1
|
Mann J, Reznik E, Santer M, Fongheiser MA, Smith N, Hirschhorn T, Zandkarimi F, Soni RK, Dafré AL, Miranda-Vizuete A, Farina M, Stockwell BR. Ferroptosis inhibition by oleic acid mitigates iron-overload-induced injury. Cell Chem Biol 2024; 31:249-264.e7. [PMID: 37944523 PMCID: PMC10922137 DOI: 10.1016/j.chembiol.2023.10.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 07/24/2023] [Accepted: 10/13/2023] [Indexed: 11/12/2023]
Abstract
Iron overload, characterized by accumulation of iron in tissues, induces a multiorgan toxicity whose mechanisms are not fully understood. Using cultured cell lines, Caenorhabditis elegans, and mice, we found that ferroptosis occurs in the context of iron-overload-mediated damage. Exogenous oleic acid protected against iron-overload-toxicity in cell culture and Caenorhabditis elegans by suppressing ferroptosis. In mice, oleic acid protected against FAC-induced liver lipid peroxidation and damage. Oleic acid changed the cellular lipid composition, characterized by decreased levels of polyunsaturated fatty acyl phospholipids and decreased levels of ether-linked phospholipids. The protective effect of oleic acid in cells was attenuated by GW6471 (PPAR-α antagonist), as well as in Caenorhabditis elegans lacking the nuclear hormone receptor NHR-49 (a PPAR-α functional homologue). These results highlight ferroptosis as a driver of iron-overload-mediated damage, which is inhibited by oleic acid. This monounsaturated fatty acid represents a potential therapeutic approach to mitigating organ damage in iron overload individuals.
Collapse
Affiliation(s)
- Josiane Mann
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Eduard Reznik
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Melania Santer
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Mark A Fongheiser
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Nailah Smith
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Tal Hirschhorn
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | | | - Rajesh Kumar Soni
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - Alcir Luiz Dafré
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Antonio Miranda-Vizuete
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Seville, Spain
| | - Marcelo Farina
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil; Department of Biological Sciences, Columbia University, New York, NY 10027, USA.
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA; Department of Chemistry, Columbia University, New York, NY 10027, USA; Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York. NY 10032, USA.
| |
Collapse
|
2
|
Lê S, Minty M, Boyer É, Blasco-Baque V, Bonnaure-Mallet M, Meuric V. [Oral microbiota and liver]. Med Sci (Paris) 2024; 40:42-48. [PMID: 38299902 DOI: 10.1051/medsci/2023194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024] Open
Abstract
The liver has many important biological functions for the body, as it is involved in the storage and distribution of nutrients (carbohydrates to glycogen, lipids to triglycerides), the digestion of fats, the synthesis of blood proteins, and the detoxification of alcohol and drugs. The liver can be affected by various diseases such as viral or drug-induced hepatitis, fibrosis and cirrhosis, in which damaged hepatocytes are progressively replaced by scar tissue.
Collapse
Affiliation(s)
- Sylvie Lê
- Département dentaire, université Paul Sabatier III (UPS), Toulouse, France - Service d'odontologie Toulouse, CHU Toulouse, Toulouse, France - UMR1297 Inserm, équipe InCOMM (Intestine ClinicOmics Metabolism & Microbiota), Institut des maladies métaboliques et cardiovasculaires (I2MC), université Paul Sabatier, Toulouse, France
| | - Matthieu Minty
- Département dentaire, université Paul Sabatier III (UPS), Toulouse, France - Service d'odontologie Toulouse, CHU Toulouse, Toulouse, France - UMR1297 Inserm, équipe InCOMM (Intestine ClinicOmics Metabolism & Microbiota), Institut des maladies métaboliques et cardiovasculaires (I2MC), université Paul Sabatier, Toulouse, France
| | - Émile Boyer
- Inserm U1317, Inrae, université de Rennes, CHU de Rennes, site Pontchaillou-Villejean, Rennes, France
| | - Vincent Blasco-Baque
- Département dentaire, université Paul Sabatier III (UPS), Toulouse, France - Service d'odontologie Toulouse, CHU Toulouse, Toulouse, France - UMR1297 Inserm, équipe InCOMM (Intestine ClinicOmics Metabolism & Microbiota), Institut des maladies métaboliques et cardiovasculaires (I2MC), université Paul Sabatier, Toulouse, France
| | - Martine Bonnaure-Mallet
- Inserm U1317, Inrae, université de Rennes, CHU de Rennes, site Pontchaillou-Villejean, Rennes, France
| | - Vincent Meuric
- Inserm U1317, Inrae, université de Rennes, CHU de Rennes, site Pontchaillou-Villejean, Rennes, France
| |
Collapse
|
3
|
Tong S, Hong Y, Xu Y, Sun Q, Ye L, Cai J, Ye Z, Chen Q, Tian D. TFR2 regulates ferroptosis and enhances temozolomide chemo-sensitization in gliomas. Exp Cell Res 2023; 424:113474. [PMID: 36702193 DOI: 10.1016/j.yexcr.2023.113474] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/04/2023] [Accepted: 01/08/2023] [Indexed: 01/24/2023]
Abstract
Glioma is a common type of brain tumor with high incidence and mortality rates. Iron plays an important role in various physiological and pathological processes. Iron entry into the cell is promoted by binding the transferrin receptor 2 (TFR2) to the iron-transferrin complex. This study was designed to assess the association between TFR2 and ferroptosis in glioma. Lipid peroxidation levels in glioma cells were assessed by determination of lipid reactive oxygen species (ROS), glutathione content, and mitochondrial membrane potential. The effect of TFR2 on TMZ sensitivity was examined by cell viability assays, flow cytometry, and colony formation assays. We found that Low TFR2 expression predicted a better prognosis for glioma patients. And overexpression of TFR2 promoted the production of reactive oxygen species and lipid peroxidation in glioma cells, thereby further promoting ferroptosis. This could be reversed by the ferroptosis inhibitors Fer-1 and DFO (both inhibitors of ferroptosis). Moreover, TFR2 potentiated the cytotoxic effect of TMZ (temozolomide) via activating ferroptosis. In conclusion, we found that TFR2 induced ferroptosis and enhanced TMZ sensitivity in gliomas. Our findings might provide a new treatment strategy for glioma patients and improve their prognosis.
Collapse
Affiliation(s)
- Shiao Tong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Yu Hong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Yang Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Qian Sun
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Liguo Ye
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Jiayang Cai
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Zhang Ye
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China.
| | - Daofeng Tian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China.
| |
Collapse
|
4
|
Protein Susceptibility to Peroxidation by 4-Hydroxynonenal in Hereditary Hemochromatosis. Int J Mol Sci 2023; 24:ijms24032922. [PMID: 36769239 PMCID: PMC9917916 DOI: 10.3390/ijms24032922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Iron overload caused by hereditary hemochromatosis (HH) increases free reactive oxygen species that, in turn, induce lipid peroxidation. Its 4-hydroxynonenal (HNE) by-product is a well-established marker of lipid peroxidation since it reacts with accessible proteins with deleterious consequences. Indeed, elevated levels of HNE are often detected in a wide variety of human diseases related to oxidative stress. Here, we evaluated HNE-modified proteins in the membrane of erythrocytes from HH patients and in organs of Hfe-/- male and female mice, a mouse model of HH. For this purpose, we used one- and two-dimensional gel electrophoresis, immunoblotting and MALDI-TOF/TOF analysis. We identified cytoskeletal membrane proteins and membrane receptors of erythrocytes bound to HNE exclusively in HH patients. Furthermore, kidney and brain of Hfe-/- mice contained more HNE-adducted protein than healthy controls. Our results identified main HNE-modified proteins suggesting that HH favours preferred protein targets for oxidation by HNE.
Collapse
|
5
|
Dissanayake R, Samarasinghe N, Waidyanatha S, Pathirana S, Neththikumara N, Dissanayake VHW, Wetthasinghe K, Gooneratne L, Wickramasinghe P. Assessment of iron overload in a cohort of Sri Lankan patients with transfusion dependent beta thalassaemia and its correlation with pathogenic variants in HBB, HFE, SLC40A1, and TFR2 genes. BMC Pediatr 2022; 22:344. [PMID: 35705926 PMCID: PMC9199146 DOI: 10.1186/s12887-022-03191-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 02/22/2022] [Indexed: 01/21/2023] Open
Abstract
Background Iron overload (IO) is a complication in transfusion dependent beta thalassaemia (TDT). Pathogenic variants in genes involving iron metabolism may confer increased risk of IO. The objective of this study was to determine the magnitude of the cardiac and hepatic IO and determine whether pathogenic variants in HFE, SLC40A1 and TFR2 genes increase the risk of IO in a cohort of TDT patients in Sri Lanka. Materials and Methods Fifty-seven (57) patients with TDT were recruited for this study. Serum ferritin was done once in 3 months for a period of one year in all. Those who were ≥ 8 years of age (40 patients) underwent T2* MRI of the liver and heart. Fifty-two (52) patients underwent next generation sequencing (NGS) to identify pathogenic variants in HBB, HFE, SLC40A1 and TFR2 genes. Results The median age of the patients of this cohort was 10 years. It comprised of 30 (52.6%) boys and 27 (47.4%) girls. The median level of serum ferritin was 2452 ng/dl. Hepatic IO was seen in 37 (92.5%) patients and cardiac IO was seen in 17 (42.5%) patients. There was no statistically significant correlation between serum ferritin and hepatic or cardiac IO. Thirty-two (61.5%), 18 (34.6%), 2 (3.8%) of patients were homozygotes, compound heterozygotes and heterozygotes for pathogenic variants in the HBB gene. Eight (15.4%) and 1 (1.9%) patients were heterozygotes for pathogenic and likely pathogenic variants of HFE genes respectively. There were no pathogenic variants for the TfR2 and SLC40A1 genes. The heterozygotes of the pathogenic variants of the HFE were not at increased risk of IO. Conclusions Cardiac T2* MRI helps to detect cardiac IO in asymptomatic patients. It is important to perform hepatic and cardiac T2* MRI to detect IO in patients with TDT. There was no statistically significant correlation between pathogenic variants of HBB and HFE genes with hepatic and cardiac IO in this cohort of patients.
Collapse
Affiliation(s)
- Ruwangi Dissanayake
- Department of Paediatrics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka. .,Lady Ridgeway Hospital for Children, Colombo, Sri Lanka.
| | | | | | - Sajeewani Pathirana
- Human Genetics Unit, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | | | | | - Kalum Wetthasinghe
- Human Genetics Unit, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Lallindra Gooneratne
- Department of Pathology, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Pujitha Wickramasinghe
- Department of Paediatrics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka.,Lady Ridgeway Hospital for Children, Colombo, Sri Lanka
| |
Collapse
|
6
|
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: 24] [Impact Index Per Article: 8.0] [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
|
7
|
Kowdley DS, Kowdley KV. Appropriate Clinical Genetic Testing of Hemochromatosis Type 2-4, Including Ferroportin Disease. Appl Clin Genet 2021; 14:353-361. [PMID: 34413666 PMCID: PMC8369226 DOI: 10.2147/tacg.s269622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/18/2021] [Indexed: 11/23/2022] Open
Abstract
Hereditary hemochromatosis (HH) is an inherited iron overload disorder due to a deficiency of hepcidin, or a failure of hepcidin to degrade ferroportin. The most common form of HH, Type 1 HH, is most commonly due to a homozygous C282Y mutation in HFE and is relatively well understood in significance and action; however, other rare forms of HH (Types 2–4) exist and are more difficult to identify and diagnose in clinical practice. In this review, we describe the clinical characteristics of HH Type 2–4 and the mutation patterns that have been described in these conditions. We also review the different methods for genetic testing available in clinical practice and a pragmatic approach to the patient with suspected non-HFE HH.
Collapse
Affiliation(s)
- Devan S Kowdley
- Liver Institute Northwest and Elson S. Floyd College of Medicine, Washington State University, Seattle, WA, USA
| | - Kris V Kowdley
- Liver Institute Northwest and Elson S. Floyd College of Medicine, Washington State University, Seattle, WA, USA
| |
Collapse
|
8
|
Wu HX, Liu JY, Yan DW, Li L, Zhuang XH, Li HY, Zhou ZG, Zhou HD. Atypical juvenile hereditary hemochromatosis onset with positive pancreatic islet autoantibodies diabetes caused by novel mutations in HAMP and overall clinical management. Mol Genet Genomic Med 2020; 8:e1522. [PMID: 33016646 PMCID: PMC7767552 DOI: 10.1002/mgg3.1522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/06/2020] [Accepted: 09/17/2020] [Indexed: 12/19/2022] Open
Abstract
Background Atypical clinical symptoms of juvenile hereditary hemochromatosis (JHH) often leads to misdiagnosis and underdiagnosis bringing ominous outcomes, even death. Methods The whole exome was sequenced and interpreted. A literature review assisted to analyze and verify the phenotype–genotype relationships. We revealed the entire process of diagnosis, treatments, and outcome of two diabetic onset of JHH families to provide new insights for genotype–phenotype relation with novel compound heterozygous mutations in the hepcidin antimicrobial peptide (HAMP, OMIM: 606464). Results Two probands were diagnosed and treated as type 1 diabetes initially because of specific symptoms and positive islet autoantibodies. Poor control of hyperglycemia and progressive symptoms occurred. Sequencing informed that the compound heterozygous and homozygous mutations c.166C>G and c.223C>T in HAMP caused type 1 diabetic‐onset JHH. The two patients accessed irregular phlebotomy treatments, and then, experienced poor prognosis. We summarized the process of overall clinical management of reported 26 cases comparing to our novel atypical diabetic onsets Juvenile Hereditary Hemochromatosis cases. Conclusion It was first reported that positive pancreatic islet autoantibodies diabetes onset of JHH resulted from loss‐of‐function mutations of HAMP, of which the atypical JHH should be differentially diagnosed with type 1 diabetes at the onset. Early administration of phlebotomy and vital organs protection and surveillance might be important for the treatment of atypical JHH.
Collapse
Affiliation(s)
- Hui-Xuan Wu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jun-Ying Liu
- Department of Endocrinology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - De-Wen Yan
- Department of Endocrinology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Long Li
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiang-Hua Zhuang
- Department of Endocrinology, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Hai-Yan Li
- Department of Endocrinology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Zhi-Guang Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hou-De Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
9
|
Regulation of Iron Homeostasis and Related Diseases. Mediators Inflamm 2020; 2020:6062094. [PMID: 32454791 PMCID: PMC7212278 DOI: 10.1155/2020/6062094] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 03/23/2020] [Indexed: 12/18/2022] Open
Abstract
The liver is the organ for iron storage and regulation; it senses circulating iron concentrations in the body through the BMP-SMAD pathway and regulates the iron intake from food and erythrocyte recovery into the bloodstream by secreting hepcidin. Under iron deficiency, hypoxia, and hemorrhage, the liver reduces the expression of hepcidin to ensure the erythropoiesis but increases the excretion of hepcidin during infection and inflammation to reduce the usage of iron by pathogens. Excessive iron causes system iron overload; it accumulates in never system and damages neurocyte leading to neurodegenerative diseases such as Parkinson's syndrome. When some gene mutations affect the perception of iron and iron regulation ability in the liver, then they decrease the expression of hepcidin, causing hereditary diseases such as hereditary hemochromatosis. This review summarizes the source and utilization of iron in the body, the liver regulates systemic iron homeostasis by sensing the circulating iron concentration, and the expression of hepcidin regulated by various signaling pathways, thereby understanding the pathogenesis of iron-related diseases.
Collapse
|
10
|
A Rare Case Of a 2-year-old Boy With Alagille Syndrome and Type 3 Hereditary Hemochromatosis With TFR2 Mutation. J Pediatr Gastroenterol Nutr 2019; 68:e68-e70. [PMID: 29985876 DOI: 10.1097/mpg.0000000000002078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
|
11
|
Dhillon BK, Chopra G, Jamwal M, Chandak GR, Duseja A, Malhotra P, Chawla YK, Garewal G, Das R. Adult onset hereditary hemochromatosis is associated with a novel recurrent Hemojuvelin (HJV) gene mutation in north Indians. Blood Cells Mol Dis 2018; 73:14-21. [DOI: 10.1016/j.bcmd.2018.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 08/23/2018] [Accepted: 08/24/2018] [Indexed: 12/26/2022]
|
12
|
The Functional Versatility of Transferrin Receptor 2 and Its Therapeutic Value. Pharmaceuticals (Basel) 2018; 11:ph11040115. [PMID: 30360575 PMCID: PMC6316356 DOI: 10.3390/ph11040115] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/19/2018] [Accepted: 10/21/2018] [Indexed: 12/11/2022] Open
Abstract
Iron homeostasis is a tightly regulated process in all living organisms because this metal is essential for cellular metabolism, but could be extremely toxic when present in excess. In mammals, there is a complex pathway devoted to iron regulation, whose key protein is hepcidin (Hepc), which is a powerful iron absorption inhibitor mainly produced by the liver. Transferrin receptor 2 (Tfr2) is one of the hepcidin regulators, and mutations in TFR2 gene are responsible for type 3 hereditary hemochromatosis (HFE3), a genetically heterogeneous disease characterized by systemic iron overload. It has been recently pointed out that Hepc production and iron regulation could be exerted also in tissues other than liver, and that Tfr2 has an extrahepatic role in iron metabolism as well. This review summarizes all the most recent data on Tfr2 extrahepatic role, taking into account the putative distinct roles of the two main Tfr2 isoforms, Tfr2α and Tfr2β. Representing Hepc modulation an effective approach to correct iron balance impairment in common human diseases, and with Tfr2 being one of its regulators, it would be worthwhile to envisage Tfr2 as a therapeutic target.
Collapse
|
13
|
Phenotypic analysis of hemochromatosis subtypes reveals variations in severity of iron overload and clinical disease. Blood 2018; 132:101-110. [PMID: 29743178 DOI: 10.1182/blood-2018-02-830562] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/03/2018] [Indexed: 12/15/2022] Open
Abstract
The clinical progression of HFE-related hereditary hemochromatosis (HH) and its phenotypic variability has been well studied. Less is known about the natural history of non-HFE HH caused by mutations in the HJV, HAMP, or TFR2 genes. The purpose of this study was to compare the phenotypic and clinical presentations of hepcidin-deficient forms of HH. A literature review of all published cases of genetically confirmed HJV, HAMP, and TFR2 HH was performed. Phenotypic and clinical data from a total of 156 patients with non-HFE HH was extracted from 53 publications and compared with data from 984 patients with HFE-p.C282Y homozygous HH from the QIMR Berghofer Hemochromatosis Database. Analyses confirmed that non-HFE forms of HH have an earlier age of onset and a more severe clinical course than HFE HH. HJV and HAMP HH are phenotypically and clinically very similar and have the most severe presentation, with cardiomyopathy and hypogonadism being particularly prevalent findings. TFR2 HH is more intermediate in its age of onset and severity. All clinical outcomes analyzed were more prevalent in the juvenile forms of HH, with the exception of arthritis and arthropathy, which were more commonly seen in HFE HH. This is the first comprehensive analysis comparing the different phenotypic and clinical aspects of the genetic forms of HH, and the results will be valuable for the differential diagnosis and management of these conditions. Importantly, our analyses indicate that factors other than iron overload may be contributing to joint pathology in patients with HFE HH.
Collapse
|
14
|
Richardson KJ, McNamee AP, Simmonds MJ. Haemochromatosis: Pathophysiology and the red blood cell1. Clin Hemorheol Microcirc 2018; 69:295-304. [DOI: 10.3233/ch-189128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
| | - Antony P. McNamee
- Biorheology Research Laboratory, Griffith University, Gold Coast, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Michael J. Simmonds
- Biorheology Research Laboratory, Griffith University, Gold Coast, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| |
Collapse
|
15
|
Abstract
Haemochromatosis is defined as systemic iron overload of genetic origin, caused by a reduction in the concentration of the iron regulatory hormone hepcidin, or a reduction in hepcidin-ferroportin binding. Hepcidin regulates the activity of ferroportin, which is the only identified cellular iron exporter. The most common form of haemochromatosis is due to homozygous mutations (specifically, the C282Y mutation) in HFE, which encodes hereditary haemochromatosis protein. Non-HFE forms of haemochromatosis due to mutations in HAMP, HJV or TFR2 are much rarer. Mutations in SLC40A1 (also known as FPN1; encoding ferroportin) that prevent hepcidin-ferroportin binding also cause haemochromatosis. Cellular iron excess in HFE and non-HFE forms of haemochromatosis is caused by increased concentrations of plasma iron, which can lead to the accumulation of iron in parenchymal cells, particularly hepatocytes, pancreatic cells and cardiomyocytes. Diagnosis is noninvasive and includes clinical examination, assessment of plasma iron parameters, imaging and genetic testing. The mainstay therapy is phlebotomy, although iron chelation can be used in some patients. Hepcidin supplementation might be an innovative future approach.
Collapse
Affiliation(s)
- Pierre Brissot
- INSERM, Univ. Rennes, INRA, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR_A 1341, UMR_S 1241, F-35000 Rennes, France
| | - Antonello Pietrangelo
- Division of Internal Medicine 2 and Center for Haemochromatosis, University Hospital of Modena, Modena, Italy
| | - Paul C. Adams
- Department of Medicine, University of Western Ontario, London, Ontario, Canada
| | - Barbara de Graaff
- Menzies Institute for Medical Research, University of Tasmania, Tasmania, Australia
| | | | - Olivier Loréal
- INSERM, Univ. Rennes, INRA, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR_A 1341, UMR_S 1241, F-35000 Rennes, France
| |
Collapse
|
16
|
|
17
|
Achi H, Moukalled N, Mahfouz R, Piperno A, Taher A. Novel mutation in the Transferrin receptor-2 in a patient with Hereditary Hemochromatosis type 3. Meta Gene 2017. [DOI: 10.1016/j.mgene.2017.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
18
|
Nistal M, Paniagua R, González-Peramato P, Reyes-Múgica M. Perspectives in Pediatric Pathology, Chapter 21. Testicular Pathology in Heritable Metabolic Disease. Pediatr Dev Pathol 2017; 19:371-382. [PMID: 25361068 DOI: 10.2350/14-06-1519-pb.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Inborn errors of metabolism have wide and profound effects in many or all organs, and especially so in those with endocrine functions. The testes are greatly affected by systemic metabolic disorders, leading to specific histological findings that generally reveal the nature of the underlying disorder. Here we describe the main testicular changes seen in the setting of metabolic disease.
Collapse
Affiliation(s)
- Manuel Nistal
- 1 Department of Pathology, Hospital La Paz, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo No. 2, Madrid 28029, Spain
| | - Ricardo Paniagua
- 2 Department of Cell Biology, Universidad de Alcala, Madrid, Spain
| | - Pilar González-Peramato
- 1 Department of Pathology, Hospital La Paz, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo No. 2, Madrid 28029, Spain
| | - Miguel Reyes-Múgica
- 3 Department of Pathology, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, One Children's Hospital Drive, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| |
Collapse
|
19
|
Pellegrino RM, Riondato F, Ferbo L, Boero M, Palmieri A, Osella L, Pollicino P, Miniscalco B, Saglio G, Roetto A. Altered Erythropoiesis in Mouse Models of Type 3 Hemochromatosis. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2408941. [PMID: 28540293 PMCID: PMC5433419 DOI: 10.1155/2017/2408941] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 03/01/2017] [Accepted: 04/04/2017] [Indexed: 01/08/2023]
Abstract
Type 3 haemochromatosis (HFE3) is a rare genetic iron overload disease which ultimately lead to compromised organs functioning. HFE3 is caused by mutations in transferrin receptor 2 (TFR2) gene that codes for two main isoforms (Tfr2α and Tfr2β). Tfr2α is one of the hepatic regulators of iron inhibitor hepcidin. Tfr2β is an intracellular isoform of the protein involved in the regulation of iron levels in reticuloendothelial cells. It has been recently demonstrated that Tfr2 is also involved in erythropoiesis. This study aims to further investigate Tfr2 erythropoietic role by evaluating the erythropoiesis of two Tfr2 murine models wherein either one or both of Tfr2 isoforms have been selectively silenced (Tfr2 KI and Tfr2 KO). The evaluations were performed in bone marrow and spleen, in 14 days' and 10 weeks' old mice, to assess erythropoiesis in young versus adult animals. The lack of Tfr2α leads to macrocytosis with low reticulocyte number and increased hemoglobin values, together with an anticipation of adult BM erythropoiesis and an increased splenic erythropoiesis. On the other hand, lack of Tfr2β (Tfr2 KI mice) causes an increased and immature splenic erythropoiesis. Taken together, these data confirm the role of Tfr2α in modulation of erythropoiesis and of Tfr2β in favoring iron availability for erythropoiesis.
Collapse
Affiliation(s)
- R. M. Pellegrino
- Department of Clinical and Biological Sciences, AOU San Luigi Gonzaga, University of Torino, Orbassano, Torino, Italy
| | - F. Riondato
- Department of Veterinary Sciences, University of Torino, Grugliasco, Torino, Italy
| | - L. Ferbo
- Department of Clinical and Biological Sciences, AOU San Luigi Gonzaga, University of Torino, Orbassano, Torino, Italy
| | - M. Boero
- Department of Clinical and Biological Sciences, AOU San Luigi Gonzaga, University of Torino, Orbassano, Torino, Italy
| | - A. Palmieri
- Department of Clinical and Biological Sciences, AOU San Luigi Gonzaga, University of Torino, Orbassano, Torino, Italy
| | - L. Osella
- Department of Veterinary Sciences, University of Torino, Grugliasco, Torino, Italy
| | - P. Pollicino
- Department of Veterinary Sciences, University of Torino, Grugliasco, Torino, Italy
| | - B. Miniscalco
- Department of Veterinary Sciences, University of Torino, Grugliasco, Torino, Italy
| | - G. Saglio
- Department of Clinical and Biological Sciences, AOU San Luigi Gonzaga, University of Torino, Orbassano, Torino, Italy
| | - A. Roetto
- Department of Clinical and Biological Sciences, AOU San Luigi Gonzaga, University of Torino, Orbassano, Torino, Italy
| |
Collapse
|
20
|
Hollerer I, Bachmann A, Muckenthaler MU. Pathophysiological consequences and benefits of HFE mutations: 20 years of research. Haematologica 2017; 102:809-817. [PMID: 28280078 PMCID: PMC5477599 DOI: 10.3324/haematol.2016.160432] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 03/01/2017] [Indexed: 12/15/2022] Open
Abstract
Mutations in the HFE (hemochromatosis) gene cause hereditary hemochromatosis, an iron overload disorder that is hallmarked by excessive accumulation of iron in parenchymal organs. The HFE mutation p.Cys282Tyr is pathologically most relevant and occurs in the Caucasian population with a carrier frequency of up to 1 in 8 in specific European regions. Despite this high prevalence, the mutation causes a clinically relevant phenotype only in a minority of cases. In this review, we summarize historical facts and recent research findings about hereditary hemochromatosis, and outline the pathological consequences of the associated gene defects. In addition, we discuss potential advantages of HFE mutations in asymptomatic carriers.
Collapse
Affiliation(s)
- Ina Hollerer
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Germany
| | | | - Martina U Muckenthaler
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Germany
| |
Collapse
|
21
|
Rishi G, Secondes ES, Wallace DF, Subramaniam VN. Normal systemic iron homeostasis in mice with macrophage-specific deletion of transferrin receptor 2. Am J Physiol Gastrointest Liver Physiol 2016; 310:G171-80. [PMID: 26608187 DOI: 10.1152/ajpgi.00291.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/23/2015] [Indexed: 01/31/2023]
Abstract
Iron is an essential element, since it is a component of many macromolecules involved in diverse physiological and cellular functions, including oxygen transport, cellular growth, and metabolism. Systemic iron homeostasis is predominantly regulated by the liver through the iron regulatory hormone hepcidin. Hepcidin expression is itself regulated by a number of proteins, including transferrin receptor 2 (TFR2). TFR2 has been shown to be expressed in the liver, bone marrow, macrophages, and peripheral blood mononuclear cells. Studies from our laboratory have shown that mice with a hepatocyte-specific deletion of Tfr2 recapitulate the hemochromatosis phenotype of the global Tfr2 knockout mice, suggesting that the hepatic expression of TFR2 is important in systemic iron homeostasis. It is unclear how TFR2 in macrophages contributes to the regulation of iron metabolism. We examined the role of TFR2 in macrophages by analysis of transgenic mice lacking Tfr2 in macrophages by crossing Tfr2(f/f) mice with LysM-Cre mice. Mice were fed an iron-rich diet or injected with lipopolysaccharide to examine the role of macrophage Tfr2 in iron- or inflammation-mediated regulation of hepcidin. Body iron homeostasis was unaffected in the knockout mice, suggesting that macrophage TFR2 is not required for the regulation of systemic iron metabolism. However, peritoneal macrophages of knockout mice had significantly lower levels of ferroportin mRNA and protein, suggesting that TFR2 may be involved in regulating ferroportin levels in macrophages. These studies further elucidate the role of TFR2 in the regulation of iron homeostasis and its role in regulation of ferroportin and thus macrophage iron homeostasis.
Collapse
Affiliation(s)
- Gautam Rishi
- Membrane Transport Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; and School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Eriza S Secondes
- Membrane Transport Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; and
| | - Daniel F Wallace
- Membrane Transport Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; and School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - V Nathan Subramaniam
- Membrane Transport Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; and School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
22
|
Ravasi G, Rausa M, Pelucchi S, Arosio C, Greni F, Mariani R, Pelloni I, Silvestri L, Pineda P, Camaschella C, Piperno A. Transferrin receptor 2 mutations in patients with juvenile hemochromatosis phenotype. Am J Hematol 2015; 90:E226-7. [PMID: 26408288 DOI: 10.1002/ajh.24202] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 08/31/2015] [Accepted: 09/17/2015] [Indexed: 01/06/2023]
Affiliation(s)
- Giulia Ravasi
- Department of Health Sciences; University of Milano-Bicocca; Monza Italy
| | - Marco Rausa
- Division of Genetics and Cell Biology; San Raffaele Scientific Institute, Vita-Salute University; Milan Italy
| | - Sara Pelucchi
- Department of Health Sciences; University of Milano-Bicocca; Monza Italy
| | | | - Federico Greni
- Department of Health Sciences; University of Milano-Bicocca; Monza Italy
| | - Raffaella Mariani
- Unit of Internal Medicine 2, Centre for Iron Metabolism Disorders, S. Gerardo Hospital; Monza Italy
| | - Irene Pelloni
- Unit of Internal Medicine 2, Centre for Iron Metabolism Disorders, S. Gerardo Hospital; Monza Italy
| | - Laura Silvestri
- Division of Genetics and Cell Biology; San Raffaele Scientific Institute, Vita-Salute University; Milan Italy
| | - Pedro Pineda
- Seccíon Endocrinología y Diabetes; Departamento de Medicina; Hospital Clínico Universidad de Chile; Santiago Chile
| | - Clara Camaschella
- Division of Genetics and Cell Biology; San Raffaele Scientific Institute, Vita-Salute University; Milan Italy
| | - Alberto Piperno
- Department of Health Sciences; University of Milano-Bicocca; Monza Italy
- Consortium of Human Molecular Genetics; Monza Italy
- Unit of Internal Medicine 2, Centre for Iron Metabolism Disorders, S. Gerardo Hospital; Monza Italy
| |
Collapse
|
23
|
Abstract
Transferrin receptor 2 (TFR2) contributes to hepcidin regulation in the liver and associates with erythropoietin receptor in erythroid cells. Nevertheless, TFR2 mutations cause iron overload (hemochromatosis type 3) without overt erythroid abnormalities. To clarify TFR2 erythroid function, we generated a mouse lacking Tfr2 exclusively in the bone marrow (Tfr2(BMKO)). Tfr2(BMKO) mice have normal iron parameters, reduced hepcidin levels, higher hemoglobin and red blood cell counts, and lower mean corpuscular volume than normal control mice, a phenotype that becomes more evident in iron deficiency. In Tfr2(BMKO) mice, the proportion of nucleated erythroid cells in the bone marrow is higher and the apoptosis lower than in controls, irrespective of comparable erythropoietin levels. Induction of moderate iron deficiency increases erythroblasts number, reduces apoptosis, and enhances erythropoietin (Epo) levels in controls, but not in Tfr2(BMKO) mice. Epo-target genes such as Bcl-xL and Epor are highly expressed in the spleen and in isolated erythroblasts from Tfr2(BMKO) mice. Low hepcidin expression in Tfr2(BMKO) is accounted for by erythroid expansion and production of the erythroid regulator erythroferrone. We suggest that Tfr2 is a component of a novel iron-sensing mechanism that adjusts erythrocyte production according to iron availability, likely by modulating the erythroblast Epo sensitivity.
Collapse
|
24
|
Bardou-Jacquet E, Ben Ali Z, Beaumont-Epinette MP, Loreal O, Jouanolle AM, Brissot P. Non-HFE hemochromatosis: pathophysiological and diagnostic aspects. Clin Res Hepatol Gastroenterol 2014; 38:143-54. [PMID: 24321703 DOI: 10.1016/j.clinre.2013.11.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 11/04/2013] [Accepted: 11/13/2013] [Indexed: 02/04/2023]
Abstract
Rare genetic iron overload diseases are an evolving field due to major advances in genetics and molecular biology. Genetic iron overload has long been confined to the classical type 1 hemochromatosis related to the HFE C282Y mutation. Breakthroughs in the understanding of iron metabolism biology and molecular mechanisms led to the discovery of new genes and subsequently, new types of hemochromatosis. To date, four types of hemochromatosis have been identified: HFE-related or type1 hemochromatosis, the most frequent form in Caucasians, and four rare types, named type 2 (A and B) hemochromatosis (juvenile hemochromatosis due to hemojuvelin and hepcidin mutation), type 3 hemochromatosis (related to transferrin receptor 2 mutation), and type 4 (A and B) hemochromatosis (ferroportin disease). The diagnosis relies on the comprehension of the involved physiological defect that can now be explored by biological and imaging tools, which allow non-invasive assessment of iron metabolism. A multidisciplinary approach is essential to support the physicians in the diagnosis and management of those rare diseases.
Collapse
Affiliation(s)
- Edouard Bardou-Jacquet
- University Hospital of Rennes, French reference center for rare iron overload diseases of genetic origin, Rennes, France; University of Rennes1, Inserm UMR 991, 35000 Rennes, France; University Hospital of Rennes, Liver disease department, Rennes, France.
| | - Zeineb Ben Ali
- University Hospital of Rennes, French reference center for rare iron overload diseases of genetic origin, Rennes, France; University Hospital of Rennes, Liver disease department, Rennes, France
| | - Marie-Pascale Beaumont-Epinette
- University Hospital of Rennes, French reference center for rare iron overload diseases of genetic origin, Rennes, France; University Hospital of Rennes, Molecular Genetics Department, Rennes, France
| | - Olivier Loreal
- University Hospital of Rennes, French reference center for rare iron overload diseases of genetic origin, Rennes, France; University of Rennes1, Inserm UMR 991, 35000 Rennes, France
| | - Anne-Marie Jouanolle
- University Hospital of Rennes, French reference center for rare iron overload diseases of genetic origin, Rennes, France; University Hospital of Rennes, Molecular Genetics Department, Rennes, France
| | - Pierre Brissot
- University Hospital of Rennes, French reference center for rare iron overload diseases of genetic origin, Rennes, France; University of Rennes1, Inserm UMR 991, 35000 Rennes, France; University Hospital of Rennes, Liver disease department, Rennes, France
| |
Collapse
|
25
|
Nai A, Pellegrino RM, Rausa M, Pagani A, Boero M, Silvestri L, Saglio G, Roetto A, Camaschella C. The erythroid function of transferrin receptor 2 revealed by Tmprss6 inactivation in different models of transferrin receptor 2 knockout mice. Haematologica 2014; 99:1016-21. [PMID: 24658816 DOI: 10.3324/haematol.2013.103143] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Transferrin receptor 2 (TFR2) is a transmembrane glycoprotein expressed in the liver and in the erythroid compartment, mutated in a form of hereditary hemochromatosis. Hepatic TFR2, together with HFE, activates the transcription of the iron-regulator hepcidin, while erythroid TFR2 is a member of the erythropoietin receptor complex. The TMPRSS6 gene, encoding the liver-expressed serine protease matriptase-2, is the main inhibitor of hepcidin and inactivation of TMPRSS6 leads to iron deficiency with high hepcidin levels. Here we evaluate the phenotype resulting from the genetic loss of Tmprss6 in Tfr2 total (Tfr2(-/-)) and liver-specific (Tfr2(LCKO)) knockout mice. Tmprss6(-/-)Tfr2(-/-) and Tmprss6(-/-)Tfr2(LCKO) mice have increased hepcidin levels and show iron-deficiency anemia like Tmprss6(-/-)mice. However, while Tmprss6(-/-)Tfr2(LCKO) are phenotypically identical to Tmprss6(-/-) mice, Tmprss6(-/-)Tfr2(-/-) mice have increased red blood cell count and more severe microcytosis than Tmprss6(-/-) mice. In addition hepcidin expression in Tmprss6(-/-)Tfr2(-/-) mice is higher than in the wild-type animals, but lower than in Tmprss6(-/-) mice, suggesting partial inhibition of the hepcidin activating pathway. Our results prove that hepatic TFR2 acts upstream of TMPRSS6. In addition Tfr2 deletion causes a relative erythrocytosis in iron-deficient mice, which likely attenuates the effect of over-expression of hepcidin in Tmprss6(-/-) mice. Since liver-specific deletion of Tfr2 in Tmprss6(-/-) mice does not modify the erythrocyte count, we speculate that loss of Tfr2 in the erythroid compartment accounts for the hematologic phenotype of Tmprss6(-/-)Tfr2(-/-) mice. We propose that TFR2 is a limiting factor for erythropoiesis, particularly in conditions of iron restriction.
Collapse
Affiliation(s)
- Antonella Nai
- Vita Salute University and San Raffaele Scientific Institute, Division of Genetics and Cell Biology, Milan, Italy
| | - Rosa M Pellegrino
- Department of Clinical and Biological Sciences, University of Torino, Italy
| | - Marco Rausa
- Vita Salute University and San Raffaele Scientific Institute, Division of Genetics and Cell Biology, Milan, Italy
| | - Alessia Pagani
- Vita Salute University and San Raffaele Scientific Institute, Division of Genetics and Cell Biology, Milan, Italy
| | - Martina Boero
- Department of Clinical and Biological Sciences, University of Torino, Italy
| | - Laura Silvestri
- Vita Salute University and San Raffaele Scientific Institute, Division of Genetics and Cell Biology, Milan, Italy
| | - Giuseppe Saglio
- Department of Clinical and Biological Sciences, University of Torino, Italy
| | - Antonella Roetto
- Department of Clinical and Biological Sciences, University of Torino, Italy
| | - Clara Camaschella
- Vita Salute University and San Raffaele Scientific Institute, Division of Genetics and Cell Biology, Milan, Italy
| |
Collapse
|
26
|
Worthen CA, Enns CA. The role of hepatic transferrin receptor 2 in the regulation of iron homeostasis in the body. Front Pharmacol 2014; 5:34. [PMID: 24639653 PMCID: PMC3944196 DOI: 10.3389/fphar.2014.00034] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 02/18/2014] [Indexed: 12/22/2022] Open
Abstract
Fine-tuning of body iron is required to prevent diseases such as iron-overload and anemia. The putative iron sensor, transferrin receptor 2 (TfR2), is expressed in the liver and mutations in this protein result in the iron-overload disease Type III hereditary hemochromatosis (HH). With the loss of functional TfR2, the liver produces about 2-fold less of the peptide hormone hepcidin, which is responsible for negatively regulating iron uptake from the diet. This reduction in hepcidin expression leads to the slow accumulation of iron in the liver, heart, joints, and pancreas and subsequent cirrhosis, heart disease, arthritis, and diabetes. TfR2 can bind iron-loaded transferrin (Tf) in the bloodstream, and hepatocytes treated with Tf respond with a 2-fold increase in hepcidin expression through stimulation of the bone morphogenetic protein (BMP)-signaling pathway. Loss of functional TfR2 or its binding partner, the original HH protein, results in a loss of this transferrin-sensitivity. While much is known about the trafficking and regulation of TfR2, the mechanism of its transferrin-sensitivity through the BMP-signaling pathway is still not known.
Collapse
Affiliation(s)
- Christal A Worthen
- Department of Cell and Developmental Biology, Oregon Health and Science University Portland, OR, USA
| | - Caroline A Enns
- Department of Cell and Developmental Biology, Oregon Health and Science University Portland, OR, USA
| |
Collapse
|
27
|
TFR2-related hereditary hemochromatosis as a frequent cause of primary iron overload in patients from Central-Southern Italy. Blood Cells Mol Dis 2014; 52:83-7. [DOI: 10.1016/j.bcmd.2013.08.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 07/23/2013] [Indexed: 11/21/2022]
|
28
|
Brissot P, Bardou-Jacquet E, Troadec MB, Mosser A, Island ML, Detivaud L, Loréal O, Jouanolle AM. Molecular diagnosis of genetic iron-overload disorders. Expert Rev Mol Diagn 2014; 10:755-63. [DOI: 10.1586/erm.10.55] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
29
|
Assi TB, Baz E. Current applications of therapeutic phlebotomy. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2014; 12 Suppl 1:s75-83. [PMID: 24120605 PMCID: PMC3934278 DOI: 10.2450/2013.0299-12] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 05/20/2013] [Indexed: 12/19/2022]
Affiliation(s)
- Tarek Bou Assi
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Centre, Beirut, Lebanon
| | - Elizabeth Baz
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Centre, Beirut, Lebanon
| |
Collapse
|
30
|
Goetze O, Schmitt J, Spliethoff K, Theurl I, Weiss G, Swinkels DW, Tjalsma H, Maggiorini M, Krayenbühl P, Rau M, Fruehauf H, Wojtal KA, Müllhaupt B, Fried M, Gassmann M, Lutz T, Geier A. Adaptation of iron transport and metabolism to acute high-altitude hypoxia in mountaineers. Hepatology 2013; 58:2153-62. [PMID: 23787477 DOI: 10.1002/hep.26581] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 06/06/2013] [Indexed: 12/19/2022]
Abstract
UNLABELLED Human iron homeostasis is regulated by intestinal iron transport, hepatic hepcidin release, and signals from pathways that consume or supply iron. The aim of this study was to characterize the adaptation of iron homeostasis under hypoxia in mountaineers at the levels of (1) hepatic hepcidin release, (2) intestinal iron transport, and (3) systemic inflammatory and erythropoietic responses. Twenty-five healthy mountaineers were studied. Blood samples and duodenal biopsies were taken at baseline of 446 m as well as on day 2 (MG2) and 4 (MG4) after rapid ascent to 4559 m. Divalent metal-ion transporter 1 (DMT-1), ferroportin 1 (FP-1) messenger RNA (mRNA), and protein expression were analyzed in biopsy specimens by quantitative reverse-transcription polymerase chain reaction (RT-PCR) and immunohistochemistry. Serum hepcidin levels were analyzed by mass spectrometry. Serum iron, ferritin, transferrin, interleukin (IL)-6, and C-reactive protein (CRP) were quantified by standard techniques. Serum erythropoietin and growth differentiation factor 15 (GDF15) levels were measured by enzyme-linked immunosorbent assay (ELISA). Under hypoxia, erythropoietin peaked at MG2 (P < 0.001) paralleled by increased GDF15 on MG2 (P < 0.001). Serum iron and ferritin levels declined rapidly on MG2 and MG4 (P < 0.001). Duodenal DMT-1 and FP-1 mRNA expression increased up to 10-fold from baseline on MG2 and MG4 (P < 0.001). Plasma CRP increased on MG2 and MG4, while IL-6 only increased on MG2 (P < 0.001). Serum hepcidin levels decreased at high altitude on MG2 and MG4 (P < 0.001). CONCLUSION This study in healthy volunteers showed that under hypoxemic conditions hepcidin is repressed and duodenal iron transport is rapidly up-regulated. These changes may increase dietary iron uptake and allow release of stored iron to ensure a sufficient iron supply for hypoxia-induced compensatory erythropoiesis.
Collapse
Affiliation(s)
- Oliver Goetze
- Division of Gastroenterology & Hepatology, University Hospital Zurich, Switzerland; Division of Hepatology, Department of Medicine II, University Hospital Würzburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Zhao N, Enns CA. N-linked glycosylation is required for transferrin-induced stabilization of transferrin receptor 2, but not for transferrin binding or trafficking to the cell surface. Biochemistry 2013; 52:3310-9. [PMID: 23556518 PMCID: PMC3656769 DOI: 10.1021/bi4000063] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Transferrin receptor 2 (TfR2) is
a member of the transferrin receptor-like
family of proteins. Mutations in TfR2 can lead to a rare form of the
iron overload disease, hereditary hemochromatosis. TfR2 is proposed
to sense body iron levels and increase the level of expression of
the iron regulatory hormone, hepcidin. Human TfR2 (hTfR2) contains
four potential Asn-linked (N-linked) glycosylation sites on its ectodomain.
The importance of glycosylation in TfR2 function has not been elucidated.
In this study, by employing site-directed mutagenesis to remove glycosylation
sites of hTfR2 individually or in combination, we found that hTfR2
was glycosylated at Asn 240, 339, and 754, while the consensus sequence
for N-linked glycosylation at Asn 540 was not utilized. Cell surface
protein biotinylation and biotin-labeled Tf indicated that in the
absence of N-linked oligosaccharides, hTfR2 still moved to the plasma
membrane and bound its ligand, holo-Tf. However, without N-linked
glycosylation, hTfR2 did not form the intersubunit disulfide bonds
as efficiently as the wild type (WT). Moreover, the unglycosylated
form of hTfR2 could not be stabilized by holo-Tf. We further provide
evidence that the unglycosylated hTfR2 behaved in manner different
from that of the WT in response to holo-Tf treatment. Thus, the putative
iron-sensing function of TfR2 could not be achieved in the absence
of N-linked oligosaccharides. On the basis of our analyses, we conclude
that unlike TfR1, N-linked glycosylation is dispensable for the cell
surface expression and holo-Tf binding, but it is required for efficient
intersubunit disulfide bond formation and holo-Tf-induced stabilization
of TfR2.
Collapse
Affiliation(s)
- Ningning Zhao
- Department of Cell and Developmental Biology, Oregon Health & Science University , Portland, Oregon 97239, United States
| | | |
Collapse
|
32
|
Abstract
INTRODUCTION The discovery of hemochromatosis genes and the availability of molecular-genetic tests considerably modified the knowledge of the disease relative to physiopathology, penetrance, and expression, and had major impact in the diagnostic settings. AREAS COVERED Hemochromatosis is a heterogenous disorder at both genetic and phenotypic level. The review discusses criteria to define patients' iron phenotype and to use molecular tests to diagnose HFE-related and non-HFE hemochromatosis. The material examined includes articles published in the journals covered by PubMed US National Library of Medicine. The author has been working in the field of iron overload diseases for several years and has contributed 18 of the papers cited in the references. EXPERT OPINION Hemochromatosis genotyping is inseparable from phenotype characterization. A full clinical assessment is needed and DNA test performed when data suggest a clear indication of suspicion of being at risk for HH. HFE testing for p.Cys282Tyr mutation and p.His63Asp variant is the first molecular diagnostic step. Genotyping for rare mutations can be offered to patients with negative first-level HFE testing who have iron overload with no other explanation and should be performed in referral centers for iron overload disorders that can provide genetic advice and in-house genotyping services.
Collapse
Affiliation(s)
- Alberto Piperno
- University of Milano-Bicocca, Centre for the Diagnosis and Treatment of Hemochromatosis and Iron Disorders, S.Gerardo Hospital, Department of Health Sciences, Monza, Italy.
| |
Collapse
|
33
|
Acikyol B, Graham RM, Trinder D, House MJ, Olynyk JK, Scott RJ, Milward EA, Johnstone DM. Brain transcriptome perturbations in the transferrin receptor 2 mutant mouse support the case for brain changes in iron loading disorders, including effects relating to long-term depression and long-term potentiation. Neuroscience 2013; 235:119-28. [PMID: 23333676 DOI: 10.1016/j.neuroscience.2013.01.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 12/14/2012] [Accepted: 01/02/2013] [Indexed: 11/16/2022]
Abstract
Iron abnormalities within the brain are associated with several rare but severe neurodegenerative conditions. There is growing evidence that more common systemic iron loading disorders such as hemochromatosis can also have important effects on the brain. To identify features that are common across different forms of hemochromatosis, we used microarray and real-time reverse transcription polymerase chain reaction (RT-PCR) to assess brain transcriptome profiles of transferrin receptor 2 mutant mice (Tfr2(mut)), a model of a rare type of hereditary hemochromatosis, relative to wildtype control mice. The results were compared with our previous findings in dietary iron-supplemented wildtype mice and Hfe(-/-) mice, a model of a common type of hereditary hemochromatosis. For transcripts showing significant changes relative to controls across all three models, there was perfect (100%) directional concordance (i.e. transcripts were increased in all models or decreased in all models). Comparison of the two models of hereditary hemochromatosis, which showed more pronounced changes than the dietary iron-supplemented mice, revealed numerous common molecular effects. Pathway analyses highlighted changes for genes relating to long-term depression (6.8-fold enrichment, p=5.4×10(-7)) and, to a lesser extent, long-term potentiation (3.7-fold enrichment, p=0.01), with generalized reductions in transcription of key genes from these pathways, which are involved in modulating synaptic strength and efficacy and are essential for memory and learning. The agreement across the models suggests the findings are robust and strengthens previous evidence that iron loading disorders affect the brain. Perturbations of brain phenomena such as long-term depression and long-term potentiation might partly explain neurologic symptoms reported for some hemochromatosis patients.
Collapse
Affiliation(s)
- B Acikyol
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Ding K, Shameer K, Jouni H, Masys DR, Jarvik GP, Kho AN, Ritchie MD, McCarty CA, Chute CG, Manolio TA, Kullo IJ. Genetic Loci implicated in erythroid differentiation and cell cycle regulation are associated with red blood cell traits. Mayo Clin Proc 2012; 87:461-74. [PMID: 22560525 PMCID: PMC3538470 DOI: 10.1016/j.mayocp.2012.01.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 01/10/2012] [Accepted: 01/19/2012] [Indexed: 01/16/2023]
Abstract
OBJECTIVE To identify common genetic variants influencing red blood cell (RBC) traits. PATIENTS AND METHODS We performed a genomewide association study from June 2008 through July 2011 of hemoglobin, hematocrit, RBC count, mean corpuscular volume, mean corpuscular hemoglobin, and mean corpuscular hemoglobin concentration in 12,486 patients of European ancestry from the electronic MEdical Records and Genomics (eMERGE) network. We developed an electronic medical record-based algorithm that included individuals who had RBC measurements obtained for clinical care and excluded values measured in the setting of hematopoietic disorders, comorbid conditions, or medications known to affect RBC production or a recent history of blood loss. RESULTS We identified 4 new genetic loci and replicated 11 loci previously reported to be associated with one or more RBC traits in individuals of European ancestry. Notably, genes present in 3 of the 4 newly identified loci (THRB, PTPLAD1, CDT1) and in 6 of the 11 replicated loci (KLF1, ALDH8A1, CCND3, SPTA1, FBXO7, TFR2/EPO) are implicated in erythroid differentiation and regulation of cell cycle in hematopoietic stem cells. CONCLUSION Genes in the erythroid differentiation and cell cycle regulation pathways influence interindividual variation in RBC indices. Our results provide insights into the molecular basis underlying variation in RBC traits.
Collapse
Key Words
- emerge, electronic medical records and genomics
- emmax, mixed-model association-expedited
- emr, electronic medical record
- eqtl, expression quantitative trait locus
- ghc, group health cooperative--university of washington
- gwas, genomewide association study
- hct, hematocrit
- hgb, hemoglobin
- ibs, identity-by-state
- ld, linkage disequilibrium
- mc, marshfield clinic
- mch, mean corpuscular hemoglobin
- mchc, mean corpuscular hemoglobin concentration
- mcv, mean corpuscular volume
- mim, mendelian inheritance of man
- nu, northwestern university
- rbc, red blood cell
- snp, single-nucleotide polymorphism
- vumc, vanderbilt university medical center
Collapse
Affiliation(s)
- Keyue Ding
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - Khader Shameer
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - Hayan Jouni
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - Daniel R. Masys
- Division of Biomedical and Health Informatics, Department of Medical Education and Biomedical Informatics, University of Washington, Seattle
| | - Gail P. Jarvik
- Department of Medicine (Medical Genetics) and Department of Genome Sciences, University of Washington, Seattle
| | - Abel N. Kho
- Department of Medicine, Northwestern University, Chicago, IL
| | - Marylyn D. Ritchie
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park
| | | | | | - Teri A. Manolio
- Office of Population Genomics, National Human Genome Research Institute, Bethesda, MD
| | - Iftikhar J. Kullo
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
- Correspondence: Address to Iftikhar J. Kullo, MD, Mayo Clinic, 200 First St SW, Rochester, MN 55905
| |
Collapse
|
35
|
Siddique A, Kowdley KV. Review article: the iron overload syndromes. Aliment Pharmacol Ther 2012; 35:876-93. [PMID: 22385471 DOI: 10.1111/j.1365-2036.2012.05051.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 06/26/2011] [Accepted: 02/08/2012] [Indexed: 12/12/2022]
Abstract
BACKGROUND Iron overload syndromes encompass a wide range of hereditary and acquired conditions. Major developments in the field of genetics and the discovery of hepcidin as a central regulator of iron homeostasis have greatly increased our understanding of the pathophysiology of iron overload syndromes. AIM To review advances in iron regulation and iron overload syndrome with special emphasis on hereditary haemochromatosis, the prototype iron overload syndrome. METHODS A PubMed search using words such as 'iron overload', 'hemochromatosis', 'HFE', 'Non-HFE', 'secondary iron overload' was undertaken. RESULTS Iron overload is associated with significant morbidity and mortality. Sensitive diagnostic tests and effective therapy are widely available and can prevent complications associated with iron accumulation in end- organs. Therapeutic phlebotomy remains the cornerstone of therapy for removal of excess body iron, but novel therapeutic agents including oral iron chelators have been developed for iron overload associated with anaemia. CONCLUSIONS Iron overload disorders are common. Inexpensive screening tests as well as confirmatory diagnostic tests are widely available. Increased awareness of the causes and importance of early diagnosis and knowledge of the appropriate use of genetic testing are encouraged. The availability of novel treatments should increase therapeutic options for patients with iron overload disorders.
Collapse
Affiliation(s)
- A Siddique
- Department of Hepatology, Virginia Mason Medical Center, Seattle, WA, USA.
| | | |
Collapse
|
36
|
Affiliation(s)
- Robert E Fleming
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, USA
| | | |
Collapse
|
37
|
Iron transport machinery of human cells: players and their interactions. CURRENT TOPICS IN MEMBRANES 2012; 69:67-93. [PMID: 23046647 DOI: 10.1016/b978-0-12-394390-3.00003-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Organisms, like cells, maintain tight control of iron. In humans as well as other mammals, control is achieved through the regulation of iron uptake into the body rather than through the excretion of iron. The mechanisms by which humans and mice regulate both iron uptake and the distribution of iron within the body and cells are reviewed. Special emphasis is given to the iron transporters involved in this process.
Collapse
|
38
|
Transferrin receptor 2 is frequently and highly expressed in glioblastomas. Transl Oncol 2011; 3:123-34. [PMID: 20360937 DOI: 10.1593/tlo.09274] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 09/15/2009] [Accepted: 11/11/2009] [Indexed: 12/19/2022] Open
Abstract
Under physiological conditions, transferrin receptor 2 (TfR2) is expressed in the liver and its balance is related to the cell cycle rather than to intracellular iron levels. We recently showed that TfR2 is highly expressed in glioblastoma cell lines. Here, we demonstrate that, in these cells, TfR2 appears to localize in lipid rafts, induces extracellular signal-regulated kinase 1/2 phosphorylation after transferrin binding, and contributes to cell proliferation, as shown by RNA silencing experiments. In vitro hypoxic conditions induce a significant TfR2 up-regulation, suggesting a role in tumor angiogenesis. As assessed by immunohistochemistry, the level of TfR2 expression in astrocytic tumors is related to histologic grade, with the highest expression observed in glioblastomas. The level of TfR2 expression represents a favorable prognostic factor, which is associated with the higher sensitivity to temozolomide of TfR2-positive tumor cells in vitro. The endothelial cells of glioblastoma vasculature also stain for TfR2, whereas those of the normal brain vessels do not. Importantly, TfR2 is expressed by the subpopulation of glioblastoma cells with properties of cancer-initiating cells. TfR2-positive glioblastoma cells retain their TfR2 expression on xenografting in immunodeficient mice. In conclusion, our observations demonstrate that TfR2 is a neoantigen for astrocytomas that seems attractive for developing target therapies.
Collapse
|
39
|
Bleackley MR, Macgillivray RTA. Transition metal homeostasis: from yeast to human disease. Biometals 2011; 24:785-809. [PMID: 21479832 DOI: 10.1007/s10534-011-9451-4] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 03/28/2011] [Indexed: 12/16/2022]
Abstract
Transition metal ions are essential nutrients to all forms of life. Iron, copper, zinc, manganese, cobalt and nickel all have unique chemical and physical properties that make them attractive molecules for use in biological systems. Many of these same properties that allow these metals to provide essential biochemical activities and structural motifs to a multitude of proteins including enzymes and other cellular constituents also lead to a potential for cytotoxicity. Organisms have been required to evolve a number of systems for the efficient uptake, intracellular transport, protein loading and storage of metal ions to ensure that the needs of the cells can be met while minimizing the associated toxic effects. Disruptions in the cellular systems for handling transition metals are observed as a number of diseases ranging from hemochromatosis and anemias to neurodegenerative disorders including Alzheimer's and Parkinson's disease. The yeast Saccharomyces cerevisiae has proved useful as a model organism for the investigation of these processes and many of the genes and biological systems that function in yeast metal homeostasis are conserved throughout eukaryotes to humans. This review focuses on the biological roles of iron, copper, zinc, manganese, nickel and cobalt, the homeostatic mechanisms that function in S. cerevisiae and the human diseases in which these metals have been implicated.
Collapse
Affiliation(s)
- Mark R Bleackley
- Department of Biochemistry and Molecular Biology, Centre for Blood Research, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T1Z3, Canada
| | | |
Collapse
|
40
|
Speeckaert MM, Speeckaert R, Delanghe JR. Biological and clinical aspects of soluble transferrin receptor. Crit Rev Clin Lab Sci 2011; 47:213-28. [DOI: 10.3109/10408363.2010.550461] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
41
|
Deugnier Y, Lainé F, Le Lan C, Bardou-Jacquet E, Jouanolle AM, Brissot P. Hémochromatoses et autres surcharges hépatiques en fer. ACTA ACUST UNITED AC 2011. [DOI: 10.1016/s1155-1976(11)40364-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
42
|
Transferrin receptor 2 is a component of the erythropoietin receptor complex and is required for efficient erythropoiesis. Blood 2010; 116:5357-67. [PMID: 20826723 DOI: 10.1182/blood-2010-04-281360] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Erythropoietin (Epo) is required for erythroid progenitor differentiation. Although Epo crosslinking experiments have revealed the presence of Epo receptor (EpoR)-associated proteins that could never be identified, EpoR is considered to be a paradigm for homodimeric cytokine receptors. We purified EpoR-binding partners and identified the type 2 transferrin receptor (TfR2) as a component of the EpoR complex corresponding to proteins previously detected in cross-linking experiments. TfR2 is involved in iron metabolism by regulating hepcidin production in liver cells. We show that TfR2 and EpoR are synchronously coexpressed during the differentiation of erythroid progenitors. TfR2 associates with EpoR in the endoplasmic reticulum and is required for the efficient transport of this receptor to the cell surface. Erythroid progenitors from TfR2(-/-)mice show a decreased sensitivity to Epo and increased circulating Epo levels. In human erythroid progenitors, TfR2 knockdown delays the terminal differentiation. Erythroid cells produce growth differentiation factor-15, a cytokine that suppresses hepatic hepcidin production in certain erythroid diseases such as thalassemia. We show that the production of growth differentiation factor-15 by erythroid cells is dependent on both Epo and TfR2. Taken together, our results show that TfR2 exhibits a non hepatic function as a component of the EpoR complex and is required for efficient erythropoiesis.
Collapse
|
43
|
Characterization of the interaction between diferric transferrin and transferrin receptor 2 by functional assays and atomic force microscopy. J Mol Biol 2010; 397:375-84. [PMID: 20096706 DOI: 10.1016/j.jmb.2010.01.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Revised: 12/29/2009] [Accepted: 01/13/2010] [Indexed: 10/19/2022]
Abstract
Transferrin receptor 2 (TfR2), a homologue of the classical transferrin receptor 1 (TfR1), is found in two isoforms, alpha and beta. Like TfR1, TfR2alpha is a type II membrane protein, but the beta form lacks transmembrane portions and therefore is likely to be an intracellular protein. To investigate the functional properties of TfR2alpha, we expressed the protein with FLAG tagging in transferrin-receptor-deficient Chinese hamster ovary cells. The association constant for the binding of diferric transferrin (Tf) to TfR2alpha is 5.6x10(6) M(-)(1), which is about 50 times lower than that for the binding of Tf to TfR1, with correspondingly reduced rates of iron uptake. Evidence for Tf internalization and recycling via TfR2alpha without degradation, as in the TfR1 pathway, was also found. The interaction of TfR2alpha with Tf was further investigated using atomic force microscopy, a powerful tool used for investigating the interaction between a ligand and its receptor at the single-molecule level on the living cell surface. Dynamic force microscopy reveals a difference in the interactions of Tf with TfR2alpha and TfR1, with Tf-TfR1 unbinding characterized by two energy barriers, while only one is present for Tf-TfR2. We speculate that this difference may reflect Tf binding to TfR2alpha by a single lobe, whereas two lobes of Tf participate in binding to TfR1. The difference in the binding properties of Tf to TfR1 and TfR2alpha may help account for the different physiological roles of the two receptors.
Collapse
|
44
|
Santos PCJL, Cançado RD, Terada CT, Rostelato S, Gonzales I, Hirata RDC, Hirata MH, Chiattone CS, Guerra-Shinohara EM. HFE gene mutations and iron status of Brazilian blood donors. Braz J Med Biol Res 2009; 43:107-14. [PMID: 20027482 DOI: 10.1590/s0100-879x2009007500031] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Accepted: 12/04/2009] [Indexed: 12/15/2022] Open
Abstract
Mutations of the HFE and TFR2 genes have been associated with iron overload. HFE and TFR2 mutations were assessed in blood donors, and the relationship with iron status was evaluated. Subjects (N = 542) were recruited at the Hemocentro da Santa Casa de São Paulo, São Paulo, Brazil. Iron status was not influenced by HFE mutations in women and was independent of blood donation frequency. In contrast, men carrying the HFE 282CY genotype had lower total iron-binding capacity (TIBC) than HFE 282CC genotype carriers. Men who donated blood for the first time and were carriers of the HFE 282CY genotype had higher transferrin saturation values and lower TIBC concentrations than those with the homozygous wild genotype for the HFE C282Y mutation. Moreover, in this group of blood donors, carriers of HFE 63DD plus 63HD genotypes had higher serum ferritin values than those with the homozygous wild genotype for HFE H63D mutation. Multiple linear regression analysis showed that HFE 282CY leads to a 17.21% increase (P = 0.018) and a 83.65% decrease (P = 0.007) in transferrin saturation and TIBC, respectively. In addition, serum ferritin is influenced by age (3.91%, P = 0.001) and the HFE 63HD plus DD genotype (55.84%, P = 0.021). In conclusion, the HFE 282Y and 65C alleles were rare, while the HFE 63D allele was frequent in Brazilian blood donors. The HFE C282Y and H63D mutations were associated with alterations in iron status in blood donors in a gender-dependent manner.
Collapse
Affiliation(s)
- P C J L Santos
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, SP, Brasil
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Calzolari A, Deaglio S, Maldi E, Cassoni P, Malavasi F, Testa U. TfR2 expression in human colon carcinomas. Blood Cells Mol Dis 2009; 43:243-9. [PMID: 19729324 DOI: 10.1016/j.bcmd.2009.08.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Accepted: 08/04/2009] [Indexed: 02/09/2023]
Abstract
Different proteins regulate iron metabolism at the level of various tissues. Among these is a second transferrin receptor (TfR2) that seems to play a key role in the regulation of iron homeostasis. Although TfR2 expression in normal tissues is restricted at the level of the liver, we observed that TfR2 is frequently expressed in cancer cell lines. Taking advantage of this observation we investigated TfR2 expression in primary colon cancers, and showed that this receptor is expressed in about 26% of cases. TfR2 expression in colon cancer is not related to histological grade, but is preferentially associated with mucinous tumors. In colon cancer cell lines, TfR2 is localized in membrane lipid rafts, induces ERK1/ERK2 phosphorylation, when activated by its ligand transferring, and is preferentially expressed during S-M phases of the cell cycle. The presence of TfR2 on the membrane of colon cancer cells may contribute the growth advantage to these cells.
Collapse
Affiliation(s)
- Alessia Calzolari
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | | | | | | | | | | |
Collapse
|
46
|
Brissot P. Les hémochromatoses. Nouvelle compréhension, nouveaux traitements. ACTA ACUST UNITED AC 2009; 33:859-67. [DOI: 10.1016/j.gcb.2009.04.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
47
|
Lee PL, Beutler E. Regulation of hepcidin and iron-overload disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2009; 4:489-515. [PMID: 19400694 DOI: 10.1146/annurev.pathol.4.110807.092205] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hepcidin, a 25-amino-acid antimicrobial peptide, is the central regulator of iron homeostasis. Hepcidin transcription is upregulated by inflammatory cytokines, iron, and bone morphogenetic proteins and is downregulated by iron deficiency, ineffective erythropoiesis, and hypoxia. The iron transporter ferroportin is the cognate receptor of hepcidin and is destroyed as a result of interaction with the peptide. Except for inherited defects of ferroportin and hepcidin itself, all forms of iron-storage disease appear to arise from hepcidin dysregulation. Studies using multiple approaches have begun to delineate the molecular mechanisms that regulate hepcidin expression, particularly at the transcriptional level. Knowledge of the regulation of hepcidin by inflammation, iron, erythropoiesis, and hypoxia will lead to an understanding of the pathogenesis of primary hemochromatosis, secondary iron overload, and anemia of inflammatory disease.
Collapse
Affiliation(s)
- Pauline L Lee
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | |
Collapse
|
48
|
Gan EK, Trinder D, Ayonrinde OT, Olynyk JK. Genetics of hereditary hemochromatosis: a clinical perspective. Expert Rev Endocrinol Metab 2009; 4:225-239. [PMID: 30743791 DOI: 10.1586/eem.09.9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Hereditary hemochromatosis due to homozygosity for the C282Y mutation in the HFE gene product is the most common autosomal recessive genetic disorder in populations of northern European descent, where it attains a maximum prevalence of approximately one in 200. Cross-sectional and longitudinal studies have revealed that clinically significant iron-overload disease develops in at least 28% of male and 1% of female HFE C282Y homozygotes. The relatively low clinical penetrance is largely unexplained. Current evidence suggests a limited role for digenic inheritance of mutations in iron homeostasis genes in modifying the penetrance of hemochromatosis. Male gender is a strong genetic factor, promoting expression of clinical disease. Dietary intake of alcohol and noncitrus fruit may also act as important environmental modifiers of penetrance. With genetic analyses becoming simpler to perform, new genetic modifiers of hepatic iron loading and liver fibrogenesis are likely to be forthcoming.
Collapse
Affiliation(s)
- Eng K Gan
- a School of Medicine & Pharmacology, Fremantle Hospital, PO Box 480, Fremantle 6959, WA, Australia.
| | - Debbie Trinder
- b School of Medicine & Pharmacology, Fremantle Hospital, PO Box 480, Fremantle 6959, WA, Australia.
| | - Oyekoya T Ayonrinde
- c School of Medicine & Pharmacology, Fremantle Hospital, PO Box 480, Fremantle 6959, WA, Australia.
| | - John K Olynyk
- d Professor, School of Medicine & Pharmacology, Fremantle Hospital, PO Box 480, Fremantle 6959, WA, Australia.
| |
Collapse
|
49
|
Hower V, Mendes P, Torti FM, Laubenbacher R, Akman S, Shulaev V, Torti SV. A general map of iron metabolism and tissue-specific subnetworks. MOLECULAR BIOSYSTEMS 2009; 5:422-43. [PMID: 19381358 PMCID: PMC2680238 DOI: 10.1039/b816714c] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Iron is required for survival of mammalian cells. Recently, understanding of iron metabolism and trafficking has increased dramatically, revealing a complex, interacting network largely unknown just a few years ago. This provides an excellent model for systems biology development and analysis. The first step in such an analysis is the construction of a structural network of iron metabolism, which we present here. This network was created using CellDesigner version 3.5.2 and includes reactions occurring in mammalian cells of numerous tissue types. The iron metabolic network contains 151 chemical species and 107 reactions and transport steps. Starting from this general model, we construct iron networks for specific tissues and cells that are fundamental to maintaining body iron homeostasis. We include subnetworks for cells of the intestine and liver, tissues important in iron uptake and storage, respectively, as well as the reticulocyte and macrophage, key cells in iron utilization and recycling. The addition of kinetic information to our structural network will permit the simulation of iron metabolism in different tissues as well as in health and disease.
Collapse
Affiliation(s)
- Valerie Hower
- Department of Cancer Biology, Wake Forest University School of Medicine, Medical Center Blvd, Winston Salem, NC 27157, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Piperno A, Mariani R, Trombini P, Girelli D. Hepcidin modulation in human diseases: From research to clinic. World J Gastroenterol 2009; 15:538-51. [PMID: 19195055 PMCID: PMC2653344 DOI: 10.3748/wjg.15.538] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
By modulating hepcidin production, an organism controls intestinal iron absorption, iron uptake and mobilization from stores to meet body iron need. In recent years there has been important advancement in our knowledge of hepcidin regulation that also has implications for understanding the physiopathology of some human disorders. Since the discovery of hepcidin and the demonstration of its pivotal role in iron homeostasis, there has been a substantial interest in developing a reliable assay of the hormone in biological fluids. Measurement of hepcidin in biological fluids can improve our understanding of iron diseases and be a useful tool for diagnosis and clinical management of these disorders. We reviewed the literature and our own research on hepcidin to give an updated status of the situation in this rapidly evolving field.
Collapse
|