1
|
Niu X, Wang M, Wang M, Liu X, Zhang Y, Zheng P, Zhang S, Liu T, Cao Z, Zhang C. Dracorhodin perochlorate sensitizes colorectal cancer to ferroptosis by activating HMOX1 and inhibiting the SLC7A11/GPX4 axis. Int Immunopharmacol 2025; 158:114827. [PMID: 40359890 DOI: 10.1016/j.intimp.2025.114827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/30/2025] [Accepted: 05/06/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Ferroptosis, an iron-dependent form of cell death mediated by lipid peroxidation, plays a critical role in tumor progression. The natural small molecule compound dracorhodin perchlorate (DP) exhibits antitumor activity, but its effects on colorectal cancer (CRC) and the underlying mechanisms remain unclear. OBJECTIVE This study aimed to elucidate the role and mechanism of DP in CRC development and ferroptosis promotion. METHODS Using RNA-Seq, molecular docking and molecular dynamics simulation, we observed ferroptosis levels and expression of HMOX1, SLC7A11, and GPX4 in CRC cells treated with DP. We also examined the impact of modulating HMOX1, SLC7A11, and GPX4 on DP-induced ferroptosis and antitumor effects. RESULTS DP inhibited various malignant behaviors of CRC cells and induced ferroptosis. Mechanistically, RNA-Seq, molecular dynamics simulations, and molecular docking studies have collectively confirmed that DP directly binds to the HO-1 molecule, thereby upregulating HO-1 expression and inducing iron overload. Additionally, DP downregulates the expression of SLC7A11 and GPX4, collectively promoting the occurrence of ferroptosis in CRC cells. The HO-1 inhibitor ZnPP and SLC7A11 overexpression significantly inhibited the antitumor activity and ferroptosis induced by DP. Hemin and ferroptosis inducers enhanced its therapeutic effectiveness. DP safely suppressed subcutaneous tumor growth and exhibited synergistic effects with cisplatin both in vitro and in vivo. HMOX1 knockdown weakened the ferroptosis induced by DP in CRC. CONCLUSIONS The findings strongly support the activation of HMOX1 by DP, downregulation of the SLC7A11/GSH/GPX4 axis, and induction of ferroptosis in CRC cells. DP inhibited CRC progression and acted synergistically when combined with cisplatin. Our research provides a scientific basis for the use of DP in the treatment of CRC and offers new insights into the application of traditional Chinese medicine in the fight against CRC.
Collapse
Affiliation(s)
- Xuben Niu
- Department of General Surgery, School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of General Surgery, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Mingkun Wang
- Department of General Surgery, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China; Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Maihuan Wang
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaoya Liu
- Department of General Surgery, School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of General Surgery, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Yun Zhang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Peng Zheng
- Department of General Surgery, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China; Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Shuomin Zhang
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Ting Liu
- The Second Medical Centre, Chinese PLA General Hospital, Beijing 100859, China
| | - Zhen Cao
- Department of General Surgery, School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of General Surgery, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China; Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China; Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, 230032, China.
| | - Chaojun Zhang
- Department of General Surgery, School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of General Surgery, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China; Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China; Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, 230032, China.
| |
Collapse
|
2
|
Dong Y, Zheng M, Ding W, Guan H, Xiao J, Li F. Nrf2 activators for the treatment of rare iron overload diseases: From bench to bedside. Redox Biol 2025; 81:103551. [PMID: 39965404 PMCID: PMC11876910 DOI: 10.1016/j.redox.2025.103551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/02/2025] [Accepted: 02/13/2025] [Indexed: 02/20/2025] Open
Abstract
Iron overload and related oxidative damage are seen in many rare diseases, due to mutation of iron homeostasis-related genes. As a core regulator on cellular antioxidant reaction, Nrf2 can also decrease systemic and cellular iron levels by regulating iron-related genes and pathways, making Nrf2 activators very good candidates for the treatment of iron overload disorders. Successful examples include the clinical use of omaveloxolone for Friedreich's Ataxia and dimethyl fumarate for relapsing-remitting multiple sclerosis. Despite these uses, the therapeutic potentials of Nrf2 activators for iron overload disorders may be overlooked in clinical practice. Therefore, this study talks about the potential use, possible mechanisms, and precautions of Nrf2 activators in treating rare iron overload diseases. In addition, a combination therapy with Nrf2 activators and iron chelators is proposed for clinical reference, aiming to facilitate the clinical use of Nrf2 activators for more iron overload disorders.
Collapse
Affiliation(s)
- Yimin Dong
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Zheng
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weizhong Ding
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hanfeng Guan
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jun Xiao
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Feng Li
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Ishikawa T, Tatsumi Y, Kato K, Hayashi Y, Imai N, Ito T, Ishizu Y, Ishigami M, Nihei W, Kato A, Hayashi H. A 70-year-old Woman with Asymptomatic Ferroportin Disease. Intern Med 2024; 63:2421-2425. [PMID: 38296485 PMCID: PMC11442921 DOI: 10.2169/internalmedicine.2392-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 11/21/2023] [Indexed: 09/03/2024] Open
Abstract
A 59-year-old Japanese woman presented with hyperferritinemia. We decided against iron removal treatment because there were no symptoms or signs of iron-induced organ damage. A follow-up study revealed a gradual increase in transferrin saturation. The patient underwent a second examination at 66 years old. A liver biopsy showed substantial iron deposits in hepatocytes and Kupffer cells but no inflammation or fibrosis. Serum hepcidin-25 levels were highly parallel with hyperferritinemia. A genetic analysis revealed a G80S mutation in SLC40A1. These features are compatible with those of ferroportin disease. The patient remained asymptomatic at 70 years old, suggesting that the iron-loading condition may have been benign.
Collapse
Affiliation(s)
- Tetsuya Ishikawa
- Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Japan
| | - Yasuaki Tatsumi
- Department of Medical Biochemistry, Faculty of Pharmaceutical Sciences, Toho University, Japan
| | - Koichi Kato
- Department of Medicine, Aichi Gakuin University School of Pharmacy, Japan
| | - Yumi Hayashi
- Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Japan
| | - Norihiro Imai
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Japan
| | - Takanori Ito
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Japan
| | - Yoji Ishizu
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Japan
| | - Masatoshi Ishigami
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Japan
| | - Wataru Nihei
- Department of Medicine, Aichi Gakuin University School of Pharmacy, Japan
| | - Ayako Kato
- Department of Medicine, Aichi Gakuin University School of Pharmacy, Japan
| | - Hisao Hayashi
- Department of Medicine, Aichi Gakuin University School of Pharmacy, Japan
| |
Collapse
|
4
|
Sui Y, Geng X, Wang Z, Zhang J, Yang Y, Meng Z. Targeting the regulation of iron homeostasis as a potential therapeutic strategy for nonalcoholic fatty liver disease. Metabolism 2024; 157:155953. [PMID: 38885833 DOI: 10.1016/j.metabol.2024.155953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/09/2024] [Accepted: 06/09/2024] [Indexed: 06/20/2024]
Abstract
With aging and the increasing incidence of obesity, nonalcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide. NAFLD mainly includes simple hepatic steatosis, nonalcoholic steatohepatitis (NASH), liver fibrosis and hepatocellular carcinoma (HCC). An imbalance in hepatic iron homeostasis is usually associated with the progression of NAFLD and induces iron overload, reactive oxygen species (ROS) production, and lipid peroxide accumulation, which leads to ferroptosis. Ferroptosis is a unique type of programmed cell death (PCD) that is characterized by iron dependence, ROS production and lipid peroxidation. The ferroptosis inhibition systems involved in NAFLD include the solute carrier family 7 member 11 (SLC7A11)/glutathione (GSH)/glutathione peroxidase 4 (GPX4) and ferroptosis suppressor protein 1 (FSP1)/coenzyme Q10 (CoQ10)/nicotinamide adenine dinucleotide phosphate (NADPH) regulatory axes. The main promotion system involved is the acyl-CoA synthetase long-chain family (ACSL4)/arachidonic lipoxygenase 15 (ALOX15) axis. In recent years, an increasing number of studies have focused on the multiple roles of iron homeostasis imbalance and ferroptosis in the progression of NAFLD. This review highlights the latest studies about iron homeostasis imbalance- and ferroptosis-associated NAFLD, mainly including the physiology and pathophysiology of hepatic iron metabolism, hepatic iron homeostasis imbalance during the development of NAFLD, and key regulatory molecules and roles of hepatic ferroptosis in NAFLD. This review aims to provide innovative therapeutic strategies for NAFLD.
Collapse
Affiliation(s)
- Yutong Sui
- Shenzhen Hospital, Southern Medical University, Shenzhen 518100, Guangdong, China
| | - Xue Geng
- Department of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang, China
| | - Ziwei Wang
- Shenzhen Hospital, Southern Medical University, Shenzhen 518100, Guangdong, China
| | - Jing Zhang
- Shenzhen Hospital, Southern Medical University, Shenzhen 518100, Guangdong, China
| | - Yanqun Yang
- Shenzhen Hospital, Southern Medical University, Shenzhen 518100, Guangdong, China.
| | - Ziyu Meng
- NHC Key Laboratory of Hormones and Development, Tianjin Medical University Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China; Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin 300134, China.
| |
Collapse
|
5
|
Hu J, Li Y, Zhang L, Peng G, Zhang F, Zhao X. Iron overload due to SLC40A1 mutation of type 4 hereditary hemochromatosis. Med Mol Morphol 2023; 56:233-238. [PMID: 37382698 DOI: 10.1007/s00795-023-00359-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 06/12/2023] [Indexed: 06/30/2023]
Abstract
Hereditary hemochromatosis type 4 is an autosomal-dominant inherited disease characterized by a mutation in the SLC40A1 gene encoding ferroportin. This condition can be further subdivided into types 4A (loss-of-function mutations) and 4B (gain-of-function mutations). To date, only a few cases of type 4B cases have been reported, and the treatment has not been clearly mentioned. Here, we report a genotype of hereditary hemochromatosis type 4B involving the heterozygous mutation c.997 T > C (p. Tyr333His) in SLC40A1. The patient was treated with red blood cell apheresis every month for 1 year, followed by oral deferasirox, and the combined therapy was found to be effective.
Collapse
Affiliation(s)
- Jing Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Yuan Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Li Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Guangxin Peng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Fengkui Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Xin Zhao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| |
Collapse
|
6
|
Hilton C, Sabaratnam R, Drakesmith H, Karpe F. Iron, glucose and fat metabolism and obesity: an intertwined relationship. Int J Obes (Lond) 2023; 47:554-563. [PMID: 37029208 PMCID: PMC10299911 DOI: 10.1038/s41366-023-01299-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 03/08/2023] [Accepted: 03/16/2023] [Indexed: 04/09/2023]
Abstract
A bidirectional relationship exists between adipose tissue metabolism and iron regulation. Total body fat, fat distribution and exercise influence iron status and components of the iron-regulatory pathway, including hepcidin and erythroferrone. Conversely, whole body and tissue iron stores associate with fat mass and distribution and glucose and lipid metabolism in adipose tissue, liver, and muscle. Manipulation of the iron-regulatory proteins erythroferrone and erythropoietin affects glucose and lipid metabolism. Several lines of evidence suggest that iron accumulation and metabolism may play a role in the development of metabolic diseases including obesity, type 2 diabetes, hyperlipidaemia and non-alcoholic fatty liver disease. In this review we summarise the current understanding of the relationship between iron homoeostasis and metabolic disease.
Collapse
Affiliation(s)
- Catriona Hilton
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK.
| | - Rugivan Sabaratnam
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Hal Drakesmith
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| |
Collapse
|
7
|
Uguen K, Ka C, Collod-Béroud G, Le Tertre M, Guellec J, Férec C, Béroud C, Callebaut I, Le Gac G. The Spectra of Disease-Causing Mutations in the Ferroportin 1 ( SLC40A1) Encoding Gene and Related Iron Overload Phenotypes (Hemochromatosis Type 4 and Ferroportin Disease). Hum Mutat 2023; 2023:5162256. [PMID: 40225168 PMCID: PMC11919020 DOI: 10.1155/2023/5162256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 05/17/2023] [Accepted: 05/26/2023] [Indexed: 04/15/2025]
Abstract
SLC40A1 is the sole iron export protein reported in mammals and is a key player in both cellular and systemic iron homeostasis. This unique iron exporter, which belongs to the major facilitator superfamily, is predominantly regulated by the hyposideremic hormone hepcidin. SLC40A1 dysfunction causes ferroportin disease, and autosomal dominant iron overload disorder characterized by cellular iron retention, principally in reticuloendothelial cells, correlating with high serum ferritin and low to normal transferrin saturation. Resistant to hepcidin, SLC40A1 mutations are rather associated with elevated plasma iron and parenchymal iron deposition, a condition that resembles HFE-related hemochromatosis and is associated with more clinical complications. With very few exceptions, only missense variations are reported at the SLC40A1 locus; this situation increasingly limits the establishment of pathogenicity. In this mutation update, we provide a comprehensive review of all the pathogenic or likely pathogenic variants, variants of unknown significance, and benign or likely benign SLC40A1 variants. The classification is essentially determined using functional, structural, segregation, and recurrence data. We furnish new information on genotype-phenotype correlations for loss-of-function, gain-of-function, and other SLC40A1 variants, confirming the existence of wide clinical heterogeneity and the potential for misdiagnosis. All information is recorded in a locus-specific online database.
Collapse
Affiliation(s)
- Kevin Uguen
- Univ Brest, Inserm, EFS, UMR1078, GGB F-29200, France
- CHRU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Laboratoire de Génétique Moléculaire et Histocompatibilité, F-29200, France
| | - Chandran Ka
- Univ Brest, Inserm, EFS, UMR1078, GGB F-29200, France
- CHRU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Laboratoire de Génétique Moléculaire et Histocompatibilité, F-29200, France
- Laboratory of Excellence GR-Ex, F-75015, France
| | | | - Marlène Le Tertre
- Univ Brest, Inserm, EFS, UMR1078, GGB F-29200, France
- CHRU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Laboratoire de Génétique Moléculaire et Histocompatibilité, F-29200, France
| | - Julie Guellec
- Univ Brest, Inserm, EFS, UMR1078, GGB F-29200, France
- CHRU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Laboratoire de Génétique Moléculaire et Histocompatibilité, F-29200, France
- Association Gaétan Saleün, F-29200, France
| | - Claude Férec
- Univ Brest, Inserm, EFS, UMR1078, GGB F-29200, France
- CHRU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Laboratoire de Génétique Moléculaire et Histocompatibilité, F-29200, France
- Association Gaétan Saleün, F-29200, France
| | - Christophe Béroud
- Aix Marseille University, INSERM, Marseille Medical Genetics, F-13005, France
| | - Isabelle Callebaut
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS 7590, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, F-75005, France
| | - Gérald Le Gac
- Univ Brest, Inserm, EFS, UMR1078, GGB F-29200, France
- CHRU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Laboratoire de Génétique Moléculaire et Histocompatibilité, F-29200, France
- Laboratory of Excellence GR-Ex, F-75015, France
| |
Collapse
|
8
|
Leischner C, Marongiu L, Piotrowsky A, Niessner H, Venturelli S, Burkard M, Renner O. Relevant Membrane Transport Proteins as Possible Gatekeepers for Effective Pharmacological Ascorbate Treatment in Cancer. Antioxidants (Basel) 2023; 12:antiox12040916. [PMID: 37107291 PMCID: PMC10135768 DOI: 10.3390/antiox12040916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/23/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Despite the increasing number of newly diagnosed malignancies worldwide, therapeutic options for some tumor diseases are unfortunately still limited. Interestingly, preclinical but also some clinical data suggest that the administration of pharmacological ascorbate seems to respond well, especially in some aggressively growing tumor entities. The membrane transport and channel proteins are highly relevant for the use of pharmacological ascorbate in cancer therapy and are involved in the transfer of active substances such as ascorbate, hydrogen peroxide, and iron that predominantly must enter malignant cells to induce antiproliferative effects and especially ferroptosis. In this review, the relevant conveying proteins from cellular surfaces are presented as an integral part of the efficacy of pharmacological ascorbate, considering the already known genetic and functional features in tumor tissues. Accordingly, candidates for diagnostic markers and therapeutic targets are mentioned.
Collapse
Affiliation(s)
- Christian Leischner
- Institute of Nutritional Sciences, Department of Nutritional Biochemistry, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
| | - Luigi Marongiu
- Institute of Nutritional Sciences, Department of Nutritional Biochemistry, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
- Department of Internal Medicine VIII, University Hospital Tuebingen, Otfried-Mueller-Straße 10, 72076 Tuebingen, Germany
| | - Alban Piotrowsky
- Institute of Nutritional Sciences, Department of Nutritional Biochemistry, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
| | - Heike Niessner
- Department of Dermatology, Division of Dermatooncology, University of Tuebingen, Liebermeisterstraße 25, 72076 Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", 72076 Tuebingen, Germany
| | - Sascha Venturelli
- Institute of Nutritional Sciences, Department of Nutritional Biochemistry, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
- Institute of Physiology, Department of Vegetative and Clinical Physiology, University of Tuebingen, Wilhelmstraße 56, 72074 Tuebingen, Germany
| | - Markus Burkard
- Institute of Nutritional Sciences, Department of Nutritional Biochemistry, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
| | - Olga Renner
- Institute of Nutritional Sciences, Department of Nutritional Biochemistry, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
| |
Collapse
|
9
|
Role of Iron in Aging Related Diseases. Antioxidants (Basel) 2022; 11:antiox11050865. [PMID: 35624729 PMCID: PMC9137504 DOI: 10.3390/antiox11050865] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/17/2022] [Accepted: 04/25/2022] [Indexed: 02/05/2023] Open
Abstract
Iron progressively accumulates with age and can be further exacerbated by dietary iron intake, genetic factors, and repeated blood transfusions. While iron plays a vital role in various physiological processes within the human body, its accumulation contributes to cellular aging in several species. In its free form, iron can initiate the formation of free radicals at a cellular level and contribute to systemic disorders. This is most evident in high iron conditions such as hereditary hemochromatosis, when accumulation of iron contributes to the development of arthritis, cirrhosis, or cardiomyopathy. A growing body of research has further identified iron’s contributory effects in neurodegenerative diseases, ocular disorders, cancer, diabetes, endocrine dysfunction, and cardiovascular diseases. Reducing iron levels by repeated phlebotomy, iron chelation, and dietary restriction are the common therapeutic considerations to prevent iron toxicity. Chelators such as deferoxamine, deferiprone, and deferasirox have become the standard of care in managing iron overload conditions with other potential applications in cancer and cardiotoxicity. In certain animal models, drugs with iron chelating ability have been found to promote health and even extend lifespan. As we further explore the role of iron in the aging process, iron chelators will likely play an increasingly important role in our health.
Collapse
|
10
|
Bonilla DA, Moreno Y, Petro JL, Forero DA, Vargas-Molina S, Odriozola-Martínez A, Orozco CA, Stout JR, Rawson ES, Kreider RB. A Bioinformatics-Assisted Review on Iron Metabolism and Immune System to Identify Potential Biomarkers of Exercise Stress-Induced Immunosuppression. Biomedicines 2022; 10:724. [PMID: 35327526 PMCID: PMC8945881 DOI: 10.3390/biomedicines10030724] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/01/2022] [Accepted: 03/09/2022] [Indexed: 02/01/2023] Open
Abstract
The immune function is closely related to iron (Fe) homeostasis and allostasis. The aim of this bioinformatics-assisted review was twofold; (i) to update the current knowledge of Fe metabolism and its relationship to the immune system, and (ii) to perform a prediction analysis of regulatory network hubs that might serve as potential biomarkers during stress-induced immunosuppression. Several literature and bioinformatics databases/repositories were utilized to review Fe metabolism and complement the molecular description of prioritized proteins. The Search Tool for the Retrieval of Interacting Genes (STRING) was used to build a protein-protein interactions network for subsequent network topology analysis. Importantly, Fe is a sensitive double-edged sword where two extremes of its nutritional status may have harmful effects on innate and adaptive immunity. We identified clearly connected important hubs that belong to two clusters: (i) presentation of peptide antigens to the immune system with the involvement of redox reactions of Fe, heme, and Fe trafficking/transport; and (ii) ubiquitination, endocytosis, and degradation processes of proteins related to Fe metabolism in immune cells (e.g., macrophages). The identified potential biomarkers were in agreement with the current experimental evidence, are included in several immunological/biomarkers databases, and/or are emerging genetic markers for different stressful conditions. Although further validation is warranted, this hybrid method (human-machine collaboration) to extract meaningful biological applications using available data in literature and bioinformatics tools should be highlighted.
Collapse
Affiliation(s)
- Diego A. Bonilla
- Research Division, Dynamical Business & Science Society—DBSS International SAS, Bogota 110311, Colombia; (Y.M.); (J.L.P.)
- Research Group in Biochemistry and Molecular Biology, Faculty of Science and Education, Universidad Distrital Francisco José de Caldas, Bogota 110311, Colombia
- Research Group in Physical Activity, Sports and Health Sciences (GICAFS), Universidad de Córdoba, Montería 230002, Colombia
- Sport Genomics Research Group, Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
| | - Yurany Moreno
- Research Division, Dynamical Business & Science Society—DBSS International SAS, Bogota 110311, Colombia; (Y.M.); (J.L.P.)
- Research Group in Biochemistry and Molecular Biology, Faculty of Science and Education, Universidad Distrital Francisco José de Caldas, Bogota 110311, Colombia
| | - Jorge L. Petro
- Research Division, Dynamical Business & Science Society—DBSS International SAS, Bogota 110311, Colombia; (Y.M.); (J.L.P.)
- Research Group in Physical Activity, Sports and Health Sciences (GICAFS), Universidad de Córdoba, Montería 230002, Colombia
| | - Diego A. Forero
- Health and Sport Sciences Research Group, School of Health and Sport Sciences, Fundación Universitaria del Área Andina, Bogotá 111221, Colombia; (D.A.F.); (C.A.O.)
| | - Salvador Vargas-Molina
- Faculty of Sport Sciences, EADE-University of Wales Trinity Saint David, 29018 Málaga, Spain;
| | - Adrián Odriozola-Martínez
- Sport Genomics Research Group, Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
- kDNA Genomics, Joxe Mari Korta Research Center, University of the Basque Country UPV/EHU, 20018 Donostia, Spain
| | - Carlos A. Orozco
- Health and Sport Sciences Research Group, School of Health and Sport Sciences, Fundación Universitaria del Área Andina, Bogotá 111221, Colombia; (D.A.F.); (C.A.O.)
| | - Jeffrey R. Stout
- Physiology of Work and Exercise Response (POWER) Laboratory, Institute of Exercise Physiology and Rehabilitation Science, University of Central Florida, Orlando, FL 32816, USA;
| | - Eric S. Rawson
- Department of Health, Nutrition and Exercise Science, Messiah University, Mechanicsburg, PA 17055, USA;
| | - Richard B. Kreider
- Exercise & Sport Nutrition Laboratory, Human Clinical Research Facility, Department of Health & Kinesiology, Texas A&M University, College Station, TX 77843, USA;
| |
Collapse
|
11
|
Landemaine A, Hamdi-Roze H, Cunat S, Loustaud-Ratti V, Causse X, Si Ahmed SN, Drénou B, Bureau C, Pelletier G, De Kerguenec C, Ganne-Carrie N, Durupt S, Laine F, Loréal O, Ropert M, Detivaud L, Morcet J, Aguilar-Martinez P, Deugnier YM, Bardou-Jacquet E. A simple clinical score to promote and enhance ferroportin disease screening. J Hepatol 2022; 76:568-576. [PMID: 34748893 DOI: 10.1016/j.jhep.2021.10.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/27/2021] [Accepted: 10/25/2021] [Indexed: 12/04/2022]
Abstract
BACKGROUND & AIMS Ferroportin disease is a rare genetic iron overload disorder which may be underdiagnosed, with recent data suggesting it occurs at a higher prevalence than suspected. Costs and the lack of defined criteria to prompt genetic testing preclude large-scale molecular screening. Hence, we aimed to develop a readily available scoring system to promote and enhance ferroportin disease screening. METHODS Our derivation cohort included probands tested for ferroportin disease from 2008 to 2016 in our rare disease network. Data were prospectively recorded. Univariate and multivariate logistic regression were used to determine significant criteria, and odds ratios were used to build a weighted score. A cut-off value was defined using a ROC curve with a predefined aim of 90% sensitivity. An independent cohort was used for cross validation. RESULTS Our derivation cohort included 1,306 patients. Mean age was 55±14 years, ferritin 1,351±1,357 μg/L, and liver iron concentration (LIC) 166±77 μmol/g. Pathogenic variants (n = 32) were identified in 71 patients. In multivariate analysis: female sex, younger age, higher ferritin, higher LIC and the absence of hypertension or diabetes were significantly associated with the diagnosis of ferroportin disease (AUROC in whole derivation cohort 0.83 [0.78-0.88]). The weighted score was based on sex, age, the presence of hypertension or diabetes, ferritin level and LIC. An AUROC of 0.83 (0.77-0.88) was obtained in the derivation cohort without missing values. Using 9.5 as a cut-off, sensitivity was 93.6 (91.7-98.3) %, specificity 49.5 (45.5-53.6) %, positive likelihood ratio 1.8 (1.6-2.0) and negative likelihood ratio 0.17 (0.04-0.37). CONCLUSION We describe a readily available score with simple criteria and good diagnostic performance that could be used to screen patients for ferroportin disease in routine clinical practice. LAY SUMMARY Increased iron burden associated with metabolic syndrome is a very common condition. Ferroportin disease is a dominant genetic iron overload disorder whose prevalence is higher than initially thought. They can be difficult to distinguish from each other, but the limited availability of genetic testing and the lack of definitive guidelines prevent adequate screening. We herein describe a simple and definitive clinical score to help clinicians decide whether to perform genetic testing.
Collapse
Affiliation(s)
- Amandine Landemaine
- Univ Rennes, CHU Rennes, INSERM CIC1414, F-35000, Rennes, France; National Reference Center for Hemochromatosis and iron metabolism disorder, CHU Rennes, F-35000, Rennes, France.
| | - Houda Hamdi-Roze
- Univ Rennes, CHU Rennes, INSERM CIC1414, F-35000, Rennes, France; National Reference Center for Hemochromatosis and iron metabolism disorder, CHU Rennes, F-35000, Rennes, France
| | - Séverine Cunat
- CHU Montpellier, Competence Center for Hemochromatosis and Iron Metabolism Disorder, Reference Center on Rare Red Cell Disorders, Montpellier, France
| | | | - Xavier Causse
- Department of Hepatology and Gastroenterology, Centre Hospitalier Régional (CHR), Orléans, France
| | - Si Nafa Si Ahmed
- Department of Hepatology and Gastroenterology, Centre Hospitalier Régional (CHR), Orléans, France
| | - Bernard Drénou
- CH Emile Muller, Department of Hematology, F-68100 Mulhouse, France
| | - Christophe Bureau
- CHU Toulouse, Liver Unit, University Hospital of Toulouse and University Paul Sabatier, Toulouse, France
| | - Gilles Pelletier
- AH-HP, Hôpital Paul Brousse, Centre Hépato-Biliaire, Villejuif, France
| | | | - Nathalie Ganne-Carrie
- AP-HP Hopitaux Universitaire Paris Saine Saint-Denis, APHP, Liver Unit, University Paris 13, Sorbonne Paris Cité; INSE RM, S1138 FunGeST F-75006, Paris, France
| | - Stéphane Durupt
- Department of Internal and Vascular Medicine, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Fabrice Laine
- Univ Rennes, CHU Rennes, INSERM CIC1414, F-35000, Rennes, France; National Reference Center for Hemochromatosis and iron metabolism disorder, CHU Rennes, F-35000, Rennes, France
| | - Olivier Loréal
- National Reference Center for Hemochromatosis and iron metabolism disorder, CHU Rennes, F-35000, Rennes, France; INSERM, Univ Rennes, CHU Rennes, UMR1241, Institut NuMeCan, Rennes, France
| | - Martine Ropert
- Univ Rennes, CHU Rennes, INSERM CIC1414, F-35000, Rennes, France; National Reference Center for Hemochromatosis and iron metabolism disorder, CHU Rennes, F-35000, Rennes, France
| | - Lenaick Detivaud
- Univ Rennes, CHU Rennes, INSERM CIC1414, F-35000, Rennes, France; National Reference Center for Hemochromatosis and iron metabolism disorder, CHU Rennes, F-35000, Rennes, France
| | - Jeff Morcet
- Univ Rennes, CHU Rennes, INSERM CIC1414, F-35000, Rennes, France
| | - Patricia Aguilar-Martinez
- CHU Montpellier, Competence Center for Hemochromatosis and Iron Metabolism Disorder, Reference Center on Rare Red Cell Disorders, Montpellier, France
| | - Yves M Deugnier
- Univ Rennes, CHU Rennes, INSERM CIC1414, F-35000, Rennes, France; National Reference Center for Hemochromatosis and iron metabolism disorder, CHU Rennes, F-35000, Rennes, France
| | - Edouard Bardou-Jacquet
- Univ Rennes, CHU Rennes, INSERM CIC1414, F-35000, Rennes, France; National Reference Center for Hemochromatosis and iron metabolism disorder, CHU Rennes, F-35000, Rennes, France; INSERM, Univ Rennes, CHU Rennes, UMR1241, Institut NuMeCan, Rennes, France
| |
Collapse
|
12
|
Vukmirovic M, Yan X, Gibson KF, Gulati M, Schupp JC, DeIuliis G, Adams TS, Hu B, Mihaljinec A, Woolard TN, Lynn H, Emeagwali N, Herzog EL, Chen ES, Morris A, Leader JK, Zhang Y, Garcia JGN, Maier LA, Collman RG, Drake WP, Becich MJ, Hochheiser H, Wisniewski SR, Benos PV, Moller DR, Prasse A, Koth LL, Kaminski N. Transcriptomics of bronchoalveolar lavage cells identifies new molecular endotypes of sarcoidosis. Eur Respir J 2021; 58:2002950. [PMID: 34083402 PMCID: PMC9759791 DOI: 10.1183/13993003.02950-2020] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 04/20/2021] [Indexed: 11/05/2022]
Abstract
BACKGROUND Sarcoidosis is a multisystem granulomatous disease of unknown origin with a variable and often unpredictable course and pattern of organ involvement. In this study we sought to identify specific bronchoalveolar lavage (BAL) cell gene expression patterns indicative of distinct disease phenotypic traits. METHODS RNA sequencing by Ion Torrent Proton was performed on BAL cells obtained from 215 well-characterised patients with pulmonary sarcoidosis enrolled in the multicentre Genomic Research in Alpha-1 Antitrypsin Deficiency and Sarcoidosis (GRADS) study. Weighted gene co-expression network analysis and nonparametric statistics were used to analyse genome-wide BAL transcriptome. Validation of results was performed using a microarray expression dataset of an independent sarcoidosis cohort (Freiburg, Germany; n=50). RESULTS Our supervised analysis found associations between distinct transcriptional programmes and major pulmonary phenotypic manifestations of sarcoidosis including T-helper type 1 (Th1) and Th17 pathways associated with hilar lymphadenopathy, transforming growth factor-β1 (TGFB1) and mechanistic target of rapamycin (MTOR) signalling with parenchymal involvement, and interleukin (IL)-7 and IL-2 with airway involvement. Our unsupervised analysis revealed gene modules that uncovered four potential sarcoidosis endotypes including hilar lymphadenopathy with increased acute T-cell immune response; extraocular organ involvement with PI3K activation pathways; chronic and multiorgan disease with increased immune response pathways; and multiorgan involvement, with increased IL-1 and IL-18 immune and inflammatory responses. We validated the occurrence of these endotypes using gene expression, pulmonary function tests and cell differentials from Freiburg. CONCLUSION Taken together, our results identify BAL gene expression programmes that characterise major pulmonary sarcoidosis phenotypes and suggest the presence of distinct disease molecular endotypes.
Collapse
Affiliation(s)
- Milica Vukmirovic
- Section of Pulmonary, Critical Care and Sleep Medicine, Dept of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Dept of Medicine, Division of Respirology, McMaster University, Hamilton, ON, Canada
- Equally contributing authors
| | - Xiting Yan
- Section of Pulmonary, Critical Care and Sleep Medicine, Dept of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Dept of Biostatistics, Yale School of Public Health, New Haven, CT, USA
- Equally contributing authors
| | - Kevin F Gibson
- Dept of Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA, US
| | - Mridu Gulati
- Section of Pulmonary, Critical Care and Sleep Medicine, Dept of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Jonas C Schupp
- Section of Pulmonary, Critical Care and Sleep Medicine, Dept of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Giuseppe DeIuliis
- Section of Pulmonary, Critical Care and Sleep Medicine, Dept of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Taylor S Adams
- Section of Pulmonary, Critical Care and Sleep Medicine, Dept of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Buqu Hu
- Section of Pulmonary, Critical Care and Sleep Medicine, Dept of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Antun Mihaljinec
- Section of Pulmonary, Critical Care and Sleep Medicine, Dept of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Tony N Woolard
- Section of Pulmonary, Critical Care and Sleep Medicine, Dept of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Heather Lynn
- Section of Pulmonary, Critical Care and Sleep Medicine, Dept of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- University of Arizona Health Sciences, Tucson, AZ, USA
| | - Nkiruka Emeagwali
- Section of Pulmonary, Critical Care and Sleep Medicine, Dept of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Erica L Herzog
- Section of Pulmonary, Critical Care and Sleep Medicine, Dept of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | - Alison Morris
- Dept of Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA, US
| | - Joseph K Leader
- Dept of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yingze Zhang
- Dept of Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA, US
| | | | | | | | | | - Michael J Becich
- Dept of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Harry Hochheiser
- Dept of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Steven R Wisniewski
- Dept of Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA, US
| | - Panayiotis V Benos
- Dept of Computational and Systems Biology and Department of Computer Science, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Antje Prasse
- Hannover Medical School (MHH), Hannover, Germany
- Fraunhofer ITEM, Hannover, Germany
| | - Laura L Koth
- University of California San Francisco, San Francisco, CA, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Dept of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
13
|
Ravasi G, Pelucchi S, Bertola F, Capelletti MM, Mariani R, Piperno A. Identification of Novel Mutations by Targeted NGS Panel in Patients with Hyperferritinemia. Genes (Basel) 2021; 12:genes12111778. [PMID: 34828384 PMCID: PMC8623017 DOI: 10.3390/genes12111778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/03/2021] [Accepted: 11/06/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Several inherited diseases cause hyperferritinemia with or without iron overload. Differential diagnosis is complex and requires an extensive work-up. Currently, a clinical-guided approach to genetic tests is performed based on gene-by-gene sequencing. Although reasonable, this approach is expensive and time-consuming and Next Generation Sequencing (NGS) technology may provide cheaper and quicker large-scale DNA sequencing. METHODS We analysed 36 patients with non-HFE-related hyperferritinemia. Liver iron concentration was measured in 33 by magnetic resonance. A panel of 25 iron related genes was designed using SureDesign software. Custom libraries were generated and then sequenced using Ion Torrent PGM. RESULTS We identified six novel mutations in SLC40A1, three novel and one known mutation in TFR2, one known mutation and a de-novo deletion in HJV, and a novel mutation in HAMP in ten patients. In silico analyses supported the pathogenic role of the mutations. CONCLUSIONS Our results support the use of an NGS-based panel in selected patients with hyperferritinemia in a tertiary center for iron metabolism disorders. However, 26 out of 36 patients did not show genetic variants that can individually explain hyperferritinemia and/or iron overload suggesting the existence of other genetic defects or gene-gene and gene-environment interactions needing further studies.
Collapse
Affiliation(s)
- Giulia Ravasi
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (G.R.); (S.P.); (M.M.C.)
| | - Sara Pelucchi
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (G.R.); (S.P.); (M.M.C.)
| | - Francesca Bertola
- Medical Genetics, S. Gerardo Hospital, ASST-Monza, 20900 Monza, Italy;
| | - Martina Maria Capelletti
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (G.R.); (S.P.); (M.M.C.)
| | - Raffaella Mariani
- Disorders of Iron Metabolism, Centre for Rare Diseases, San Gerardo Hospital, ASST-Monza, 20900 Monza, Italy;
| | - Alberto Piperno
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (G.R.); (S.P.); (M.M.C.)
- Medical Genetics, S. Gerardo Hospital, ASST-Monza, 20900 Monza, Italy;
- Disorders of Iron Metabolism, Centre for Rare Diseases, San Gerardo Hospital, ASST-Monza, 20900 Monza, Italy;
- Correspondence: ; Tel.: +39-03-9233-3461
| |
Collapse
|
14
|
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
|
15
|
Maio N, Zhang DL, Ghosh MC, Jain A, SantaMaria AM, Rouault TA. Mechanisms of cellular iron sensing, regulation of erythropoiesis and mitochondrial iron utilization. Semin Hematol 2021; 58:161-174. [PMID: 34389108 DOI: 10.1053/j.seminhematol.2021.06.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 12/11/2022]
Abstract
To maintain an adequate iron supply for hemoglobin synthesis and essential metabolic functions while counteracting iron toxicity, humans and other vertebrates have evolved effective mechanisms to conserve and finely regulate iron concentration, storage, and distribution to tissues. At the systemic level, the iron-regulatory hormone hepcidin is secreted by the liver in response to serum iron levels and inflammation. Hepcidin regulates the expression of the sole known mammalian iron exporter, ferroportin, to control dietary absorption, storage and tissue distribution of iron. At the cellular level, iron regulatory proteins 1 and 2 (IRP1 and IRP2) register cytosolic iron concentrations and post-transcriptionally regulate the expression of iron metabolism genes to optimize iron availability for essential cellular processes, including heme biosynthesis and iron-sulfur cluster biogenesis. Genetic malfunctions affecting the iron sensing mechanisms or the main pathways that utilize iron in the cell cause a broad range of human diseases, some of which are characterized by mitochondrial iron accumulation. This review will discuss the mechanisms of systemic and cellular iron sensing with a focus on the main iron utilization pathways in the cell, and on human conditions that arise from compromised function of the regulatory axes that control iron homeostasis.
Collapse
Affiliation(s)
- Nunziata Maio
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - De-Liang Zhang
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Manik C Ghosh
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Anshika Jain
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Anna M SantaMaria
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Tracey A Rouault
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
16
|
Kang W, Barad A, Clark AG, Wang Y, Lin X, Gu Z, O'Brien KO. Ethnic Differences in Iron Status. Adv Nutr 2021; 12:1838-1853. [PMID: 34009254 PMCID: PMC8483971 DOI: 10.1093/advances/nmab035] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023] Open
Abstract
Iron is unique among all minerals in that humans have no regulatable excretory pathway to eliminate excess iron after it is absorbed. Iron deficiency anemia occurs when absorbed iron is not sufficient to meet body iron demands, whereas iron overload and subsequent deposition of iron in key organs occur when absorbed iron exceeds body iron demands. Over time, iron accumulation in the body can increase risk of chronic diseases, including cirrhosis, diabetes, and heart failure. To date, only ∼30% of the interindividual variability in iron absorption can be captured by iron status biomarkers or iron regulatory hormones. Much of the regulation of iron absorption may be under genetic control, but these pathways have yet to be fully elucidated. Genome-wide and candidate gene association studies have identified several genetic variants that are associated with variations in iron status, but the majority of these data were generated in European populations. The purpose of this review is to summarize genetic variants that have been associated with alterations in iron status and to highlight the influence of ethnicity on the risk of iron deficiency or overload. Using extant data in the literature, linear mixed-effects models were constructed to explore ethnic differences in iron status biomarkers. This approach found that East Asians had significantly higher concentrations of iron status indicators (serum ferritin, transferrin saturation, and hemoglobin) than Europeans, African Americans, or South Asians. African Americans exhibited significantly lower hemoglobin concentrations compared with other ethnic groups. Further studies of the genetic basis for ethnic differences in iron metabolism and on how it affects disease susceptibility among different ethnic groups are needed to inform population-specific recommendations and personalized nutrition interventions for iron-related disorders.
Collapse
Affiliation(s)
- Wanhui Kang
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Alexa Barad
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Andrew G Clark
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA,Department of Computational Biology, Cornell University, Ithaca, NY, USA
| | - Yiqin Wang
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Xu Lin
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Zhenglong Gu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | | |
Collapse
|
17
|
Nishina S, Tomiyama Y, Ikuta K, Tatsumi Y, Toki Y, Kato A, Kato K, Yoshioka N, Sasaki K, Hara Y, Hino K. Long-term phlebotomy successfully alleviated hepatic iron accumulation in a ferroportin disease patient with a mutation in SLC40A1: a case report. BMC Gastroenterol 2021; 21:111. [PMID: 33673803 PMCID: PMC7934381 DOI: 10.1186/s12876-021-01674-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 02/17/2021] [Indexed: 02/08/2023] Open
Abstract
Background Hereditary hemochromatosis is a heterogenous group of inherited iron-overload conditions that is characterized by increased intestinal absorption and deposition in vital organs. Hepcidin is a soluble regulator that acts to attenuate both intestinal iron absorption and iron release from reticuloendothelial macrophages through internalization of ferroportin-1, an iron exporter. Ferroportin disease is hereditary hemochromatosis which is affected by SLC40A1, a gene coding ferroportin-1, and phenotypically classified into two forms (classical and nonclassical). In nonclassical form, ferroportin mutations are responsible for a gain of function with full iron export capability but insensitivity to downregulation by hepcidin. Here, we report a case of nonclassical ferroportin disease. Case presentation A 46-year-old Japanese man showed elevated serum iron (284 μg/dl), ferritin (1722 ng/ml), transferrin saturation ratio (91.3%), and hepcidin-25 level (139.6 ng/ml). Magnetic resonance imaging (MRI) demonstrated a marked reduction in the signal intensity of the liver in T1- and T2-weighted images. The liver histology exhibited a large amount of iron that had accumulated predominantly in hepatocytes. We identified a heterozygous 1520A > G (p.H507R) mutation in the SLC40A1 gene. Phlebotomy (400 ml at a time) was monthly performed for 3 years in this patient. Importantly, the serum hepcidin level (1.0 ng/ml) was normal when the serum ferritin level was normal and hepatic iron accumulation was remarkably reduced after 3 years of phlebotomy. Conclusions The present case demonstrated for the first time that there was a correlation between hepatic iron levels as measured by MRI and serum hepcidin levels through long-term phlebotomy in a patient with ferroportin disease with the p.H507R mutation of in SLC40A1.
Collapse
Affiliation(s)
- Sohji Nishina
- Department of Hepatology and Pancreatology, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan
| | - Yasuyuki Tomiyama
- Department of Hepatology and Pancreatology, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan
| | - Katsuya Ikuta
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan.,Japanese Red Cross Hokkaido Blood Center, Sapporo, Japan
| | - Yasuaki Tatsumi
- Laboratory of Medicine, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan
| | - Yasumichi Toki
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Ayako Kato
- Laboratory of Medicine, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan
| | - Koichi Kato
- Laboratory of Medicine, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan
| | - Naoko Yoshioka
- Department of Hepatology and Pancreatology, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan
| | - Kyo Sasaki
- Department of Hepatology and Pancreatology, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan
| | - Yuichi Hara
- Department of Hepatology and Pancreatology, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan
| | - Keisuke Hino
- Department of Hepatology and Pancreatology, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan.
| |
Collapse
|
18
|
Le Tertre M, Ka C, Raud L, Berlivet I, Gourlaouen I, Richard G, Uguen K, Chen JM, Férec C, Fichou Y, Le Gac G. Splicing analysis of SLC40A1 missense variations and contribution to hemochromatosis type 4 phenotypes. Blood Cells Mol Dis 2020; 87:102527. [PMID: 33341511 DOI: 10.1016/j.bcmd.2020.102527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 02/09/2023]
Abstract
Hemochromatosis type 4, or ferroportin disease, is considered as the second leading cause of primary iron overload after HFE-related hemochromatosis. The disease, which is predominantly associated with missense variations in the SLC40A1 gene, is characterized by wide clinical heterogeneity. We tested the possibility that some of the reported missense mutations, despite their positions within exons, cause splicing defects. Fifty-eight genetic variants were selected from the literature based on two criteria: a precise description of the nucleotide change and individual evidence of iron overload. The selected variants were investigated by different in silico prediction tools and prioritized for midigene splicing assays. Of the 15 variations tested in vitro, only two were associated with splicing changes. We confirm that the c.1402G>A transition (p.Gly468Ser) disrupts the exon 7 donor site, leading to the use of an exonic cryptic splicing site and the generation of a truncated reading frame. We observed, for the first time, that the p.Gly468Ser substitution has no effect on the ferroportin iron export function. We demonstrate alternative splicing of exon 5 in different cell lines and show that the c.430A>G (p.Asn144Asp) variant promotes exon 5 inclusion. This could be part of a gain-of-function mechanism. We conclude that splicing mutations rarely contribute to hemochromatosis type 4 phenotypes. An in-depth investigation of exon 5 auxiliary splicing sequences may help to elucidate the mechanism by which splicing regulatory proteins regulate the production of the full length SLC40A1 transcript and to clarify its physiological importance.
Collapse
Affiliation(s)
- Marlène Le Tertre
- Univ Brest, Inserm, EFS, UMR1078, GGB, F-29200, France; CHRU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Laboratoire de Génétique Moléculaire et Histocompatibilité, F-29200, France
| | - Chandran Ka
- Univ Brest, Inserm, EFS, UMR1078, GGB, F-29200, France; CHRU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Laboratoire de Génétique Moléculaire et Histocompatibilité, F-29200, France; Laboratory of Excellence GR-Ex, F-75015, France
| | - Loann Raud
- Univ Brest, Inserm, EFS, UMR1078, GGB, F-29200, France; Association Gaétan Saleün, F-29200, France
| | | | - Isabelle Gourlaouen
- Univ Brest, Inserm, EFS, UMR1078, GGB, F-29200, France; Laboratory of Excellence GR-Ex, F-75015, France
| | | | - Kévin Uguen
- Univ Brest, Inserm, EFS, UMR1078, GGB, F-29200, France; CHRU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Laboratoire de Génétique Moléculaire et Histocompatibilité, F-29200, France
| | - Jian-Min Chen
- Univ Brest, Inserm, EFS, UMR1078, GGB, F-29200, France
| | - Claude Férec
- Univ Brest, Inserm, EFS, UMR1078, GGB, F-29200, France; CHRU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Laboratoire de Génétique Moléculaire et Histocompatibilité, F-29200, France; Association Gaétan Saleün, F-29200, France
| | - Yann Fichou
- Univ Brest, Inserm, EFS, UMR1078, GGB, F-29200, France; Laboratory of Excellence GR-Ex, F-75015, France
| | - Gérald Le Gac
- Univ Brest, Inserm, EFS, UMR1078, GGB, F-29200, France; CHRU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Laboratoire de Génétique Moléculaire et Histocompatibilité, F-29200, France; Laboratory of Excellence GR-Ex, F-75015, France.
| |
Collapse
|
19
|
Wu Q, Hao Q, Li H, Wang B, Wang P, Jin X, Yu P, Gao G, Chang Y. Brain iron deficiency and affected contextual fear memory in mice with conditional Ferroportin1 ablation in the brain. FASEB J 2020; 35:e21174. [DOI: 10.1096/fj.202000167rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 11/11/2022]
Affiliation(s)
- Qiong Wu
- Laboratory of Molecular Iron Metabolism College of Life Science Hebei Normal University Shijiazhuang China
- College of Basic Medicine Hebei University of Chinese Medicine Shijiazhuang China
- Hebei Key Laboratory of Chinese Medicine Research on Cardio‐Cerebrovascular Disease Shijiazhuang China
| | - Qian Hao
- Laboratory of Molecular Iron Metabolism College of Life Science Hebei Normal University Shijiazhuang China
| | - Haiyan Li
- Laboratory of Molecular Iron Metabolism College of Life Science Hebei Normal University Shijiazhuang China
| | - Bo Wang
- Laboratory of Molecular Iron Metabolism College of Life Science Hebei Normal University Shijiazhuang China
| | - Peina Wang
- Laboratory of Molecular Iron Metabolism College of Life Science Hebei Normal University Shijiazhuang China
| | - Xiaofang Jin
- Laboratory of Molecular Iron Metabolism College of Life Science Hebei Normal University Shijiazhuang China
| | - Peng Yu
- Laboratory of Molecular Iron Metabolism College of Life Science Hebei Normal University Shijiazhuang China
| | - Guofen Gao
- Laboratory of Molecular Iron Metabolism College of Life Science Hebei Normal University Shijiazhuang China
| | - Yan‐Zhong Chang
- Laboratory of Molecular Iron Metabolism College of Life Science Hebei Normal University Shijiazhuang China
| |
Collapse
|
20
|
Viveiros A, Panzer M, Baumgartner N, Schaefer B, Finkenstedt A, Henninger B, Theurl I, Nachbaur K, Weiss G, Haubner R, Decristoforo C, Tilg H, Zoller H. Reduced iron export associated with hepcidin resistance can explain the iron overload spectrum in ferroportin disease. Liver Int 2020; 40:1941-1951. [PMID: 32450003 PMCID: PMC7496278 DOI: 10.1111/liv.14539] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Ferroportin disease (FD) and hemochromatosis type 4 (HH4) are associated with variants in the ferroportin-encoding gene SLC40A1. Both phenotypes are characterized by iron overload despite being caused by distinct variants that either mediate reduced cellular iron export in FD or resistance against hepcidin-induced inactivation of ferroportin in HH4. The aim of this study was to assess if reduced iron export also confers hepcidin resistance and causes iron overload in FD associated with the R178Q variant. METHODS The ferroportin disease variants R178Q andA77D and the HH4-variant C326Y were overexpressed in HEK-293T cells and subcellular localization was characterized by confocal microscopy and flow cytometry. Iron export and cytosolic ferritin were measured as markers of iron transport and radioligand binding studies were performed. The hepcidin-ferroportin axis was assessed by ferritin/hepcidin correlation in patients with different iron storage diseases. RESULTS In the absence of hepcidin, the R178Q and A77D variants exported less iron when compared to normal and C326Y ferroportin. In the presence of hepcidin, the R178Q and C326Y, but not the A77D-variant, exported more iron than cells expressing normal ferroportin. Regression analysis of serum hepcidin and ferritin in patients with iron overload are compatible with hepcidin deficiency in HFE hemochromatosis and hepcidin resistance in R178Q FD. CONCLUSIONS These results support a novel concept that in certain FD variants reduced iron export and hepcidin resistance could be interlinked. Evasion of mutant ferroportin from hepcidin-mediated regulation could result in uncontrolled iron absorption and iron overload despite reduced transport function.
Collapse
Affiliation(s)
- André Viveiros
- Department of Medicine IMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Marlene Panzer
- Department of Medicine IMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Nadja Baumgartner
- Department of Medicine IMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Benedikt Schaefer
- Department of Medicine IMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Armin Finkenstedt
- Department of Medicine IMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Benjamin Henninger
- Department of RadiologyMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Igor Theurl
- Department of Medicine IIMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Karin Nachbaur
- Department of Medicine IMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Günter Weiss
- Department of Medicine IIMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Roland Haubner
- Department of Nuclear MedicineMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Clemens Decristoforo
- Department of Nuclear MedicineMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Herbert Tilg
- Department of Medicine IMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Heinz Zoller
- Department of Medicine IMedical University and University Hospital of InnsbruckInnsbruckAustria
| |
Collapse
|
21
|
Type 4B hereditary hemochromatosis due to heterozygous p.D157A mutation in SLC40A1 complicated with hypopituitarism. Med Mol Morphol 2020; 54:60-67. [PMID: 32607777 DOI: 10.1007/s00795-020-00259-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/13/2020] [Indexed: 10/24/2022]
Abstract
Hemochromatosis is a clinical syndrome characterized by iron overload in various organs. We present here a case of type 4 hereditary hemochromatosis due to heterozygous mutation in SLC40A1 gene (p.D157A). SLC40A1 encodes ferroportin, a macromolecule only known as iron exporter from mammalian cells. He first presented symptoms correlated with hypopituitarism. Furthermore, marked hyperferritinemia and high transferrin saturation were revealed in combination with the findings of iron overload in the liver, spleen and pituitary gland by computed tomography and magnetic resonance imaging. Liver biopsy revealed iron deposition in both hepatocytes and Kupffer cells. SLC40A1 mutations are considered to cause wide heterogeneity by various ferroportin mutations. Thus, clinicopathological examinations seem to be very important for diagnosing phenotype of type 4 hemochromatosis in addition to the gene analysis. We diagnosed him as type 4B hereditary hemochromatosis (ferroportin-associated hemochromatosis) by the findings of high transferrin saturation and iron deposition in hepatocytes, and then started iron chelating treatment. We should suspect the possibility of hereditary hemochromatosis even in Japanese with severe iron overload. Although the same mutation in SLC40A1 gene (p.D157A) had been reported to cause "loss of function" phenotype, we considered that the mutation of our case caused "gain of function" phenotype.
Collapse
|
22
|
Rajamanickam K, Leela V, Suganya G, Basha SH, Parthiban M, Pazhanivel N, Mangala Gowri A. Expression of iron regulatory proteins in full-term swine placenta. Reprod Domest Anim 2020; 55:931-942. [PMID: 32449967 DOI: 10.1111/rda.13730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/16/2020] [Accepted: 05/17/2020] [Indexed: 11/29/2022]
Abstract
In swine, even though the pregnant sows were with iron abundance, the inborn iron reserve of piglets was compromised. This indicates the insufficiency of molecular machinery involved in local placental iron flux. Here, we investigated the expression of iron regulatory proteins like hepcidin and ferroportin and also their association with iron reserve, inflammation and oxidative stress in placenta of full-term pregnant sows (n = 6). Amplification and sequencing of placental DNA confirmed the presence of hepcidin (MN579557) and ferroportin (MN565887) sequences and their 100% identity with existing GenBank data. Real-time amplification of placental mRNA revealed significant higher expression of hepcidin (p < .05) than ferroportin. Western blot analysis of placental tissues revealed specific bands for both hepcidin (~8 kDa) and ferroportin (~62 kDa) molecules. Immunohistochemistry revealed the immunoreactivity for both proteins in the cytoplasm and membrane of trophoblastic cells of the placenta. Hepcidin and ferroportin expressions were positively associated with placental non-haem iron reserve (p < .0001; p = .033), lipid peroxidation (p = .0060; p < .0001) and reactive oxygen species level (p = .0092; p = .0292). Hepcidin expression was positively associated with interleukin - 6 (p = .0002) and interferon gamma (p < .0001) expressions but ferroportin expression was negatively associated with interleukin-6 (p = .0005), interleukin-1β (p = .0226) and interferon gamma (p = .0059) expressions. This indicates hepcidin and ferroportin may have a role in controlling the local placental iron flux by acting as a molecular bridge between iron trafficking and inflammation.
Collapse
Affiliation(s)
- Kandasamy Rajamanickam
- Department of Veterinary Physiology, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Venkatasubramanian Leela
- Department of Veterinary Physiology, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Gopalakrishnan Suganya
- Department of Veterinary Physiology, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Sabiha Hayath Basha
- Centre for Stem Cell Research and Regenerative Medicine, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Manoharan Parthiban
- Department of Animal Biotechnology, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Natesan Pazhanivel
- Department of Veterinary Pathology, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Angappan Mangala Gowri
- Centre for Stem Cell Research and Regenerative Medicine, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| |
Collapse
|
23
|
Saraf SL, Gordeuk VR. Iron. ESSENTIAL AND TOXIC TRACE ELEMENTS AND VITAMINS IN HUMAN HEALTH 2020:83-102. [DOI: 10.1016/b978-0-12-805378-2.00006-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
24
|
Yin X, Zhang Y, Gao H, Jin QL, Wen XY. A case report of hereditary hemochromatosis caused by mutation of SLC40A1 gene. Medicine (Baltimore) 2019; 98:e17526. [PMID: 31689754 PMCID: PMC6946363 DOI: 10.1097/md.0000000000017526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 08/16/2019] [Accepted: 09/11/2019] [Indexed: 12/31/2022] Open
Abstract
RATIONALE Hereditary hemochromatosis (HH) is a frequent autosomal recessive disease. The pathogenesis of disease is excessive intestinal absorption of dietary iron, resulting in pathologically high iron storage in tissues and organs. As a systemic disease, it has several manifestations including cirrhosis, diabetes mellitus, cardiomyopathy, joint disease. However, a proportion of patients are asymptomatic. PATIENT CONCERNS A 34-year-old man who had abnormal liver function for 9 months without specific symptoms. He underwent various tests, including liver biopsy and genetic testing, which eventually ruled out common liver diseases and identified iron metabolic abnormalities. In addition, we confirmed the pathogenic genes by sequencing the genes of him and his families. DIAGNOSIS Combined with the symptoms, auxiliary examinations and sequencing results, the patient was diagnosed as HH. INTERVENTIONS The patient was given a low iron diet and phlebotomy therapy interval 2 weeks until the ferritin is <100 mg/L. OUTCOMES The patient' condition is stable during the follow-up period. LESSONS When clinicians are confronted with unexplained liver dysfunction, the possibility of the HH should be considered. Liver biopsy and gene sequencing are helpful in diagnosis. Phlebotomy treatment is the most economical and practical treatment for HH at present, but it should vary from person to person.
Collapse
Affiliation(s)
- Xin Yin
- Department of Hepatology, The First Hospital of Jilin University, Changchun
| | - Yu Zhang
- Department of Hepatology, The First Hospital of Jilin University, Changchun
| | - Hui Gao
- Department of Digestive disease, Tai’an Municipal Hospital, Tai’an, China
| | - Qing-long Jin
- Department of Hepatology, The First Hospital of Jilin University, Changchun
| | - Xiao-yu Wen
- Department of Hepatology, The First Hospital of Jilin University, Changchun
| |
Collapse
|
25
|
Vlasveld LT, Janssen R, Bardou-Jacquet E, Venselaar H, Hamdi-Roze H, Drakesmith H, Swinkels DW. Twenty Years of Ferroportin Disease: A Review or An Update of Published Clinical, Biochemical, Molecular, and Functional Features. Pharmaceuticals (Basel) 2019; 12:ph12030132. [PMID: 31505869 PMCID: PMC6789780 DOI: 10.3390/ph12030132] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/14/2019] [Accepted: 08/20/2019] [Indexed: 12/14/2022] Open
Abstract
Iron overloading disorders linked to mutations in ferroportin have diverse phenotypes in vivo, and the effects of mutations on ferroportin in vitro range from loss of function (LOF) to gain of function (GOF) with hepcidin resistance. We reviewed 359 patients with 60 ferroportin variants. Overall, macrophage iron overload and low/normal transferrin saturation (TSAT) segregated with mutations that caused LOF, while GOF mutations were linked to high TSAT and parenchymal iron accumulation. However, the pathogenicity of individual variants is difficult to establish due to the lack of sufficiently reported data, large inter-assay variability of functional studies, and the uncertainty associated with the performance of available in silico prediction models. Since the phenotypes of hepcidin-resistant GOF variants are indistinguishable from the other types of hereditary hemochromatosis (HH), these variants may be categorized as ferroportin-associated HH, while the entity ferroportin disease may be confined to patients with LOF variants. To further improve the management of ferroportin disease, we advocate for a global registry, with standardized clinical analysis and validation of the functional tests preferably performed in human-derived enterocytic and macrophagic cell lines. Moreover, studies are warranted to unravel the definite structure of ferroportin and the indispensable residues that are essential for functionality.
Collapse
Affiliation(s)
- L Tom Vlasveld
- Department of Internal Medicine, Haaglanden MC-Bronovo, 2597AX The Hague, The Netherlands
| | - Roel Janssen
- Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Edouard Bardou-Jacquet
- Liver Diseases Department, French Reference Centre for Rare Iron Overload Diseases of Genetic Origin, University Hospital Pontchaillou, 35033 Rennes, France
| | - Hanka Venselaar
- Centre for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud, University Medical Center, P.O. Box 9191, 6500 HB Nijmegen, The Netherlands
| | - Houda Hamdi-Roze
- Molecular Genetics Department, French Reference Centre for Rare Iron Overload Diseases of Genetic Origin, University Hospital Pontchaillou, 35033 Rennes, France
| | - Hal Drakesmith
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX39DS, UK
| | - Dorine W Swinkels
- Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
26
|
Ferroportin Disease Caused by a Heterozygous Variant p.Cys326Phe in the SLC40A1 Gene and the Efficacy of Therapeutic Phlebotomy in Children. J Pediatr Hematol Oncol 2019; 41:e325-e328. [PMID: 30130274 DOI: 10.1097/mph.0000000000001301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Therapeutic phlebotomy is recommended for treating hereditary hemochromatosis. However, the procedure and its efficacy for children remain unclear. We describe a young female patient with ferroportin disease, which was confirmed from excess iron deposition within hepatocytes and by identifying a heterozygous variant p.Cys326Phe in SLC40A1. She had been followed without phlebotomy. Liver histology at age 13 years revealed iron deposition progression. Phlebotomy was initiated and her iron markers and imaging findings improved without severe adverse effects. Therapeutic phlebotomy for children is effective and well-tolerated and should be considered as early as possible after a hemochromatosis diagnosis.
Collapse
|
27
|
Viveiros A, Schaefer B, Tilg H, Zoller H. Iron Matryoshka-Haemochromatosis nested in Ferroportin Disease? Liver Int 2019; 39:1014-1015. [PMID: 31127686 DOI: 10.1111/liv.14061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 01/23/2019] [Accepted: 01/26/2019] [Indexed: 02/13/2023]
Affiliation(s)
- André Viveiros
- Department of Medicine I, Medical University of Innsbruck, Innsbruck, Austria
| | - Benedikt Schaefer
- Department of Medicine I, Medical University of Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Department of Medicine I, Medical University of Innsbruck, Innsbruck, Austria
| | - Heinz Zoller
- Department of Medicine I, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
28
|
Zhang W, Xu A, Li Y, Zhao S, Zhou D, Wu L, Zhang B, Zhao X, Wang Y, Wang X, Duan W, Wang Q, Nan Y, You H, Jia J, Ou X, Huang J. A novel SLC40A1 p.Y333H mutation with gain of function of ferroportin: A recurrent cause of haemochromatosis in China. Liver Int 2019; 39:1120-1127. [PMID: 30500107 DOI: 10.1111/liv.14013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/01/2018] [Accepted: 11/19/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Haemochromatosis type 4, also known as ferroportin disease, is an autosomal dominant genetic disorder caused by pathogenic mutations in the SLC40A1 gene, which encodes ferroportin 1 (FPN1). We have identified a novel SLC40A1 p.Y333H mutation in our previous study. In the present study, we tried to investigate the frequency and pathogenicity of the SLC40A1 p.Y333H mutation in haemochromatosis in China. METHODS Patients were analysed for SLC40A1 p.Y333H as well as mutations in the other classic haemochromatosis-related genes by Sanger sequencing. To analyse iron export capacity of the SLC40A1 p.Y333H mutant, the 293T cells were transfected with the SLC40A1 p.Y333H construct and then treated with hepcidin after exposure to ferric ammonium citrate. Cellular localization of mutant FPN1, expression of FPN1 and intracellular ferritin were analysed by immunofluorescence and Western blotting. RESULTS Of 22 unrelated cases with primary iron overload, three cases (3/22, 13.6%) harboured the SLC40A1 p.Y333H, with no missense mutations identified in any other classical haemochromatosis-related genes including HFE, HJV, HAMP and TFR2. Pedigree analysis showed that three probands and the son of one proband had haemochromatosis of stage 3, while the son of another proband with age of 16 showed elevated transferrin saturation but normal serum ferritin level. In vitro studies showed the mutant p.Y333H ferroportin was resistant to hepcidin, affecting the subsequent internalization and degradation of FPN1, and was associated with ferroportin gain of function. CONCLUSIONS The SLC40A1 p.Y333H mutation is associated with gain of function of ferroportin, representing one of the major aetiological factors of haemochromatosis in China.
Collapse
Affiliation(s)
- Wei Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Anjian Xu
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yanmeng Li
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Suxian Zhao
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Donghu Zhou
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lina Wu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Bei Zhang
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xinyan Zhao
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Yu Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Xiaoming Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Weijia Duan
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Qianyi Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Yuemin Nan
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hong You
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Xiaojuan Ou
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jian Huang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
29
|
Haschka D, Petzer V, Kocher F, Tschurtschenthaler C, Schaefer B, Seifert M, Sopper S, Sonnweber T, Feistritzer C, Arvedson TL, Zoller H, Stauder R, Theurl I, Weiss G, Tymoszuk P. Classical and intermediate monocytes scavenge non-transferrin-bound iron and damaged erythrocytes. JCI Insight 2019; 4:98867. [PMID: 30996139 PMCID: PMC6538345 DOI: 10.1172/jci.insight.98867] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/14/2019] [Indexed: 12/12/2022] Open
Abstract
Myelomonocytic cells are critically involved in iron turnover as aged RBC recyclers. Human monocytes are divided in 3 subpopulations of classical, intermediate, and nonclassical cells, differing in inflammatory and migratory phenotype. Their functions in iron homeostasis are, however, unclear. Here, we asked whether the functional diversity of monocyte subsets translates into differences in handling physiological and pathological iron species. By microarray data analysis and flow cytometry we identified a set of iron-related genes and proteins upregulated in classical and, in part, intermediate monocytes. These included the iron exporter ferroportin (FPN1), ferritin, transferrin receptor, putative transporters of non-transferrin-bound iron (NTBI), and receptors for damaged erythrocytes. Consequently, classical monocytes displayed superior scavenging capabilities of potentially toxic NTBI, which were augmented by blocking iron export via hepcidin. The same subset and, to a lesser extent, the intermediate population, efficiently cleared damaged erythrocytes in vitro and mediated erythrophagocytosis in vivo in healthy volunteers and patients having received blood transfusions. To summarize, our data underline the physiologically important function of the classical and intermediate subset in clearing NTBI and damaged RBCs. As such, these cells may play a nonnegligible role in iron homeostasis and limit iron toxicity in iron overload conditions. Human classical and intermediate monocytes mediate clearance of non-transferrin-bound iron and erythrophagocytosis.
Collapse
Affiliation(s)
| | | | | | | | - Benedikt Schaefer
- Department of Internal Medicine I, Medical University of Innsbruck, Innsbruck, Austria
| | | | | | | | | | - Tara L Arvedson
- Department of Oncology, Amgen Inc., Thousand Oaks, California, USA
| | - Heinz Zoller
- Department of Internal Medicine I, Medical University of Innsbruck, Innsbruck, Austria
| | | | | | | | | |
Collapse
|
30
|
Lv T, Zhang W, Xu A, Li Y, Zhou D, Zhang B, Li X, Zhao X, Wang Y, Wang X, Duan W, Wang Q, Xu H, Zheng J, Zhao R, Zhu L, Dong Y, Lu L, Chen Y, Long J, Zheng S, Wang W, You H, Jia J, Ou X, Huang J. Non- HFE mutations in haemochromatosis in China: combination of heterozygous mutations involving HJV signal peptide variants. J Med Genet 2018; 55:650-660. [PMID: 30166352 DOI: 10.1136/jmedgenet-2018-105348] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/22/2018] [Accepted: 07/08/2018] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Hereditary haemochromatosis (HH) caused by a homozygous p.C282Y mutation in haemochromatosis (HFE) gene has been well documented. However, less is known about the causative non-HFE mutation. We aimed to assess mutation patterns of haemochromatosis-related genes in Chinese patients with primary iron overload. METHODS Patients were preanalysed for mutations in the classic HH-related genes: HFE, HJV, HAMP, TFR2 and SLC40A1. Whole exome sequencing was conducted for cases with variants in HJV signal peptide region. Representative variants were analysed for biological function. RESULTS None of the cases analysed harboured the HFE p.C282Y; however, 21 of 22 primary iron-overload cases harboured at least one non-synonymous variant in the non-HFE genes. Specifically, p.E3D or p.Q6H variants in the HJV signal peptide region were identified in nine cases (40.9%). In two of three probands with the HJV p.E3D, exome sequencing identified accompanying variants in BMP/SMAD pathway genes, including TMPRSS6 p.T331M and BMP4 p.R269Q, and interestingly, SUGP2 p.R639Q was identified in all the three cases. Pedigree analysis showed a similar pattern of combination of heterozygous mutations in cases with HJV p.E3D or p.Q6H, with SUGP2 p.R639Q or HJV p.C321X being common mutation. In vitro siRNA interference of SUGP2 showed a novel role of downregulating the BMP/SMAD pathway. Site-directed mutagenesis of HJV p.Q6H/p.C321X in cell lines resulted in loss of membrane localisation of mutant HJV, and downregulation of p-SMAD1/5 and HAMP. CONCLUSION Compound heterozygous mutations of HJV or combined heterozygous mutations of BMP/SMAD pathway genes, marked by HJV variants in the signal peptide region, may represent a novel aetiological factor for HH.
Collapse
Affiliation(s)
- Tingxia Lv
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Wei Zhang
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Anjian Xu
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Yanmeng Li
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Donghu Zhou
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Bei Zhang
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Xiaojin Li
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Xinyan Zhao
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Yu Wang
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Xiaoming Wang
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Weijia Duan
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Qianyi Wang
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Hexiang Xu
- Department of Infectious Diseases, The Third Affiliated Hospital of Anhui Medical University, The First People's Hospital of Hefei, Hefei, China
| | - JiShun Zheng
- Department of Infectious Diseases, The Third Affiliated Hospital of Anhui Medical University, The First People's Hospital of Hefei, Hefei, China
| | - Rongrong Zhao
- Department of Infectious Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Longdong Zhu
- Department of Infectious Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yuwei Dong
- Department of Gastroenterology, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Lungen Lu
- Department of Gastroenterology, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yongpeng Chen
- Department of Infectious Diseases, Institute of Hepatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiang Long
- Department of Oncology Minimally Invasive Interventional Radiology, Beijing You-An Hospital, Capital Medical University, Shanghai, China
| | - Sujun Zheng
- Artificial Liver Center, Beijing You-An Hospital, Capital Medical University, Beijing, China
| | - Wei Wang
- Department of Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hong You
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jidong Jia
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Xiaojuan Ou
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jian Huang
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| |
Collapse
|
31
|
Hayashi H, Yano M, Urawa N, Mizutani A, Hamaoka S, Araki J, Kojima Y, Naito Y, Kato A, Tatsumi Y, Kato K. A 10-year Follow-up Study of a Japanese Family with Ferroportin Disease A: Mild Iron Overload with Mild Hyperferritinemia Co-occurring with Hyperhepcidinemia May Be Benign. Intern Med 2018; 57:2865-2871. [PMID: 29780118 PMCID: PMC6207810 DOI: 10.2169/internalmedicine.0481-17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
This is a 10-year follow-up study of a family with ferroportin disease A. The proband, a 59-year-old man showed no noteworthy findings with the exception of an abnormal iron level. The proband's 90-year-old father showed reduced abilities in gait and cognition; however, with the exception of his iron level, his biochemistry results were almost normal. Brain imaging showed age-matched atrophy and iron deposition. In both patients, the serum levels of ferritin and hepcidin25, and liver computed tomography scores declined over a 10-year period. These changes were mainly due to a habitual change to a low-iron diet. The iron disorder in this family was not associated with major organ damage.
Collapse
Affiliation(s)
- Hisao Hayashi
- Department of Medicine, Aichi-Gakuin University School of Pharmacy, Japan
| | - Motoyoshi Yano
- Department of Gastroenterology, Yokkaichi Municipal Hospital, Japan
| | - Naohito Urawa
- Department of Hepatology, Ise Red Cross Hospital, Japan
| | | | - Shima Hamaoka
- Department of Hepatology, Ise Red Cross Hospital, Japan
| | - Jun Araki
- Department of Hepatology, Ise Red Cross Hospital, Japan
| | - Yuji Kojima
- Department of Hepatology, Ise Red Cross Hospital, Japan
| | - Yutaka Naito
- Department of Neurology, Ise Red Cross Hospital, Japan
| | - Ayako Kato
- Department of Medicine, Aichi-Gakuin University School of Pharmacy, Japan
| | - Yasuaki Tatsumi
- Department of Medicine, Aichi-Gakuin University School of Pharmacy, Japan
| | - Koichi Kato
- Department of Medicine, Aichi-Gakuin University School of Pharmacy, Japan
| |
Collapse
|
32
|
Ka C, Guellec J, Pepermans X, Kannengiesser C, Ged C, Wuyts W, Cassiman D, de Ledinghen V, Varet B, de Kerguenec C, Oudin C, Gourlaouen I, Lefebvre T, Férec C, Callebaut I, Le Gac G. The SLC40A1 R178Q mutation is a recurrent cause of hemochromatosis and is associated with a novel pathogenic mechanism. Haematologica 2018; 103:1796-1805. [PMID: 30002125 PMCID: PMC6278975 DOI: 10.3324/haematol.2018.189845] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/06/2018] [Indexed: 12/29/2022] Open
Abstract
Hemochromatosis type 4 is one of the most common causes of primary iron overload, after HFE-related hemochromatosis. It is an autosomal dominant disorder, primarily due to missense mutations in SLC40A1. This gene encodes ferroportin 1 (FPN1), which is the sole iron export protein reported in mammals. Not all heterozygous missense mutations in SLC40A1 are disease-causing. Due to phenocopies and an increased demand for genetic testing, rare SLC40A1 variations are fortuitously observed in patients with a secondary cause of hyperferritinemia. Structure/function analysis is the most effective way of establishing causality when clinical and segregation data are lacking. It can also provide important insights into the mechanism of iron egress and FPN1 regulation by hepcidin. The present study aimed to determine the pathogenicity of the previously reported p.Arg178Gln variant. We present the biological, clinical, histological and radiological findings of 22 patients from six independent families of French, Belgian or Iraqi decent. Despite phenotypic variability, all patients with p.Arg178Gln had elevated serum ferritin concentrations and normal to low transferrin saturation levels. In vitro experiments demonstrated that the p.Arg178Gln mutant reduces the ability of FPN1 to export iron without causing protein mislocalization. Based on a comparative model of the 3D structure of human FPN1 in an outward facing conformation, we argue that p.Arg178 is part of an interaction network modulating the conformational changes required for iron transport. We conclude that p.Arg178Gln represents a new category of loss-of-function mutations and that the study of “gating residues” is necessary in order to fully understand the action mechanism of FPN1.
Collapse
Affiliation(s)
- Chandran Ka
- UMR1078, INSERM, Université Bretagne Loire - Université de Bretagne Occidentale, Etablissement Français du Sang - Bretagne, Institut Brestois Santé-Agro-Matière, Brest, France.,Laboratoire de Génétique Moléculaire et Histocompatibilité, CHRU de Brest, Hôpital Morvan, France.,Laboratory of Excellence GR-Ex, Paris, France
| | - Julie Guellec
- UMR1078, INSERM, Université Bretagne Loire - Université de Bretagne Occidentale, Etablissement Français du Sang - Bretagne, Institut Brestois Santé-Agro-Matière, Brest, France.,Laboratory of Excellence GR-Ex, Paris, France.,Association Gaetan Saleun, Brest, France
| | - Xavier Pepermans
- Center for Human Genetics, University Hospital of St-Luc, Brussels, Belgium
| | - Caroline Kannengiesser
- Laboratory of Excellence GR-Ex, Paris, France.,UMR1149, INSERM, Centre de Recherche sur l'Inflammation, Université Paris Diderot, AP-HP, Hôpital Bichat, Département de Génétique, France.,On behalf of the French National Network for the Molecular Diagnosis of Inherited Iron Overload Disorders (J. Rochette, E. Cadet, C. Kannengiesser, H. Puy, C. Ged, H. de Verneuil, G. Le Gac, C. Férec, S. Pissard, V. Gérolami), Brest, France
| | - Cécile Ged
- On behalf of the French National Network for the Molecular Diagnosis of Inherited Iron Overload Disorders (J. Rochette, E. Cadet, C. Kannengiesser, H. Puy, C. Ged, H. de Verneuil, G. Le Gac, C. Férec, S. Pissard, V. Gérolami), Brest, France.,INSERM U1035, BMGIC, CHU de Bordeaux, Laboratoire de Biochimie et Biologie Moléculaire, France
| | - Wim Wuyts
- Department of Medical Genetics, University and University Hospital of Antwerp, Edegem, Belgium
| | - David Cassiman
- Department of Gastroenterology-Hepatology and Metabolic Center, University Hospital of Leuven, Belgium
| | - Victor de Ledinghen
- Department of Gastroenterology and Digestive Oncology, University Hospital of Bordeaux, France
| | - Bruno Varet
- Université Paris Descartes et AP-HP, Hôpital Necker, Service d'Hématologie, France
| | | | - Claire Oudin
- UMR1149, INSERM, Centre de Recherche sur l'Inflammation, Université Paris Diderot, AP-HP, Hôpital Bichat, Département de Génétique, France
| | - Isabelle Gourlaouen
- UMR1078, INSERM, Université Bretagne Loire - Université de Bretagne Occidentale, Etablissement Français du Sang - Bretagne, Institut Brestois Santé-Agro-Matière, Brest, France.,Laboratory of Excellence GR-Ex, Paris, France
| | - Thibaud Lefebvre
- UMR1149, INSERM, Centre de Recherche sur l'Inflammation, Université Paris Diderot, AP-HP, Hôpital Bichat, Département de Génétique, France
| | - Claude Férec
- UMR1078, INSERM, Université Bretagne Loire - Université de Bretagne Occidentale, Etablissement Français du Sang - Bretagne, Institut Brestois Santé-Agro-Matière, Brest, France.,Laboratoire de Génétique Moléculaire et Histocompatibilité, CHRU de Brest, Hôpital Morvan, France.,On behalf of the French National Network for the Molecular Diagnosis of Inherited Iron Overload Disorders (J. Rochette, E. Cadet, C. Kannengiesser, H. Puy, C. Ged, H. de Verneuil, G. Le Gac, C. Férec, S. Pissard, V. Gérolami), Brest, France
| | - Isabelle Callebaut
- UMR7590, CNRS, Sorbonne Universités, Université Pierre et Marie Curie-Paris, France
| | - Gérald Le Gac
- UMR1078, INSERM, Université Bretagne Loire - Université de Bretagne Occidentale, Etablissement Français du Sang - Bretagne, Institut Brestois Santé-Agro-Matière, Brest, France .,Laboratoire de Génétique Moléculaire et Histocompatibilité, CHRU de Brest, Hôpital Morvan, France.,Laboratory of Excellence GR-Ex, Paris, France.,On behalf of the French National Network for the Molecular Diagnosis of Inherited Iron Overload Disorders (J. Rochette, E. Cadet, C. Kannengiesser, H. Puy, C. Ged, H. de Verneuil, G. Le Gac, C. Férec, S. Pissard, V. Gérolami), Brest, France
| |
Collapse
|
33
|
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: 39] [Impact Index Per Article: 5.6] [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
|
34
|
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
|
35
|
Characterization of three novel pathogenic SLC40A1 mutations and genotype/phenotype correlations in 7 Italian families with type 4 hereditary hemochromatosis. Biochim Biophys Acta Mol Basis Dis 2018; 1864:464-470. [DOI: 10.1016/j.bbadis.2017.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/23/2017] [Accepted: 11/13/2017] [Indexed: 11/19/2022]
|
36
|
Tortosa V, Bonaccorsi di Patti MC, Brandi V, Musci G, Polticelli F. An improved structural model of the human iron exporter ferroportin. Insight into the role of pathogenic mutations in hereditary hemochromatosis type 4. BIO-ALGORITHMS AND MED-SYSTEMS 2017. [DOI: 10.1515/bams-2017-0029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
AbstractFerroportin (Fpn) is a membrane protein representing the major cellular iron exporter, essential for metal translocation from cells into plasma. Despite its pivotal role in human iron homeostasis, many questions on Fpn structure and biology remain unanswered. In this work, we present two novel and more reliable structural models of human Fpn (hFpn; inward-facing and outward-facing conformations) obtained using as templates the recently solved crystal structures of a bacterial homologue of hFpn,
Collapse
|
37
|
Abstract
Trace elements are chemical elements needed in minute amounts for normal physiology. Some of the physiologically relevant trace elements include iodine, copper, iron, manganese, zinc, selenium, cobalt and molybdenum. Of these, some are metals, and in particular, transition metals. The different electron shells of an atom carry different energy levels, with those closest to the nucleus being lowest in energy. The number of electrons in the outermost shell determines the reactivity of such an atom. The electron shells are divided in sub-shells, and in particular the third shell has s, p and d sub-shells. Transition metals are strictly defined as elements whose atom has an incomplete d sub-shell. This incomplete d sub-shell makes them prone to chemical reactions, particularly redox reactions. Transition metals of biologic importance include copper, iron, manganese, cobalt and molybdenum. Zinc is not a transition metal, since it has a complete d sub-shell. Selenium, on the other hand, is strictly speaking a nonmetal, although given its chemical properties between those of metals and nonmetals, it is sometimes considered a metalloid. In this review, we summarize the current knowledge on the inborn errors of metal and metalloid metabolism.
Collapse
Affiliation(s)
- Carlos R. Ferreira
- Division of Genetics and Metabolism, Children’s National Health System, Washington, DC, USA
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Section on Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - William A. Gahl
- Section on Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
38
|
Pietrangelo A. Ferroportin disease: pathogenesis, diagnosis and treatment. Haematologica 2017; 102:1972-1984. [PMID: 29101207 PMCID: PMC5709096 DOI: 10.3324/haematol.2017.170720] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/25/2017] [Indexed: 12/14/2022] Open
Abstract
Ferroportin Disease (FD) is an autosomal dominant hereditary iron loading disorder associated with heterozygote mutations of the ferroportin-1 (FPN) gene. It represents one of the commonest causes of genetic hyperferritinemia, regardless of ethnicity. FPN1 transfers iron from the intestine, macrophages and placenta into the bloodstream. In FD, loss-of-function mutations of FPN1 limit but do not impair iron export in enterocytes, but they do severely affect iron transfer in macrophages. This leads to progressive and preferential iron trapping in tissue macrophages, reduced iron release to serum transferrin (i.e. inappropriately low transferrin saturation) and a tendency towards anemia at menarche or after intense bloodletting. The hallmark of FD is marked iron accumulation in hepatic Kupffer cells. Numerous FD-associated mutations have been reported worldwide, with a few occurring in different populations and some more commonly reported (e.g. Val192del, A77D, and G80S). FPN1 polymorphisms also represent the gene variants most commonly responsible for hyperferritinemia in Africans. Differential diagnosis includes mainly hereditary hemochromatosis, the syndrome commonly due to either HFE or TfR2, HJV, HAMP, and, in rare instances, FPN1 itself. Here, unlike FD, hyperferritinemia associates with high transferrin saturation, iron-spared macrophages, and progressive parenchymal cell iron load. Abdominal magnetic resonance imaging (MRI), the key non-invasive diagnostic tool for the diagnosis of FD, shows the characteristic iron loading SSL triad (spleen, spine and liver). A non-aggressive phlebotomy regimen is recommended, with careful monitoring of transferrin saturation and hemoglobin due to the risk of anemia. Family screening is mandatory since siblings and offspring have a 50% chance of carrying the pathogenic mutation.
Collapse
Affiliation(s)
- Antonello Pietrangelo
- Center for Hemochromatosis, Department of Internal Medicine II, University of Modena and Reggio Emilia Policlinico, Modena, Italy
| |
Collapse
|
39
|
Wallace DF, McDonald CJ, Ostini L, Iser D, Tuckfield A, Subramaniam VN. The dynamics of hepcidin-ferroportin internalization and consequences of a novel ferroportin disease mutation. Am J Hematol 2017; 92:1052-1061. [PMID: 28681497 DOI: 10.1002/ajh.24844] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/29/2017] [Accepted: 06/30/2017] [Indexed: 12/31/2022]
Abstract
The hepcidin-ferroportin axis underlies the pathophysiology of many iron-associated disorders and is a key target for the development of therapeutics for treating iron-associated disorders. The aims of this study were to investigate the dynamics of hepcidin-mediated ferroportin internalization and the consequences of a novel disease-causing mutation on ferroportin function. Specific reagents for ferroportin are limited; we developed and characterized antibodies against the largest extracellular loop of ferroportin and developed a novel cell-based assay for studying hepcidin-ferroportin function. We show that hepcidin-mediated ferroportin internalization is a rapid process and could be induced using low concentrations of hepcidin. Targeted next-generation sequencing utilizing an iron metabolism gene panel developed in our group identified a novel ferroportin p.D84E variant in a patient with iron overload. Wild-type and mutant ferroportin constructs were generated, transfected into HEK293 cells and analysed using an all-in-one flow-cytometry-based assay to study the effects on hepcidin-mediated internalization and iron transport. Consistent with the classical phenotype of ferroportin disease, the p.D84E mutation results in an inability to transport iron and hepcidin insensitivity. These results validate a recently proposed 3D-structural model of ferroportin and highlight the significance of this variant in the structure and function of ferroportin. Our novel ferroportin antibody and assay will be valuable tools for investigating the regulation of hepcidin/ferroportin function and the development of novel approaches for the therapeutic modulation of iron homeostasis.
Collapse
Affiliation(s)
- Daniel F. Wallace
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences. Queensland University of Technology (QUT); Brisbane Queensland Australia
- Membrane Transport Laboratory; QIMR Berghofer Medical Research Institute; Brisbane Queensland Australia
| | - Cameron J. McDonald
- Membrane Transport Laboratory; QIMR Berghofer Medical Research Institute; Brisbane Queensland Australia
| | - Lesa Ostini
- Membrane Transport Laboratory; QIMR Berghofer Medical Research Institute; Brisbane Queensland Australia
| | - David Iser
- Department of Gastroenterology; St Vincent's Hospital; Fitzroy Victoria Australia
| | | | - V. Nathan Subramaniam
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences. Queensland University of Technology (QUT); Brisbane Queensland Australia
- Membrane Transport Laboratory; QIMR Berghofer Medical Research Institute; Brisbane Queensland Australia
| |
Collapse
|
40
|
Zhang LY, Li XF, Liao XD, Zhang LY, Lu L, Luo XG. Effect of iron source on iron absorption and gene expression of iron transporters in the ligated duodenal loops of broilers. J Anim Sci 2017; 95:1587-1597. [PMID: 28464091 DOI: 10.2527/jas.2016.1147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
This experiment was conducted to investigate the effect of iron source on Fe absorption and the gene expression of divalent metal transporter 1 (DMT1) and ferroportin 1 (FPN1) in the ligated duodenal loops of broilers. The in situ ligated duodenal loops from Fe-deficient broiler chicks (28-d-old) were perfused with Fe solutions containing 0 to 14.33 mmol Fe/L from 1 of the following: Fe sulfate (FeSO∙7HO), Fe methionine with weak chelation strength (Fe-Met W; chelation strength is expressed as quotient of formation [Q] value, Q = 1.37), Fe proteinate with moderate chelation strength (Fe-Prot M; Q = 43.6), and Fe proteinate with extremely strong chelation strength (Fe-Prot ES; Q = 8,590) for up to 30 min. The gene expression of DMT1 and FPN1 in the duodenal loops from the control group and the groups treated with 3.58 mmol Fe/L from 1 of 4 Fe sources was analyzed. The absorption kinetics of Fe from different Fe sources in the duodenum followed a saturated carrier-dependent transport process. The maximum transport rate (J) values in the duodenum were greater ( < 0.03) for Fe-Prot ES and Fe-Prot M than for Fe-Met W and FeSO∙7HO. The Fe perfusion inhibited ( < 0.05) the mRNA expression of but enhanced ( < 0.0008) the mRNA expression of in the duodenum and had no effect ( > 0.14) on the protein expression levels of the 2 transporters. These results indicated that organic Fe sources with greater Q values showed higher Fe absorption; however, all Fe sources followed the same saturated carrier-dependent transport process in the duodenum, and DMT1 and FPN1 might participate in Fe absorption in the duodenum of broilers regardless of Fe source.
Collapse
|
41
|
Zhang W, Lv T, Huang J, Ou X. Type 4B hereditary hemochromatosis associated with a novel mutation in the SLC40A1 gene: A case report and a review of the literature. Medicine (Baltimore) 2017; 96:e8064. [PMID: 28930842 PMCID: PMC5617709 DOI: 10.1097/md.0000000000008064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 08/09/2017] [Accepted: 08/22/2017] [Indexed: 12/04/2022] Open
Abstract
RATIONALE Hereditary hemochromatosis can be divided into HFE- and non-HFE-related based on genetic mutations in different genes. HFE-related hemochromatosis is the most common inherited genetic disease in European populations but rare in Asia-pacific region. Recently, non-HFE-related hemochromatosis has been reported in patients from the Asian countries. PATIENT CONCERNS We report the case of a 48-year-old Chinese Han woman who presented with abnormal liver function, diabetes mellitus, hyperferritinemia, and high transferrin saturation, with severe iron overload in parenchymal cells, Kupffer cells, and periportal fibrosis on liver biopsy. No secondary factor for iron overload was identified. DIAGNOSES Sanger sequencing was conducted for the screening of mutation in the hereditary hemochromatosis related genes. The functional effect of a splicing mutation, SLC40A1 IVS 3+10 del gtt, was assessed by reverse-polymerase chain reaction analysis for SLC40A1 mRNA level, and by immunohistochemistry analysis of liver biopsy for ferroportin expression and cellular localization. OUTCOMES A novel splicing mutation IVS 3+10 del gtt was identified in the SLC40A1 gene. Functional analysis showed that IVS 3+10 del gtt in the SLC40A1 gene lead to a substantial reduction in the basal levels of SLC40A1 mRNA and increased membrane localization of ferroportin. Finally, the patient was diagnosed as ferroportin disease (type 4B hemochromatosis). LESSONS The present study is the first report to identify a classical splicing mutation in the SLC40A1 gene in type 4B hemochromatosis, and provide further evidence of the prevalence of type 4 hereditary hemochromatosis in Asian countries.
Collapse
Affiliation(s)
- Wei Zhang
- Liver Research Center, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University
- National Clinical Research Center for Digestive Diseases
| | - Tingxia Lv
- Liver Research Center, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University
- National Clinical Research Center for Digestive Diseases
- Experimental Center, Beijing Friendship Hospital, Capital Medical University; Beijing, China
| | - Jian Huang
- Liver Research Center, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University
- National Clinical Research Center for Digestive Diseases
- Experimental Center, Beijing Friendship Hospital, Capital Medical University; Beijing, China
| | - Xiaojuan Ou
- Liver Research Center, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University
- National Clinical Research Center for Digestive Diseases
| |
Collapse
|
42
|
Rockfield S, Raffel J, Mehta R, Rehman N, Nanjundan M. Iron overload and altered iron metabolism in ovarian cancer. Biol Chem 2017; 398:995-1007. [PMID: 28095368 DOI: 10.1515/hsz-2016-0336] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 01/09/2017] [Indexed: 12/28/2022]
Abstract
Iron is an essential element required for many processes within the cell. Dysregulation in iron homeostasis due to iron overload is detrimental. This nutrient is postulated to contribute to the initiation of cancer; however, the mechanisms by which this occurs remain unclear. Defining how iron promotes the development of ovarian cancers from precursor lesions is essential for developing novel therapeutic strategies. In this review, we discuss (1) how iron overload conditions may initiate ovarian cancer development, (2) dysregulated iron metabolism in cancers, (3) the interplay between bacteria, iron, and cancer, and (4) chemotherapeutic strategies targeting iron metabolism in cancer patients.
Collapse
|
43
|
Expression of hepcidin and ferroportin in full term placenta of pregnant cows. Theriogenology 2017; 103:90-97. [PMID: 28780484 DOI: 10.1016/j.theriogenology.2017.07.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/04/2017] [Accepted: 07/25/2017] [Indexed: 12/29/2022]
Abstract
Hepcidin (HEP) and ferroportin (FPN) play a central role in systemic iron homeostasis. The HEP/FPN axis controls both extracellular iron concentration and total body iron levels. HEP is synthesized mainly by hepatocytes and controls the absorption of dietary iron and the distribution of iron to the various cell types; its synthesis is regulated by both iron and innate immunity. FPN is a membrane protein and the major exporter of iron from mammalian cells, including iron recycling macrophages, iron absorbing duodenal enterocytes, and iron storing hepatocytes. HEP limits the pool of extracellular iron by binding FPN and mediating its degradation, thus preventing its release from intracellular sources. Here we investigated, for the first time, the molecular and morphological expression of HEP and FPN in placenta of pregnant cows at term. Their expression has been evaluated investigating their mRNAs by reverse transcriptase PCR (RT-PCR). Sequencing of related amplicons revealed a 100% identity with HEP and FPN sequences from Bos taurus as reported in the GeneBank (mRNASequence ID: NM_001114508.2 and ID: NM_001077970.1, respectively). HEP and FPN proteins have also been revealed by Western blot analysis and immunohistochemistry. The strongest immunoreactivity for both proteins was observed in the cytoplasm of the trophoblastic cells of the villi and the caruncular crypts of the placentome. Hep mRNA was more representative in caruncular rather cotyledonar areas; on the contrary, Fpn mRNA was more expressed in cotyledonar rather than in caruncular areas. Transcripts of ferritin, transferrin and its receptor have been also documented by real time RT-PCR. HEP and FPN placental proteins may play a dual role. HEP/FPN axis seems to have a central role in infections, with microorganisms within macrophages or that survive in the bloodstream or other cellular spaces. In addition, HEP may be responsible for iron flux regulation as a molecular bridge for iron trafficking and response to infection. FPN may also have a significant role for embryonic development, growth and organogenesis.
Collapse
|
44
|
Sabelli M, Montosi G, Garuti C, Caleffi A, Oliveto S, Biffo S, Pietrangelo A. Human macrophage ferroportin biology and the basis for the ferroportin disease. Hepatology 2017; 65:1512-1525. [PMID: 28027576 PMCID: PMC5413859 DOI: 10.1002/hep.29007] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 12/15/2016] [Indexed: 01/01/2023]
Abstract
Ferroportin (FPN1) is the sole iron exporter in mammals, but its cell-specific function and regulation are still elusive. This study examined FPN1 expression in human macrophages, the cells that are primarily responsible on a daily basis for plasma iron turnover and are central in the pathogenesis of ferroportin disease (FD), the disease attributed to lack-of-function FPN1 mutations. We characterized FPN1 protein expression and traffic by confocal microscopy, western blotting, gel filtration, and immunoprecipitation studies in macrophages from control blood donors (donor) and patients with either FPN1 p.A77D, p.G80S, and p.Val162del lack-of-function or p.A69T gain-of-function mutations. We found that in normal macrophages, FPN1 cycles in the early endocytic compartment does not multimerize and is promptly degraded by hepcidin (Hepc), its physiological inhibitor, within 3-6 hours. In FD macrophages, endogenous FPN1 showed a similar localization, except for greater accumulation in lysosomes. However, in contrast with previous studies using overexpressed mutant protein in cell lines, FPN1 could still reach the cell surface and be normally internalized and degraded upon exposure to Hepc. However, when FD macrophages were exposed to large amounts of heme iron, in contrast to donor and p.A69T macrophages, FPN1 could no longer reach the cell surface, leading to intracellular iron retention. CONCLUSION FPN1 cycles as a monomer within the endocytic/plasma membrane compartment and responds to its physiological inhibitor, Hepc, in both control and FD cells. However, in FD, FPN1 fails to reach the cell surface when cells undergo high iron turnover. Our findings provide a basis for the FD characterized by a preserved iron transfer in the enterocytes (i.e., cells with low iron turnover) and iron retention in cells exposed to high iron flux, such as liver and spleen macrophages. (Hepatology 2017;65:1512-1525).
Collapse
Affiliation(s)
- Manuela Sabelli
- Division of Internal Medicine 2 and Center for HemochromatosisUniversity Hospital of ModenaModenaItaly
| | - Giuliana Montosi
- Division of Internal Medicine 2 and Center for HemochromatosisUniversity Hospital of ModenaModenaItaly
| | - Cinzia Garuti
- Division of Internal Medicine 2 and Center for HemochromatosisUniversity Hospital of ModenaModenaItaly
| | - Angela Caleffi
- Division of Internal Medicine 2 and Center for HemochromatosisUniversity Hospital of ModenaModenaItaly
| | | | - Stefano Biffo
- INGM, ‘Romeo ed Enrica Invernizzi’MilanoItaly
- Department of BiosciencesUniversity of MilanMilanItaly
| | - Antonello Pietrangelo
- Division of Internal Medicine 2 and Center for HemochromatosisUniversity Hospital of ModenaModenaItaly
| |
Collapse
|
45
|
Nairz M, Theurl I, Swirski FK, Weiss G. "Pumping iron"-how macrophages handle iron at the systemic, microenvironmental, and cellular levels. Pflugers Arch 2017; 469:397-418. [PMID: 28251312 PMCID: PMC5362662 DOI: 10.1007/s00424-017-1944-8] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/25/2017] [Accepted: 01/29/2017] [Indexed: 12/12/2022]
Abstract
Macrophages reside in virtually every organ. First arising during embryogenesis, macrophages replenish themselves in the adult through a combination of self-renewal and influx of bone marrow-derived monocytes. As large phagocytic cells, macrophages participate in innate immunity while contributing to tissue-specific homeostatic functions. Among the key metabolic tasks are senescent red blood cell recycling, free heme detoxification, and provision of iron for de novo hemoglobin synthesis. While this systemic mechanism involves the shuttling of iron between spleen, liver, and bone marrow through the concerted function of defined macrophage populations, similar circuits appear to exist within the microenvironment of other organs. The high turnover of iron is the prerequisite for continuous erythropoiesis and tissue integrity but challenges macrophages’ ability to maintain cellular iron homeostasis and immune function. This review provides a brief overview of systemic, microenvironmental, and cellular aspects of macrophage iron handling with a focus on exciting and unresolved questions in the field.
Collapse
Affiliation(s)
- Manfred Nairz
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Anichstr. 35, 6020, Innsbruck, Austria. .,Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA. .,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Igor Theurl
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Anichstr. 35, 6020, Innsbruck, Austria
| | - Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Guenter Weiss
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Anichstr. 35, 6020, Innsbruck, Austria.
| |
Collapse
|
46
|
|
47
|
Cikomola JC, Vandepoele K, Katchunga PB, Kishabongo AS, Padalko EY, Speeckaert MM, Delanghe JR. The association between fructosamine-3 kinase 900C/G polymorphism, transferrin polymorphism and human herpesvirus-8 infection in diabetics living in South Kivu. Acta Trop 2016; 163:14-9. [PMID: 27461879 DOI: 10.1016/j.actatropica.2016.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 07/12/2016] [Accepted: 07/17/2016] [Indexed: 02/08/2023]
Abstract
Prevalences of human herpesvirus-8 (HHV-8) infection and diabetes mellitus are very common in certain parts of Africa, containing iron-rich soils. We hypothesized that some genetic factors could have a link with susceptibility to HHV-8 infection. We focused on ferroportin Q248H mutation (rs11568350), transferrin (TF) polymorphism and fructosamine-3 kinase (FN3K) 900C/G polymorphism (rs1056534). The study population consisted of 210 type 2 diabetic adults and 125 healthy controls recruited in Bukavu (South Kivu). In the whole study population (diabetics+healthy controls), ferroportin Q248H mutation was detected in 47 subjects (14.0%) with 43 heterozygotes and 4 homozygotes. TF phenotype frequencies were 88.1% (CC), 10.4% (CD) and 1.5% (BC). Genotype frequencies of FN3K 900C/G polymorphism were respectively 9,3% (CC), 43.3% (GC) and 47.4% (GG). Prevalence of HHV8-infection in the study population was 77.3%. HHV-8 infection rate and HHV-8 IgG antibody titer were significantly higher in diabetics then in controls (p<0.0001). Significant differences were observed in HHV-8 infection rate and in HHV-8 IgG antibody titer according to FN3K rs1056534 (p<0.05 and p<0.05, respectively) and TF polymorphism (p<0.05 and p=0.005, respectively). No significant differences in HHV-8 infection rate and in HHV-8 IgG antibody titer were observed in the ferroportin Q248H mutation carriers (rs11568350) in comparison with ferroportin wild type. In a multiple regression analysis, FN3K rs1056534, TF polymorphism and presence of diabetes mellitus were predictors for HHV-8 infection. In contrast to these findings, ferroportin Q248H mutation (rs11568350) did not influence the susceptibility for an HHV-8 infection in sub-Saharan Africans.
Collapse
|
48
|
Rametta R, Dongiovanni P, Pelusi S, Francione P, Iuculano F, Borroni V, Fatta E, Castagna A, Girelli D, Fargion S, Valenti L. Hepcidin resistance in dysmetabolic iron overload. Liver Int 2016; 36:1540-8. [PMID: 26998752 DOI: 10.1111/liv.13124] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/12/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Dysmetabolic iron overload syndrome (DIOS) is a frequent condition predisposing to metabolic, cardiovascular and hepatic damage, whose pathogenesis remains poorly defined. Aim of this study was to characterize iron metabolism in DIOS. METHODS We evaluated 18 patients with DIOS, compared to 18 with nonalcoholic fatty liver and 23 healthy individuals with normal iron status, and 10 patients with hereditary haemochromatosis by a 24-h oral iron tolerance test with hepcidin measurement and iron metabolism modelling under normal iron stores. RESULTS Dysmetabolic iron overload syndrome patients had higher peak transferrin saturation and area under the-curve of transferrin saturation than subjects with normal iron status, but lower values than haemochromatosis patients (P < 0.05 for all). Conversely, they had higher peak circulating hepcidin levels and area under the curve of hepcidin than the other groups (P < 0.05 for all). This was independent age, sex, haemoglobin, ferritin, and transferrin saturation levels (P = 0.0002). Hepcidin increase in response to the rise in transferrin saturation (hepcidin release index) was not impaired in DIOS patients. Viceversa, the ability of the hepcidin spike to control the rise in transferrin saturation at the beginning of the test (hepcidin resistance index) was impaired in DIOS (P = 0.0002). In DIOS patients, the hepcidin resistance index was correlated with ferritin levels at diagnosis (P = 0.016). CONCLUSIONS Dysmetabolic iron overload syndrome is associated with a subtle impairment in the ability of the iron hormone hepcidin to restrain iron absorption following an iron challenge, suggesting a hepcidin resistance state. Further studies are required to better characterize the molecular mechanism underpinning this new iron metabolism alteration.
Collapse
Affiliation(s)
- Raffaela Rametta
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Paola Dongiovanni
- Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy
| | - Serena Pelusi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Paolo Francione
- Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy
| | - Federica Iuculano
- Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy
| | - Vittorio Borroni
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Erika Fatta
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Annalisa Castagna
- Policlinico G.B. Rossi, Department of Medicine, Università di Verona, Verona, Italy
| | - Domenico Girelli
- Policlinico G.B. Rossi, Department of Medicine, Università di Verona, Verona, Italy
| | - Silvia Fargion
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.,Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy. .,Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy.
| |
Collapse
|
49
|
Lv T, Li X, Zhang W, Zhao X, Ou X, Huang J. Recent advance in the molecular genetics of Wilson disease and hereditary hemochromatosis. Eur J Med Genet 2016; 59:532-539. [PMID: 27592149 DOI: 10.1016/j.ejmg.2016.08.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 07/12/2016] [Accepted: 08/31/2016] [Indexed: 02/07/2023]
Abstract
Metabolic liver diseases such as Wilson disease (WD) and hereditary hemochromatosis (HH) possess complicated pathogenesis and typical hereditary characteristics with the hallmarks of a deficiency in metal metabolism. Mutations in genes encoding ATPase, Cu + transporting, beta polypeptide (ATP7B) and hemochromatosis (HFE) or several non-HFE genes are considered to be causative for WD and HH, respectively. Although the identification of novel mutations in ATP7B for WD and HFE or the non-HFE genes for HH has increased, especially with the application of whole genome sequencing technology in recent years, the biological function of the identified mutations, as well as genotype-phenotype correlations remain to be explored. Further analysis of the causative gene mutation would be critical to clarify the mechanisms underlying specific disease phenotypes. In this review, we therefore summarize the recent advances in the molecular genetics of WD and HH including the updated mutation spectrums and the correlation between genotype and phenotype, with an emphasis on biological functional studies of the individual mutations identified in WD and HH. The weakness of the current functional studies and analysis for the clinical association of the individual mutation was also discussed. These works are essential for the understanding of the association between genotypes and phenotypes of these inherited metabolic liver diseases.
Collapse
Affiliation(s)
- Tingxia Lv
- Liver Research Center, Experimental Center, Beijing Friendship Hospital, Capital Medical University, 95 Yong-an Road, Xuan-wu District, Beijing, 100050, China.
| | - Xiaojin Li
- Liver Research Center, Experimental Center, Beijing Friendship Hospital, Capital Medical University, 95 Yong-an Road, Xuan-wu District, Beijing, 100050, China.
| | - Wei Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, 95 Yong-an Road, Xuan-wu District, Beijing, 100050, China.
| | - Xinyan Zhao
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, 95 Yong-an Road, Xuan-wu District, Beijing, 100050, China.
| | - Xiaojuan Ou
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, 95 Yong-an Road, Xuan-wu District, Beijing, 100050, China.
| | - Jian Huang
- Liver Research Center, Experimental Center, Beijing Friendship Hospital, Capital Medical University, 95 Yong-an Road, Xuan-wu District, Beijing, 100050, China.
| |
Collapse
|
50
|
Ong SY, Nicoll AJ, Delatycki MB. How should hyperferritinaemia be investigated and managed? Eur J Intern Med 2016; 33:21-7. [PMID: 27346296 DOI: 10.1016/j.ejim.2016.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/18/2016] [Accepted: 05/12/2016] [Indexed: 12/15/2022]
Abstract
Hyperferritinaemia is commonly found in clinical practice. In assessing the cause of hyperferritinaemia, it is important to identify if there is true iron overload or not as hyperferritinaemia may be seen in other conditions such as excess alcohol intake, inflammation and non-alcoholic fatty liver disease. Assessment of whether the serum ferritin level is elevated or not should take into account body mass index, gender and age. This review article provides an overview of the different causes of hyperferritinaemia, differentiating those due to iron overload from those not due to iron overload, and provides an algorithm for clinicians to use in clinical practice to carry out appropriate investigations and management.
Collapse
Affiliation(s)
- Sim Y Ong
- Bruce Lefroy Centre, Murdoch Childrens Research Institute, Flemington Road, Parkville, Victoria 3052, Australia; The University of Melbourne, Parkville, Victoria 3010, Australia; Department of Gastroenterology and Hepatology, Royal Melbourne Hospital, 300 Grattan Street, Parkville, Victoria 3050, Australia.
| | - Amanda J Nicoll
- The University of Melbourne, Parkville, Victoria 3010, Australia; Department of Gastroenterology and Hepatology, Royal Melbourne Hospital, 300 Grattan Street, Parkville, Victoria 3050, Australia; Department of Gastroenterology, Eastern Health, Arnold Street, Box Hill, Victoria 3128, Australia; Monash University, Clayton, Victoria 3800, Australia.
| | - Martin B Delatycki
- Bruce Lefroy Centre, Murdoch Childrens Research Institute, Flemington Road, Parkville, Victoria 3052, Australia; The University of Melbourne, Parkville, Victoria 3010, Australia; Clinical Genetics, Austin Health, 145 Studley Road, Heidelberg, Victoria 3084, Australia.
| |
Collapse
|