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

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

THE ROLE OF HEMOJUVELIN IN IRON DEFICIENCY AND OVERLOAD

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

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

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

A mutation in the TMPRSS6

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

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

Iron-refractory iron deficiency anemia: new molecular mechanisms

Clinical Spectrum and Genetic Mechanism of GLUT1-DS. Yasushi ITO (Tokyo Women s Medical University, Japan)

Corporate Medical Policy Genetic Testing for Hereditary Hemochromatosis

Corporate Medical Policy

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

Thalassemias:general aspects and molecular pathology

Research and Development

Iron deficiency anemia and porphyrias

Role of hepcidin in the pathophysiology and diagnosis of anemia

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

Tumor suppressor genes D R. S H O S S E I N I - A S L

Microcytic Hypochromic Anemia An Approach to Diagnosis

Metabolic Liver Diseases

Phenylketonuria (PKU) the Biochemical Basis. Biol 405 Molecular Medicine

Congenital Dyserythropoietic anemias: where we are

CANCER GENETICS PROVIDER SURVEY

Single Gene (Monogenic) Disorders. Mendelian Inheritance: Definitions. Mendelian Inheritance: Definitions

BIOL2005 WORKSHEET 2008

Report of Beta Thalassemia in Newar Ethnicity

Dr. Ayman Mohsen Mashi, MBBS Consultant Hematology & Blood Transfusion Department Head, Laboratory & Blood Bank King Fahad Central Hospital, Gazan,

Thalassemias. Emanuela Veras, M.D. 01/08/2006

YEAR III Pharm.D Dr. V. Chitra

Biology 2C03: Genetics What is a Gene?

Identifying Mutations Responsible for Rare Disorders Using New Technologies

Proposal form for the evaluation of a genetic test for NHS Service Gene Dossier

Investigating Seven Recently Identified Genes in 100 Iranian Families with Autosomal Recessive Non-syndromic Hearing Loss

variant led to a premature stop codon p.k316* which resulted in nonsense-mediated mrna decay. Although the exact function of the C19L1 is still

Metabolic Liver Disease

Insulin Resistance. Biol 405 Molecular Medicine

HEREDITARY HEMOCHROMATOSIS

RED BLOOD CELLS AND IRON: best presentations from 21 st EHA Meeting Copenhagen

MEDICAL GENOMICS LABORATORY. Next-Gen Sequencing and Deletion/Duplication Analysis of NF1 Only (NF1-NG)

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

Genome 371, Autumn 2018 Quiz Section 9: Genetics of Cancer Worksheet

Deciphering Developmental Disorders (DDD) DDG2P

Significance of the MHC

Human Genetics (Learning Objectives)

Reporting TP53 gene analysis results in CLL

Repeated IV doses of iron provides effective supplemental treatment of restless legs syndrome

Dan Koller, Ph.D. Medical and Molecular Genetics

The Foundations of Personalized Medicine

Computational Systems Biology: Biology X

A rare thing may be just like any other but it is also paradoxically nothing like any of them.

NGS panels in clinical diagnostics: Utrecht experience. Van Gijn ME PhD Genome Diagnostics UMCUtrecht

Serum hepcidin levels and iron parameters in children with iron deficiency

Hands-On Ten The BRCA1 Gene and Protein

Management of anemia in CKD

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

Unraveling Mechanisms Regulating Systemic Iron Homeostasis

MRC-Holland MLPA. Description version 19;

Merging single gene-level CNV with sequence variant interpretation following the ACMGG/AMP sequence variant guidelines

Diagnostic difficulties in prevention and control program for thalassemia in Thailand: atypical thalassemia carriers

Figure 1: Transmission of Wing Shape & Body Color Alleles: F0 Mating. Figure 1.1: Transmission of Wing Shape & Body Color Alleles: Expected F1 Outcome

Author's response to reviews

DOES THE BRCAX GENE EXIST? FUTURE OUTLOOK

High Hemoglobin F in a Saudi Child Presenting with Pancytopenia

Journal of American Science 2018;14(10)

BONE MARROW PERIPHERAL BLOOD Erythrocyte

JULY 21, Genetics 101: SCN1A. Katie Angione, MS CGC Certified Genetic Counselor CHCO Neurology

SALSA MLPA KIT P060-B2 SMA

The coiled-coil domain containing protein CCDC40 is essential for motile cilia function and left-right axis formation

6.1 Extended family screening

Genome-wide association study identifies variants in TMPRSS6 associated with hemoglobin levels.

Lecture 17: Human Genetics. I. Types of Genetic Disorders. A. Single gene disorders

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

Proposal form for the evaluation of a genetic test for NHS Service Gene Dossier

The laws of Heredity. Allele: is the copy (or a version) of the gene that control the same characteristics.

Wilson Disease Robert Baumgartner, Kira Melamud and Amelia Wnorowski

PALB2 c g>c is. VARIANT OF UNCERTAIN SIGNIFICANCE (VUS) CGI s summary of the available evidence is in Appendices A-C.

Genotype-Phenotype in Egyptian Patients with Nephropathic Cystinosis. (December 2012 report)

Hemosiderin. Livia Vida 2018

MRC-Holland MLPA. Description version 07; 26 November 2015

Red cell disorder. Dr. Ahmed Hasan

Protocol. Genetic Testing for Hereditary Hemochromatosis

Anaemia in Pregnancy

Understanding genetics, mutation and other details. Stanley F. Nelson, MD 6/29/18

George R. Honig Junius G. Adams III. Human Hemoglobin. Genetics. Springer-Verlag Wien New York

Hemochromatosis - Genetic Inheritance

* imagine if the Hb is free ( e.g. hemolysis ) in the plasma what happens?

Germline Testing for Hereditary Cancer with Multigene Panel

Next Generation Sequencing as a tool for breakpoint analysis in rearrangements of the globin-gene clusters

MOLECULAR DIAGNOSIS for X-LINKED INTELLECTUAL DISABILITY

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

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

Chromosomal Mutations

Molecular Oncology, oncology parameters see each test

Basic Definitions. Dr. Mohammed Hussein Assi MBChB MSc DCH (UK) MRCPCH

Endocrine Surgery. Characteristics of the Germline MEN1 Mutations in Korea: A Literature Review ORIGINAL ARTICLE. The Korean Journal of INTRODUCTION

Non-Mendelian inheritance

Metabolic Liver Disease: What s New in Diagnosis and Therapy?

Next-Generation Biomarkers for Iron Status

Approach to the child with anemia. Nittaya Wisanuyothin,MD. Pediatrics Department, Maharat Nakhonratchasima Hospital

Transcription:

NIH Public Access Author Manuscript Published in final edited form as: Nat Genet. 2008 May ; 40(5): 569 571. doi:10.1038/ng.130. Mutations in TMPRSS6 cause iron-refractory iron deficiency anemia (IRIDA) Karin E Finberg 1,2,14, Matthew M Heeney 2,3,15, Dean R Campagna 4,15, Yeşim Aydınok 5, Howard A Pearson 6, Kip R Hartman 7, Mary M Mayo 8, Stewart M Samuel 9, John J Strouse 10, Kyriacos Markianos 11,12, Nancy C Andrews 2,12,14,16, and Mark D Fleming 4,13,16 1 Pathology Service, Massachusetts General Hospital, Boston, Massachusetts 02114, USA. 2 Division of Hematology/Oncology, Children's Hospital Boston, Boston, Massachusetts 02115, USA. 3 Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA. 4 Department of Pathology, Children's Hospital Boston, Boston, Massachusetts 02115, USA. 5 Department of Pediatric Hematology, Ege University Faculty of Medicine, 35100 Bornova, Izmir, Turkey. 6 Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut 06520, USA. 7 Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA. 8 Department of Pathology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, USA. 9 Pediatric Health Associates, P.C., Plainview, New York 11803, USA. 10 Division of Pediatric Hematology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA. 11 Program in Genomics, Children's Hospital Boston, Boston, Massachusetts 02115, USA. 12 Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA. 13 Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115, USA. 2008 Nature Publishing Group Correspondence should be addressed to N.C.A. (Nancy.Andrews@duke.edu) and M.D.F. (Mark.Fleming@childrens.harvard.edu).. 14 Present addresses: Department of Pathology, Duke University School of Medicine, Durham, North Carolina 27710, USA (K.E.F.) and Department of Pediatrics and Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA (N.C.A.) 15 These authors contributed equally to this work. 16 These authors contributed equally to this work. AUTHOR CONTRIBUTIONS K.E.F. obtained institutional review board approval and consents, designed, conducted and interpreted results from the segregation studies and sequence analysis, and prepared the manuscript. M.M.H. obtained institutional review board approval and consents and coordinated clinical sample acquisition and clinical data analysis. D.R.C. conducted and interpreted results from the sequencing analysis and assisted with the segregation studies and other technical aspects of the project. Y.A., H.A.P., K.R.H., M.M.M. S.M.S. and J.J.S. were clinical collaborators who provided samples from affected individuals, phenotypic information and results of laboratory testing. K.M. interpreted results from the segregation studies and sequencing analysis. N.C.A. and M.D.F. obtained institutional review board approval and consents, supervised the design of experiments and data interpretation, and prepared the manuscript. N.C.A. also provided samples from affected individuals, phenotypic information and results of laboratory testing. Note: Supplementary information is available on the Nature Genetics website.

Finberg et al. Page 2 Abstract Iron deficiency is usually attributed to chronic blood loss or inadequate dietary intake. Here, we show that iron deficiency anemia refractory to oral iron therapy can be caused by germline mutations in TMPRSS6, which encodes a type II transmembrane serine protease produced by the liver that regulates the expression of the systemic iron regulatory hormone hepcidin. These findings demonstrate that TMPRSS6 is essential for normal systemic iron homeostasis in humans. We and others have identified families with multiple individuals with iron deficiency anemia unresponsive to oral iron therapy but partially responsive to parenteral iron administration, suggesting that some cases of iron deficiency may be genetically determined 1 6. We refer to this phenotype as iron-refractory iron deficiency anemia (IRIDA), the key features of which are: a congenital hypochromic, microcytic anemia, a very low mean corpuscular erythrocyte volume, a low transferrin saturation, abnormal iron absorption characterized by no hematological improvement following treatment with oral iron, and abnormal iron utilization characterized by a sluggish, incomplete response to parenteral iron (Table 1). To determine the genetic basis of IRIDA, we studied five multiplex kindreds. Acquired causes of iron deficiency and other inherited causes of microcytosis were rigorously excluded (Supplementary Note online). In all five families, recessive transmission was suggested by the absence of the phenotype in the parents of affected sibling pairs; one kindred was also notable for parental consanguinity (Table 1 and Supplementary Fig. 1 online). We excluded several genes involved in intestinal iron absorption and/or systemic iron utilization, including CYBRD1, HAMP, SLC11A2 and SLC40A1, as IRIDA candidates through haplotype analysis using flanking microsatellite markers and/or by sequencing coding regions and intron exon boundaries (data not shown). Other investigators recently described a Sardinian kindred with autosomal recessive IRIDA linked to chromosome 22q12 13 (R. Galanello, M. Cau, M.A. Melis, F. Deidda, A. Cao and M. Cazzola, unpublished data; M.A. Melis, M. Cau, R. Congiu, G. Sole, A. Cao and R. Galanello, unpublished data). The phenotype in the families we studied was similarly compatible with linkage to 22q12 13 (Supplementary Fig. 1 and Supplementary Methods online). TMPRSS6, located within the critical interval, encodes a type II transmembrane serine protease (also known as matriptase-2) that is expressed primarily in the liver 7 (Supplementary Fig. 2 online), and we considered it an excellent positional candidate gene, as a recessive mutation in the mouse ortholog (Tmprss6) leads to anemia as a result of defective dietary iron uptake (E. Beutler, P. Lee, T. Gelbart, X. Du and B. Beutler, unpublished data). We analyzed all TMPRSS6 coding regions and intron exon boundaries and identified sequence variants in each of the five multiplex IRIDA kindreds (Table 1, Fig. 1, Supplementary Methods and Supplementary Table 1 online). Affected individuals harbored frame-shift mutations, splice junction mutations or missense mutations altering residues conserved in TMPRSS6 homologs from humans to fugu (Supplementary Fig. 3 online). In three of the four kindreds in which the phase of chromosomal segregation was known, we identified bialleic mutations. In the fourth family, we found a mutation only on the paternal allele; however, we did not exclude the presence of other types of mutations, such as large deletions, that would not be detectable by sequencing. Additionally, in the fifth kindred, for which DNA was available from only the affected individuals, we found a nonconservative missense mutation in both siblings. We also examined two individuals with sporadic IRIDA and found nonsense, frameshift or splice junction mutations in both (Table 1). None of the disease-associated variants were present in the NCBI and Ensembl SNP

Finberg et al. Page 3 databases or in 100 control chromosomes (Table 1 and data not shown). These findings conclusively show that mutations in TMPRSS6 cause IRIDA. All of the TMPRSS6 mutations that we identified in individuals with IRIDA lie distal to exon 8, in regions that encode several conserved structural domains, most notably a trypsinlike serine protease domain (Fig. 1). This catalytic domain is highly similar to that of other type II transmembrane serine proteases, particularly the S1 family of trypsin-like serine proteases. Whether it is the putative catalytic activity of TMPRSS6 that is essential for normal systemic iron homeostasis, rather than another function of the molecule, is uncertain. Nonetheless, the finding of individuals with IRIDA harboring homozygous frameshift mutations predicted to disrupt only the catalytic domain suggests that this portion of the molecule is important for iron homeostasis. Of note, the Tmprss6 mouse mutant also results from a splice site mutation that disrupts the catalytic domain (E. Beutler, P. Lee, T. Gelbart, X. Du and B. Beutler, unpublished data). To gain insight into the pathophysiology of IRIDA, we determined levels of hepcidin, a hormone produced by the liver that regulates intestinal iron absorption and macrophage iron release 8. Under normal circumstances, hepcidin is induced by iron overload and repressed by iron deficiency and anemia. Although urinary hepcidin levels are typically undetectable in individuals with iron deficiency 9, in the five affected individuals from three IRIDA kindreds we examined, urinary hepcidin/creatinine ratios were either within or above the normal range (Supplementary Note and Supplementary Table 2 online). The finding of inappropriately elevated urinary hepcidin levels in individuals with IRIDA provides insight into the pathophysiology of the disorder, as it may explain the failure to absorb dietary iron despite systemic iron deficiency, as well as the coexistent failure to respond to parenteral iron administered as iron-dextran, which must be processed and exported by macrophages before utilization for erythropoiesis. How TMPRSS6 mutations lead to inappropriately elevated hepcidin levels remains unclear. The simplest explanation would be that TMPRSS6 normally cleaves a protein that acts in or on hepatocytes to negatively regulate hepcidin production, secretion or clearance. Studies in the Tmprss6 mouse mutant suggest that TMPRSS6 is a negative regulator of hepcidin transcription (E. Beutler, P. Lee, T. Gelbart, X. Du and B. Beutler, unpublished data). This transcriptional effect might be achieved by cleaving a protein that upregulates a pathway that normally represses hepcidin transcription, or that down-regulates a pathway that normally activates hepcidin transcription. The identification of TMPRSS6 mutations in individuals with IRIDA has broad implications for clinical disorders of iron metabolism. Mutations or polymorphisms in TMPRSS6 may contribute to iron deficiency anemia in individuals with or without other predisposing factors. Furthermore, the finding that TMPRSS6 regulates hepcidin levels in humans may have potential applications for treatment of iron disorders. For example, inhibition of the putative protease function of TMPRSS6 might be a potential treatment for disorders in which hepcidin is inappropriately low, such as primary hemochromatosis and iron loading anemias. Similarly, treatment with agonists or with the endogenous substrate of TMPRSS6 might be employed in the anemia of chronic disease, in which hepcidin is inappropriately high. Supplementary Material Refer to Web version on PubMed Central for supplementary material.

Finberg et al. Page 4 Acknowledgments References We thank the families for their invaluable contribution to this study. We are indebted to E. Neufeld for ongoing mentorship. We thank C. Trenor, A. Donovan, I. Rubio-Aliaga and other members of the Andrews laboratory for their contributions to the early stages of this project. We thank A.J. Iafrate and J. Miller for technical advice and assistance. K.E.F. was supported by T32 CA009216 awarded to the Department of Pathology, Massachusetts General Hospital. This work was also supported by R01 DK080011 (M.D.F.), K12 HL087164 (M.M.H.), R01 DK066373 (N.C.A.) and DK053813 (N.C.A.). 1. Andrews NC. Yale J. Biol. Med. 1997; 70:219 226. [PubMed: 9544492] 2. Brown AC, et al. Am. J. Hematol. 1988; 27:1 6. [PubMed: 3354554] 3. Buchanan GR, Sheehan RG. J. Pediatr. 1981; 98:723 728. [PubMed: 7229750] 4. Hartman KR, Barker JA. Am. J. Hematol. 1996; 51:269 275. [PubMed: 8602626] 5. Mayo MM, Samuel SM. Clin. Lab. Sci. 2001; 14:135 138. [PubMed: 11517621] 6. Pearson HA, Lukens JN. J. Pediatr. Hematol. Oncol. 1999; 21:412 417. [PubMed: 10524456] 7. Velasco G, Cal S, Quesada V, Sanchez LM, Lopez-Otin C. J. Biol. Chem. 2002; 277:37637 37646. [PubMed: 12149247] 8. Nemeth E, Ganz T. Annu. Rev. Nutr. 2006; 26:323 342. [PubMed: 16848710] 9. Kemna EH, Tjalsma H, Podust VN, Swinkels DW. Clin. Chem. 2007; 53:620 628. [PubMed: 17272487]

Finberg et al. Page 5 Figure 1. Schematic representation of TMPRSS6 mutations and corresponding TMPRSS6 domains. The missense, nonsense, frameshift and splice junction mutations present in five familial and two sporadic cases of IRIDA are diagrammed adjacent to the affected TMPRSS6 domain (Table 1). The transmembrane (TM), complement factor C1r/C1s, urchin embryonic growth factor and bone morphogenetic protein (CUB), LDL-receptor class A (L) and trypsin-like serine protease domains are shown in green, blue, yellow and red, respectively.

Finberg et al. Page 6 Table 1 parameters of IRIDA index cases with TMPRSS6 mutations Mutation 1 Mutation 2 estry Consanguinity Sex Age at evaluation RBC 10 12 /l Hb (g/dl) MCV (fl) Retics (%) Transferrin saturation (%) Nucleotide/amino acid change Domain Nucleotide/amino acid change Domain a,b Protease kish Yes M 6 y 5.0 8.8 58 n.d. 2 1906_1907insGC (K636fs) a,b Protease 1906_1907insGC (K636fs) European No F 13 mo. n.d. 9.2 65 1.0 10 1813delG (A605fs) a Protease IVS13+1G>A a LDLRA a,b Protease Not identified n.a. rian No M 17 mo. 4.2 7.0 49 n.d. 5 IVS16+1G>C European No F 11 y 4.9 8.2 56 1.6 3 1324G>A (G442R) a CUB 1561G>A (D521N) a LDLRA b Protease Not identified n.a. merican No M 7 y 5.1 7.5 49 0.6 4 2320C>T (R774C) a,b Protease rian No F 3 y 5.0 9.7 61 0.5 4 IVS15-1G>C a,b Protease IVS15-1G>C merican No M 15 mo. 5.0 7.9 53 0.8 2 1065C>A b (Y355X) CUB 1383delA b (E461fs) LDLRA additional affected sibling who harbors the identical mutation(s) as the index case. Each mutation was present in heterozygous form in affected individuals, with the t in the affected individuals from Kindreds A and F, which were present in homozygous form. Hematological parameters from kindreds B 2,6, D 5 and E 4 have been aturation (%) was calculated by dividing the serum iron level by the total iron binding capacity and multiplying by 100. RBC, red blood cell count; Hb, hemoglobin; MCV,, reticulocytes, CUB, complement factor C1r/C1s, urchin embryonic growth factor, and bone morphogenetic protein; LDLRA, LDL-receptor class A; n.d., not determined; b, MCV and transferrin saturation were below the respective reference ranges provided by the referring hospital laboratory. ol chromosomes from individuals of self-reported Caucasian ancestry. ol chromosomes from individuals of African-American ancestry.