austria.
| |
Collapse
|
21
|
Hepcidin Therapeutics. Pharmaceuticals (Basel) 2018; 11:ph11040127. [PMID: 30469435 PMCID: PMC6316648 DOI: 10.3390/ph11040127] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 11/15/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022] Open
Abstract
Hepcidin is a key hormonal regulator of systemic iron homeostasis and its expression is induced by iron or inflammatory stimuli. Genetic defects in iron signaling to hepcidin lead to “hepcidinopathies” ranging from hereditary hemochromatosis to iron-refractory iron deficiency anemia, which are disorders caused by hepcidin deficiency or excess, respectively. Moreover, dysregulation of hepcidin is a pathogenic cofactor in iron-loading anemias with ineffective erythropoiesis and in anemia of inflammation. Experiments with preclinical animal models provided evidence that restoration of appropriate hepcidin levels can be used for the treatment of these conditions. This fueled the rapidly growing field of hepcidin therapeutics. Several hepcidin agonists and antagonists, as well as inducers and inhibitors of hepcidin expression have been identified to date. Some of them were further developed and are currently being evaluated in clinical trials. This review summarizes the state of the art.
Collapse
|
22
|
Abstract
The liver orchestrates systemic iron balance by producing and secreting hepcidin. Known as the iron hormone, hepcidin induces degradation of the iron exporter ferroportin to control iron entry into the bloodstream from dietary sources, iron recycling macrophages, and body stores. Under physiologic conditions, hepcidin production is reduced by iron deficiency and erythropoietic drive to increase the iron supply when needed to support red blood cell production and other essential functions. Conversely, hepcidin production is induced by iron loading and inflammation to prevent the toxicity of iron excess and limit its availability to pathogens. The inability to appropriately regulate hepcidin production in response to these physiologic cues underlies genetic disorders of iron overload and deficiency, including hereditary hemochromatosis and iron-refractory iron deficiency anemia. Moreover, excess hepcidin suppression in the setting of ineffective erythropoiesis contributes to iron-loading anemias such as β-thalassemia, whereas excess hepcidin induction contributes to iron-restricted erythropoiesis and anemia in chronic inflammatory diseases. These diseases have provided key insights into understanding the mechanisms by which the liver senses plasma and tissue iron levels, the iron demand of erythrocyte precursors, and the presence of potential pathogens and, importantly, how these various signals are integrated to appropriately regulate hepcidin production. This review will focus on recent insights into how the liver senses body iron levels and coordinates this with other signals to regulate hepcidin production and systemic iron homeostasis.
Collapse
|
23
|
Song H, Zhang S, Sun X, Liu J, Wu Y, Guo W, Wang F, Ou X, Cong M, Jin E, Li W, Liu S. Distinct Iron Deposition Profiles of Liver Zones in Various Models with Iron Homeostasis Disorders. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1800866. [PMID: 30479929 PMCID: PMC6247051 DOI: 10.1002/advs.201800866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/19/2018] [Indexed: 06/09/2023]
Abstract
Determination of iron accumulation is crucial in diagnosing the occurrence and progression of many liver- and iron-related diseases. Thus far, little is known about the profiles of iron deposition in different liver zones, particularly under conditions with disordered iron homeostasis. Here, uneven iron distribution in livers of patients with hereditary hemochromatosis (HH) is uncovered, showing the region with the highest iron concentration near the entrance site of the portal vein and hepatic artery in contrast to the sites with the lowest iron concentration close to the distal edge. Distinct iron distribution profiles are also found throughout liver zones in wild-type mice and various mouse models with iron metabolism disorders, including hemochromatosis (Hfe-/- ), iron deficiency, and inflammation. Of note, similar findings observed in HH patients are further demonstrated in Hfe-/- mice. Moreover, the zones with greater iron accumulation appear to be more sensitive to iron changes, e.g., there is iron increase upon iron overload and iron loss in response to iron deficiency. Mechanistic investigation manifests that these differential iron changes in liver zones are subjected to the regulation by the hepcidin-ferroportin axis. Additionally, the data corroborate the reliability of magnetic resonance imaging (MRI) in recognizing the differential iron deposition profiles among liver zones.
Collapse
Affiliation(s)
- Haoyang Song
- Anhui Province Key Laboratory of Embryo Development and Reproductive RegulationAnhui Province Key Laboratory of Environmental Hormone and ReproductionFuyang Normal UniversityFuyang236037China
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085China
| | - Shuping Zhang
- Institute for Medical Engineering and ScienceMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Xia Sun
- Radiology DepartmentBeijing Friendship HospitalCapital Medical UniversityBeijing100050China
| | - Jing Liu
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085China
| | - Yakun Wu
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085China
- University of Chinese Academy of SciencesBeijing100049China
| | - Wenli Guo
- College of FisheriesHenan Normal UniversityXinxiang453007China
- QIMR Berghofer Medical Research InstituteBrisbane4029Australia
| | - Fudi Wang
- Department of NutritionNutrition Discovery Innovation CenterInstitute of Nutrition and Food SafetySchool of Public HealthSchool of MedicineZhejiang UniversityHangzhou310085China
| | - Xiaojuan Ou
- Liver Research CenterBeijing Friendship HospitalCapital Medical UniversityBeijing100050China
| | - Min Cong
- Liver Research CenterBeijing Friendship HospitalCapital Medical UniversityBeijing100050China
| | - Erhu Jin
- Radiology DepartmentBeijing Friendship HospitalCapital Medical UniversityBeijing100050China
| | - Wenyong Li
- Anhui Province Key Laboratory of Embryo Development and Reproductive RegulationAnhui Province Key Laboratory of Environmental Hormone and ReproductionFuyang Normal UniversityFuyang236037China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
24
|
Pandey S, Pandey SK, Shah V. Role of HAMP Genetic Variants on Pathophysiology of Iron Deficiency Anemia. Indian J Clin Biochem 2018; 33:479-482. [PMID: 30319197 DOI: 10.1007/s12291-017-0707-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/16/2017] [Indexed: 12/21/2022]
Abstract
Hepcidin is a 25-amino acid peptide hormone produced by hepatocytes and plays a key role in body iron metabolism. Hepcidin deficiency is the cause of iron overload in hereditary hemochromatosis, iron-loading anemia, and its excess is associated with anemia of inflammation, chronic disease and iron deficiency anemia (IDA). The aims of this study was to evaluate HAMP gene mutation, namely IVS2 + 1(-G) (c.148-150 + 1del) and Gly71 Asp (c.212G > A (rs104894696) association with iron status in IDA conditions. Our study participants were 500 IDA patients and 550 age and sex-matched healthy controls. Hepcidin, ferritin and CRP analysis was done by ELISA method while ESR analysis was done according to Wintrobe method. CBC analysis was done by auto-analyzer. Two mutations in the HAMP genes were analysed by PCR RFLP method. Among the IDA patients, 7 were heterozygous for Met50del IVS2 + 1(-G) mutation. Nine IDA patients were heterozygous for G71D G-A mutation and homozygous were not identified in both mutations.Controls were showing heterozygous frequency 1.8 and 2.1% of Met50del IVS2 + 1(-G) and G71D G-A mutations respectively. Mutation of HAMP (Met50del IVS2 + 1(-G) and G71D G-A) were clinically associated with IDA and act as modulator of disease.
Collapse
Affiliation(s)
- S Pandey
- Centre for Biotechnology Studies, Awadhesh Pratap Singh University, Rewa, M.P. 486003 India
| | - S K Pandey
- Centre for Biotechnology Studies, Awadhesh Pratap Singh University, Rewa, M.P. 486003 India
| | - V Shah
- Centre for Biotechnology Studies, Awadhesh Pratap Singh University, Rewa, M.P. 486003 India
| |
Collapse
|
25
|
Yin X, Wu Q, Monga J, Xie E, Wang H, Wang S, Zhang H, Wang ZY, Zhou T, Shi Y, Rogers J, Lin H, Min J, Wang F. HDAC1 Governs Iron Homeostasis Independent of Histone Deacetylation in Iron-Overload Murine Models. Antioxid Redox Signal 2018; 28:1224-1237. [PMID: 29113455 DOI: 10.1089/ars.2017.7161] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
AIMS Iron-overload disorders are common and could lead to significant morbidity and mortality worldwide. Due to limited treatment options, there is a great need to develop novel strategies to remove the excess body iron. To discover potential epigenetic modulator in hepcidin upregulation and subsequently decreasing iron burden, we performed an epigenetic screen. The in vivo effects of the identified compounds were further tested in iron-overload mouse models, including Hfe-/-, Hjv-/-, and hepatocyte-specific Smad4 knockout (Smad4fl/fl;Alb-Cre+) mice. RESULTS Entinostat (MS-275), the clinical used histone deacetylase 1 (HDAC1) inhibitor, was identified the most potent hepcidin agonist. Consistently, Hdac1-deficient mice also presented higher hepcidin levels than wild-type controls. Notably, the long-term treatment with entinostat in Hfe-/- mice significantly alleviated iron overload through upregulating hepcidin transcription. In contrast, entinostat showed no effect on hepcidin expression and iron levels in Smad4fl/fl;Alb-Cre+ mice. Further mechanistic studies revealed that HDAC1 suppressed expression of hepcidin through interacting with SMAD4 rather than deacetylation of SMAD4 or histone-H3 on the hepcidin promoter. INNOVATION The findings uncovered HDAC1 as a novel hepcidin suppressor through complexing with SMAD4 but not deacetylation of either histone 3 or SMAD4. In addition, our study suggested a novel implication of entinostat in treating iron-overload disorders. CONCLUSIONS Based on our results, we conclude that entinostat strongly activated hepcidin in vivo and in vitro. HDAC1 could serve as a novel hepcidin suppressor by binding to SMAD4, effect of which is independent of BMP/SMAD1/5/8 signaling. Antioxid. Redox Signal. 28, 1224-1237.
Collapse
Affiliation(s)
- Xiangju Yin
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China .,2 Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University , Zhengzhou, China .,3 Institute of Resources and Environment, Henan Polytechnic University , Jiaozuo, China
| | - Qian Wu
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China .,2 Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University , Zhengzhou, China
| | - Jitender Monga
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Enjun Xie
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Hao Wang
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China .,2 Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University , Zhengzhou, China
| | - Shufen Wang
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Huizhen Zhang
- 2 Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University , Zhengzhou, China
| | - Zhan-You Wang
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Tianhua Zhou
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Yujun Shi
- 4 Laboratory of Pathology, West China Hospital, Sichuan University , Chengdu, China
| | - Jack Rogers
- 5 Neurochemistry Laboratory, Departments of Psychiatry and Pediatrics, Massachusetts General Hospital and Harvard Medical School , Charlestown, Massachusetts
| | - Hening Lin
- 6 Department of Chemistry and Chemical Biology, Howard Hughes Medical Institute, Cornell University , Ithaca, New York
| | - Junxia Min
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Fudi Wang
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China .,2 Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University , Zhengzhou, China
| |
Collapse
|
26
|
Abstract
Hepcidin agonists are a new class of compounds that regulate blood iron levels, limit iron absorption, and could improve the treatment of hemochromatosis, β-thalassemia, polycythemia vera, and other disorders in which disrupted iron homeostasis causes or contributes to disease. Hepcidin agonists also have the potential to prevent severe complications of siderophilic infections in patients with iron overload or chronic liver disease. This review highlights the preclinical studies that support the development of hepcidin agonists for the treatment of these disorders.
Collapse
|
27
|
Vela D. Balance of cardiac and systemic hepcidin and its role in heart physiology and pathology. J Transl Med 2018; 98:315-326. [PMID: 29058707 DOI: 10.1038/labinvest.2017.111] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/23/2017] [Accepted: 08/24/2017] [Indexed: 02/07/2023] Open
Abstract
Hepcidin is the main regulator of iron metabolism in tissues. Its serum levels are mostly correlated with the levels of hepcidin expression from the liver, but local hepcidin can be important for the physiology of other organs as well. There is an increasing evidence that this is the case with cardiac hepcidin. This has been confirmed by studies with models of ischemic heart disease and other heart pathologies. In this review the discussion dissects the role of cardiac hepcidin in cellular homeostasis. This review is complemented with examination of the role of systemic hepcidin in heart disease and its use as a biochemical marker. The relationship between systemic vs local hepcidin in the heart is important because it can help us understand how the fine balance between the actions of two hepcidins affects heart function. Manipulating the axis systemic/cardiac hepcidin could serve as a new therapeutic strategy in heart diseases.
Collapse
Affiliation(s)
- Driton Vela
- Department of Physiology, Faculty of Medicine, University of Prishtina, Prishtina, Kosova
| |
Collapse
|
28
|
Scotet V, Saliou P, Uguen M, L'Hostis C, Merour MC, Triponey C, Chanu B, Nousbaum JB, Le Gac G, Ferec C. Do pregnancies reduce iron overload in HFE hemochromatosis women? results from an observational prospective study. BMC Pregnancy Childbirth 2018; 18:53. [PMID: 29454332 PMCID: PMC5816504 DOI: 10.1186/s12884-018-1684-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 02/06/2018] [Indexed: 12/17/2022] Open
Abstract
Background HFE hemochromatosis is an inborn error of iron metabolism linked to a defect in the regulation of hepcidin synthesis. This autosomal recessive disease typically manifests later in women than men. Although it is commonly stated that pregnancy is, with menses, one of the factors that offsets iron accumulation in women, no epidemiological study has yet supported this hypothesis. The aim of our study was to evaluate the influence of pregnancy on expression of the predominant HFE p.[Cys282Tyr];[Cys282Tyr] genotype. Methods One hundred and forty p.Cys282Tyr homozygous women enrolled in a phlebotomy program between 2004 and 2011 at a blood centre in western Brittany (France) were included in the study. After checking whether the disease expression was delayed in women than in men in our study, the association between pregnancy and iron overload was assessed using multivariable regression analysis. Results Our study confirms that women with HFE hemochromatosis were diagnosed later than men cared for during the same period (52.6 vs. 47.4 y., P < 0.001). Compared to no pregnancy, having at least one pregnancy was not associated with lower iron markers. In contrast, the amount of iron removed by phlebotomies appeared significantly higher in women who had at least one pregnancy (eβ = 1.50, P = 0.047). This relationship disappeared after adjustment for confounding factors (eβ = 1.35, P = 0.088). Conclusions Our study shows that pregnancy status has no impact on iron markers level, and is not in favour of pregnancy being a protective factor in progressive iron accumulation. Our results are consistent with recent experimental data suggesting that the difference in disease expression observed between men and women may be explained by other factors such as hormones.
Collapse
Affiliation(s)
- Virginie Scotet
- UMR1078 "Génétique, Génomique Fonctionnelle et Biotechnologies", Inserm, EFS, Université de Brest, ISBAM, 22 avenue Camille Desmoulins, 29200, Brest, France.
| | - Philippe Saliou
- UMR1078 "Génétique, Génomique Fonctionnelle et Biotechnologies", Inserm, EFS, Université de Brest, ISBAM, 22 avenue Camille Desmoulins, 29200, Brest, France.,Laboratoire d'Hygiene et de Sante Publique, Hopital Morvan, Brest, France
| | - Marianne Uguen
- UMR1078 "Génétique, Génomique Fonctionnelle et Biotechnologies", Inserm, EFS, Université de Brest, ISBAM, 22 avenue Camille Desmoulins, 29200, Brest, France
| | - Carine L'Hostis
- UMR1078 "Génétique, Génomique Fonctionnelle et Biotechnologies", Inserm, EFS, Université de Brest, ISBAM, 22 avenue Camille Desmoulins, 29200, Brest, France
| | | | - Céline Triponey
- Etablissement Français du Sang - Bretagne, Site de Brest, Brest, France
| | - Brigitte Chanu
- Etablissement Français du Sang - Bretagne, Site de Brest, Brest, France
| | - Jean-Baptiste Nousbaum
- UMR1078 "Génétique, Génomique Fonctionnelle et Biotechnologies", Inserm, EFS, Université de Brest, ISBAM, 22 avenue Camille Desmoulins, 29200, Brest, France.,Service d'Hepato-Gastroenterologie, Hopital La Cavale Blanche, Brest, France
| | - Gerald Le Gac
- UMR1078 "Génétique, Génomique Fonctionnelle et Biotechnologies", Inserm, EFS, Université de Brest, ISBAM, 22 avenue Camille Desmoulins, 29200, Brest, France.,Laboratoire de Genetique Moleculaire et d'Histocompatibilite, Hopital Morvan, Brest, France
| | - Claude Ferec
- UMR1078 "Génétique, Génomique Fonctionnelle et Biotechnologies", Inserm, EFS, Université de Brest, ISBAM, 22 avenue Camille Desmoulins, 29200, Brest, France.,Etablissement Français du Sang - Bretagne, Site de Brest, Brest, France.,Laboratoire de Genetique Moleculaire et d'Histocompatibilite, Hopital Morvan, Brest, France
| |
Collapse
|
29
|
Abstract
Stress erythropoiesis (SE) is characterized by an imbalance in erythroid proliferation and differentiation under increased demands of erythrocyte generation and tissue oxygenation. β-thalassemia represents a chronic state of SE, called ineffective erythropoiesis (IE), exhibiting an expansion of erythroid-progenitor pool and deposition of alpha chains on erythrocyte membranes, causing cell death and anemia. Concurrently, there is a decrease in hepcidin expression and a subsequent state of iron overload. There are substantial investigative efforts to target increased iron absorption under IE. There are also avenues for targeting cell contact and signaling within erythroblastic islands under SE, for therapeutic benefits.
Collapse
|
30
|
Wang C, Fang Z, Zhu Z, Liu J, Chen H. Reciprocal regulation between hepcidin and erythropoiesis and its therapeutic application in erythroid disorders. Exp Hematol 2017; 52:24-31. [DOI: 10.1016/j.exphem.2017.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 05/03/2017] [Accepted: 05/04/2017] [Indexed: 12/16/2022]
|
31
|
Wang H, An P, Xie E, Wu Q, Fang X, Gao H, Zhang Z, Li Y, Wang X, Zhang J, Li G, Yang L, Liu W, Min J, Wang F. Characterization of ferroptosis in murine models of hemochromatosis. Hepatology 2017; 66:449-465. [PMID: 28195347 PMCID: PMC5573904 DOI: 10.1002/hep.29117] [Citation(s) in RCA: 446] [Impact Index Per Article: 63.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 02/09/2017] [Indexed: 12/11/2022]
Abstract
UNLABELLED Ferroptosis is a recently identified iron-dependent form of nonapoptotic cell death implicated in brain, kidney, and heart pathology. However, the biological roles of iron and iron metabolism in ferroptosis remain poorly understood. Here, we studied the functional role of iron and iron metabolism in the pathogenesis of ferroptosis. We found that ferric citrate potently induces ferroptosis in murine primary hepatocytes and bone marrow-derived macrophages. Next, we screened for ferroptosis in mice fed a high-iron diet and in mouse models of hereditary hemochromatosis with iron overload. We found that ferroptosis occurred in mice fed a high-iron diet and in two knockout mouse lines that develop severe iron overload (Hjv-/- and Smad4Alb/Alb mice) but not in a third line that develops only mild iron overload (Hfe-/- mice). Moreover, we found that iron overload-induced liver damage was rescued by the ferroptosis inhibitor ferrostatin-1. To identify the genes involved in iron-induced ferroptosis, we performed microarray analyses of iron-treated bone marrow-derived macrophages. Interestingly, solute carrier family 7, member 11 (Slc7a11), a known ferroptosis-related gene, was significantly up-regulated in iron-treated cells compared with untreated cells. However, genetically deleting Slc7a11 expression was not sufficient to induce ferroptosis in mice. Next, we studied iron-treated hepatocytes and bone marrow-derived macrophages isolated from Slc7a11-/- mice fed a high-iron diet. CONCLUSION We found that iron treatment induced ferroptosis in Slc7a11-/- cells, indicating that deleting Slc7a11 facilitates the onset of ferroptosis specifically under high-iron conditions; these results provide compelling evidence that iron plays a key role in triggering Slc7a11-mediated ferroptosis and suggest that ferroptosis may be a promising target for treating hemochromatosis-related tissue damage. (Hepatology 2017;66:449-465).
Collapse
Affiliation(s)
- Hao Wang
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Peng An
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Enjun Xie
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Qian Wu
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Xuexian Fang
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Hong Gao
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Zhuzhen Zhang
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Yuzhu Li
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Xudong Wang
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Jiaying Zhang
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Guoli Li
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Lei Yang
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Wei Liu
- Department of Biochemistry and Molecular Biology, Program in Molecular and Cell Biology, School of MedicineZhejiang UniversityHangzhouChina
| | - Junxia Min
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Fudi Wang
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| |
Collapse
|
32
|
Shah NR. Advances in iron chelation therapy: transitioning to a new oral formulation. Drugs Context 2017; 6:212502. [PMID: 28706555 PMCID: PMC5499896 DOI: 10.7573/dic.212502] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 05/02/2017] [Indexed: 01/19/2023] Open
Abstract
Iron overload is a concern for patients who require repeated red-blood-cell transfusions due to conditions such as sickle cell disease, thalassemia, or myelodysplastic syndromes. The recommended treatment for removing excess iron in these patients is iron chelation therapy. Currently available iron chelators include deferoxamine, which is administered by injection, and deferasirox and deferiprone, both of which are administered orally. Adherence to iron chelator therapy is an important consideration and may be affected by side effects. A new formulation of deferasirox, a film-coated tablet (FCT), has the potential to improve adherence by offering greater flexibility in administration compared with the original formulation of deferasirox, a dispersible tablet (DT) for oral suspension. This review provides an overview of the currently available iron chelator formulations, with a focus on a comparison between deferasirox DT for oral suspension and deferasirox FCT. The new formulation may be associated with fewer side effects and has increased bioavailability. In addition, alternative strategies for iron chelation, such as combining two different iron chelators, will be discussed.
Collapse
|
33
|
Nairz M, Schroll A, Haschka D, Dichtl S, Tymoszuk P, Demetz E, Moser P, Haas H, Fang FC, Theurl I, Weiss G. Genetic and Dietary Iron Overload Differentially Affect the Course of Salmonella Typhimurium Infection. Front Cell Infect Microbiol 2017; 7:110. [PMID: 28443246 PMCID: PMC5387078 DOI: 10.3389/fcimb.2017.00110] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 03/20/2017] [Indexed: 12/14/2022] Open
Abstract
Genetic and dietary forms of iron overload have distinctive clinical and pathophysiological features. HFE-associated hereditary hemochromatosis is characterized by overwhelming intestinal iron absorption, parenchymal iron deposition, and macrophage iron depletion. In contrast, excessive dietary iron intake results in iron deposition in macrophages. However, the functional consequences of genetic and dietary iron overload for the control of microbes are incompletely understood. Using Hfe+/+ and Hfe-/- mice in combination with oral iron overload in a model of Salmonella enterica serovar Typhimurium infection, we found animals of either genotype to induce hepcidin antimicrobial peptide expression and hypoferremia following systemic infection in an Hfe-independent manner. As predicted, Hfe-/- mice, a model of hereditary hemochromatosis, displayed reduced spleen iron content, which translated into improved control of Salmonella replication. Salmonella adapted to the iron-poor microenvironment in the spleens of Hfe-/- mice by inducing the expression of its siderophore iron-uptake machinery. Dietary iron loading resulted in higher bacterial numbers in both WT and Hfe-/- mice, although Hfe deficiency still resulted in better pathogen control and improved survival. This suggests that Hfe deficiency may exert protective effects in addition to the control of iron availability for intracellular bacteria. Our data show that a dynamic adaptation of iron metabolism in both immune cells and microbes shapes the host-pathogen interaction in the setting of systemic Salmonella infection. Moreover, Hfe-associated iron overload and dietary iron excess result in different outcomes in infection, indicating that tissue and cellular iron distribution determines the susceptibility to infection with specific pathogens.
Collapse
Affiliation(s)
- Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of InnsbruckInnsbruck, Austria
| | - Andrea Schroll
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of InnsbruckInnsbruck, Austria
| | - David Haschka
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of InnsbruckInnsbruck, Austria
| | - Stefanie Dichtl
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of InnsbruckInnsbruck, Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of InnsbruckInnsbruck, Austria
| | - Egon Demetz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of InnsbruckInnsbruck, Austria
| | - Patrizia Moser
- Department of Pathology, Medical University of InnsbruckInnsbruck, Austria
| | - Hubertus Haas
- Division of Molecular Microbiology, Biocenter, Medical University of InnsbruckInnsbruck, Austria
| | - Ferric C Fang
- Department of Laboratory Medicine, University of WashingtonSeattle, WA, USA.,Department of Microbiology, University of WashingtonSeattle, WA, USA
| | - Igor Theurl
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of InnsbruckInnsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of InnsbruckInnsbruck, Austria
| |
Collapse
|
34
|
|
35
|
Sebastiani G, Wilkinson N, Pantopoulos K. Pharmacological Targeting of the Hepcidin/Ferroportin Axis. Front Pharmacol 2016; 7:160. [PMID: 27445804 PMCID: PMC4914558 DOI: 10.3389/fphar.2016.00160] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 05/31/2016] [Indexed: 12/22/2022] Open
Abstract
The iron regulatory hormone hepcidin limits iron fluxes to the bloodstream by promoting degradation of the iron exporter ferroportin in target cells. Hepcidin insufficiency causes hyperabsorption of dietary iron, hyperferremia and tissue iron overload, which are hallmarks of hereditary hemochromatosis. Similar responses are also observed in iron-loading anemias due to ineffective erythropoiesis (such as thalassemias, dyserythropoietic anemias and myelodysplastic syndromes) and in chronic liver diseases. On the other hand, excessive hepcidin expression inhibits dietary iron absorption and leads to hypoferremia and iron retention within tissue macrophages. This reduces iron availability for erythroblasts and contributes to the development of anemias with iron-restricted erythropoiesis (such as anemia of chronic disease and iron-refractory iron-deficiency anemia). Pharmacological targeting of the hepcidin/ferroportin axis may offer considerable therapeutic benefits by correcting iron traffic. This review summarizes the principles underlying the development of hepcidin-based therapies for the treatment of iron-related disorders, and discusses the emerging strategies for manipulating hepcidin pathways.
Collapse
Affiliation(s)
- Giada Sebastiani
- Department of Medicine, McGill UniversityMontreal, QC, Canada; Division of Gastroenterology, Royal Victoria HospitalMontreal, QC, Canada
| | - Nicole Wilkinson
- Lady Davis Institute for Medical Research, Jewish General Hospital Montreal, QC, Canada
| | - Kostas Pantopoulos
- Department of Medicine, McGill UniversityMontreal, QC, Canada; Lady Davis Institute for Medical Research, Jewish General HospitalMontreal, QC, Canada
| |
Collapse
|
36
|
Patchen B, Koppe T, Cheng A, Seo YA, Wessling-Resnick M, Fraenkel PG. Dietary supplementation with ipriflavone decreases hepatic iron stores in wild type mice. Blood Cells Mol Dis 2016; 60:36-43. [PMID: 27519943 DOI: 10.1016/j.bcmd.2016.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 05/04/2016] [Accepted: 05/04/2016] [Indexed: 01/19/2023]
Abstract
Hepcidin, a peptide produced in the liver, decreases intestinal iron absorption and macrophage iron release by causing degradation of the iron exporter, ferroportin. Because its levels are inappropriately low in patients with iron overload syndromes, hepcidin is a potential drug target. We previously conducted a chemical screen that revealed ipriflavone, an orally available small molecule, as a potent inducer of hepcidin expression. To evaluate ipriflavone's effect on iron homeostasis, we placed groups of 5-week old wild type or thalassemia intermedia (Hbb(Th3+/-)) mice on a soy-free, iron-sufficient diet, AIN-93G containing 220mg iron and 0-750mgipriflavone/kg of food for 50days. Ipriflavone 500mg/kg significantly reduced liver iron stores and intestinal ferroportin expression in WT mice, while increasing the ratio of hepcidin transcript levels to liver iron stores. Ipriflavone supplementation in Hbb(Th3+/-) mice failed to alleviate iron overload and was associated with a milder reduction in intestinal ferroportin and a failure to alter the ratio of hepcidin transcript levels to liver iron stores or splenic expression of the hepcidin-regulatory hormone, erythroferrone. These data suggest that dietary supplementation with ipriflavone alone would not be sufficient to treat iron overload in thalassemia intermedia.
Collapse
Affiliation(s)
- Bonnie Patchen
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, MA
| | - Tiago Koppe
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, MA
| | - Aaron Cheng
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, MA
| | - Young Ah Seo
- Departments of Genetics and Complex Diseases and Nutrition, Harvard School of Public Health, Boston, MA
| | - Marianne Wessling-Resnick
- Departments of Genetics and Complex Diseases and Nutrition, Harvard School of Public Health, Boston, MA
| | - Paula G Fraenkel
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, MA
| |
Collapse
|
37
|
Abstract
Iron is required for most forms of organisms, and it is the most essential element for the functions of many iron-containing proteins involved in oxygen transport, cellular respiration, DNA replication, and so on. Disorders of iron metabolism are associated with diverse diseases, including anemias (e.g., iron-deficiency anemia and anemia of chronic diseases) and iron overload diseases, such as hereditary hemochromatosis and β-thalassemia. Hepcidin (encoded by Hamp gene) is a peptide hormone synthesized by hepatocytes, and it plays an important role in regulating the systematic iron homeostasis. As the systemic iron regulator, hepcidin, not only controls dietary iron absorption and iron egress out of iron storage cells, but also induces iron redistribution in various organs. Deregulated hepcidin is often seen in a variety of iron-related diseases including anemias and iron overload disorders. In the case of iron overload disorders (e.g., hereditary hemochromatosis and β-thalassemia), hepatic hepcidin concentration is significantly reduced.Since hepcidin deregulation is responsible for iron disorder-associated diseases, the purpose of this review is to summarize the recent findings on therapeutics targeting hepcidin.Continuous efforts have been made to search for hepcidin mimics and chemical compounds that could be used to increase hepcidin level. Here, a literature search was conducted in PubMed, and research papers relevant to hepcidin regulation or hepcidin-centered therapeutic work were reviewed. On the basis of literature search, we recapitulated recent findings on therapeutic studies targeting hepcidin, including agonists and antagonists to modulate hepcidin expression or its downstream signaling. We also discussed the molecular mechanisms by which hepcidin level and iron metabolism are modulated.Elevating hepcidin concentration is an optimal strategy to ameliorate iron overload diseases, and also to relieve β-thalassemia phenotypes by improving ineffective erythropoiesis. Relative to the current conventional therapies, such as phlebotomy and blood transfusion, therapeutics targeting hepcidin would open a new avenue for treatment of iron-related diseases.
Collapse
Affiliation(s)
- Jing Liu
- From the State Key Laboratory of Environmental Chemistry and Ecotoxicology (JL, SL), Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China; Department of Medicine (BS), University of California, Los Angeles, CA; Department of Cardiovascular Disease (HY), Beijing Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing; and Gansu University of Traditional Chinese Medicine (HY), Lanzhou, China
| | | | | | | |
Collapse
|
38
|
Camacho A, Simão M, Ea HK, Cohen-Solal M, Richette P, Branco J, Cancela ML. Iron overload in a murine model of hereditary hemochromatosis is associated with accelerated progression of osteoarthritis under mechanical stress. Osteoarthritis Cartilage 2016; 24:494-502. [PMID: 26403062 DOI: 10.1016/j.joca.2015.09.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 08/21/2015] [Accepted: 09/11/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Hereditary hemochromatosis (HH) is a disease caused by mutations in the Hfe gene characterised by systemic iron overload and associated with an increased prevalence of osteoarthritis (OA) but the role of iron overload in the development of OA is still undefined. To further understand the molecular mechanisms involved we have used a murine model of HH and studied the progression of experimental OA under mechanical stress. DESIGN OA was surgically induced in the knee joints of 10-week-old C57BL6 (wild-type) mice and Hfe-KO mice. OA progression was assessed using histology, micro CT, gene expression and immunohistochemistry at 8 weeks after surgery. RESULTS Hfe-KO mice showed a systemic iron overload and an increased iron accumulation in the knee synovial membrane following surgery. The histological OA score was significantly higher in the Hfe-KO mice at 8 weeks after surgery. Micro CT study of the proximal tibia revealed increased subchondral bone volume and increased trabecular thickness. Gene expression and immunohistochemical analysis showed a significant increase in the expression of matrix metallopeptidase 3 (MMP-3) in the joints of Hfe-KO mice compared with control mice at 8 weeks after surgery. CONCLUSIONS HH was associated with an accelerated development of OA in mice. Our findings suggest that synovial iron overload has a definite role in the progression of HH-related OA.
Collapse
Affiliation(s)
- A Camacho
- Department of Orthopedics, Centro Hospitalar Lisboa Central, Lisboa, Portugal; PhD Program in Medicine, NOVA Medical School, University Nova de Lisboa, Lisbon, Portugal; Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal.
| | - M Simão
- Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal; PhD Program in Biomedical Sciences, University of Algarve, Faro, Portugal
| | - H-K Ea
- Inserm 1132, Hôpital Lariboisière, Paris, France; Université Paris Diderot, UFR médicale, Assistance Publique-Hôpitaux de Paris, Hôpital Lariboisière, Fédération de Rhumatologie, Paris, France
| | - M Cohen-Solal
- Inserm 1132, Hôpital Lariboisière, Paris, France; Université Paris Diderot, UFR médicale, Assistance Publique-Hôpitaux de Paris, Hôpital Lariboisière, Fédération de Rhumatologie, Paris, France
| | - P Richette
- Inserm 1132, Hôpital Lariboisière, Paris, France; Université Paris Diderot, UFR médicale, Assistance Publique-Hôpitaux de Paris, Hôpital Lariboisière, Fédération de Rhumatologie, Paris, France
| | - J Branco
- Department of Rheumatology, Hospital Egas Moniz, Centro Hospitalar Lisboa Ocidental EPE, Lisbon, Portugal; CEDOC - Chronic Diseases Research Center, NOVA Medical School, University Nova de Lisboa, Lisbon, Portugal
| | - M L Cancela
- Department of Biomedical Sciences and Medicine (DCBM), University of Algarve, Faro, Portugal; Centre of Marine Sciences (CCMAR), University of Algarve, Faro, Portugal
| |
Collapse
|
39
|
Abstract
In this issue of Blood, Kautz et al show that the ablation of the erythroid-derived factor erythroferrone (ERFE), which has been shown to be highly expressed in β-thalassemic mice, restores hepcidin levels and corrects iron overload. However, correction of hepcidin levels in those mice does not improve anemia of β-thalassemia.
Collapse
|
40
|
Lehtihet M, Bonde Y, Beckman L, Berinder K, Hoybye C, Rudling M, Sloan JH, Konrad RJ, Angelin B. Circulating Hepcidin-25 Is Reduced by Endogenous Estrogen in Humans. PLoS One 2016; 11:e0148802. [PMID: 26866603 PMCID: PMC4750915 DOI: 10.1371/journal.pone.0148802] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 12/17/2015] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Hepcidin reduces iron absorption by binding to the intestinal iron transporter ferroportin, thereby causing its degradation. Although short-term administration of testosterone or growth hormone (GH) has been reported to decrease circulating hepcidin levels, little is known about how hepcidin is influenced in human endocrine conditions associated with anemia. RESEARCH DESIGN AND METHODS We used a sensitive and specific dual-monoclonal antibody sandwich immunoassay to measure hepcidin-25 in patients (a) during initiation of in vitro fertilization when endogenous estrogens were elevated vs. suppressed, (b) with GH deficiency before and after 12 months substitution treatment, (c) with hyperthyroidism before and after normalization, and (d) with hyperprolactinemia before and after six months of treatment with a dopamine agonist. RESULTS In response to a marked stimulation of endogenous estrogen production, median hepcidin levels decreased from 4.85 to 1.43 ng/mL (p < 0.01). Hyperthyroidism, hyperprolactinemia, or GH substitution to GH-deficient patients did not influence serum hepcidin-25 levels. CONCLUSIONS In humans, gonadotropin-stimulated endogenous estrogen markedly decreases circulating hepcidin-25 levels. No clear and stable correlation between iron biomarkers and hepcidin-25 was seen before or after treatment of hyperthyroidism, hyperprolactinemia or growth hormone deficiency.
Collapse
Affiliation(s)
- Mikael Lehtihet
- Department of Endocrinology, Metabolism and Diabetes, Departments of Medicine and Molecular Medicine and Surgery, Karolinska Institutet at Karolinska University Hospital, S-141 86 Stockholm, Sweden
- * E-mail:
| | - Ylva Bonde
- Department of Endocrinology, Metabolism and Diabetes, Departments of Medicine and Molecular Medicine and Surgery, Karolinska Institutet at Karolinska University Hospital, S-141 86 Stockholm, Sweden
- Molecular Nutrition Unit, Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institutet at Karolinska University Hospital, S-141 86 Stockholm, Sweden
| | - Lena Beckman
- Department of Endocrinology, Metabolism and Diabetes, Departments of Medicine and Molecular Medicine and Surgery, Karolinska Institutet at Karolinska University Hospital, S-141 86 Stockholm, Sweden
- Molecular Nutrition Unit, Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institutet at Karolinska University Hospital, S-141 86 Stockholm, Sweden
| | - Katarina Berinder
- Department of Endocrinology, Metabolism and Diabetes, Departments of Medicine and Molecular Medicine and Surgery, Karolinska Institutet at Karolinska University Hospital, S-141 86 Stockholm, Sweden
| | - Charlotte Hoybye
- Department of Endocrinology, Metabolism and Diabetes, Departments of Medicine and Molecular Medicine and Surgery, Karolinska Institutet at Karolinska University Hospital, S-141 86 Stockholm, Sweden
| | - Mats Rudling
- Department of Endocrinology, Metabolism and Diabetes, Departments of Medicine and Molecular Medicine and Surgery, Karolinska Institutet at Karolinska University Hospital, S-141 86 Stockholm, Sweden
- Molecular Nutrition Unit, Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institutet at Karolinska University Hospital, S-141 86 Stockholm, Sweden
| | - John H. Sloan
- Lilly Research Laboratories, Eli Lilly & Co, Indianapolis, Indiana, United States of America
| | - Robert J. Konrad
- Lilly Research Laboratories, Eli Lilly & Co, Indianapolis, Indiana, United States of America
| | - Bo Angelin
- Department of Endocrinology, Metabolism and Diabetes, Departments of Medicine and Molecular Medicine and Surgery, Karolinska Institutet at Karolinska University Hospital, S-141 86 Stockholm, Sweden
- Molecular Nutrition Unit, Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institutet at Karolinska University Hospital, S-141 86 Stockholm, Sweden
| |
Collapse
|
41
|
Engert A, Balduini C, Brand A, Coiffier B, Cordonnier C, Döhner H, de Wit TD, Eichinger S, Fibbe W, Green T, de Haas F, Iolascon A, Jaffredo T, Rodeghiero F, Salles G, Schuringa JJ. The European Hematology Association Roadmap for European Hematology Research: a consensus document. Haematologica 2016; 101:115-208. [PMID: 26819058 PMCID: PMC4938336 DOI: 10.3324/haematol.2015.136739] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 01/27/2016] [Indexed: 01/28/2023] Open
Abstract
The European Hematology Association (EHA) Roadmap for European Hematology Research highlights major achievements in diagnosis and treatment of blood disorders and identifies the greatest unmet clinical and scientific needs in those areas to enable better funded, more focused European hematology research. Initiated by the EHA, around 300 experts contributed to the consensus document, which will help European policy makers, research funders, research organizations, researchers, and patient groups make better informed decisions on hematology research. It also aims to raise public awareness of the burden of blood disorders on European society, which purely in economic terms is estimated at €23 billion per year, a level of cost that is not matched in current European hematology research funding. In recent decades, hematology research has improved our fundamental understanding of the biology of blood disorders, and has improved diagnostics and treatments, sometimes in revolutionary ways. This progress highlights the potential of focused basic research programs such as this EHA Roadmap.The EHA Roadmap identifies nine 'sections' in hematology: normal hematopoiesis, malignant lymphoid and myeloid diseases, anemias and related diseases, platelet disorders, blood coagulation and hemostatic disorders, transfusion medicine, infections in hematology, and hematopoietic stem cell transplantation. These sections span 60 smaller groups of diseases or disorders.The EHA Roadmap identifies priorities and needs across the field of hematology, including those to develop targeted therapies based on genomic profiling and chemical biology, to eradicate minimal residual malignant disease, and to develop cellular immunotherapies, combination treatments, gene therapies, hematopoietic stem cell treatments, and treatments that are better tolerated by elderly patients.
Collapse
Affiliation(s)
| | | | - Anneke Brand
- Leids Universitair Medisch Centrum, Leiden, the Netherlands
| | | | | | | | | | | | - Willem Fibbe
- Leids Universitair Medisch Centrum, Leiden, the Netherlands
| | - Tony Green
- Cambridge Institute for Medical Research, United Kingdom
| | - Fleur de Haas
- European Hematology Association, The Hague, the Netherlands
| | | | | | | | - Gilles Salles
- Hospices Civils de Lyon/Université de Lyon, Pierre-Bénite, France
| | | |
Collapse
|
42
|
Barton JC, Edwards CQ, Acton RT. HFE gene: Structure, function, mutations, and associated iron abnormalities. Gene 2015; 574:179-92. [PMID: 26456104 PMCID: PMC6660136 DOI: 10.1016/j.gene.2015.10.009] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 10/04/2015] [Accepted: 10/06/2015] [Indexed: 01/05/2023]
Abstract
The hemochromatosis gene HFE was discovered in 1996, more than a century after clinical and pathologic manifestations of hemochromatosis were reported. Linked to the major histocompatibility complex (MHC) on chromosome 6p, HFE encodes the MHC class I-like protein HFE that binds beta-2 microglobulin. HFE influences iron absorption by modulating the expression of hepcidin, the main controller of iron metabolism. Common HFE mutations account for ~90% of hemochromatosis phenotypes in whites of western European descent. We review HFE mapping and cloning, structure, promoters and controllers, and coding region mutations, HFE protein structure, cell and tissue expression and function, mouse Hfe knockouts and knockins, and HFE mutations in other mammals with iron overload. We describe the pertinence of HFE and HFE to mechanisms of iron homeostasis, the origin and fixation of HFE polymorphisms in European and other populations, and the genetic and biochemical basis of HFE hemochromatosis and iron overload.
Collapse
Affiliation(s)
- James C Barton
- Southern Iron Disorders Center, Birmingham, AL, USA and Department of Medicine; University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Corwin Q Edwards
- Department of Medicine, Intermountain Medical Center and University of Utah, Salt Lake City, UT, USA.
| | - Ronald T Acton
- Southern Iron Disorders Center, Birmingham, AL, USA and Department of Medicine; Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
43
|
Lipopolysaccharides upregulate hepcidin in neuron via microglia and the IL-6/STAT3 signaling pathway. Mol Neurobiol 2015; 50:811-20. [PMID: 24659348 DOI: 10.1007/s12035-014-8671-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 02/28/2014] [Indexed: 12/13/2022]
Abstract
Neuroinflammation is closely related to brain iron homeostasis. Our previous study demonstrated that lipopolysaccharides (LPS) can regulate expression of iron-regulatory peptide hepcidin; however, the mechanism is undefined. Here, we demonstrated that intracerebroventricular injection of LPS in rat brain upregulated hepcidin and downregulated ferroportin 1 in the cortex and substantia nigra. LPS increased hepcidin expression in neurons only when they were co-cultured with BV-2 microglia, and the upregulation was suppressed by IL-6 neutralizing antibody in vitro. In addition, IL-6 but not IL-1α, IL-1β, or tumor necrosis factor-alpha increased hepcidin expression and signal transducer and activator of transcription 3 (STAT3) phosphorylation in cortical neurons and MES23.5 dopaminergic neurons. These effects were blocked by the STAT3 inhibitor, stattic. Our results show that neurons are the major source of increased hepcidin expression in response to LPS challenge but microglia play a key mediator role by releasing IL-6 and recruiting the STAT3 pathway. We conclude that LPS upregulates hepcidin expression in neurons via microglia and the IL-6/STAT3 signaling pathway.
Collapse
|
44
|
Chen B, Li GF, Shen Y, Huang XI, Xu YJ. Reducing iron accumulation: A potential approach for the prevention and treatment of postmenopausal osteoporosis. Exp Ther Med 2015; 10:7-11. [PMID: 26170904 DOI: 10.3892/etm.2015.2484] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 04/27/2015] [Indexed: 01/21/2023] Open
Abstract
Postmenopausal osteoporosis (PMOP) is a systemic bone metabolism disease, characterized by progressive bone loss following menopause and a subsequent increase in fracture risk. Estrogen deficiency as a result of menopause is known to increase bone resorption and accelerate bone loss. Furthermore, postmenopausal women may exhibit iron accumulation, in addition to estrogen deficiency. Elevated iron levels are a risk factor for PMOP in postmenopausal women, and reducing the iron overload has been demonstrated to benefit bone cell metabolism in vitro and improve the bone in vivo by normalizing osteoclastic bone resorption and formation. The identification of hepcidin was a key development in the field of iron metabolism in the previous decade. We hypothesize that hepcidin may aid in the prevention and treatment of PMOP due to its capacity to control body iron stores and its intrinsic effects on osteoblast function. The aim of the current review was to highlight the role of iron accumulation in the pathogenesis of PMOP and to evaluate the possible use of hepcidin as a potential therapy for this condition.
Collapse
Affiliation(s)
- Bin Chen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Guang-Fei Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Ying Shen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - X I Huang
- Division of Rheumatology, NYU Hospital for Joint Diseases, New York, NY 10003, USA
| | - You-Jia Xu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| |
Collapse
|
45
|
Saliou P, Le Gac G, Mérour MC, Tripogney C, Chanu B, Gourlaouen I, Nousbaum JB, Férec C, Scotet V. HFE hemochromatosis: influence of dietary iron intake on the iron overload of C282Y homozygous patients. Ann Hematol 2015; 94:1225-7. [DOI: 10.1007/s00277-015-2343-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/16/2015] [Indexed: 12/13/2022]
|
46
|
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: 29] [Impact Index Per Article: 3.2] [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
|
47
|
Bulycheva E, Rauner M, Medyouf H, Theurl I, Bornhäuser M, Hofbauer LC, Platzbecker U. Myelodysplasia is in the niche: novel concepts and emerging therapies. Leukemia 2014; 29:259-68. [PMID: 25394715 PMCID: PMC4320287 DOI: 10.1038/leu.2014.325] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 09/25/2014] [Indexed: 12/28/2022]
Abstract
Myelodysplastic syndromes (MDSs) represent clonal disorders mainly of the elderly that are characterized by ineffective hematopoiesis and an increased risk of transformation into acute myeloid leukemia. The pathogenesis of MDS is thought to evolve from accumulation and selection of specific genetic or epigenetic events. Emerging evidence indicates that MDS is not solely a hematopoietic disease but rather affects the entire bone marrow microenvironment, including bone metabolism. Many of these cells, in particular mesenchymal stem and progenitor cells (MSPCs) and osteoblasts, express a number of adhesion molecules and secreted factors that regulate blood regeneration throughout life by contributing to hematopoietic stem and progenitor cell (HSPC) maintenance, self-renewal and differentiation. Several endocrine factors, such as erythropoietin, parathyroid hormone and estrogens, as well as deranged iron metabolism modulate these processes. Thus, interactions between MSPC and HSPC contribute to the pathogenesis of MDS and associated pathologies. A detailed understanding of these mechanisms may help to define novel targets for diagnosis and possibly therapy. In this review, we will discuss the scientific rationale of ‘osteohematology' as an emerging research field in MDS and outline clinical implications.
Collapse
Affiliation(s)
- E Bulycheva
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl-Gustav-Carus, Technische Universität, Dresden, Germany
| | - M Rauner
- Medizinische Klinik und Poliklinik III, Universitätsklinikum Carl-Gustav-Carus, Technische Universität, Dresden, Germany
| | - H Medyouf
- Georg-Speyer-Haus, Institut for Tumor Biology and Experimental Therapy, 60596, Frankfurt am Main, Germany
| | - I Theurl
- Department of Internal Medicine VI, Medical University of Innsbruck, Innsbruck, Austria
| | - M Bornhäuser
- 1] Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl-Gustav-Carus, Technische Universität, Dresden, Germany [2] Center for Regenerative Therapies Dresden, Technical University, Dresden, Germany
| | - L C Hofbauer
- 1] Medizinische Klinik und Poliklinik III, Universitätsklinikum Carl-Gustav-Carus, Technische Universität, Dresden, Germany [2] Center for Regenerative Therapies Dresden, Technical University, Dresden, Germany
| | - U Platzbecker
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl-Gustav-Carus, Technische Universität, Dresden, Germany
| |
Collapse
|
48
|
Abstract
Hepcidin, the liver-produced peptide hormone, is a principal regulator of iron homeostasis. Abnormal hepcidin production has emerged as a causative factor in several common iron disorders. Hepcidin insufficiency results in iron overload in hereditary hemochromatosis and iron-loading anemias, whereas hepcidin excess causes or contributes to the development of iron-restricted anemias in inflammatory diseases, infections, some cancers and chronic kidney disease. Not surprisingly, hepcidin and related pathways have become the target for the development of novel therapeutics for iron disorders. In this review, we will summarize the strategies and development programs that have been devised for agonizing or antagonizing hepcidin and its receptor ferroportin.
Collapse
|
49
|
Gaun V, Patchen B, Volovetz J, Zhen AW, Andreev A, Pollastri MP, Fraenkel PG. A chemical screen identifies small molecules that regulate hepcidin expression. Blood Cells Mol Dis 2014; 53:231-40. [PMID: 24998898 DOI: 10.1016/j.bcmd.2014.06.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 06/09/2014] [Accepted: 06/09/2014] [Indexed: 12/19/2022]
Abstract
Hepcidin, a peptide hormone produced in the liver, decreases intestinal iron absorption and macrophage iron release via effects on ferroportin. Bone morphogenic protein and Stat3 signaling regulate Hepcidin's transcription. Hepcidin is a potential drug target for patients with iron overload syndromes because its levels are inappropriately low in these individuals. To generate a tool for identifying small molecules that modulate Hepcidin expression, we stably transfected human hepatocytes (HepG2) cells with a reporter construct containing 2.7kb of the human Hepcidin promoter upstream of a firefly reporter gene. We used high throughput methods to screen 10,169 chemicals in duplicate for their effect on Hepcidin expression and cell viability. Regulators were identified as chemicals that caused a change >3 standard deviations above or >1 standard deviation below the mean of the other chemicals (z-score >3 or <1), while not adversely affecting cell viability, quantified by fluorescence assay. Following validation assays, we identified 16 chemicals in a broad range of functional classes that promote Hepcidin expression. All of the chemicals identified increased expression of bone morphogenic protein-dependent and/or Stat3-dependent genes, however none of them strongly increased phosphorylation of Smad1,5,8 or Stat3.
Collapse
Affiliation(s)
- Vera Gaun
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, United States; Department of Medicine, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Bonnie Patchen
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, United States; Department of Medicine, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Josephine Volovetz
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, United States; Department of Medicine, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Aileen W Zhen
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, United States; Department of Medicine, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Aleksandr Andreev
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, United States; Department of Medicine, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Michael P Pollastri
- Department of Chemistry and Chemical Biology, Northeastern University, 417 Egan Research Center, 120 Forsyth Street, Boston, MA 02115, United States
| | - Paula G Fraenkel
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, United States; Department of Medicine, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States.
| |
Collapse
|
50
|
Poli M, Asperti M, Ruzzenenti P, Regoni M, Arosio P. Hepcidin antagonists for potential treatments of disorders with hepcidin excess. Front Pharmacol 2014; 5:86. [PMID: 24808863 PMCID: PMC4009444 DOI: 10.3389/fphar.2014.00086] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/07/2014] [Indexed: 12/20/2022] Open
Abstract
The discovery of hepcidin clarified the basic mechanism of the control of systemic iron homeostasis. Hepcidin is mainly produced by the liver as a propeptide and processed by furin into the mature active peptide. Hepcidin binds ferroportin, the only cellular iron exporter, causing the internalization and degradation of both. Thus hepcidin blocks iron export from the key cells for dietary iron absorption (enterocytes), recycling of hemoglobin iron (the macrophages) and the release of storage iron from hepatocytes, resulting in the reduction of systemic iron availability. The BMP/HJV/SMAD pathway is the major regulator of hepcidin expression that responds to iron status. Also inflammation stimulates hepcidin via the IL6/STAT3 pathway with a support of an active BMP/HJV/SMAD pathway. In some pathological conditions hepcidin level is inadequately elevated and reduces iron availability in the body, resulting in anemia. These conditions occur in the genetic iron refractory iron deficiency anemia and the common anemia of chronic disease (ACD) or anemia of inflammation. Currently, there is no definite treatment for ACD. Erythropoiesis-stimulating agents and intravenous iron have been proposed in some cases but they are scarcely effective and may have adverse effects. Alternative approaches aimed to a pharmacological control of hepcidin expression have been attempted, targeting different regulatory steps. They include hepcidin sequestering agents (antibodies, anticalins, and aptamers), inhibitors of BMP/SMAD or of IL6/STAT3 pathway or of hepcidin transduction (siRNA/shRNA) or ferroportin stabilizers. In this review we summarized the biochemical interactions of the proteins involved in the BMP/HJV/SMAD pathway and its natural inhibitors, the murine and rat models with high hepcidin levels currently available and finally the progresses in the development of hepcidin antagonists, with particular attention to the role of heparins and heparin sulfate proteoglycans in hepcidin expression and modulation of the BMP6/SMAD pathway.
Collapse
Affiliation(s)
- Maura Poli
- Molecular Biology Laboratory, Department of Molecular and Translational Medicine, University of Brescia Brescia, Italy
| | - Michela Asperti
- Molecular Biology Laboratory, Department of Molecular and Translational Medicine, University of Brescia Brescia, Italy
| | - Paola Ruzzenenti
- Molecular Biology Laboratory, Department of Molecular and Translational Medicine, University of Brescia Brescia, Italy
| | - Maria Regoni
- Molecular Biology Laboratory, Department of Molecular and Translational Medicine, University of Brescia Brescia, Italy
| | - Paolo Arosio
- Molecular Biology Laboratory, Department of Molecular and Translational Medicine, University of Brescia Brescia, Italy
| |
Collapse
|