1
|
Wu L, Zhang Y, Wang G, Ren J. Molecular Mechanisms and Therapeutic Targeting of Ferroptosis in Doxorubicin-Induced Cardiotoxicity. JACC Basic Transl Sci 2024; 9:811-826. [PMID: 39070280 PMCID: PMC11282888 DOI: 10.1016/j.jacbts.2023.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 07/30/2024]
Abstract
Ferroptosis, an iron-dependent form of regulated cell death, has received increasing attention for its pathophysiologic contribution to the onset and development of doxorubicin-induced cardiotoxicity. Moreover, modulation of ferroptosis with specific inhibitors may provide new therapeutic opportunities for doxorubicin-induced cardiotoxicity. Here, we will review the molecular mechanisms and therapeutic promise of targeting ferroptosis in doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Lin Wu
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Yingmei Zhang
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Guizhen Wang
- Department of Emergency, Shanghai Tenth People’s Hospital, School of Medicine Tongji University, Shanghai, China
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| |
Collapse
|
2
|
Zhang J, Song Y, Li Y, Lin HB, Fang X. Iron homeostasis in the heart: Molecular mechanisms and pharmacological implications. J Mol Cell Cardiol 2023; 174:15-24. [PMID: 36375319 DOI: 10.1016/j.yjmcc.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/13/2022]
Abstract
Iron is necessary for the life of practically all living things, yet it may also harm people toxically. Accordingly, humans and other mammals have evolved an effective and tightly regulatory system to maintain iron homeostasis in healthy tissues, including the heart. Iron deficiency is common in patients with heart failure, and is associated with worse prognosis in this population; while the prevalence of iron overload-related cardiovascular disorders is also increasing. Therefore, enhancing the therapy of patients with cardiovascular disorders requires a thorough understanding of iron homeostasis. Here, we give readers an overview of the fundamental mechanisms governing systemic iron homeostasis as well as the most recent knowledge about the intake, storage, use, and export of iron from the heart. Genetic mouse models used for investigation of iron metabolism in various in vivo scenarios are summarized and highlighted. We also go through different clinical conditions and therapeutic approaches that target cardiac iron dyshomeostasis. Finally, we conclude the review by outlining the present knowledge gaps and important open questions in this field in order to guide future research on cardiac iron metabolism.
Collapse
Affiliation(s)
- Jiawei Zhang
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Yijing Song
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - You Li
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Han-Bin Lin
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xuexian Fang
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China; Key Laboratory of Elemene Class Anti-cancer Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China.
| |
Collapse
|
3
|
Fang X, Ardehali H, Min J, Wang F. The molecular and metabolic landscape of iron and ferroptosis in cardiovascular disease. Nat Rev Cardiol 2023; 20:7-23. [PMID: 35788564 PMCID: PMC9252571 DOI: 10.1038/s41569-022-00735-4] [Citation(s) in RCA: 382] [Impact Index Per Article: 382.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/30/2022] [Indexed: 02/08/2023]
Abstract
The maintenance of iron homeostasis is essential for proper cardiac function. A growing body of evidence suggests that iron imbalance is the common denominator in many subtypes of cardiovascular disease. In the past 10 years, ferroptosis, an iron-dependent form of regulated cell death, has become increasingly recognized as an important process that mediates the pathogenesis and progression of numerous cardiovascular diseases, including atherosclerosis, drug-induced heart failure, myocardial ischaemia-reperfusion injury, sepsis-induced cardiomyopathy, arrhythmia and diabetic cardiomyopathy. Therefore, a thorough understanding of the mechanisms involved in the regulation of iron metabolism and ferroptosis in cardiomyocytes might lead to improvements in disease management. In this Review, we summarize the relationship between the metabolic and molecular pathways of iron signalling and ferroptosis in the context of cardiovascular disease. We also discuss the potential targets of ferroptosis in the treatment of cardiovascular disease and describe the current limitations and future directions of these novel treatment targets.
Collapse
Affiliation(s)
- Xuexian Fang
- grid.410595.c0000 0001 2230 9154Department of Nutrition and Toxicology, School of Public Health, State Key Laboratory of Experimental Hematology, Hangzhou Normal University, Hangzhou, China ,grid.13402.340000 0004 1759 700XThe Fourth Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China ,grid.412017.10000 0001 0266 8918The First Affiliated Hospital, The Second Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Hossein Ardehali
- grid.16753.360000 0001 2299 3507Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL USA
| | - Junxia Min
- The Fourth Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Fourth Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China. .,The First Affiliated Hospital, The Second Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China.
| |
Collapse
|
4
|
The Toxic Influence of Excess Free Iron on Red Blood Cells in the Biophysical Experiment: An In Vitro Study. J Toxicol 2022; 2022:7113958. [PMID: 35256882 PMCID: PMC8898121 DOI: 10.1155/2022/7113958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/10/2022] [Indexed: 11/18/2022] Open
Abstract
Iron is needed for life-essential processes, but free iron overload causes dangerous clinical consequences. The study of the role of red blood cells (RBCs) in the influence of excess free iron in the blood on the pathological consequences in an organism is relevant. Here, in a direct biophysical experiment in vitro, we studied the action of free iron overload on the packed red blood cell (pRBC) characteristics. In experiments, we incubated pRBCs with the ferrous sulfate solution (Fe2+). Wе used free iron in a wide range of concentrations. High Fe2+ concentrations made us possible to establish the pattern of the toxic effect of excess iron on pRBCs during a reduced incubation time in a biophysical experiment in vitro. It was found that excess free iron causes changes in pRBC morphology, the appearance of bridges between cells, and the formation of clots, increasing the membrane stiffness and methemoglobin concentration. We created a kinetic model of changes in the hemoglobin derivatives. The complex of simultaneous distortions of pRBCs established in our experiments can be taken into account when studying the mechanism of the toxic influence of excess free iron in the blood on pathological changes in an organism.
Collapse
|
5
|
Vinchi F. Non-Transferrin-Bound Iron in the Spotlight: Novel Mechanistic Insights into the Vasculotoxic and Atherosclerotic Effect of Iron. Antioxid Redox Signal 2021; 35:387-414. [PMID: 33554718 PMCID: PMC8328045 DOI: 10.1089/ars.2020.8167] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 01/06/2021] [Accepted: 01/25/2021] [Indexed: 12/11/2022]
Abstract
Significance: While atherosclerosis is an almost inevitable consequence of aging, food preferences, lack of exercise, and other aspects of the lifestyle in many countries, the identification of new risk factors is of increasing importance to tackle a disease, which has become a major health burden for billions of people. Iron has long been suspected to promote the development of atherosclerosis, but data have been conflicting, and the contribution of iron is still debated controversially. Recent Advances: Several experimental and clinical studies have been recently published about this longstanding controversial problem, highlighting the critical need to unravel the complexity behind this topic. Critical Issues: The aim of the current review is to provide an overview of the current knowledge about the proatherosclerotic impact of iron, and discuss the emerging role of non-transferrin-bound iron (NTBI) as driver of vasculotoxicity and atherosclerosis. Finally, I will provide detailed mechanistic insights on the cellular processes and molecular pathways underlying iron-exacerbated atherosclerosis. Overall, this review highlights a complex framework where NTBI acts at multiple levels in atherosclerosis by altering the serum and vascular microenvironment in a proatherogenic and proinflammatory manner, affecting the functionality and survival of vascular cells, promoting foam cell formation and inducing angiogenesis, calcification, and plaque destabilization. Future Directions: The use of additional iron markers (e.g., NTBI) may help adequately predict predisposition to cardiovascular disease. Clinical studies are needed in the aging population to address the atherogenic role of iron fluctuations within physiological limits and the therapeutic value of iron restriction approaches. Antioxid. Redox Signal. 35, 387-414.
Collapse
Affiliation(s)
- Francesca Vinchi
- Iron Research Program, Lindsley F. Kimball Research Institute (LFKRI), New York Blood Center (NYBC), New York, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, New York, USA
| |
Collapse
|
6
|
You P, Li X, Wang Z, Wang H, Dong B, Li Q. Characterization of Brain Iron Deposition Pattern and Its Association With Genetic Risk Factor in Alzheimer's Disease Using Susceptibility-Weighted Imaging. Front Hum Neurosci 2021; 15:654381. [PMID: 34163341 PMCID: PMC8215439 DOI: 10.3389/fnhum.2021.654381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 04/27/2021] [Indexed: 11/30/2022] Open
Abstract
The presence of iron is an important factor for normal brain functions, whereas excessive deposition of iron may impair normal cognitive function in the brain and lead to Alzheimer’s disease (AD). MRI has been widely applied to characterize brain structural and functional changes caused by AD. However, the effectiveness of using susceptibility-weighted imaging (SWI) for the analysis of brain iron deposition is still unclear, especially within the context of early AD diagnosis. Thus, in this study, we aim to explore the relationship between brain iron deposition measured by SWI with the progression of AD using various feature selection and classification methods. The proposed model was evaluated on a 69-subject SWI imaging dataset consisting of 24 AD patients, 21 mild cognitive impairment patients, and 24 normal controls. The identified AD progression-related regions were then compared with the regions reported from previous genetic association studies, and we observed considerable overlap between these two. Further, we have identified a new potential AD-related gene (MEF2C) closely related to the interaction between iron deposition and AD progression in the brain.
Collapse
Affiliation(s)
- Peiting You
- Beijing International Center for Mathematical Research, Peking University, Beijing, China.,Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Xiang Li
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Zhijiang Wang
- Peking University Institute of Mental Health (Sixth Hospital), Beijing, China.,National Clinical Research Center for Mental Disorders and Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, China.,Beijing Municipal Key Laboratory for Translational Research on Diagnosis and Treatment of Dementia, Beijing, China
| | - Huali Wang
- Peking University Institute of Mental Health (Sixth Hospital), Beijing, China.,National Clinical Research Center for Mental Disorders and Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, China.,Beijing Municipal Key Laboratory for Translational Research on Diagnosis and Treatment of Dementia, Beijing, China
| | - Bin Dong
- Beijing International Center for Mathematical Research, Peking University, Beijing, China
| | - Quanzheng Li
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
7
|
Iron overload-induced oxidative stress in myelodysplastic syndromes and its cellular sequelae. Crit Rev Oncol Hematol 2021; 163:103367. [PMID: 34058341 DOI: 10.1016/j.critrevonc.2021.103367] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 03/30/2021] [Accepted: 05/03/2021] [Indexed: 12/14/2022] Open
Abstract
The myelodysplastic syndromes (MDS) are clonal hematopoietic stem cell disorders. MDS patients often require red blood cell transfusions, resulting in iron overload (IOL). IOL increases production of reactive oxygen species (ROS), oxygen free radicals. We review and illustrate how IOL-induced ROS influence cellular activities relevant to MDS pathophysiology. ROS damage lipids, nucleic acids in mitochondrial and nuclear DNA, structural proteins, transcription factors and enzymes. Cellular consequences include decreased metabolism and tissue and organ dysfunction. In hematopoietic stem cells (HSC), consequences of ROS include decreased glycolysis, shifting the cell from anaerobic to aerobic metabolism and causing HSC to exit the quiescent state, leading to HSC exhaustion or senescence. ROS oxidizes DNA bases, resulting in accumulation of mutations. Membrane oxidation alters fluidity and permeability. In summary, evidence indicates that IOL-induced ROS alters cellular signaling pathways resulting in toxicity to organs and hematopoietic cells, in keeping with adverse clinical outcomes in MDS.
Collapse
|
8
|
Bone Microthrombus Promotes Bone Loss in Iron Accumulation Rats. Curr Med Sci 2020; 40:943-950. [PMID: 32980898 DOI: 10.1007/s11596-020-2251-8] [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: 12/20/2019] [Accepted: 06/10/2020] [Indexed: 10/23/2022]
Abstract
In the present study, we investigated the changes of the coagulation state, bone microthrombus, microvascular bed and bone density levels in iron accumulation rats. Meanwhile,the effect of anticoagulation therapy on bone mineral density was further investigated. We established two groups: a control (Ctrl) group and an iron intervention (FAC) group. Changes in coagulation function, peripheral blood cell counts, bone microthrombus, bone vessels and bone mineral density were compared between the two groups. We designed the non-treatment group and treatment group to study the changes of bone mineral density by preventing microthrombus formation with the anticoagulant fondaparinux. We found that the fibrinogen and D-dimer contents were significantly higher, whereas the thrombin time (TT) and prothrombin time (PT) were significantly shorter in the FAC group. After ink staining, the microvascular bed in the FAC group was significantly reduced compared with that in the Ctrl group. HE and Martius Scarlet Blue (MSB) staining showed microthrombus in the bone marrow of the iron accumulation rats. Following anticoagulation therapy, the bone microcirculation vascular bed areas in the treatment group rats were significantly increased. Furthermore, the bone mineral density was increased in the treatment group compared with that in the non-treatment group. Through experiments, we found that the blood in iron accumulation rat was relatively hypercoagulable; moreover, there was microthrombus in the bone marrow, and the bone vascular bed was reduced. Additionally, anticoagulation was helpful for improving bone microcirculation, reducing microthrombus and decreasing bone loss.
Collapse
|
9
|
Demetz E, Tymoszuk P, Hilbe R, Volani C, Haschka D, Heim C, Auer K, Lener D, Zeiger LB, Pfeifhofer-Obermair C, Boehm A, Obermair GJ, Ablinger C, Coassin S, Lamina C, Kager J, Petzer V, Asshoff M, Schroll A, Nairz M, Dichtl S, Seifert M, von Raffay L, Fischer C, Barros-Pinkelnig M, Brigo N, Valente de Souza L, Sopper S, Hirsch J, Graber M, Gollmann-Tepeköylü C, Holfeld J, Halper J, Macheiner S, Gostner J, Vogel GF, Pechlaner R, Moser P, Imboden M, Marques-Vidal P, Probst-Hensch NM, Meiselbach H, Strauch K, Peters A, Paulweber B, Willeit J, Kiechl S, Kronenberg F, Theurl I, Tancevski I, Weiss G. The haemochromatosis gene Hfe and Kupffer cells control LDL cholesterol homeostasis and impact on atherosclerosis development. Eur Heart J 2020; 41:3949-3959. [PMID: 32227235 DOI: 10.1093/eurheartj/ehaa140] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 10/16/2019] [Accepted: 02/18/2020] [Indexed: 12/12/2022] Open
Abstract
AIMS Imbalances of iron metabolism have been linked to the development of atherosclerosis. However, subjects with hereditary haemochromatosis have a lower prevalence of cardiovascular disease. The aim of our study was to understand the underlying mechanisms by combining data from genome-wide association study analyses in humans, CRISPR/Cas9 genome editing, and loss-of-function studies in mice. METHODS AND RESULTS Our analysis of the Global Lipids Genetics Consortium (GLGC) dataset revealed that single nucleotide polymorphisms (SNPs) in the haemochromatosis gene HFE associate with reduced low-density lipoprotein cholesterol (LDL-C) in human plasma. The LDL-C lowering effect could be phenocopied in dyslipidaemic ApoE-/- mice lacking Hfe, which translated into reduced atherosclerosis burden. Mechanistically, we identified HFE as a negative regulator of LDL receptor expression in hepatocytes. Moreover, we uncovered liver-resident Kupffer cells (KCs) as central players in cholesterol homeostasis as they were found to acquire and transfer LDL-derived cholesterol to hepatocytes in an Abca1-dependent fashion, which is controlled by iron availability. CONCLUSION Our results disentangle novel regulatory interactions between iron metabolism, KC biology and cholesterol homeostasis which are promising targets for treating dyslipidaemia but also provide a mechanistic explanation for reduced cardiovascular morbidity in subjects with haemochromatosis.
Collapse
Affiliation(s)
- Egon Demetz
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Richard Hilbe
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Chiara Volani
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - David Haschka
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Christiane Heim
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Kristina Auer
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Daniela Lener
- Department of Internal Medicine III, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Lucas B Zeiger
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Christa Pfeifhofer-Obermair
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Anna Boehm
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Gerald J Obermair
- Department of Physiology and Medical Physics, Medical University of Innsbruck, Fritz-Pregl-Straße 3, 6020 Innsbruck, Austria
- Division of Physiology, Karl Landsteiner University of Health Sciences, Dr.-Karl-Dorrek-Straße 30, 3500 Krems, Austria
| | - Cornelia Ablinger
- Department of Physiology and Medical Physics, Medical University of Innsbruck, Fritz-Pregl-Straße 3, 6020 Innsbruck, Austria
| | - Stefan Coassin
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstraße 41, 6020 Innsbruck, Austria
| | - Claudia Lamina
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstraße 41, 6020 Innsbruck, Austria
| | - Juliane Kager
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Verena Petzer
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Malte Asshoff
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Andrea Schroll
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Manfred Nairz
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Stefanie Dichtl
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Laura von Raffay
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Christine Fischer
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Marina Barros-Pinkelnig
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Natascha Brigo
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Lara Valente de Souza
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Sieghart Sopper
- Department of Internal Medicine V, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Jakob Hirsch
- Department of Cardiac Surgery, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Michael Graber
- Department of Cardiac Surgery, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Can Gollmann-Tepeköylü
- Department of Cardiac Surgery, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Johannes Holfeld
- Department of Cardiac Surgery, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Julia Halper
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Sophie Macheiner
- Department of Internal Medicine I, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Johanna Gostner
- Division of Medical Biochemistry, Medical University of Innsbruck, Innrain 80/IV, 6020 Innsbruck, Austria
| | - Georg F Vogel
- Department of Pediatrics I, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Raimund Pechlaner
- Department of Neurology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Patrizia Moser
- Department of Pathology, Innsbruck University Hospital, Anichstraße 35, 6020 Innsbruck, Austria
| | - Medea Imboden
- Swiss Tropical and Public Health Institute, Socinstraße 57, 4051 Basel, Switzerland
- Department of Public Health, University of Basel, Bernoullistraße 28, 4056 Basel, Switzerland
| | - Pedro Marques-Vidal
- Department of Internal Medicine, Lausanne University Hospital, Rue du Bugnon 46, 1011 Lausanne, Switzerland
| | - Nicole M Probst-Hensch
- Swiss Tropical and Public Health Institute, Socinstraße 57, 4051 Basel, Switzerland
- Department of Public Health, University of Basel, Bernoullistraße 28, 4056 Basel, Switzerland
| | - Heike Meiselbach
- Department of Nephrology and Hypertension, University Hospital Erlangen, Maximiliansplatz 2, 91054 Erlangen, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-Universität, Marchioninistraße 15, 81377 Munich, Germany
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
- German Center for Diabetes Research, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
- German Center for Cardiovascular Research, Lazarettstraße 36, 80636 Munich, Germany
| | - Bernhard Paulweber
- First Department of Medicine, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria
| | - Johann Willeit
- Department of Neurology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Stefan Kiechl
- Department of Neurology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Florian Kronenberg
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstraße 41, 6020 Innsbruck, Austria
| | - Igor Theurl
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Ivan Tancevski
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Guenter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
10
|
Vinchi F, Porto G, Simmelbauer A, Altamura S, Passos ST, Garbowski M, Silva AMN, Spaich S, Seide SE, Sparla R, Hentze MW, Muckenthaler MU. Atherosclerosis is aggravated by iron overload and ameliorated by dietary and pharmacological iron restriction. Eur Heart J 2020; 41:2681-2695. [PMID: 30903157 DOI: 10.1093/eurheartj/ehz112] [Citation(s) in RCA: 176] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 10/10/2018] [Accepted: 03/07/2019] [Indexed: 12/20/2022] Open
Abstract
AIMS Whether and how iron affects the progression of atherosclerosis remains highly debated. Here, we investigate susceptibility to atherosclerosis in a mouse model (ApoE-/- FPNwt/C326S), which develops the disease in the context of elevated non-transferrin bound serum iron (NTBI). METHODS AND RESULTS Compared with normo-ferremic ApoE-/- mice, atherosclerosis is profoundly aggravated in iron-loaded ApoE-/- FPNwt/C326S mice, suggesting a pro-atherogenic role for iron. Iron heavily deposits in the arterial media layer, which correlates with plaque formation, vascular oxidative stress and dysfunction. Atherosclerosis is exacerbated by iron-triggered lipid profile alterations, vascular permeabilization, sustained endothelial activation, elevated pro-atherogenic inflammatory mediators, and reduced nitric oxide availability. NTBI causes iron overload, induces reactive oxygen species production and apoptosis in cultured vascular cells, and stimulates massive MCP-1-mediated monocyte recruitment, well-established mechanisms contributing to atherosclerosis. NTBI-mediated toxicity is prevented by transferrin- or chelator-mediated iron scavenging. Consistently, a low-iron diet and iron chelation therapy strongly improved the course of the disease in ApoE-/- FPNwt/C326S mice. Our results are corroborated by analyses of serum samples of haemochromatosis patients, which show an inverse correlation between the degree of iron depletion and hallmarks of endothelial dysfunction and inflammation. CONCLUSION Our data demonstrate that NTBI-triggered iron overload aggravates atherosclerosis and unravel a causal link between NTBI and the progression of atherosclerotic lesions. Our findings support clinical applications of iron restriction in iron-loaded individuals to counteract iron-aggravated vascular dysfunction and atherosclerosis.
Collapse
Affiliation(s)
- Francesca Vinchi
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Otto Meyerhof Zentrum, Im Neuenheimer Feld 350, 69120 Heidelberg, Germany.,Iron Homeostasis Group, Molecular Medicine Partnership Unit (MMPU), Heidelberg University, Im Neuenheimer Feld 350, 69120, Heidelberg & European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany.,New York Blood Center (NYBC), Iron Research Program, Lindsley F. Kimball Research Institute (LFKRI), 310 East 67th Street, 10065, New York, NY, USA.,Institute of Medical Biometry and Informatics (IMBI), University Hospital Heidelberg, Im Neuenheimer Feld 130.3, 69120 Heidelberg, Germany
| | - Graca Porto
- Centro Hospitalar do Porto-Hospital Santo António, Largo do Prof. Abel Slazar, 4099-001 Porto, Portugal.,Instituto de Biologia Molecular e Celular & Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen, 4200-135 Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Andreas Simmelbauer
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Otto Meyerhof Zentrum, Im Neuenheimer Feld 350, 69120 Heidelberg, Germany.,Iron Homeostasis Group, Molecular Medicine Partnership Unit (MMPU), Heidelberg University, Im Neuenheimer Feld 350, 69120, Heidelberg & European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Sandro Altamura
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Otto Meyerhof Zentrum, Im Neuenheimer Feld 350, 69120 Heidelberg, Germany.,Iron Homeostasis Group, Molecular Medicine Partnership Unit (MMPU), Heidelberg University, Im Neuenheimer Feld 350, 69120, Heidelberg & European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany.,Institute of Medical Biometry and Informatics (IMBI), University Hospital Heidelberg, Im Neuenheimer Feld 130.3, 69120 Heidelberg, Germany
| | - Sara T Passos
- New York Blood Center (NYBC), Iron Research Program, Lindsley F. Kimball Research Institute (LFKRI), 310 East 67th Street, 10065, New York, NY, USA
| | - Maciej Garbowski
- Hematology Department, University College London Cancer Institute, London, aul O'Gorman Bld, 72 Huntley Street, WC1E 6DD, London, UK
| | - André M N Silva
- Departamento de Quimica e Bioquimica, REQUIMITE-LAQV, Faculdade de Ciencias, University of Porto, Rua Do Campo Alegre, 4169-007 Porto, Portugal
| | - Sebastian Spaich
- Department of Cardiology, Angiology and Pneumonology, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Svenja E Seide
- Institute of Medical Biometry and Informatics (IMBI), University Hospital Heidelberg, Im Neuenheimer Feld 130.3, 69120 Heidelberg, Germany
| | - Richard Sparla
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Otto Meyerhof Zentrum, Im Neuenheimer Feld 350, 69120 Heidelberg, Germany
| | - Matthias W Hentze
- Iron Homeostasis Group, Molecular Medicine Partnership Unit (MMPU), Heidelberg University, Im Neuenheimer Feld 350, 69120, Heidelberg & European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany.,European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Martina U Muckenthaler
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Otto Meyerhof Zentrum, Im Neuenheimer Feld 350, 69120 Heidelberg, Germany.,Iron Homeostasis Group, Molecular Medicine Partnership Unit (MMPU), Heidelberg University, Im Neuenheimer Feld 350, 69120, Heidelberg & European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany.,European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| |
Collapse
|
11
|
Sumi MP, Guru SA, Sahu S, Khan B, Mp G, Saxena A. “Role of HFE gene in coronary artery disease” – A study from India. CLINICAL EPIDEMIOLOGY AND GLOBAL HEALTH 2020. [DOI: 10.1016/j.cegh.2019.10.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
12
|
Cornelissen A, Guo L, Sakamoto A, Virmani R, Finn AV. New insights into the role of iron in inflammation and atherosclerosis. EBioMedicine 2019; 47:598-606. [PMID: 31416722 PMCID: PMC6796517 DOI: 10.1016/j.ebiom.2019.08.014] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/24/2022] Open
Abstract
Iron is fundamental for life-essential processes. However, it can also cause oxidative damage, which is thought to trigger numerous pathologies, including cardiovascular diseases. The role of iron in the pathogenesis of atherosclerosis is still not completely understood. Macrophages are both key players in the handling of iron throughout the body and in the onset, progression and destabilization of atherosclerotic plaques. Iron itself might impact atherosclerosis through its effects on macrophages. However, while targeting iron metabolism within macrophages may have some beneficial effects on preventing atherosclerotic plaque progression there may also be negative consequences. Thus, the prevailing view of iron being capable of accelerating the progression of coronary disease through lipid peroxidation may not fully take into account the multi-faceted role of iron in pathogenesis of atherosclerosis. In this review, we will summarize the current understanding of iron metabolism in the context of the complex interplay between iron, inflammation, and atherosclerosis.
Collapse
Affiliation(s)
- Anne Cornelissen
- CVPath Institute, Gaithersburg, MD, USA; University Hospital RWTH Aachen, Department of Cardiology, Aachen, Germany.
| | - Liang Guo
- CVPath Institute, Gaithersburg, MD, USA.
| | | | | | - Aloke V Finn
- CVPath Institute, Gaithersburg, MD, USA; University of Maryland, School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
13
|
Abstract
Ferritin and increased iron stores first appeared on the list of cardiovascular risk factors more than 30 years ago and their causal role in the pathogenesis of atherosclerosis has been heavily discussed since the early 1990s. It seems that besides traditional factors such as hyperlipoproteinemia, hypertension, diabetes mellitus, obesity, physical inactivity, smoking and family history, high iron stores represent an additional parameter that could modify individual cardiovascular risk. The role of iron in the pathogenesis of atherosclerosis was originally primarily associated with its ability to catalyze the formation of highly reactive free oxygen radicals and the oxidation of atherogenic lipoproteins. Later, it became clear that the mechanism is more complex. Atherosclerosis is a chronic fibroproliferative inflammatory process and iron, through increased oxidation stress as well as directly, can control both native and adaptive immune responses. Within the arterial wall, iron affects all of the cell types that participate in the atherosclerotic process (monocytes/macrophages, endothelial cells, vascular smooth muscle cells and platelets). Most intracellular iron is bound in ferritin, whereas redox-active iron forms labile iron pool. Pro-inflammatory and anti-inflammatory macrophages within arterial plaque differ with regard to the amount of intracellular iron and most probably with regard to their labile iron pool. Yet, the relation between plasma ferritin and intracellular labile iron pool has not been fully clarified. Data from population studies document that the consumption of meat and lack of physical activity contribute to increased iron stores. Patients with hereditary hemochromatosis, despite extreme iron storage, do not show increased manifestation of atherosclerosis probably due to the low expression of hepcidin in macrophages.
Collapse
Affiliation(s)
- P Kraml
- Second Department of Internal Medicine, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic.
| |
Collapse
|
14
|
Zhong Y, Lin SL, Schooling CM. The effect of hematocrit and hemoglobin on the risk of ischemic heart disease: A Mendelian randomization study. Prev Med 2016; 91:351-355. [PMID: 27609746 DOI: 10.1016/j.ypmed.2016.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 08/31/2016] [Accepted: 09/04/2016] [Indexed: 11/24/2022]
Abstract
Hematocrit and hemoglobin affect viscosity, and have been considered as risk factors for ischemic heart disease (IHD), although observations are inconsistent; randomized controlled trials targeting hematocrit or hemoglobin have not been definitive. To clarify their role, the risk of IHD was assessed according to genetically determined hematocrit and hemoglobin. We applied single nucleotide polymorphisms (SNPs) strongly determining hematocrit and hemoglobin, from a genome wide association study, to a large case (64,746) control (130,681) study of coronary artery disease, CARDIoGRAMplusC4D, to obtain unconfounded estimates using instrumental variable analysis by combining the Wald estimators for each SNP taking into account any correlation between SNPs using weighted generalized linear regression. Hematocrit was positively associated with IHD, odds ratio (OR) 1.07 per %, 95% confidence interval (CI) 1.03 to 1.11, before and after excluding SNPs from gene regions directly functionally relevant to IHD. However, hematocrit was not associated with IHD (OR 0.99, 0.94 to 1.04) after also excluding SNPs associated with lipids at genome wide significance. Hemoglobin was not associated with IHD (OR 1.06 per g/dL, 0.97 to 1.15) which was similar (OR 1.02, 0.94 to 1.11) after excluding SNPs from gene regions directly functionally relevant to IHD. Hemoglobin was negatively associated with IHD after also excluding SNPs associated with lipids at genome wide significance (OR 0.86, 0.78 to 0.94). In conclusion, hematocrit shares genetic determinants with IHD, but whether the genes contribute to IHD via hematocrit or other mechanisms is not entirely clear. Higher Hemoglobin is unlikely to cause IHD.
Collapse
Affiliation(s)
- Y Zhong
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - S L Lin
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - C M Schooling
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; CUNY School of Public Health and Health Policy, New York, USA.
| |
Collapse
|
15
|
Yunker LM, Parboosingh JS, Conradson HE, Faris P, Bridge PJ, Buithieu J, Title LM, Charbonneau F, Verma S, Lonn EM, Anderson TJ. The effect of iron status on vascular health. Vasc Med 2016; 11:85-91. [PMID: 16886838 DOI: 10.1191/1358863x06vm656oa] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The effect of increased iron stores on the progression of atherosclerosis and endothelial health remains inconclusive. This study was designed to evaluate the relationship between hemochromatosis genotypes, serum ferritin levels and presymptomatic vascular abnormalities in a cohort of healthy subjects. Carotid intima-media thickness (CIMT) and brachial flow-mediated vasodilation (FMD) were assessed by high-resolution ultrasound in 907 male (47 ± 10 years) participants enrolled in the Firefighters and their Endothelium (FATE) study. Analyses of the hemochromatosis C282Y, H63D and S65C alleles were simultaneously determined by a single nucleotide polymorphism (SNP) primer extension method. It was found that brachial FMD was not related to serum ferritin or hemochromatosis genotype status. The presence of a hemochromatosis-associated genotype ( n = 18) or heterozygosity for the C282Y genotype ( n = 98) was not associated with an increased mean CIMT. After adjustment for conventional risk factors, serum ferritin was also not associated with mean CIMT. In conclusion, neither ferritin nor a hemochromatosis genotype was related to brachial endothelial function or carotid atherosclerosis. The present study does not support the hypothesis that mild to moderately increased iron stores are associated with enhanced atherosclerosis risk.
Collapse
Affiliation(s)
- Leanne M Yunker
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Heath KM, Axton JH, McCullough JM, Harris N. The evolutionary adaptation of the C282Y mutation to culture and climate during the European Neolithic. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2016; 160:86-101. [PMID: 26799452 PMCID: PMC5066702 DOI: 10.1002/ajpa.22937] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 12/20/2015] [Accepted: 12/20/2015] [Indexed: 01/10/2023]
Abstract
OBJECTIVES The C282Y allele is the major cause of hemochromatosis as a result of excessive iron absorption. The mutation arose in continental Europe no earlier than 6,000 years ago, coinciding with the arrival of the Neolithic agricultural revolution. Here we hypothesize that this new Neolithic diet, which originated in the sunny warm and dry climates of the Middle East, was carried by migrating farmers into the chilly and damp environments of Europe where iron is a critical micronutrient for effective thermoregulation. We argue that the C282Y allele was an adaptation to this novel environment. MATERIALS AND METHODS To address our hypothesis, we compiled C282Y allele frequencies, known Neolithic sites in Europe and climatic data on temperature and rainfall for statistical analysis. RESULTS Our findings indicate that the geographic cline for C282Y frequency in Europe increases as average temperatures decrease below 16°C, a critical threshold for thermoregulation, with rainy days intensifying the trend. DISCUSSION The results indicate that the deleterious C282Y allele, responsible for most cases of hemochromatosis, may have evolved as a selective advantage to culture and climate during the European Neolithic.
Collapse
Affiliation(s)
- Kathleen M. Heath
- Department of Earth and Environmental SystemsIndiana State UniversityTerre HauteIN47809
| | - Jacob H. Axton
- Department of BiologyIndiana State UniversityTerre HauteIN47809
| | | | - Nathan Harris
- Department of AnthropologyUniversity of UtahSalt Lake CityUT84112
| |
Collapse
|
17
|
Saremi L, Saremi M, Lotfipanah S, İmani S, Fu J, Zhang T. Correlation between HFE gene polymorphisms and increased risk of coronary artery disease among patients with type 2 diabetes in Iran. Turk J Med Sci 2016; 46:590-6. [PMID: 27513231 DOI: 10.3906/sag-1409-88] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 02/26/2015] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND/AIM Diabetes mellitus is a risk factor for cardiovascular diseases (CVDs), which are among the major causes of deaths in type 2 diabetes (T2D). The purpose of the present study was to determine the association of C282Y and H63D mutations in the HFE gene with increased risk of coronary artery disease (CAD) in T2D patients. MATERIALS AND METHODS Two hundred and ninety individuals were divided into two groups: a case group and a control group. Genomic DNA of peripheral venous blood cells was extracted and the HFE gene mutations were analyzed using the PCR-RFLP technique. RESULTS Data analysis revealed a significant difference between the allele frequencies of H63D and C282Y mutations between the case group and the controls (P < 0.05). The relationships between the GA and GG genotypes in C282Y and H63D mutations in terms of fasting blood sugar (FBS), lipid profile (total cholesterol, triglycerides, high-density lipoproteins (HDL), low-density lipoproteins), body mass index (BMI), HbA1c, micro albuminuria, and creatine levels did not show a significant differences between the two groups (P > 0.05). Using a logistic regression model, BMI, FBS, HDL, and total cholesterol levels were significantly different with independent predictors of CVD (P < 0.05). CONCLUSION Our results revealed a significant correlation between C282Y and H63D mutations and the development of CAD in T2D patients.
Collapse
Affiliation(s)
- Leila Saremi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Marzieh Saremi
- Department of Microbiology, Qom branch, Islamic Azad University, Qom, Iran
| | - Shirin Lotfipanah
- Shahid Mofatteh Teacher Education College, Farhangian University, Qatran, Iran
| | - Saber İmani
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine,Luzhou Medical College, Luzhou, Sichuan, P.R. China
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine,Luzhou Medical College, Luzhou, Sichuan, P.R. China
| | - Tianyu Zhang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health,Chinese Academy of Sciences, Guangzhou, P.R. China
| |
Collapse
|
18
|
Bardou-Jacquet E, Morcet J, Manet G, Lainé F, Perrin M, Jouanolle AM, Guyader D, Moirand R, Viel JF, Deugnier Y. Decreased cardiovascular and extrahepatic cancer-related mortality in treated patients with mild HFE hemochromatosis. J Hepatol 2015; 62:682-9. [PMID: 25450707 DOI: 10.1016/j.jhep.2014.10.025] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 10/06/2014] [Accepted: 10/08/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Mortality studies in patients with hemochromatosis give conflicting results especially with respect to extrahepatic causes of death. Our objective was to assess mortality and causes of death in a cohort of patients homozygous for the C282Y mutation in the HFE gene, diagnosed since the availability of HFE testing. METHODS We studied 1085 C282Y homozygotes, consecutively diagnosed from 1996 to 2009, and treated according to current recommendations. Mortality and causes of death were obtained from death certificates and compared to those of the general population. Standardized mortality ratios (SMRs) were used to assess specific causes of death and the Cox model was used to identify prognostic factors for death. RESULTS Patients were followed for 8.3±3.9 years. Overall the SMR was the same as in the general population (0.94 CI: 0.71-1.22). Patients with serum ferritin⩾2000 μg/L had increased liver-related deaths (SMR: 23.9 CI: 13.9-38.2), especially due to hepatic cancer (SMR: 49.1 CI: 24.5-87.9). Patients with serum ferritin between normal and 1000 μg/L had a lower mortality than the general population (SMR: 0.27 CI: 0.1-0.5), due to a decreased mortality, related to reduced cardiovascular events and extrahepatic cancers in the absence of increased liver-related mortality. Age, diabetes, alcohol consumption, and hepatic fibrosis were independent prognostic factors of death. CONCLUSIONS In treated HFE hemochromatosis, only patients with serum ferritin higher than 2000 μg/L have an increased mortality, mainly related to liver diseases. Those with mild iron burden have a decreased overall mortality in relation to reduced cardiovascular and extrahepatic cancer-related events. These results support a beneficial effect of early and sustained management of patients with iron excess, even when mild.
Collapse
Affiliation(s)
- Edouard Bardou-Jacquet
- CHU Rennes, Service des maladies du foie, Rennes, France; INSERM, U991, Hôpital Pontchaillou, Rennes, France.
| | - Jeff Morcet
- INSERM, CIC 1414, Hôpital Pontchaillou, Rennes, France
| | | | - Fabrice Lainé
- CHU Rennes, Service des maladies du foie, Rennes, France; INSERM, CIC 1414, Hôpital Pontchaillou, Rennes, France
| | | | | | - Dominique Guyader
- CHU Rennes, Service des maladies du foie, Rennes, France; INSERM, U991, Hôpital Pontchaillou, Rennes, France; University of Rennes 1, UFR médecine, Rennes, France
| | - Romain Moirand
- CHU Rennes, Service des maladies du foie, Rennes, France; INSERM, U991, Hôpital Pontchaillou, Rennes, France; University of Rennes 1, UFR médecine, Rennes, France
| | - Jean-François Viel
- University of Rennes 1, UFR médecine, Rennes, France; CHU Rennes, Service d'épidémiologie et de santé publique, Hôpital Pontchaillou, Rennes, France
| | - Yves Deugnier
- CHU Rennes, Service des maladies du foie, Rennes, France; INSERM, U991, Hôpital Pontchaillou, Rennes, France; INSERM, CIC 1414, Hôpital Pontchaillou, Rennes, France; University of Rennes 1, UFR médecine, Rennes, France
| |
Collapse
|
19
|
Molekulargenetische Analyse und klinische Aspekte bei Patienten mit hereditärer Hämochromatose. DER ORTHOPADE 2014; 43:772-9. [DOI: 10.1007/s00132-014-2318-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
20
|
Vinchi F, Muckenthaler MU, Da Silva MC, Balla G, Balla J, Jeney V. Atherogenesis and iron: from epidemiology to cellular level. Front Pharmacol 2014; 5:94. [PMID: 24847266 PMCID: PMC4017151 DOI: 10.3389/fphar.2014.00094] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/14/2014] [Indexed: 12/12/2022] Open
Abstract
Iron accumulates in human atherosclerotic lesions but whether it is a cause or simply a downstream consequence of the atheroma formation has been an open question for decades. According to the so called "iron hypothesis," iron is believed to be detrimental for the cardiovascular system, thus promoting atherosclerosis development and progression. Iron, in its catalytically active form, can participate in the generation of reactive oxygen species and induce lipid-peroxidation, triggering endothelial activation, smooth muscle cell proliferation and macrophage activation; all of these processes are considered to be proatherogenic. On the other hand, the observation that hemochromatotic patients, affected by life-long iron overload, do not show any increased incidence of atherosclerosis is perceived as the most convincing evidence against the "iron hypothesis." Epidemiological studies and data from animal models provided conflicting evidences about the role of iron in atherogenesis. Therefore, more careful studies are needed in which issues like the source and the compartmentalization of iron will be addressed. This review article summarizes what we have learnt about iron and atherosclerosis from epidemiological studies, animal models and cellular systems and highlights the rather contributory than innocent role of iron in atherogenesis.
Collapse
Affiliation(s)
- Francesca Vinchi
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg Heidelberg, Germany ; Molecular Medicine and Partnership Unit, University of Heidelberg Heidelberg, Germany
| | - Martina U Muckenthaler
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg Heidelberg, Germany ; Molecular Medicine and Partnership Unit, University of Heidelberg Heidelberg, Germany
| | - Milene C Da Silva
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg Heidelberg, Germany ; Molecular Medicine and Partnership Unit, University of Heidelberg Heidelberg, Germany
| | - György Balla
- MTA-DE Vascular Biology, Thrombosis and Hemostasis Research Group, Hungarian Academy of Sciences Debrecen, Hungary ; Department of Pediatrics, University of Debrecen Debrecen, Hungary
| | - József Balla
- Department of Medicine, University of Debrecen Debrecen, Hungary
| | - Viktória Jeney
- MTA-DE Vascular Biology, Thrombosis and Hemostasis Research Group, Hungarian Academy of Sciences Debrecen, Hungary ; Department of Medicine, University of Debrecen Debrecen, Hungary
| |
Collapse
|
21
|
Cash WJ, O'Neill S, O'Donnell ME, McCance DR, Young IS, McEneny J, McDougall NI, Callender ME. Endothelial function, antioxidant status and vascular compliance in newly diagnosed HFE C282Y homozygotes. Adv Med Sci 2014; 59:28-33. [PMID: 24797970 DOI: 10.1016/j.advms.2013.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 07/01/2013] [Indexed: 01/19/2023]
Abstract
PURPOSE This pilot study was aimed to establish techniques for assessing and observing trends in endothelial function, antioxidant status and vascular compliance in newly diagnosed HFE haemochromatosis during the first year of venesection. PATIENTS/METHODS Untreated newly diagnosed HFE haemochromatosis patients were tested for baseline liver function, iron indices, lipid profile, markers of endothelial function, anti-oxidant status and vascular compliance. Following baseline assessment, subjects attended at 6-weeks and at 3, 6, 9 and 12-months for follow-up studies. RESULTS Ten patients were recruited (M=8, F=2, mean age=51 years). Venesection significantly increased high density lipoproteins at 12-months (1.25 mmol/L vs. 1.37 mmol/L, p=0.01). However, venesection did not significantly affect lipid hydroperoxides, intracellular and vascular cell adhesion molecules or high sensitivity C-reactive protein (0.57 μmol/L vs. 0.51 μmol/L, p=0.45, 427.4 ng/ml vs. 307.22 ng/ml, p=0.54, 517.70 ng/ml vs. 377.50 ng/ml, p=0.51 and 290.75 μg/dL vs. 224.26 μg/dL, p=0.25). There was also no significant effect of venesection on anti-oxidant status or pulse wave velocity (9.65 m/s vs. 8.74 m/s, p=0.34). CONCLUSIONS Venesection significantly reduced high density lipoproteins but was not associated with significant changes in endothelial function, anti-oxidant status or vascular compliance. Larger studies using this established methodology are required to clarify this relationship further.
Collapse
Affiliation(s)
- William J Cash
- Department of Hepatology (Liver Unit), Royal Victoria Hospital, Belfast, UK.
| | - Stephen O'Neill
- Department of Vascular and Endovascular Surgery, Royal Victoria Hospital, Belfast, UK
| | - Mark E O'Donnell
- Department of Vascular and Endovascular Surgery, Royal Victoria Hospital, Belfast, UK
| | - David R McCance
- Department of Endocrinology and Diabetes, Royal Victoria Hospital, Belfast, UK
| | - Ian S Young
- Department of Clinical Biochemistry, Royal Victoria Hospital, Belfast, UK; Department of Medicine, Queen's University Belfast, Belfast, UK
| | - Jane McEneny
- Department of Medicine, Queen's University Belfast, Belfast, UK
| | - Neil I McDougall
- Department of Hepatology (Liver Unit), Royal Victoria Hospital, Belfast, UK
| | | |
Collapse
|
22
|
Cash WJ, O'Neill S, O'Donnell ME, McCance DR, Young IS, McEneny J, Cadden IS, McDougall NI, Callender ME. Disordered vascular compliance in haemochromatosis. Ir J Med Sci 2013; 183:303-9. [PMID: 24000100 DOI: 10.1007/s11845-013-1010-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 08/21/2013] [Indexed: 01/21/2023]
Abstract
BACKGROUND A relationship may exist between body iron stores, endothelial dysfunction and overall cardiovascular risk. AIMS To compare vascular compliance, biochemical endothelial function and antioxidant status between patients with homozygous hereditary haemochromatosis and healthy controls. METHODS Haemochromatosis patients and healthy controls were recruited. Measures of vascular compliance were assessed by applanation tonometry. Serological markers of endothelial function (plasma lipid hydroperoxides, cell adhesion molecules), antioxidant levels (ascorbate, lipid soluble antioxidants) and high-sensitivity C-reactive protein (CRP) were also measured. RESULTS Thirty-five hereditary haemochromatosis patients (ten females, mean age 54.6) and 36 controls (27 female, mean age 54.0) were recruited. Haemochromatosis patients had significantly higher systolic and diastolic blood pressures. Pulse wave velocity (PWV) was significantly higher in male haemochromatosis patients (9.90 vs. 8.65 m/s, p = 0.048). Following adjustment for age and blood pressure, male haemochromatosis patients continued to have a trend for higher PWVs (+1.37 m/s, p = 0.058). Haemochromatosis patients had significantly lower levels of ascorbate (46.11 vs. 72.68 μmol/L, p = 0.011), retinol (1.17 vs. 1.81 μmol/L, p = 0.001) and g-tocopherol (2.51 vs. 3.14 μmol/L, p = 0.011). However, there was no difference in lipid hydroperoxides (0.46 vs. 0.47 nmol/L, p = 0.94), cell adhesion molecule levels (ICAM: 348.12 vs. 308.03 ng/mL, p = 0.32 and VCAM: 472.78 vs. 461.31 ng/mL, p = 0.79) or high-sensitivity CRP (225.01 vs. 207.13 mg/L, p = 0.32). CONCLUSIONS Haemochromatosis is associated with higher PWVs in males and diminished antioxidants across the sexes but no evidence of endothelial dysfunction or increased lipid peroxidation.
Collapse
Affiliation(s)
- W J Cash
- Department of Hepatology (Liver Unit), Queens University Belfast, Mullhouse Building, Grosvenor Road, Belfast, BT12 6AB, Northern Ireland, UK,
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Hagemeier J, Geurts JJG, Zivadinov R. Brain iron accumulation in aging and neurodegenerative disorders. Expert Rev Neurother 2013; 12:1467-80. [PMID: 23237353 DOI: 10.1586/ern.12.128] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Over the decades, various studies have established an association between accumulation of iron and both aging and neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease. Excess levels of iron can lead to increased oxidative stress through Fenton chemistry, and depletion of iron can similarly have deleterious effects. In addition, metal ions are known to be involved in both Alzheimer's disease and Parkinson's disease protein aggregation. Metal ion chelators have been extensively investigated in preclinical models, and may prove to be appropriate for modulating brain iron levels in age-related neurodegenerative disorders. Investigating age-related iron deposition is vital, and can possibly aid in determining at-risk groups and diagnosing neurodegenerative diseases at an early stage. Novel imaging methods have enabled researchers to examine iron deposition in vivo, and offer a noninvasive method of monitoring the progression of accumulation, and possible therapeutic effects of chelating compounds.
Collapse
Affiliation(s)
- Jesper Hagemeier
- Buffalo Neuroimaging Analysis Center, Department of Neurology, University at Buffalo, 100 High Street, Buffalo, NY 14203, USA
| | | | | |
Collapse
|
24
|
Alwan NA, Lawlor DA, McArdle HJ, Greenwood DC, Cade JE. Exploring the relationship between maternal iron status and offspring's blood pressure and adiposity: a Mendelian randomization study. Clin Epidemiol 2012; 4:193-200. [PMID: 22942651 PMCID: PMC3422112 DOI: 10.2147/clep.s33833] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Iron deficiency is the most common micronutrient deficiency worldwide. Experimental animal studies suggest that mothers deficient in iron during pregnancy are more likely to have offspring who become obese with high blood pressure. C282Y mutation carriers are more likely to have higher iron stores. Methods We undertook an instrumental variable (IV) analysis, using maternal C282Y as an indicator for the mother’s iron status, to examine its association with offspring blood pressure (BP), waist circumference (WC), and body mass index (BMI), and compared the results to that of ordinary least squares (OLS) regression. Offspring of a sub-cohort of mothers from the UK Women’s Cohort Study (UKWCS) were recruited in 2009–2010 (n = 348, mean age = 41 years). Their blood pressure, height, and weight were measured at their local general medical practice, and they were asked to self-measure their waist circumference. About half were offspring of C282Y carriers. Maternal ferritin was used as a biomarker of maternal iron status. Results Maternal C282Y was strongly associated with maternal ferritin (mean difference per allele = 84 g/L, 95% confidence interval: 31–137, P = 0.002). Using IV analyses, maternal ferritin was not linked to offspring’s BP, BMI, or WC. The first stage F-statistic for the strength of the instrument was 10 (Kleibergen–Paap rk LM P = 0.009). Maternal ferritin was linked to offspring diastolic BP, WC, and BMI in univariable, but not in multivariable OLS analysis. There was no difference between the OLS and the IV models coefficients for any of the outcomes considered. Conclusion We found no association between maternal iron status and adult offspring’s BP and adiposity using both multivariable OLS and IV modeling. To our knowledge, this is the first study examining this relationship. Further exploration in larger studies that have genetic variation assessed in both mother and offspring should be considered.
Collapse
Affiliation(s)
- Nisreen A Alwan
- Nutritional Epidemiology Group, School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | | | | | | | | |
Collapse
|
25
|
Elmberg M, Hultcrantz R, Simard JF, Stål P, Pehrsson K, Askling J. Risk of ischaemic heart disease and cardiomyopathy in patients with haemochromatosis and in their first-degree relatives: a nationwide, population-based study. J Intern Med 2012; 272:45-54. [PMID: 22026548 DOI: 10.1111/j.1365-2796.2011.02475.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Iron-loaded macrophages increase atherosclerosis formation. Genetic haemochromatosis (GH) is an autosomal recessive disease characterized by iron overload, for example in the myocardium, but the reticuloendothelial system is depleted of iron. In contrast to the elevated risk of cardiomyopathy in GH, the risk of ischaemic heart disease (IHD) may therefore not be increased. Little is known of these risks among heterozygotes also being first-degree relatives (FDRs), thus sharing other factors for phenotypic expression of GH. OBJECTIVE To assess the risks of IHD and cardiomyopathy among haemochromatosis patients and their FDRs. DESIGN Population-based cohort study. SETTING AND SUBJECTS A total of 3531 haemochromatosis patients and 11 794 FDRs were identified using nationwide, population-based health and census registers. Matched (1:10) population controls were randomly selected. Individuals with a record of IHD and cardiomyopathy during 1997-2005 were identified through linkage with the National Patient Register. Relative risks were estimated using Cox proportional hazard regression. RESULTS Of the 3531 patients, 259 were diagnosed with IHD compared with 3077 of the 37 369 controls [hazard ratio (HR) = 1.17; 95% CI, 1.03-1.33]. Based on 30 patients versus 115 controls, the HR for cardiomyopathy was 3.21 (95% CI, 2.15-4.81). Of 11 794 FDRs of haemochromatosis patients, 582 were registered with IHD compared with 6197 among FDRs of controls (HR = 1.05; 95% CI, 0.97-1.15). Based on 28 FDRs of patients versus 291 FDRs of controls registered with cardiomyopathy, the HR for cardiomyopathy was 1.06 (95% CI, 0.72-1.56). CONCLUSIONS In patients with haemochromatosis, the increased risk of cardiomyopathy is much more pronounced than that of IHD, which is barely elevated. FDRs of haemochromatosis patients are not at increased risk of cardiomyopathy or IHD.
Collapse
Affiliation(s)
- M Elmberg
- Division of Gastroenterology and Hepatology, Karolinska University Hospital Clinical, Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
26
|
Micronutrient (Zn, Cu, Fe)-gene interactions in ageing and inflammatory age-related diseases: implications for treatments. Ageing Res Rev 2012; 11:297-319. [PMID: 22322094 DOI: 10.1016/j.arr.2012.01.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 01/20/2012] [Accepted: 01/23/2012] [Indexed: 02/07/2023]
Abstract
In ageing, alterations in inflammatory/immune response and antioxidant capacity lead to increased susceptibility to diseases and loss of mobility and agility. Various essential micronutrients in the diet are involved in age-altered biological functions. Micronutrients (zinc, copper, iron) play a pivotal role either in maintaining and reinforcing the immune and antioxidant performances or in affecting the complex network of genes (nutrigenomic approach) involved in encoding proteins for a correct inflammatory/immune response. By the other side, the genetic inter-individual variability may affect the absorption and uptake of the micronutrients (nutrigenetic approach) with subsequent altered effects on inflammatory/immune response and antioxidant activity. Therefore, the individual micronutrient-gene interactions are fundamental to achieve healthy ageing. In this review, we report and discuss the role of micronutrients (Zn, Cu, Fe)-gene interactions in relation to the inflammatory status and the possibility of a supplement in the event of a micronutrient deficiency or chelation in presence of micronutrient overload in relation to specific polymorphisms of inflammatory proteins or proteins related of the delivery of the micronutriemts to various organs and tissues. In this last context, we report the protein-metal speciation analysis in order to have, coupled with micronutrient-gene interactions, a more complete picture of the individual need in micronutrient supplementation or chelation to achieve healthy ageing and longevity.
Collapse
|
27
|
Hayes VW, DePalma RG, Zacharski LR. Menstrual Suppression, Iron Homeostasis, and Disease Risk. J Nurse Pract 2011. [DOI: 10.1016/j.nurpra.2011.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
28
|
Adler G, Clark JS, Łoniewska B, Ciechanowicz A. Prevalence of 845G>A HFE mutation in Slavic populations: an east-west linear gradient in South Slavs. Croat Med J 2011; 52:351-7. [PMID: 21674831 PMCID: PMC3118720 DOI: 10.3325/cmj.2011.52.351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
AIM To compare A allele frequencies of the 845G>A mutation of 10 Slavic populations in central, eastern, and southern Europe between each other and with other European populations. METHODS The 845G>A mutation from the DNA of 400 Polish neonates collected in 2005-2006 was analyzed by polymerase chain reaction-restriction fragment length polymorphism. The data were compared with reports from other countries. RESULTS We identified 381 GG homozygotes, 18 GA heterozygotes, and 1 AA homozygote. The 845A allele frequency was 2.5%, which makes the summary figure for Poland from this and previous studies 3.5%. The average prevalence for Poland and other West Slavic countries was 3.6%, similar to Russia (inhabited by the East Slavs, 3.5%). The average prevalence in South Slavic countries was 2.2%, gradually decreasing from 3.6% in Slovenia to 0% in Bulgaria, with a longitudinal linear gradient (adjusted R(2)=0.976, P<0.001). CONCLUSIONS The West and East Slavs, together with Finland, Estonia, Germany, Austria, Hungary, Slovenia, and Croatia, form a group with 845A allele frequencies between 3% and 4%. In the South Slavs, there is a gradual decline in the prevalence of 845A allele from northwest to southeast, with a surprisingly exact east-west linear gradient.
Collapse
Affiliation(s)
- Grazyna Adler
- Pomeranian Medical University, Department of Medical Biology, Szczecin, Poland.
| | | | | | | |
Collapse
|
29
|
Shi Y, Zhou L, Huang LH, Lian YT, Zhang XM, Guo H, Wu TC, Cheng LX, He MA. Plasma ferritin levels, genetic variations in HFE gene, and coronary heart disease in Chinese: a case-control study. Atherosclerosis 2011; 218:386-90. [PMID: 21696736 DOI: 10.1016/j.atherosclerosis.2011.05.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2011] [Revised: 05/03/2011] [Accepted: 05/24/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND The association between body iron stores and coronary heart disease (CHD) was inconsistent. We sought to explore this association in Chinese Han population and further examine the association of the variations in hemochromatosis (HFE) gene and CHD risk. METHODS We conducted a case-control study including 1334 CHD patients and 1334 age- and sex-frequency matched controls. The plasma ferritin levels were measured by enzyme linked immunosorbent assay. Genotypes of the tagging single nucleotide polymorphisms (tagSNPs) were determined by TaqMan SNP allelic discrimination. RESULTS The plasma ferritin levels in CHD cases (197.9μg/L [2.7-932.9μg/L]) were higher than those in controls (179.9μg/L [21.1-878.2μg/L]; P=0.028). The odds ratios (ORs) across the tertiles of plasma ferritin levels were 1.0 (reference), 0.93 (0.76-1.13), and 1.23 (1.02-1.48; P for trend=0.028). Adjustment for the traditional risk factors attenuated the associations to null (P for trend=0.22). Compared with the TT genotype of tagSNP rs9366637, subjects with C allele had higher risk of CHD (OR=1.35 for TC and 1.76 for CC; P=0.001 and <0.001 respectively). After adjustment for the conventional risk factors the results remained unchanged. We did not find significantly different plasma ferritin levels among different genotypes of rs9366637 (P=0.52). CONCLUSIONS The plasma ferritin levels were not significantly associated with CHD risk. However, the SNP rs9366637 in HFE gene was associated with higher CHD risk in Chinese Han population. The underlie mechanism remained to be elucidated in further studies.
Collapse
Affiliation(s)
- Y Shi
- Department of Cardiology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Ruiz-Jaramillo MDLC, Guizar-Mendoza JM, Amador-Licona N, Gutierrez-Navarro MDJ, Hernandez-Gonzalez MA, Dubey-Ortega LA, Solorio-Meza SE. Iron overload as cardiovascular risk factor in children and adolescents with renal disease. Nephrol Dial Transplant 2011; 26:3268-73. [DOI: 10.1093/ndt/gfr044] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|
31
|
Colli ML, Gross JL, Canani LH. Mutation H63D in the HFE gene confers risk for the development of type 2 diabetes mellitus but not for chronic complications. J Diabetes Complications 2011; 25:25-30. [PMID: 20097100 DOI: 10.1016/j.jdiacomp.2009.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Revised: 12/01/2009] [Accepted: 12/10/2009] [Indexed: 12/27/2022]
Abstract
PURPOSE To evaluate the frequency of mutations in the HFE gene (C282Y and H63D) in type 2 diabetes mellitus (DM) patients and their possible association with diabetic chronic complications. METHODS A case-control study with 723 subjects was performed. All diabetic subjects (n=519) underwent a clinical and laboratory evaluation. Diabetic retinopathy (DR) was evaluated by an ophthalmologist. Diabetic nephropathy (DN) was categorized by urinary albumin excretion (UAE) as normoalbuminuria (n=247), microalbuminuria (n=68), macroalbuminuria (n=70), or the presence of end-stage renal disease (dialysis; n=134). Data available for blood donors (n=204) were limited to age, sex, body mass index, and absence of previous diagnosis of diabetes and normal fasting plasma glucose. The mutations C282Y and H63D in the HFE gene were genotyped based on PCR protocols and digested with the restriction enzymes SnabI (C282Y) and MboI (H63D). RESULTS There was an association of type 2 DM with H63D polymorphism (genotypes HD/DD: OR=1.7, 95% CI=1.2-2.6), but not with C282Y polymorphism (OR=0.7, 955 CI=0.4-1.4). In respect to the chronic complications, there was no difference in the prevalence of DR, DN, or ischemic heart disease among the different genotypes. CONCLUSIONS Mutation H63D in the HFE gene was associated with a higher risk of type 2 DM, but did not appear to confer risk for diabetic chronic complications. The mutation C282Y was not associated with diabetes or its chronic complications.
Collapse
Affiliation(s)
- Máikel L Colli
- Endocrine Division, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.
| | | | | |
Collapse
|
32
|
Olsson KS, Konar J, Dufva IH, Ricksten A, Raha-Chowdhury R. Was the C282Y mutation an Irish Gaelic mutation that the Vikings helped disseminate? HLA haplotype observations of hemochromatosis from the west coast of Sweden. Eur J Haematol 2010; 86:75-82. [PMID: 20946107 DOI: 10.1111/j.1600-0609.2010.01536.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
UNLABELLED The HLA-related hemochromatosis mutation C282Y is thought to have originated in Ireland in a person with HLA-A3-B14 and was spread by Vikings. Irish people with two HLA-A3 alleles had a high risk of hemochromatosis. In this study, from west Sweden, we wanted to test these hypotheses. METHODS HFE mutations in controls, bone marrow donors with HLA-A3/A3 and patients with hemochromatosis. HLA haplotypes, extended haplotype analysis and pedigree studies. RESULTS The allelic C282Y frequency 0.04, (CI 0.01-0.07) was lower (P < 0.001) in Sweden than in Ireland 0.10 (CI 0.08-0.11), and Swedish bone marrow donors with HLA-A3/A3 (n = 77) had a low risk of hemochromatosis. HLA haplotypes available from 239/262 (91.5%) proband patients homozygous for C282Y showed a dominance of A3-B7 and A3-B14 both in linkage disequilibrium with controls (P < 0.001). Pedigree studies extended into the 17th century supported a local founder effect of A3-B14 in the county of Bohuslän. The A3-B14 haplotype may well be the original and A3-B7 the result of centromeric recombinations. The haplotype diversity and recombination events were not different from a Celtic series. These findings do not support the hypothesis of the C282Y mutation being of an Irish Celtic origin. CONCLUSIONS The C282Y frequency shows a west to east decline from Ireland through the north of Europe. Vikings may have been involved in the spread of C282Y, but the mutation is probably older and may have been spread in Europe by earlier seafarers.
Collapse
Affiliation(s)
- Karl Sigvard Olsson
- Section of Hematology and Coagulation, Department of Medicine, Sahlgrenska University Hospital, Göteborg, Sweden.
| | | | | | | | | |
Collapse
|
33
|
Pardo Silva MC, Njajou OT, Alizadeh BZ, Hofman A, Witteman JCM, van Duijn CM, Janssens ACJW. HFE gene mutations increase the risk of coronary heart disease in women. Eur J Epidemiol 2010; 25:643-9. [PMID: 20640879 PMCID: PMC2931632 DOI: 10.1007/s10654-010-9489-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2009] [Accepted: 07/05/2010] [Indexed: 01/14/2023]
Abstract
The purpose of the present study is to examine HFE gene mutations in relation to newly diagnosed (incident) coronary heart disease (CHD). In a population-based follow-up study of 7,983 individuals aged 55 years and older, we compared the risk of incident CHD between HFE carriers and non-carriers, overall and stratified by sex and smoking status. HFE mutations were significantly associated with an increased risk of incident CHD in women but not in men (hazard ratio [HR] for women = 1.7, 95% confidence interval [CI] 1.2-2.4 versus HR for men = 0.9, 95% CI 0.7-1.2). This increased CHD risk associated with HFE mutations in women was statistically significant in never smokers (HR = 1.8, 95% CI 1.1-2.8) and current smokers (HR = 3.1, 95% CI 1.4-7.1), but not in former smokers (HR = 1.3, 95% CI 0.7-2.4). HFE mutations are associated with increased risk of incident CHD in women.
Collapse
Affiliation(s)
- M. Carolina Pardo Silva
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, P. O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Omer T. Njajou
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, P. O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Behrooz Z. Alizadeh
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, P. O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, P. O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Jacqueline C. M. Witteman
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, P. O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Cornelia M. van Duijn
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, P. O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - A. Cecile J. W. Janssens
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, P. O. Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
34
|
Neghina AM, Anghel A. Hemochromatosis genotypes and risk of iron overload--a meta-analysis. Ann Epidemiol 2010; 21:1-14. [PMID: 20800508 DOI: 10.1016/j.annepidem.2010.05.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 05/23/2010] [Accepted: 05/24/2010] [Indexed: 11/18/2022]
Abstract
PURPOSE The incomplete phenotypic penetrance of high iron Fe genotypes in relation to hemochromatosis poses a practical problem in the interpretation of the genotyping results by clinicians. We carried out meta-analyses of the associations between hemochromatosis genotypes C282Y/C282Y, C282Y/H63D, C282Y/wild-type, H63D/H63D, H63D/wild-type, versus wild-type/wild-type and iron overload, both provisional (elevated serum iron markers) and documented (elevated serum iron markers associated with evidence of iron excess based on liver biopsy and/or quantitative phlebotomy). METHODS After reviewing 3572 article titles and evaluating 92 articles in detail, odds ratios were pooled from 43 study populations (9986 cases and 25,492 controls) using a random-effects model. RESULTS Homozygosity for either variant or compound heterozygosity was associated with both provisional and documented iron overload. Single heterozygosity conferred no risk for elevated hepatic iron index and/or mobilizable iron by quantitative phlebotomy. In patients with clinical hereditary hemochromatosis, no evidence of provisional and documented iron overload with transferrin saturation (TS) values greater than 55% was evidenced for C282Y and H63D single heterozygotes whereas documented iron overload including TS of 45% to 50% was weakly associated with C282Y/wild-type genotype; H63D/H63D genotype was not associated with documented iron overload in patients with TS values of 45% to 50%. CONCLUSIONS The results, mainly from case-control studies, cannot necessarily be extrapolated to the general population.
Collapse
Affiliation(s)
- Adriana Maria Neghina
- Biochemistry Department, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania.
| | | |
Collapse
|
35
|
Hemochromatosis (HFE) genotype and atherosclerosis: Increased susceptibility to iron-induced vascular damage in C282Y carriers? Atherosclerosis 2010; 211:520-5. [DOI: 10.1016/j.atherosclerosis.2010.03.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Revised: 03/11/2010] [Accepted: 03/11/2010] [Indexed: 11/23/2022]
|
36
|
Multiple loci influence erythrocyte phenotypes in the CHARGE Consortium. Nat Genet 2009; 41:1191-8. [PMID: 19862010 PMCID: PMC2778265 DOI: 10.1038/ng.466] [Citation(s) in RCA: 285] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Accepted: 09/01/2009] [Indexed: 12/13/2022]
Abstract
Measurements of erythrocytes within the blood are important clinical traits and can indicate various hematological disorders. We report here genome-wide association studies (GWAS) for six erythrocyte traits, including hemoglobin concentration (Hb), hematocrit (Hct), mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH), mean corpuscular hemoglobin concentration (MCHC) and red blood cell count (RBC). We performed an initial GWAS in cohorts of the CHARGE Consortium totaling 24,167 individuals of European ancestry and replication in additional independent cohorts of the HaemGen Consortium totaling 9,456 individuals. We identified 23 loci significantly associated with these traits in a meta-analysis of the discovery and replication cohorts (combined P values ranging from 5 x 10(-8) to 7 x 10(-86)). Our findings include loci previously associated with these traits (HBS1L-MYB, HFE, TMPRSS6, TFR2, SPTA1) as well as new associations (EPO, TFRC, SH2B3 and 15 other loci). This study has identified new determinants of erythrocyte traits, offering insight into common variants underlying variation in erythrocyte measures.
Collapse
|
37
|
Marx JJ, Kartikasari AE, Georgiou NA. Can Iron Chelators Influence the Progression of Atherosclerosis? Hemoglobin 2009; 32:123-34. [DOI: 10.1080/03630260701726871] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
38
|
Is serum ferritin within the reference range a risk predictor of cardiovascular disease? A population-based, long-term study comprising 2874 subjects. Br J Nutr 2009; 102:594-600. [DOI: 10.1017/s000711450922085x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
39
|
Sun Q, Ma J, Rifai N, Franco OH, Rexrode KM, Hu FB. Excessive body iron stores are not associated with risk of coronary heart disease in women. J Nutr 2008; 138:2436-41. [PMID: 19022969 PMCID: PMC2635523 DOI: 10.3945/jn.108.097766] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The positive association between body iron stores and risk of coronary heart disease (CHD) initially observed among a Finnish male population has not been corroborated by studies conducted in other populations. The soluble transferrin receptor (sTfR):ferritin ratio has been suggested to be a better index than ferritin to measure body iron stores. Because sTfR is sensitive to iron deficiency, this ratio can distinguish individuals with similar ferritin levels with respect to their iron status. To evaluate this novel index in relation to CHD risk, we prospectively identified and confirmed 242 incident CHD cases and randomly selected 483 controls matched for age, smoking, and fasting status among women that provided blood samples in the Nurses' Health Study during 9 y of follow-up. In both crude and multivariate analyses, neither the sTfR:ferritin ratio nor ferritin was significantly associated with an elevated risk of CHD. After multivariate adjustment for established and potential CHD risk factors, compared with women in the lowest quartile of the sTfR:ferritin ratio, women in the 2nd to 4th quartiles had relative risks (RR) (95% CI) of 1.39 (0.82, 2.36), 1.12 (0.66, 1.91), and 1.13 (0.65, 1.97; P-trend = 0.61), respectively. The multivariate RR (95% CI) for ferritin were 1.05 (0.62, 1.77), 1.19 (0.69, 2.03), and 1.05 (0.60, 1.85; P-trend = 0.90) across quartiles. Our data do not support the hypothesis that excessive body iron stores are associated with risk of CHD.
Collapse
Affiliation(s)
- Qi Sun
- Department of Nutrition, Harvard School of Public Health, Boston, MA 02115, USA.
| | - Jing Ma
- Department of Nutrition, Harvard School of Public Health, Boston, MA 02115; Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; Department of Laboratory Medicine, Children's Hospital and Harvard Medical School, Boston, MA 02115; Unilever Corporate Research, Colworth Park, Sharnbrook, UK MK44 1LQ; Channing Laboratory, Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and Department of Epidemiology, Harvard School of Public Health, Boston, MA 02115
| | - Nader Rifai
- Department of Nutrition, Harvard School of Public Health, Boston, MA 02115; Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; Department of Laboratory Medicine, Children's Hospital and Harvard Medical School, Boston, MA 02115; Unilever Corporate Research, Colworth Park, Sharnbrook, UK MK44 1LQ; Channing Laboratory, Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and Department of Epidemiology, Harvard School of Public Health, Boston, MA 02115
| | - Oscar H. Franco
- Department of Nutrition, Harvard School of Public Health, Boston, MA 02115; Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; Department of Laboratory Medicine, Children's Hospital and Harvard Medical School, Boston, MA 02115; Unilever Corporate Research, Colworth Park, Sharnbrook, UK MK44 1LQ; Channing Laboratory, Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and Department of Epidemiology, Harvard School of Public Health, Boston, MA 02115
| | - Kathryn M. Rexrode
- Department of Nutrition, Harvard School of Public Health, Boston, MA 02115; Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; Department of Laboratory Medicine, Children's Hospital and Harvard Medical School, Boston, MA 02115; Unilever Corporate Research, Colworth Park, Sharnbrook, UK MK44 1LQ; Channing Laboratory, Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and Department of Epidemiology, Harvard School of Public Health, Boston, MA 02115
| | - Frank B. Hu
- Department of Nutrition, Harvard School of Public Health, Boston, MA 02115; Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; Department of Laboratory Medicine, Children's Hospital and Harvard Medical School, Boston, MA 02115; Unilever Corporate Research, Colworth Park, Sharnbrook, UK MK44 1LQ; Channing Laboratory, Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and Department of Epidemiology, Harvard School of Public Health, Boston, MA 02115
| |
Collapse
|
40
|
Dunn T, Blankenship D, Beal N, Allen R, Schechter E, Moore W, Perveen G, Eichner J. HFE mutations in heart disease. Heart Vessels 2008; 23:348-55. [DOI: 10.1007/s00380-008-1047-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2007] [Accepted: 02/05/2008] [Indexed: 11/30/2022]
|
41
|
Lubitz SA, Goldbarg SH, Mehta D. Sudden cardiac death in infiltrative cardiomyopathies: sarcoidosis, scleroderma, amyloidosis, hemachromatosis. Prog Cardiovasc Dis 2008; 51:58-73. [PMID: 18634918 DOI: 10.1016/j.pcad.2007.10.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sarcoidosis, scleroderma, amyloidosis, and hemachromatosis are systemic infiltrative disorders that commonly affect the heart. Owing to their potential for diffuse organ involvement, these diseases may present with myriad clinical manifestations. Conduction system abnormalities are common, and each of these disorders has been associated with sudden cardiac death. In this review, we summarize the epidemiology, clinical features, diagnosis, and treatment of each of these entities. We place special emphasis on existing literature as it pertains to risk stratification and therapy aimed at the prevention of sudden cardiac death in these infiltrative cardiomyopathies.
Collapse
Affiliation(s)
- Steven A Lubitz
- Zena and Michael Wiener Cardiovascular Institute, Mount Sinai School of Medicine, NY, USA
| | | | | |
Collapse
|
42
|
Pedersen P, Melsen GV, Milman N. Frequencies of the haemochromatosis gene (HFE) variants C282Y, H63D and S65C in 6,020 ethnic Danish men. Ann Hematol 2008; 87:735-40. [PMID: 18542962 DOI: 10.1007/s00277-008-0506-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2008] [Accepted: 04/29/2008] [Indexed: 10/22/2022]
Abstract
The objective was to assess the frequencies of haemochromatosis (HFE) gene mutations or variants C282Y, H63D and S65C in ethnic Danes. This is a prospective epidemiologic population study. A cohort of 6,020 Danish men aged 30-50 years was screened for HFE C282Y (c845G-->A), H63D (c187C-->G) and S65C (c193A-->T) gene variants, assessed on saliva or blood samples by restriction fragment length polymorphism (RFLP) analysis. The C282Y gene variant allele was present in 5.6%, H63D in 12.8% and S65C in 1.8% of the chromosomes. In the entire series, we observed 1.4% H63D/C282Y, 0.1% S65C/C282Y and 0.4% H63D/S65C compound heterozygotes. The C282Y allele frequency in Denmark is of similar order as reported in other Scandinavian countries: Iceland 5.1%, Faeroe Islands 6.6%, Norway 6.8% and Sweden 5.8%. Also, the H63D frequency in Denmark is close to the frequencies in other Scandinavian countries: Iceland 10.9%, Faeroe Islands 15.2%, Norway 11.4% and Sweden 12.1%.
Collapse
Affiliation(s)
- Palle Pedersen
- Department of Clinical Biochemistry, Naestved Hospital, Ringstedgade 61, 4700 Naestved, Denmark.
| | | | | |
Collapse
|
43
|
Ayonrinde OT, Milward EA, Chua ACG, Trinder D, Olynyk JK. Clinical Perspectives on Hereditary Hemochromatosis. Crit Rev Clin Lab Sci 2008; 45:451-84. [DOI: 10.1080/10408360802335716] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
44
|
Franchini M, Targher G, Montagnana M, Lippi G. Iron and thrombosis. Ann Hematol 2007; 87:167-73. [PMID: 18066546 PMCID: PMC2226003 DOI: 10.1007/s00277-007-0416-1] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Accepted: 11/11/2007] [Indexed: 11/28/2022]
Abstract
Although essential for cell physiology, an increase or depletion of body iron has harmful effects on health. Apart from iron deficiency anemia and iron overload-related organ tissue damage, there are increasing evidences that body iron status is implicated in atherosclerotic cardiovascular diseases. The hypothesis formulated in 1981 that iron depletion may protect against cardiovascular events is intriguing and has generated a significant debate in the last two decades. Indeed, to study this phenomenon, several investigators have tried to design appropriate experimental and clinical studies and to identify useful biochemical and genetic markers of iron status. The results of the literature on the effect of iron deficiency and overload on vascular health are critically reviewed in this study from a pathogenic and clinical point of view.
Collapse
Affiliation(s)
- Massimo Franchini
- Servizio di Immunoematologia e Trasfusione, Centro Emofilia, Azienda Ospedaliera di Verona, Verona, Italy.
| | | | | | | |
Collapse
|
45
|
|
46
|
DePalma RG, Hayes VW, Zacharski LR. Bloodletting: Past and Present. J Am Coll Surg 2007; 205:132-44. [PMID: 17617342 DOI: 10.1016/j.jamcollsurg.2007.01.071] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Revised: 01/15/2007] [Accepted: 01/30/2007] [Indexed: 01/14/2023]
Affiliation(s)
- Ralph G DePalma
- Patient Care Services VA Central Office, Washington, DC, USA
| | | | | |
Collapse
|
47
|
Janower S, Rosmorduc O, Cohen A. [Cardiac involvement in hemochromatosis]. Presse Med 2007; 36:1301-12. [PMID: 17574380 DOI: 10.1016/j.lpm.2006.12.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2006] [Accepted: 12/31/2006] [Indexed: 10/22/2022] Open
Abstract
Cardiac involvement in hemochromatosis affects mainly the myocardium: iron overload of the myocytes reduces left ventricular distensibility. Heart failure is the most frequent manifestation of cardiac involvement. Diagnosis of cardiac involvement depends essentially on Doppler echocardiography showing abnormal left ventricular filling and, later, ventricular dilatation with left ventricular systolic dysfunction. Magnetic resonance imaging can quantify intrahepatic and intramyocardial iron levels. Age at onset of symptoms and specific organ involvement in hemochromatosis depend on the type of mutation. The two principal means of treatment by iron depletion are phlebotomy in primary hemochromatosis and excretion of iron by chemical chelation in secondary hemochromatosis. Early diagnosis and iron depletion improve survival by reducing organ iron overload, especially in the liver and the myocardium. Recent guidelines issued by Anaes (national agency for health evaluation) make it possible to identify risk factors for complications early, to determine disease stage, and to provide appropriate management as a function of disease severity.
Collapse
Affiliation(s)
- Sandra Janower
- Service de cardiologie, Hôpital Saint-Antoine, AP-HP, et Université Pierre et Marie Curie, Paris VI
| | | | | |
Collapse
|
48
|
van der A DL, Peeters PHM, Grobbee DE, Roest M, Marx JJM, Voorbij HM, van der Schouw YT. HFE mutations and risk of coronary heart disease in middle-aged women. Eur J Clin Invest 2006; 36:682-90. [PMID: 16968463 DOI: 10.1111/j.1365-2362.2006.01711.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Although heterozygosity for the C282Y mutation in the HFE gene has been associated with an increased risk of cardiovascular events, epidemiological studies remain inconclusive. The aim of the present study was to obtain further evidence as to whether HFE mutations are associated with risk of coronary heart disease (CHD) in middle-aged women. We used data of a cohort of 15 236 Dutch middle-aged women to investigate whether C282Y carriers and H63D carriers are at increased risk of coronary heart disease compared with non-carriers. MATERIALS AND METHODS Women were included in the study between 1993 and 1997 and were followed until 1 January 2000 for cardiovascular events. HFE genotyping was performed on all 211 coronary heart disease cases and a randomly selected sample from the baseline cohort (n = 1526). A weighted Cox proportional hazards model was used to estimate crude, age-adjusted and multivariate adjusted hazard ratios for C282Y and H63D carriership in relation to coronary heart disease. RESULTS Compared with non-carriers, those that carried the C282Y allele were not at increased risk for CHD (HR = 1.25, 95% CI = 0.74-2.09). Neither did we find an association between the H63D mutation and CHD risk (HR = 0.73, 95% CI = 0.43-1.24). CONCLUSIONS Our results are in accordance with similar studies to date, for which we present a meta-analysis. HFE mutations appear not to affect the risk of coronary heart disease.
Collapse
|
49
|
Zintzaras E, Kitsios G. Identification of chromosomal regions linked to premature myocardial infarction: a meta-analysis of whole-genome searches. J Hum Genet 2006; 51:1015-1021. [PMID: 17024316 DOI: 10.1007/s10038-006-0053-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2006] [Accepted: 08/13/2006] [Indexed: 10/24/2022]
Abstract
Myocardial infarction (MI) is a complication of coronary artery disease and the leading cause of death in the Western world. MI is considered a distinct phenotype with an increased genetic component for its premature type. MI's exact inheritance pattern is still unknown. Genome searches for identifying susceptibility loci for premature MI produced inconclusive or inconsistent results. Thus, a genome search meta-analysis (GSMA) was applied to available genome search data on premature MI. GSMA is a non-parametric method to identify genetic regions that rank high, on average in terms of linkage statistics across genome searches unweighted or weighted by study size. The significance of each region's average and heterogeneity, unadjusted or adjusted by neighbouring average simulated ranks, was calculated using a Monte Carlo test. The meta-analysis involved five genome searches in Caucasians. Eight regions (6p22.3-6p21.1, 14p13-14q13.1, 13q33.1-13q34, 5p15.33-5p15.1, 8q13.2-8q22.2, 1p36.21-1p35.2, 12q24.31-12q24.33, 8q24.21-8q24.3) were found to have significant average rank by either unweighted or weighted analyses. In addition, region 8q24.21-8q24.3 produced significant low heterogeneity (P (unadjusted)=0.03 and P (adjusted)=0.05). Four regions (6p22.3-6p21.1, 14p13-14q13.1, 8q13.2-8q22.2, 8q24.21-8q24.3) were not identified by the individual studies. The meta-analysis suggests that these four regions should be further investigated for genes that confer susceptibility to MI.
Collapse
Affiliation(s)
- Elias Zintzaras
- Department of Biomathematics, University of Thessaly School of Medicine, Papakyriazi 22, 41222, Larissa, Greece.
| | - Georgios Kitsios
- Department of Biomathematics, University of Thessaly School of Medicine, Papakyriazi 22, 41222, Larissa, Greece
| |
Collapse
|
50
|
Metzler B, Jehle J, Theurl I, Ludwiczek S, Obrist P, Pachinger O, Weiss G. Short term protective effects of iron in a murine model of ischemia/reperfusion. Biometals 2006; 20:205-15. [PMID: 16927172 DOI: 10.1007/s10534-006-9034-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2005] [Accepted: 07/24/2006] [Indexed: 10/24/2022]
Abstract
The role of iron in the pathogenesis of cardio-vascular disorders is still controversial. We studied the effects of iron perturbations on myocardial injury upon temporary ischemia/reperfusion. C57BL/6J male mice were injected with iron dextran for 2 weeks while controls received saline. Mice were then subjected to 30 min of myocardial ischemia and subsequent reperfusion for 6-24 h. Tissue damage was quantified histologically and by troponin T determination. The expressions of tumor necrosis factor-alpha (TNF-alpha), superoxide dismutase (SOD) and inducible nitric oxide synthase (iNOS) were investigated in non-ischemic and ischemic regions of both groups. After myocardial ischemia and reperfusion, troponin T levels, as a marker of myocardial damage, were significantly reduced in iron-treated mice as compared to control mice (P < 0.05). Under the same conditions the infarction area and damage score were significantly lower in iron-treated animals. In parallel, TNF-alpha and SOD expressions were increased in infarcted regions of iron-treated mice as compared to controls, whereas myocardial iNOS expression was significantly lower in iron-treated mice. Although, iron challenge increased radical formation and TNF-alpha expression in vivo, this did not result in myocardial damage which may be linked to the parallel induction of SOD. Importantly, iron treatment inhibited iNOS expression. Since, an increased nitric oxide (NO) formation has been linked to cardiac damage after acute myocardial infarction, iron may exert short time cardio-protective effects after induction of ischemia/reperfusion via decreasing iNOS formation.
Collapse
Affiliation(s)
- Bernhard Metzler
- Division of Cardiology, Medical University, A-6020, Innsbruck, Austria
| | | | | | | | | | | | | |
Collapse
|