The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters.

Similar documents
Iron age: novel targets for iron overload

HHS Public Access Author manuscript Ann N Y Acad Sci. Author manuscript; available in PMC 2017 March 01.

Fourth European Symposium on Rare Anaemias. Vita-Salute University - San Raffaele Scientific Institute, Milano

2011 ASH Annual Meeting Targeting the Hepcidin Pathway with RNAi Therapeutics for the Treatment of Anemia. December 12, 2011

Discovery. Hepcidin Today. Hepcidin: discovery June 2000: Man: Plasma ultrafiltrate Liver Expressed Antimicrobial Peptide

Iron and hepcidin: a story of recycling and balance

The Hepcidin-Ferroportin System as a Therapeutic Target in Anemias and Iron Overload Disorders

NIH Public Access Author Manuscript Nat Genet. Author manuscript; available in PMC 2011 May 30.

Iron: a global issue in hematology. Clara Camaschella, MD

BMP6 treatment compensates for the molecular defect and ameliorates hemochromatosis in Hfe knockout mice

In adults, the predominant Hb (HbA) molecule has four chains: two α and two β chains. In thalassemias, the synthesis of either the α or the β chains

THE ROLE OF HEMOJUVELIN IN IRON DEFICIENCY AND OVERLOAD

Corporate Medical Policy

Iron overload in transfusion-dependent survivors of hemoglobin Bart s hydrops fetalis

Report of Beta Thalassemia in Newar Ethnicity

Reducing TMPRSS6 ameliorates hemochromatosis and β-thalassemia in mice

Unraveling Mechanisms Regulating Systemic Iron Homeostasis

Microcytic Hypochromic Anemia An Approach to Diagnosis

Oral Exfoliative Cytology In Beta Thalassaemia Patients Undergoing Repeated Blood Transfusions

Review Article Crosstalk between Iron Metabolism and Erythropoiesis

Year 2003 Paper two: Questions supplied by Tricia

Into the matrix: regulation of the iron regulatory hormone hepcidin by matriptase-2

Is the acronym IRIDA acceptable for slow responders to iron in the presence of TMPRSS6 mutations?

Part I. Pathophysiology and management of Thalassemia Intermedia. M. Domenica Cappellini Fondazione IRCCS Policlinico University of Milan

Biochimica et Biophysica Acta

Role of hepcidin in the pathophysiology and diagnosis of anemia

Red cell disorder. Dr. Ahmed Hasan

Thalassemia Maria Luz Uy del Rosario, M.D.

CLINICAL AND LABORATORY PATTERNS OF HEREDITARY HAEMOLYTIC ANEMIAS IN CHILDREN FROM CENTRAL REGION OF ROMANIA

Disclosures and Funding

Relationship Between Serum Hepcidin and Ferritin Levels in Patients With Thalassemia Major and Intermedia in Southern Iran

Next-Generation Biomarkers for Iron Status

RBCs Disorders 2. Dr. Nabila Hamdi MD, PhD

Metabolismo del ferro in condizioni normali e patologiche

Innovative therapies for ß-hemoglobinopathies

Management of anemia in CKD

Genetics of Thalassemia

A group of inherited disorders characterized by reduced or absent amounts of hemoglobin, the oxygencarrying protein inside the red blood cells.

3) The sheer number of and inconsistency between different animal models used make the paper difficult to follow and may impact data interpretation:

Role of Serum Hepcidin levels in the Diagnosis of Iron Deficiency Anemia in Children in Saudi Arabia

INTERELATIONSHIP BETWEEN IDA AND VITAMIN D DEFICIENCY IS NOW ESTABLISHED

Pedro A. Martinez, PhD December 7 th, 2015

Guideline developed by Shelley Crary, MD, MS,* in collaboration with the ANGELS team. Last reviewed by Shelley Crary, MD, MS, January 19, 2017.

Systemic iron homeostasis is dependent on the hepatic

Characterization of iron metabolism and erythropoiesis in erythrocyte membrane defects and thalassemia traits

Hepcidin as a therapeutic tool to limit iron overload and improve anemia in β-thalassemic mice

Hemosiderin. Livia Vida 2018

Iron deficiency anemia and porphyrias

BMP6 Treatment Compensates for the Molecular Defect and Ameliorates Hemochromatosis in Hfe Knockout Mice

THALASSEMIA AND COMPREHENSIVE CARE

The Nucleated Red Blood Cell (NRBC) Count in Thalassaemia Syndromes Paolo Danise and Giovanni Amendola

Recent Advances in Erythroid Iron Homeostasis: Implications for Pathophysiology of Microcytic Anemias

Ten years of Highlights from EHA: Red cells and Iron

Transferrin Receptors and Hematopoiesis: Review

Anemia s. Troy Lund MSMS PhD MD

Thalassemias:general aspects and molecular pathology

Pedro A. Martinez, PhD June 10 th, 2016

Iron-Sensing Proteins that Regulate Hepcidin and Enteric Iron Absorption

The Interaction of Alcohol and Iron-Overload in the in-vivo Regulation of Iron Responsive Genes

RBCs Disorders 2. Dr. Nabila Hamdi MD, PhD

YEAR III Pharm.D Dr. V. Chitra

Hepcidin and iron regulation, 10 years later

Manipulation of the hepcidin pathway for therapeutic purposes

Evidence for the Safety of Fortifying Flour with Iron in the Presence of Thalassemia and Other Blood Disorders

Iron and hepcidin: a story of recycling and balance

Extra Notes 3. Warm. In the core (center) of the body, where the temperature is 37 C.

Short-term administration of JAK2 inhibitors reduces splenomegaly in mouse models of b-thalassemia intermedia and major

Hematopoiesis, The hematopoietic machinery requires a constant supply iron, vitamin B 12, and folic acid.

Anemia in -thalassemia patients targets hepatic hepcidin transcript levels independently of iron metabolism genes controlling hepcidin expression

Iron Overload Disorders and Iron Chelation Therapy

Corporate Medical Policy Genetic Testing for Hereditary Hemochromatosis

6.1 Extended family screening

Hereditary Hemochromatosis: What Have We Learnt from Population Studies Professor John K. Olynyk

Faculty of Medicine Dr. Tariq Aladily

Nuovi Approcci alla Ferrochelazione

SICKLE CELL DISEASE. Dr. MUBARAK ABDELRAHMAN MD PEDIATRICS AND CHILD HEALTH. Assistant Professor FACULTY OF MEDICINE -JAZAN

Clinical Characteristics of Pediatric Thalassemia in Korea: A Single Institute Experience

Serum soluble transferrin receptor in hypochromic microcytic anaemia

New therapeutic targets in transfusion-dependent and -independent thalassemia

Ferrata Storti Foundation

Plenary Paper. Modified activin receptor IIB ligand trap mitigates ineffective erythropoiesis and disease complications in murine b-thalassemia

Managing peri-operative anaemiathe Papworth way. Dr Andrew A Klein Royal Papworth Hospital Cambridge UK

ANEMIA & HEMODIALYSIS

4 Jumana Jihad Dr. Ahmad Mansour Dr. Ahmad Mansour

Quantification of serum hepcidin as a potential biomarker in diagnosis of iron deficiency anaemia

I. Definitions. V. Evaluation A. History B. Physical Exam C. Laboratory evaluation D. Bone marrow examination E. Specialty referrals

Introduction 5/2/2013 IRON RELATED GENES AND OXIDATIVE STRESS IN NON- ALCOHOLIC STEATOHEPATITIS. Iron Physiology. Iron Physiology

HFE mutations in idiopathic erythrocytosis

Hepcidin Levels and Iron Status in β- Thalassemia Major Patients with Hepatitis C Virus Infection

Iron-refractory iron deficiency anemia: new molecular mechanisms

Utilizing Sysmex RET He to Evaluate Anemia in Cancer Patients

Réunion annuelle de pathologie digestive. Iron Metabolism

Assessing Iron Deficiency in Adults. Chris Theberge. Iron (Fe) deficiency remains as one of the major global public health problems for

Hypoxia-inducible factor regulates hepcidin via erythropoietin-induced erythropoiesis

Novità nelle MDS. Matteo G Della Porta. Cancer Center IRCCS Humanitas Research Hospital & Humanitas University Rozzano Milano, Italy

Iron Metabolism in Thalassemia and Sickle Cell Disease.

Drugs Used in Anemia

Chapter 2. ERYTHROPOIESIS and ANEMIA

The Regulation of Hepcidin and Its Effects on Systemic and Cellular Iron Metabolism

How to Write a Life Care Plan for a Child with Hemoglobinopathy

Transcription:

Combination therapy with a Tmprss6 RNAi-therapeutic and the oral iron chelator deferiprone additively diminishes secondary iron overload in a mouse model of β-thalassemia intermedia The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters. Citation Published Version Accessed Citable Link Terms of Use Schmidt, Paul J, Tim Racie, Mark Westerman, Kevin Fitzgerald, James S Butler, and Mark D Fleming. 2015. Combination therapy with a Tmprss6 RNAi-therapeutic and the oral iron chelator deferiprone additively diminishes secondary iron overload in a mouse model of β-thalassemia intermedia. American Journal of Hematology 90 (4): 310-313. doi:10.1002/ajh.23934. http://dx.doi.org/10.1002/ajh.23934. doi:10.1002/ajh.23934 July 4, 2018 2:39:34 PM EDT http://nrs.harvard.edu/urn-3:hul.instrepos:15034885 This article was downloaded from Harvard University's DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http://nrs.harvard.edu/urn-3:hul.instrepos:dash.current.terms-ofuse#laa (Article begins on next page)

Combination therapy with a Tmprss6 RNAi-therapeutic and the oral iron chelator deferiprone additively diminishes secondary iron overload in a mouse model of b-thalassemia intermedia AJH Paul J. Schmidt, 1 * Tim Racie, 2 Mark Westerman, 3 Kevin Fitzgerald, 2 James S. Butler, 2 and Mark D. Fleming 1 b-thalassemias result from diminished b-globin synthesis and are associated with ineffective erythropoiesis and secondary iron overload caused by inappropriately low levels of the iron regulatory hormone hepcidin. The serine protease TMPRSS6 attenuates hepcidin production in response to iron stores. Hepcidin induction reduces iron overload and mitigates anemia in murine models of b-thalassemia intermedia. To further interrogate the efficacy of an RNAi-therapeutic downregulating Tmprss6, b-thalassemic Hbb th3/1 animals on an iron replete, an iron deficient, or an iron replete diet also containing the iron chelator deferiprone were treated with Tmprss6 sirna. We demonstrate that the total body iron burden is markedly improved in Hbb th3/1 animals treated with sirna and chelated with oral deferiprone, representing a significant improvement compared to either compound alone. These data indicate that sirna suppression of Tmprss6, in conjunction with oral iron chelation therapy, may prove superior for treatment of anemia and secondary iron loading seen in b-thalassemia intermedia. Am. J. Hematol. 90:310 313, 2015. VC 2015 The Authors. American Journal of Hematology Published by Wiley Periodicals, Inc. Introduction The thalassemias result from a quantitative defect in the production of a- or b-globin chains. b-thalassemia is caused by the partial or complete lack of b-globin synthesis resulting in a variably severe anemia and/or red blood cell abnormalities. In its most severe form, b-thalassemia major, affected patients have profound secondary iron overload largely due the requirement for chronic red blood cell transfusional support. Patients with b-thalassemia intermedia, a less severe form, do not require chronic transfusions (i.e., nontransfusion dependent thalassemia), but still develop iron overload as a consequence of chronic suppression of the synthesis of the iron regulatory hormone hepcidin by ineffective erythropoiesis [1,2]. In both cases, complications of iron overload, including liver disease, cardiomyopathy, and endocrinopathies, are the most significant contributors to morbidity and mortality in these individuals. A mouse model of b-thalassemia intermedia, Hbb th3/1, has a heterozygous deletion of both the b-minor and b-major hemoglobin genes [3]. Similar to their human counterparts, these animals have moderately severe, transfusion-independent hypochromic, microcytic anemia associated and ineffective erythropoeisis resulting in splenomegaly, hepcidin suppression, and secondary iron overload [4 7]. TMPRSS6 (matriptase-2) is a serine protease expressed predominantly in the liver that down-regulates hepcidin transcription by inhibiting an iron-responsive bone morphogenetic protein-mothers against decapentaplegic (BMP-SMAD) signaling pathway [8]. Germline mutations in TMPRSS6 in both mice and humans result in iron-refractory iron deficiency anemia (IRIDA) due to chronic inappropriate overexpression of hepcidin relative to liver iron stores [9]. Modulating hepcidin expression by targeting the BMP-SMAD signaling pathway in hepatocytes may be an effective modality for treating iron disorders, particularly those associated with a relative hepcidin deficiency or excess. For example, deletion of Tmprss6 in the Hfe 2/2 mouse model of hereditary hemochromatosis results in elevated expression of hepcidin, leading to an amelioration of the iron overload phenotype [10]. Likewise, induction of hepcidin or iron deficiency in b-thalassemic animals using several different methods not only mitigates the iron overload, but also ameliorates the anemia through an incompletely understood mechanism [11 13]. Most recently, we, and others, have demonstrated that suppression of Tmprss6 mrna expression with lipid nanoparticle (LNP)-formulated sirnas or antisense oligonucleotides (ASOs) increases hepcidin, leading to diminished iron uptake and recycling and improved erythropoiesis and anemia in b-thalassemic This is an open access article under the terms of the Creative Commons Attribution NonCommercial License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes. Additional Supporting Information may be found in the online version of this article. 1 Department of Pathology, Boston Children s Hospital and Harvard Medical School, Boston, Massachusetts; 2 Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts; 3 Intrinsic Lifesciences, Inc., LaJolla, California Conflict of interest: TR, JSB, and KF are employees and/or stockowners of Alnylam Pharmaceuticals; MDF receives research funding from Alnylam Pharmaceuticals; MW is a founder, chief executive officer, and stock owner in Intrinsic LifeSciences; PJS has no relevant conflict of interest. *Correspondence to: Paul J. Schmidt, Ph.D. Department of Pathology, Boston Children s Hospital, Enders Research Building, Room 1110, 320 Longwood Ave., Boston, MA 02115. E-mail: paul.schmidt@childrens.harvard.edu Contract grant sponsor: Alnylam Pharmaceuticals. Received for publication: 22 December 2014; Accepted: 24 December 2014 Am. J. Hematol. 90:310 313, 2015. Published online: 2 January 2015 in Wiley Online Library (wileyonlinelibrary.com). DOI: 10.1002/ajh.23934 VC 2015 The Authors. American Journal of Hematology Published by Wiley Periodicals, Inc. 310 American Journal of Hematology, Vol. 90, No. 4, April 2015 doi:10.1002/ajh.23934

Tmprss6 sirna and Deferiprone Combination Therapy in Murine b-thalassemia Intermedia Figure 1. Iron parameters in Hbb th3/1 treated with LNP-Tmprss6 sirna and dietary iron deficiency or chelation. Six-week old female animals were injected with LNP-siRNA formulations (1 mg/kg) every other week for 6 weeks and then killed 14 days after the final injection. On the day of the first injection, animals were placed on a control (50 ppm iron), iron deficient (3 5 ppm iron), or control diet supplemented with deferiprone (50 ppm iron, 0.125% deferiprone). A: Liver Tmprss6 mrna assessed by quantitative real-time PCR and normalized to b-actin (Actb). B: ELISA serum hepcidin analysis (ng/ml). C: Serum transferrin saturation (%). D: Serum iron (lg/dl). E: Nonheme liver iron (lg/g). F: 3,3 0 -diaminobenzidine (DAB)-enhanced iron stain of liver sections. G: Total spleen iron (lg). Error bars are 6SEM. (n 5 426 for each group) Student s t-test P-values: ****P < 0.001, ***P < 0.005, **P <0.01, and *P < 0.05. [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.] animals [14,15]. These preclinical data suggest that modulation of hepcidin expression could be a substitute for or an adjunct to iron chelation therapy in patients with thalassemia. To further investigate this possibility, we treated Hbb th3/1 animals with an LNP-formulated RNAi therapeutic targeting Tmprss6 in combination with deferiprone, an orally bioavailable iron chelator [16]. We found that sirna combined with oral deferiprone therapy is superior to monotherapy with dietary iron deficiency, iron chelator, or Tmprss6 sirna for the reduction of hepatic iron stores, while still maintaining some of the beneficial effects on anemia. Methods Other than as noted below, all mouse strains and methods are identical to those previously described [14]. Mice were born and housed in the barrier facility at Boston Children s Hospital under approved protocols, and were raised on Prolab RMH 3000 diet (Lab Diet, 380 ppm iron) until 6 weeks of age, when the diet was switched to an iron replete (50 ppm) control diet, iron deficient (3 5 ppm) diet, or an iron replete (50ppm) diet with 0.125% (1.25 g/kg) deferiprone (Harlan diet # s, TD120277, TD120276, and TD120278, respectively). Serum hepcidin levels were measured with a novel murine-specific competitive ELISA [17]. Results LNP-Tmprss6 sirna plus oral deferiprone therapy diminishes secondary iron overload in murine b-thalassemia intermedia We determined whether modulating systemic iron stores by dietary restriction or pharmacological chelation could add to the beneficial effects of Tmprss6 sirna therapy on Hbb th3/1 anemia and iron phenotypes. To do so, we treated wild type and Hbb th3/1 animals with biweekly LNP-Tmprss6 sirna injections in conjunction with an iron doi:10.1002/ajh.23934 American Journal of Hematology, Vol. 90, No. 4, April 2015 311

Schmidt et al. RESEARCH ARTICLE Figure 2. Erythropoietic parameters in Hbb th3/1 animals treated with LNP-Tmprss6 sirna and dietary iron deficiency or chelation. Animals were treated as in Figure 1. A: Hemoglobin (g/dl). B: Red blood cell count. C: Mean red blood cell volume (fl). D: RBC morphology on Wright s stained peripheral blood smears. E: Fraction of reticulocytes (%). F: Absolute reticulocyte count. G: Serum erythropoietin levels (pg/ml). H: Spleen weight (g). Error bars are 6SEM. (n 5 426 for each group) Student s t-test P-values: ****P < 0.001, ***P < 0.005, **P <0.01, and *P < 0.05. [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.] replete synthetic control diet (50 ppm iron), a low iron diet (3 5 ppm iron), or an iron replete diet containing the orally bioavailable chelator deferiprone (50 ppm iron, 0.125% wt/wt chelator). In wild-type animals, we observed qualitatively similar effects on gene expression and iron metabolism, including Tmprss6 suppression and the induction of systemic iron deficiency, as we did previously (Supporting Information Fig. 1) [14]. As expected, LNP-Tmprss6 treatment of Hbb th3/1 animals on all diets suppressed Tmprss6 mrna expression and up-regulated serum hepcidin (Fig. 1A,B). Furthermore, treated animals had decreased transferrin saturation and serum iron (Fig. 1C,D), with the largest and most significant changes seen in animals on the diet containing deferiprone. When compared to control sirna-treated animals on the same diet, nonheme liver iron was diminished only in animals treated with deferiprone (Fig. 1E), possibly due to the relatively low iron (50 ppm), which is only approximately two times the recommended adult murine daily requirement, in the formulated test diets compared to conventional mouse chow (380 ppm iron) that we employed previously [14]. In comparison to the control diet, LNP-Tmprss6 treated animals on either an iron deficient diet or diet with added chelator had lower liver iron levels, with the latter having a greater effect. In Hbb th3/1 animals, this change in liver iron can be visualized on DAB-enhanced Perls stained tissue sections as decreased periportal hepatocellular iron (Fig. 1F). Total spleen iron (Fig. 1G) decreased significantly in treated Hbb th3/1 animals on the iron replete and low iron diets, but only trended downward in animals fed chlelator-enhanced chow, possibly due to effects on erythroid iron utilization or RBC turnover. LNP-Tmprss6 sirna plus oral deferiprone therapy improves erythropoiesis and splenomegaly in murine b-thalassemia intermedia In earlier studies, we observed that Tmprss6 sirna treatment induces a mild microcytic, hypochromic anemia in wild-type animals [14]. We observed the same phenotype here, regardless of the diet employed (Supporting Information Fig. 2). However, in slight contrast to published studies [11], treatment of Hbb th3/1 mice with an 312 American Journal of Hematology, Vol. 90, No. 4, April 2015 doi:10.1002/ajh.23934

Tmprss6 sirna and Deferiprone Combination Therapy in Murine b-thalassemia Intermedia iron deficient diet alone did not improve their anemia. Furthermore, with 50 ppm iron in the diet, treatment of Hbb th3/1 mice with Tmprss6 sirna improved the anemia only when combined with deferiprone therapy (Fig. 2A). Both of these results are also likely attributable to the relatively low baseline levels of iron in the control and chelator-enhanced test diets compared to historical controls. This caveat notwithstanding, other RBC and erythropoietic parameters, characteristic of a response to iron restriction, are seen in LNP- Tmprss6 treated animals on each of the test diets. All experimental sirna-treated thalassemic groups exhibited an increase in RBC numbers (Fig. 2B) associated with a decreased mean cell volume (MCV, Fig. 2C). These changes are readily appreciated on peripheral blood smears, which demonstrate replacement of the anisopoikilocytosis of the baseline phenotype with a relatively monotonous population of evenly sized and shaped hypochromic microcytes (Fig. 2D). Despite the relative beneficial effect of the addition of deferiprone to LNP-Tmprss6 therapy on the Hbb th3/1 anemia, some parameters suggest that the addition of chelator at the dose employed mitigates some of the other positive effects of hepcidin-induced iron restriction. For example, while the fraction of RBCs that are reticulocytes (Fig. 2E) is decreased, there is no decrease in the absolute number of reticulocytes (Fig. 2F). Furthermore, although the decrease in serum erythropoietin is most significant in deferiprone-treated animals, changes in spleen size are not as profound as in animals on an iron deficient diet alone (Fig. 2G,H). Finally, consistent with the reduction in splenomegaly, there was partial restoration of splenic architecture in LNP-Tmprss6 treated animals also maintained on deferiprone (Supporting Information Fig. 3) Discussion Taken together, these data demonstrate that combined LNP-Tmprss6 sirna and oral deferiprone chelator therapy is superior in reducing hepatic iron stores compared to dietary iron deficiency with or without LNP-Tmprss6 treatment, or chelator therapy alone. However, in this murine model and at these sirna and chelator doses, the addition of chelation appears to partially attenuate several of the ancillary beneficial effects, including amelioration of the anemia and splenomegaly, observed in previous studies. Although this could be a direct effect of the chelator on erythropoiesis, it is likely that many of the deviations are due to the more iron-restricted baseline dietary conditions under which the current experiments were conducted. For example, it is possible that the relatively high concentrations of iron found in most mouse chows are inhibitory to thalassemic erythropoiesis, thus worsening the baseline phenotype and exaggerating any positive effects of hepcidin induction therapy. On the other hand, there is also evidence that too much hepcidin and iron restriction is equally detrimental to thalassemic hematological phenotypes [11]. For this reason, it will be prudent ultimately, if this is to be translated to the clinic, to titrate both modalities to each patient to achieve individualized chelation and hematological effects. In addition to the beneficial effects on the anemia, combination therapy with long-acting hepcidin induction therapy, such as TMPRSS6 sirna, has several distinct theoretical advantages over chelation alone. First, hepcidin-induced down-regulation of ferroportin sequesters iron in macrophages may limit its toxicity by reducing plasma free (e.g., nontransferrin bound) iron, particularly when plasma chelators are at their nadir between doses. Second, the effect of hepcidin on ferroportin expression in duodenal enterocytes responsible for iron absorption limits the ongoing absorption of iron an effect not achieved by chelators alone. This mechanism of action will likely be critical in individuals with nontransfusional iron overload, where intestinal absorption due to hepcidin suppression is the key pathogenetic mechanism. Nevertheless, even in the transfusion-dependent patient, it has been shown that hepcidin levels, which are ordinarily increased, vary pre- and post-transfusion; hepcidin levels increase after transfusion, presumably due to the suppression of hepcidin synthesis by anemia, hypoxia, and ineffective hematopoiesis that increase as the hemoglobin drops in the interval between transfusions [2,18]. Thus, while the physiological changes induced by hepcidin induction therapy, as with TMPRSS6 sirna, are ideally suited to the patient with transfusion-independent, iron-loading anemias, they may equally benefit patients on chronic transfusion programs. Acknowledgment Authors would like to thank Dean Campagna and Daniel Kierstead for excellent technical assistance and members of the Fleming laboratory for helpful discussions. Author Contributions PJS, JSB, KF and MDF conceived and designed the murine experiments; PJS analyzed the mouse data and wrote the manuscript; TR completed the tail injections; and JSB, KF and MDF designed research and wrote the manuscript. MW and colleagues developed and performed the serum hepcidin assays. References 1. Papanikolaou G, Tzilianos M, Christakis JI, et al. Hepcidin in iron overload disorders. Blood 2005;105:4103 4105. 2. Origa R, Galanello R, Ganz T, et al. Liver iron concentrations and urinary hepcidin in betathalassemia. Haematologica 2007;92:583 588. 3. Yang B, Kirby S, Lewis J, et al. A mouse model for beta 0-thalassemia. Proc Natl Acad Sci USA 1995;92:11608 11612. 4. Gardenghi S, Marongiu MF, Ramos P, et al. Ineffective erythropoiesis in beta-thalassemia is characterized by increased iron absorption mediated by down-regulation of hepcidin and up-regulation of ferroportin. Blood 2007;109: 5027 5035. 5. De Franceschi L, Daraio F, Filippini A, et al. Liver expression of hepcidin and other iron genes in two mouse models of beta-thalassemia. Haematologica 2006;91:1336 1342. 6. Adamsky K, Weizer O, Amariglio N, et al. Decreased hepcidin mrna expression in thalassemic mice. Br J Haematol 2004;124:123 124. 7. Weizer-Stern O, Adamsky K, Amariglio N, et al. mrna expression of iron regulatory genes in beta-thalassemia intermedia and betathalassemia major mouse models. Am J Hematol 2006;81:479 483. 8. Silvestri L, Pagani A, Nai A, et al. The serine protease matriptase-2 (TMPRSS6) inhibits hepcidin activation by cleaving membrane hemojuvelin. Cell Metab 2008;8:502 511. 9. Finberg KE, Heeney MM, Campagna DR, et al. Mutations in TMPRSS6 cause iron-refractory iron deficiency anemia (IRIDA). Nat Genet 2008;40:569 571. 10. Finberg KE, Whittlesey RL, Andrews NC. Tmprss6 is a genetic modifier of the Hfehemochromatosis phenotype in mice. Blood 2011;117:4590 4599. 11. Gardenghi S, Ramos P, Marongiu MF, et al. Hepcidin as a therapeutic tool to limit iron overload and improve anemia in beta-thalassemic mice. J Clin Invest 2010;120:4466 4477. 12. Nai A, Pagani A, Mandelli G, et al. Deletion of TMPRSS6 attenuates the phenotype in a mouse model of beta-thalassemia. Blood 2012;119: 5021 5029. 13. Li H, Rybicki AC, Suzuka SM, et al. Transferrin therapy ameliorates disease in beta-thalassemic mice. Nat Med 2010;16:177 182. 14. Schmidt PJ, Toudjarska I, Sendamarai AK, et al. An RNAi therapeutic targeting Tmprss6 decreases iron overload in Hfe(2/2) mice and ameliorates anemia and iron overload in murine betathalassemia intermedia. Blood 2013;121:1200 1208. 15. Guo S, Casu C, Gardenghi S, et al. Reducing TMPRSS6 ameliorates hemochromatosis and beta-thalassemia in mice. J Clin Invest 2013;123: 1531 1541. 16. Fisher SA, Brunskill SJ, Doree C, et al. Oral deferiprone for iron chelation in people with thalassaemia. Cochrane Database Syst Rev 2013; 8:CD004839[WorldC at] 17. Gutschow P, Schmidt PJ, Han H, et al. A competitive enzyme-linked immunosorbent assay specific for murine hepcidin-1: Correlation with hepatic mrna expression in established and novel models of dysregulated iron homeostasis. Haematologica 2014 Nov 25. pii: haematol.2014.116723. [Epub ahead of print], in press. [WorldCat] 18. Pasricha SR, Frazer DM, Bowden DK, Anderson GJ. Transfusion suppresses erythropoiesis and increases hepcidin in adult patients with betathalassemia major: A longitudinal study. Blood 2013;122:124 133. doi:10.1002/ajh.23934 American Journal of Hematology, Vol. 90, No. 4, April 2015 313