1
|
Kesharwani P, Dash D, Koiri RK. Deciphering the role of hepcidin in iron metabolism and anemia management. J Trace Elem Med Biol 2025; 87:127591. [PMID: 39813816 DOI: 10.1016/j.jtemb.2025.127591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 12/09/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025]
Abstract
One of the most common diseases worldwide is anemia, which is characterized by insufficient erythrocyte production. Numerous complex factors, such as chronic diseases, genetic mutations, and nutritional inadequacies, contribute to this widespread syndrome. This review focuses specifically on anemias caused by defective hepcidin production. Hepcidin, a peptide hormone produced primarily by liver cells, plays a crucial role in regulating iron levels by controlling its absorption. Hepcidin's mechanism of action involves binding to the ferroportin iron transporter, causing its internalization. Disturbances in iron metabolism can have far-reaching consequences, affecting not only the blood but also organs like the liver, kidneys, and brain. Iron homeostasis is crucial for maintaining optimal physiological function. Several blood-based markers are employed to assess iron stores. However, these markers have inherent limitations. Hepcidin, a key regulator of iron metabolism, plays a pivotal role in preventing iron release into the plasma from absorptive enterocytes and macrophages. Elucidating the structure and function of hepcidin is essential for understanding its role in iron homeostasis, which has significant implications for the diagnosis and management of various anemia subtypes. A well-established correlation exists between hepcidin dysregulation and iron deficiency. Despite its potential as a biomarker, the clinical application of hepcidin is hindered by the lack of a commercially available, clinically validated assay. This review aims to provide a comprehensive overview of hepcidin's role in regulating blood iron concentrations and elucidate its implications in the pathogenesis of various anemia subtypes, paving the way for its future applications in research and clinical practice.
Collapse
Affiliation(s)
- Palak Kesharwani
- Biochemistry Laboratory, Department of Zoology, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh 470003, India
| | - Debabrata Dash
- Biochemistry Laboratory, Department of Zoology, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh 470003, India
| | - Raj Kumar Koiri
- Biochemistry Laboratory, Department of Zoology, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh 470003, India.
| |
Collapse
|
2
|
Allegra S, Comità S, Roetto A, De Francia S. Sex and Gender Differences in Iron Chelation. Biomedicines 2024; 12:2885. [PMID: 39767791 PMCID: PMC11673655 DOI: 10.3390/biomedicines12122885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES In the absence of physiological mechanisms to excrete excessive iron, the administration of iron chelation therapy is necessary. Age and hormones have an impact on the absorption, distribution, metabolism, and excretion of the medications used to treat iron excess, resulting in notable sex- and gender-related variances. METHODS Here, we aimed to review the literature on sex and gender in iron overload assessment and treatment. RESULTS The development of iron chelators has shown to be a successful therapy for lowering the body's iron levels and averting the tissue damage and organ failure that follows. Numerous studies have described how individual factors can impact chelation treatment, potentially impact therapeutic response, and/or result in inadequate chelation or elevated toxicity; however, most of these data have not considered male and female patients as different groups, and particularly, the effect of hormonal variations in women have never been considered. CONCLUSIONS An effective iron chelation treatment should take into account sex and gender differences.
Collapse
Affiliation(s)
- Sarah Allegra
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Gonzaga University Hospital, 10043 Orbassano, Italy; (S.C.); (A.R.); (S.D.F.)
| | | | | | | |
Collapse
|
3
|
Characterization and Toxicity Analysis of Lab-Created Respirable Coal Mine Dust from the Appalachians and Rocky Mountains Regions. MINERALS 2022. [DOI: 10.3390/min12070898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Coal mine workers are continuously exposed to respirable coal mine dust (RCMD) in workplaces, causing severe lung diseases. RCMD characteristics and their relations with dust toxicity need further research to understand the adverse exposure effects to RCMD. The geographic clustering of coal workers’ pneumoconiosis (CWP) suggests that RCMD in the Appalachian region may exhibit more toxicity than other geographic regions such as the Rocky Mountains. This study investigates the RCMD characteristics and toxicity based on geographic location. Dissolution experiments in simulated lung fluids (SLFs) and in vitro responses were conducted to determine the toxicity level of samples collected from five mines in the Rocky Mountains and Appalachian regions. Dust characteristics were investigated using Fourier-transform infrared spectroscopy, scanning electron microscopy, the BET method, total microwave digestion, X-ray diffraction, and X-ray photoelectron spectroscopy. Inductively coupled plasma mass spectrometry was conducted to determine the concentration of metals dissolved in the SLFs. Finer particle sizes and higher mineral and elemental contents were found in samples from the Appalachian regions. Si, Al, Fe, Cu, Sr, and Pb were found in dissolution experiments, but no trends were found indicating higher dissolutions in the Appalachian region. In vitro studies indicated a proinflammatory response in epithelial and macrophage cells, suggesting their possible participation in pneumoconiosis and lung diseases development.
Collapse
|
4
|
Bjørklund G, Peana M, Pivina L, Dosa A, Aaseth J, Semenova Y, Chirumbolo S, Medici S, Dadar M, Costea DO. Iron Deficiency in Obesity and after Bariatric Surgery. Biomolecules 2021; 11:biom11050613. [PMID: 33918997 PMCID: PMC8142987 DOI: 10.3390/biom11050613] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/10/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023] Open
Abstract
Iron deficiency (ID) is particularly frequent in obese patients due to increased circulating levels of acute-phase reactant hepcidin and adiposity-associated inflammation. Inflammation in obese subjects is closely related to ID. It induces reduced iron absorption correlated to the inhibition of duodenal ferroportin expression, parallel to the increased concentrations of hepcidin. Obese subjects often get decreased inflammatory response after bariatric surgery, accompanied by decreased serum hepcidin and therefore improved iron absorption. Bariatric surgery can induce the mitigation or resolution of obesity-associated complications, such as hypertension, insulin resistance, diabetes mellitus, and hyperlipidemia, adjusting many parameters in the metabolism. However, gastric bypass surgery and sleeve gastrectomy can induce malabsorption and may accentuate ID. The present review explores the burden and characteristics of ID and anemia in obese patients after bariatric surgery, accounting for gastric bypass technique (Roux-en-Y gastric bypass-RYGB) and sleeve gastrectomy (SG). After bariatric surgery, obese subjects' iron status should be monitored, and they should be motivated to use adequate and recommended iron supplementation.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Toften 24, 8610 Mo i Rana, Norway
- Correspondence: (G.B.); (M.P.)
| | - Massimiliano Peana
- Department of Chemistry and Pharmacy, University of Sassari, Via Vienna 2, 07100 Sassari, Italy;
- Correspondence: (G.B.); (M.P.)
| | - Lyudmila Pivina
- Department of Neurology, Ophthalmology and Otolaryngology, Semey Medical University, 071400 Semey, Kazakhstan; (L.P.); (Y.S.)
- CONEM Kazakhstan Environmental Health and Safety Research Group, Semey Medical University, 071400 Semey, Kazakhstan
| | - Alexandru Dosa
- Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania; (A.D.); (D.-O.C.)
| | - Jan Aaseth
- Research Department, Innlandet Hospital Trust, 2380 Brumunddal, Norway;
| | - Yuliya Semenova
- Department of Neurology, Ophthalmology and Otolaryngology, Semey Medical University, 071400 Semey, Kazakhstan; (L.P.); (Y.S.)
- CONEM Kazakhstan Environmental Health and Safety Research Group, Semey Medical University, 071400 Semey, Kazakhstan
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy;
- CONEM Scientific Secretary, 37134 Verona, Italy
| | - Serenella Medici
- Department of Chemistry and Pharmacy, University of Sassari, Via Vienna 2, 07100 Sassari, Italy;
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj 31975/148, Iran;
| | - Daniel-Ovidiu Costea
- Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania; (A.D.); (D.-O.C.)
| |
Collapse
|
5
|
Tan Z, Lu P, Adewole D, Diarra M, Gong J, Yang C. Iron requirement in the infection of Salmonella and its relevance to poultry health. J APPL POULTRY RES 2021. [DOI: 10.1016/j.japr.2020.09.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
|
6
|
Zhang YY, Stockmann R, Ng K, Ajlouni S. Opportunities for plant-derived enhancers for iron, zinc, and calcium bioavailability: A review. Compr Rev Food Sci Food Saf 2020; 20:652-685. [PMID: 33443794 DOI: 10.1111/1541-4337.12669] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/08/2020] [Accepted: 10/20/2020] [Indexed: 12/31/2022]
Abstract
Understanding of the mechanism of interactions between dietary elements, their salts, and complexing/binding ligands is vital to manage both deficiency and toxicity associated with essential element bioavailability. Numerous mineral ligands are found in both animal and plant foods and are known to exert bioactivity via element chelation resulting in modulation of antioxidant capacity or micobiome metabolism among other physiological outcomes. However, little is explored in the context of dietary mineral ligands and element bioavailability enhancement, particularly with respect to ligands from plant-derived food sources. This review highlights a novel perspective to consider various plant macro/micronutrients as prospective bioavailability enhancing ligands of three essential elements (Fe, Zn, and Ca). We also delineate the molecular mechanisms of the ligand-binding interactions underlying mineral bioaccessibility at the luminal level. We conclude that despite current understandings of some of the structure-activity relationships associated with strong mineral-ligand binding, the physiological links between ligands as element carriers and uptake at targeted sites throughout the gastrointestinal (GI) tract still require more research. The binding behavior of potential ligands in the human diet should be further elucidated and validated using pharmacokinetic approaches and GI models.
Collapse
Affiliation(s)
- Yianna Y Zhang
- School of Agriculture and Food, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia.,CSIRO Agriculture & Food, Werribee, VIC, Australia
| | | | - Ken Ng
- School of Agriculture and Food, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Said Ajlouni
- School of Agriculture and Food, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
7
|
Ocular siderosis: a misdiagnosed cause of visual loss due to ferrous intraocular foreign bodies-epidemiology, pathogenesis, clinical signs, imaging and available treatment options. Doc Ophthalmol 2020; 142:133-152. [PMID: 32949328 PMCID: PMC7943509 DOI: 10.1007/s10633-020-09792-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/02/2020] [Indexed: 01/06/2023]
Abstract
Purpose The purpose of this paper is to provide a meaningful literature review about the epidemiology, pathogenesis, clinical signs, imaging and treatment of ocular siderosis (OS). Methods A computerized search from inception up to March 2020 of the online electronic database PubMed was performed using the following search strings: “ocular siderosis” and “siderosis bulbi”. The reference list in each article was analysed for additional relevant publications. Results OS is an uncommon cause of visual loss due to a retained ferrous intraocular foreign body (IOFB). It may develop from 18 days to years after a penetrating trauma that usually occurs during hammering. On average, patients are 22–25 years old, and the vast majority are male. The most common cause of OS development is delayed presentation by the patient or missed diagnosis of IOFB after trauma. The pathophysiology is not fully understood; nevertheless, iron deposition causes hydroxyl radical formation, which damages photoreceptors and retinal pigment epithelium. Moreover, iron damages retinal vessels with consequent inner retinal layers degeneration. The most frequent signs are iris heterochromia, pupillary mydriasis, cataract development and retinal arteriolar narrowing with pigmentary retinal degeneration. Electroretinogram signs, in particular, b-wave amplitude reduction, arise earlier than clinical signs. Orbital CT scans and ultrasonography play an essential role in detecting IOFBs. Treatment depends on the IOFB location and OS development. However, it is crucial to remove the IOFB after OS development because visual acuity and clinical signs may improve. Anterior segment IOFBs can be dislodged using an intraocular magnet (IOM) or forceps through limbal paracentesis. In contrast, posterior segment IOFBs require a pars plana vitrectomy and IOM or forceps to be removed through an enlarged sclerotomy or the limbus. Conclusion Recommending the usage of protective glasses and spreading knowledge about OS may further benefit patient care.
Collapse
|
8
|
Schiavi A, Strappazzon F, Ventura N. Mitophagy and iron: two actors sharing the stage in age-associated neuronal pathologies. Mech Ageing Dev 2020; 188:111252. [PMID: 32330468 DOI: 10.1016/j.mad.2020.111252] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 12/12/2022]
Abstract
Aging is characterized by the deterioration of different cellular and organismal structures and functions. A typical hallmark of the aging process is the accumulation of dysfunctional mitochondria and excess iron, leading to a vicious cycle that promotes cell and tissue damage, which ultimately contribute to organismal aging. Accordingly, altered mitochondrial quality control pathways such as mitochondrial autophagy (mitophagy) as well as altered iron homeostasis, with consequent iron overload, can accelerate the aging process and the development and progression of different age-associated disorders. In this review we first briefly introduce the aging process and summarize molecular mechanisms regulating mitophagy and iron homeostasis. We then provide an overview on how dysfunction of these two processes impact on aging and age-associated neurodegenerative disorders with a focus on Alzheimer's disease, Parkinson's disease and Amyotrophic Lateral Sclerosis. Finally, we summarize some recent evidence showing mechanistic links between iron metabolism and mitophagy and speculate on how regulating the crosstalk between the two processes may provide protective effects against aging and age-associated neuronal pathologies.
Collapse
Affiliation(s)
- Alfonso Schiavi
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany; IUF- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | | | - Natascia Ventura
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany; IUF- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.
| |
Collapse
|
9
|
Bsteh G, Haschka D, Tymoszuk P, Berek K, Petzer V, Hegen H, Wurth S, Auer M, Zinganell A, Pauli FD, Deisenhammer F, Weiss G, Berger T. Serum hepcidin levels in multiple sclerosis. Mult Scler J Exp Transl Clin 2019; 5:2055217319885984. [PMID: 31723437 PMCID: PMC6831977 DOI: 10.1177/2055217319885984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 10/06/2019] [Accepted: 10/07/2019] [Indexed: 12/22/2022] Open
Abstract
Background Brain iron accumulation is associated with multiple sclerosis (MS). Hepcidin
is the master regulator of iron homeostasis and distribution. Dysregulation
of hepcidin is a feature of different chronic inflammatory diseases but has
not been investigated in MS so far. Objective The aim of this study was to determine serum hepcidin levels of MS patients
and healthy volunteers serving as controls and to investigate possible
relations between hepcidin levels, disease activity and disease course. Methods In a cross-sectional design, we measured serum hepcidin levels in 71 MS
patients and 16 healthy controls (HC). MS patients were sub-grouped in
active relapsing–remitting MS (aRRMS), inactive (i)RRMS, active progressive
MS (aPMS) and inactive (i)PMS. Blood parameters were measured by standard
laboratory methods. Results Median hepcidin levels were 26.9 ng/ml (confidence interval (CI) 22.8; 30.9)
in MS and 17.3 ng/ml (CI 12.8; 23.4) in HC with significant age and sex
effects. Hepcidin correlates were in line with hepcidin as an indicator of
iron stores. After correction for age and sex, hepcidin was neither
associated with MS subgroups nor degree of disability and occurrence of
relapses. Conclusions Serum hepcidin levels are not associated with disease activity and disease
course in MS.
Collapse
Affiliation(s)
- Gabriel Bsteh
- Department of Neurology, Medical University of Vienna, Austria
| | | | - Piotr Tymoszuk
- Department of Internal Medicine II, Medical University of Innsbruck, Austria
| | - Klaus Berek
- Department of Neurology, Medical University of Innsbruck, Austria
| | - Verena Petzer
- Department of Internal Medicine II, Medical University of Innsbruck, Austria
| | | | | | | | | | | | | | - Guenter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Austria
| |
Collapse
|
10
|
Jahng JWS, Alsaadi RM, Palanivel R, Song E, Hipolito VEB, Sung HK, Botelho RJ, Russell RC, Sweeney G. Iron overload inhibits late stage autophagic flux leading to insulin resistance. EMBO Rep 2019; 20:e47911. [PMID: 31441223 PMCID: PMC6776927 DOI: 10.15252/embr.201947911] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 07/18/2019] [Accepted: 07/26/2019] [Indexed: 12/21/2022] Open
Abstract
Iron overload, a common clinical occurrence, is implicated in the metabolic syndrome although the contributing pathophysiological mechanisms are not fully defined. We show that prolonged iron overload results in an autophagy defect associated with accumulation of dysfunctional autolysosomes and loss of free lysosomes in skeletal muscle. These autophagy defects contribute to impaired insulin-stimulated glucose uptake and insulin signaling. Mechanistically, we show that iron overload leads to a decrease in Akt-mediated repression of tuberous sclerosis complex (TSC2) and Rheb-mediated mTORC1 activation on autolysosomes, thereby inhibiting autophagic-lysosome regeneration. Constitutive activation of mTORC1 or iron withdrawal replenishes lysosomal pools via increased mTORC1-UVRAG signaling, which restores insulin sensitivity. Induction of iron overload via intravenous iron-dextran delivery in mice also results in insulin resistance accompanied by abnormal autophagosome accumulation, lysosomal loss, and decreased mTORC1-UVRAG signaling in muscle. Collectively, our results show that chronic iron overload leads to a profound autophagy defect through mTORC1-UVRAG inhibition and provides new mechanistic insight into metabolic syndrome-associated insulin resistance.
Collapse
Affiliation(s)
| | | | | | - Erfei Song
- Department of BiologyYork UniversityTorontoONCanada
| | | | | | - Roberto Jorge Botelho
- Department of Chemistry and Biology and the Molecular Science Graduate ProgramRyerson UniversityTorontoONCanada
| | | | - Gary Sweeney
- Department of BiologyYork UniversityTorontoONCanada
| |
Collapse
|
11
|
Qian ZM, Ke Y. Hepcidin and its therapeutic potential in neurodegenerative disorders. Med Res Rev 2019; 40:633-653. [PMID: 31471929 DOI: 10.1002/med.21631] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/18/2019] [Accepted: 08/05/2019] [Indexed: 12/12/2022]
Abstract
Abnormally high brain iron, resulting from the disrupted expression or function of proteins involved in iron metabolism in the brain, is an initial cause of neuronal death in neuroferritinopathy and aceruloplasminemia, and also plays a causative role in at least some of the other neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, and Friedreich's ataxia. As such, iron is believed to be a novel target for pharmacological intervention in these disorders. Reducing iron toward normal levels or hampering the increases in iron associated with age in the brain is a promising therapeutic strategy for all iron-related neurodegenerative disorders. Hepcidin is a crucial regulator of iron homeostasis in the brain. Recent studies have suggested that upregulating brain hepcidin levels can significantly reduce brain iron content through the regulation of iron transport protein expression in the blood-brain barrier and in neurons and astrocytes. In this review, we focus on the discussion of the therapeutic potential of hepcidin in iron-associated neurodegenerative diseases and also provide a systematic overview of recent research progress on how misregulated brain iron metabolism is involved in the development of multiple neurodegenerative disorders.
Collapse
Affiliation(s)
- Zhong-Ming Qian
- Institute of Translational & Precision Medicine, Nantong University, Nantong, Jiangsu, China.,Laboratory of Neuropharmacology, School of Pharmacy & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ya Ke
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| |
Collapse
|
12
|
Qian ZM, Ke Y. Brain iron transport. Biol Rev Camb Philos Soc 2019; 94:1672-1684. [PMID: 31190441 DOI: 10.1111/brv.12521] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/22/2019] [Accepted: 04/24/2019] [Indexed: 12/19/2022]
Abstract
Brain iron is a crucial participant and regulator of normal physiological activity. However, excess iron is involved in the formation of free radicals, and has been associated with oxidative damage to neuronal and other brain cells. Abnormally high brain iron levels have been observed in various neurodegenerative diseases, including neurodegeneration with brain iron accumulation, Alzheimer's disease, Parkinson's disease and Huntington's disease. However, the key question of why iron levels increase in the relevant regions of the brain remains to be answered. A full understanding of the homeostatic mechanisms involved in brain iron transport and metabolism is therefore critical not only for elucidating the pathophysiological mechanisms responsible for excess iron accumulation in the brain but also for developing pharmacological interventions to disrupt the chain of pathological events occurring in these neurodegenerative diseases. Numerous studies have been conducted, but to date no effort to synthesize these studies and ideas into a systematic and coherent summary has been made, especially concerning iron transport across the luminal (apical) membrane of the capillary endothelium and the membranes of different brain cell types. Herein, we review key findings on brain iron transport, highlighting the mechanisms involved in iron transport across the luminal (apical) as well as the abluminal (basal) membrane of the blood-brain barrier, the blood-cerebrospinal fluid barrier, and iron uptake and release in neurons, oligodendrocytes, astrocytes and microglia within the brain. We offer suggestions for addressing the many important gaps in our understanding of this important topic, and provide new insights into the potential causes of abnormally increased iron levels in regions of the brain in neurodegenerative disorders.
Collapse
Affiliation(s)
- Zhong-Ming Qian
- Institute of Translational & Precision Medicine, Nantong University, Nantong, 226019, China.,Laboratory of Neuropharmacology, School of Pharmacy, & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 201203, China
| | - Ya Ke
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
13
|
Gill D, Benyamin B, Moore LSP, Monori G, Zhou A, Koskeridis F, Evangelou E, Laffan M, Walker AP, Tsilidis KK, Dehghan A, Elliott P, Hyppönen E, Tzoulaki I. Associations of genetically determined iron status across the phenome: A mendelian randomization study. PLoS Med 2019; 16:e1002833. [PMID: 31220083 PMCID: PMC6586257 DOI: 10.1371/journal.pmed.1002833] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 05/21/2019] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Iron is integral to many physiological processes, and variations in its levels, even within the normal range, can have implications for health. The objective of this study was to explore the broad clinical effects of varying iron status. METHODS AND FINDINGS Genome-wide association study (GWAS) summary data obtained from 48,972 European individuals (55% female) across 19 cohorts in the Genetics of Iron Status Consortium were used to identify 3 genetic variants (rs1800562 and rs1799945 in the hemochromatosis gene [HFE] and rs855791 in the transmembrane protease serine 6 gene [TMPRSS6]) that associate with increased serum iron, ferritin, and transferrin saturation and decreased transferrin levels, thus serving as instruments for systemic iron status. Phenome-wide association study (PheWAS) of these instruments was performed on 424,439 European individuals (54% female) in the UK Biobank who were aged 40-69 years when recruited from 2006 to 2010, with their genetic data linked to Hospital Episode Statistics (HES) from April, 1995 to March, 2016. Two-sample summary data mendelian randomization (MR) analysis was performed to investigate the effect of varying iron status on outcomes across the human phenome. MR-PheWAS analysis for the 3 iron status genetic instruments was performed separately and then pooled by meta-analysis. Correction was made for testing of multiple correlated phenotypes using a 5% false discovery rate (FDR) threshold. Heterogeneity between MR estimates for different instruments was used to indicate possible bias due to effects of the genetic variants through pathways unrelated to iron status. There were 904 distinct phenotypes included in the MR-PheWAS analyses. After correcting for multiple testing, the 3 genetic instruments for systemic iron status demonstrated consistent evidence of a causal effect of higher iron status on decreasing risk of traits related to anemia (iron deficiency anemia: odds ratio [OR] scaled to a standard deviation [SD] increase in genetically determined serum iron levels 0.72, 95% confidence interval [CI] 0.64-0.81, P = 4 × 10-8) and hypercholesterolemia (hypercholesterolemia: OR 0.88, 95% CI 0.83-0.93, P = 2 × 10-5) and increasing risk of traits related to infection of the skin and related structures (cellulitis and abscess of the leg: OR 1.25, 95% CI 1.10-1.42, P = 6 × 10-4). The main limitations of this study relate to possible bias from pleiotropic effects of the considered genetic variants and misclassification of diagnoses in the HES data. Furthermore, this work only investigated participants with European ancestry, and the findings may not be applicable to other ethnic groups. CONCLUSIONS Our findings offer novel, to our knowledge, insight into previously unreported effects of iron status, highlighting a potential protective effect of higher iron status on hypercholesterolemia and a detrimental role on risk of skin and skin structure infections. Given the modifiable and variable nature of iron status, these findings warrant further investigation.
Collapse
Affiliation(s)
- Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- * E-mail:
| | - Beben Benyamin
- Australian Centre for Precision Health, University of South Australia, Adelaide, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Luke S. P. Moore
- National Institute for Health Research Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, Imperial College London, United Kingdom
- Chelsea & Westminster NHS Foundation Trust, London, United Kingdom
- Imperial Biomedical Research Centre, Imperial College London and Imperial College NHS Healthcare Trust, London, United Kingdom
| | - Grace Monori
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Ang Zhou
- Australian Centre for Precision Health, University of South Australia, Adelaide, Australia
| | - Fotios Koskeridis
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Mike Laffan
- Centre for Haematology, Imperial College London, United Kingdom
| | - Ann P. Walker
- Population Science & Experimental Medicine, Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Konstantinos K. Tsilidis
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Medical Research Council-Public Health England Centre for Environment, School of Public Health, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Imperial Biomedical Research Centre, Imperial College London and Imperial College NHS Healthcare Trust, London, United Kingdom
- Medical Research Council-Public Health England Centre for Environment, School of Public Health, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
- Health Data Research UK-London, London, United Kingdom
| | - Elina Hyppönen
- Australian Centre for Precision Health, University of South Australia, Adelaide, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
- Population, Policy and Practice, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
- Medical Research Council-Public Health England Centre for Environment, School of Public Health, Imperial College London, London, United Kingdom
| |
Collapse
|
14
|
Wang Y, Wu Y, Li T, Wang X, Zhu C. Iron Metabolism and Brain Development in Premature Infants. Front Physiol 2019; 10:463. [PMID: 31105583 PMCID: PMC6494966 DOI: 10.3389/fphys.2019.00463] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 04/04/2019] [Indexed: 12/12/2022] Open
Abstract
Iron is important for a remarkable array of essential functions during brain development, and it needs to be provided in adequate amounts, especially to preterm infants. In this review article, we provide an overview of iron metabolism and homeostasis at the cellular level, as well as its regulation at the mRNA translation level, and we emphasize the importance of iron for brain development in fetal and early life in preterm infants. We also review the risk factors for disrupted iron metabolism that lead to high risk of developing iron deficiency and subsequent adverse effects on neurodevelopment in preterm infants. At the other extreme, iron overload, which is usually caused by excess iron supplementation in iron-replete preterm infants, might negatively impact brain development or even induce brain injury. Maintaining the balance of iron during the fetal and neonatal periods is important, and thus iron status should be monitored routinely and evaluated thoroughly during the neonatal period or before discharge of preterm infants so that iron supplementation can be individualized.
Collapse
Affiliation(s)
- Yafeng Wang
- Department of Neonatology (NICU), Children’s Hospital Affiliated Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Clinical Neuroscience, Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Yanan Wu
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tao Li
- Department of Neonatology (NICU), Children’s Hospital Affiliated Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Clinical Neuroscience, Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Physiology, Sahlgrenska Academy, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Clinical Neuroscience, Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
15
|
Tuning the Anti(myco)bacterial Activity of 3-Hydroxy-4-pyridinone Chelators through Fluorophores. Pharmaceuticals (Basel) 2018; 11:ph11040110. [PMID: 30347802 PMCID: PMC6316862 DOI: 10.3390/ph11040110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 10/17/2018] [Accepted: 10/18/2018] [Indexed: 02/08/2023] Open
Abstract
Controlling the sources of Fe available to pathogens is one of the possible strategies that can be successfully used by novel antibacterial drugs. We focused our interest on the design of chelators to address Mycobacterium avium infections. Taking into account the molecular structure of mycobacterial siderophores and considering that new chelators must be able to compete for Fe(III), we selected ligands of the 3-hydroxy-4-pyridinone class to achieve our purpose. After choosing the type of chelating unit it was also our objective to design chelators that could be monitored inside the cell and for that reason we designed chelators that could be functionalized with fluorophores. We didn’t realize at the time that the incorporation a fluorophore, to allow spectroscopic detection, would be so relevant for the antimycobacterial effect or to determine the affinity of the chelators towards biological membranes. From a biophysical perspective, this is a fascinating illustration of the fact that functionalization of a molecule with a particular label may lead to a change in its membrane permeation properties and result in a dramatic change in biological activity. For that reason we believe it is interesting to give a critical account of our entire work in this area and justify the statement “to label means to change”. New perspectives regarding combined therapeutic approaches and the use of rhodamine B conjugates to target closely related problems such as bacterial resistance and biofilm production are also discussed.
Collapse
|
16
|
Hagen WR, Hagedoorn PL, Honarmand Ebrahimi K. The workings of ferritin: a crossroad of opinions. Metallomics 2018; 9:595-605. [PMID: 28573266 DOI: 10.1039/c7mt00124j] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Biochemistry of the essential element iron is complicated by radical chemistry associated with Fe(ii) ions and by the extremely low solubility of the Fe(iii) ion in near-neutral water. To mitigate these problems cells from all domains of life synthesize the protein ferritin to take up and oxidize Fe(ii) and to form a soluble storage of Fe(iii) from which iron can be made available for physiology. A long history of studies on ferritin has not yet resulted in a generally accepted mechanism of action of this enzyme. In fact strong disagreement exists between extant ideas on several key steps in the workings of ferritin. The scope of this review is to explain the experimental background of these controversies and to indicate directions towards their possible resolution.
Collapse
Affiliation(s)
- Wilfred R Hagen
- Delft University of Technology, Department of Biotechnology, van der Maasweg 9, 2629 HZ Delft, The Netherlands.
| | | | | |
Collapse
|
17
|
Oxidative stress and neurodegeneration: the involvement of iron. Biometals 2018; 31:715-735. [PMID: 30014355 DOI: 10.1007/s10534-018-0126-2] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 07/04/2018] [Indexed: 12/14/2022]
Abstract
Many evidences indicate that oxidative stress plays a significant role in a variety of human disease states, including neurodegenerative diseases. Iron is an essential metal for almost all living organisms due to its involvement in a large number of iron-containing proteins and enzymes, though it could be also toxic. Actually, free iron excess generates oxidative stress, particularly in brain, where anti-oxidative defences are relatively low. Its accumulation in specific regions is associated with pathogenesis in a variety of neurodegenerative diseases (i.e., Parkinson's disease, Alzheimer's disease, Huntington's chorea, Amyotrophic Lateral Sclerosis and Neurodegeneration with Brain Iron Accumulation). Anyway, the extent of toxicity is dictated, in part, by the localization of the iron complex within the cell (cytosolic, lysosomal and mitochondrial), its biochemical form, i.e., ferritin or hemosiderin, as well as the ability of the cell to prevent the generation and propagation of free radical by the wide range of antioxidants and cytoprotective enzymes in the cell. Particularly, ferrous iron can act as a catalyst in the Fenton reaction that potentiates oxygen toxicity by generating a wide range of free radical species, including hydroxyl radicals (·OH). The observation that patients with neurodegenerative diseases show a dramatic increase in their brain iron content, correlated with the production of reactive oxigen species in these areas of the brain, conceivably suggests that disturbances in brain iron homeostasis may contribute to the pathogenesis of these disorders. The aim of this review is to describe the chemical features of iron in human beings and iron induced toxicity in neurodegenerative diseases. Furthermore, the attention is focused on metal chelating drugs therapeutic strategies.
Collapse
|
18
|
Milic S, Mikolasevic I, Orlic L, Devcic E, Starcevic-Cizmarevic N, Stimac D, Kapovic M, Ristic S. The Role of Iron and Iron Overload in Chronic Liver Disease. Med Sci Monit 2016; 22:2144-51. [PMID: 27332079 PMCID: PMC4922827 DOI: 10.12659/msm.896494] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The liver plays a major role in iron homeostasis; thus, in patients with chronic liver disease, iron regulation may be disturbed. Higher iron levels are present not only in patients with hereditary hemochromatosis, but also in those with alcoholic liver disease, nonalcoholic fatty liver disease, and hepatitis C viral infection. Chronic liver disease decreases the synthetic functions of the liver, including the production of hepcidin, a key protein in iron metabolism. Lower levels of hepcidin result in iron overload, which leads to iron deposits in the liver and higher levels of non-transferrin-bound iron in the bloodstream. Iron combined with reactive oxygen species leads to an increase in hydroxyl radicals, which are responsible for phospholipid peroxidation, oxidation of amino acid side chains, DNA strain breaks, and protein fragmentation. Iron-induced cellular damage may be prevented by regulating the production of hepcidin or by administering hepcidin agonists. Both of these methods have yielded successful results in mouse models.
Collapse
Affiliation(s)
- Sandra Milic
- Department of Gastroenterology, UHC Rijeka, Rijeka, Croatia
| | | | - Lidija Orlic
- Department of Nephrology, Dialysis and Kidney Transplantation, UHC Rijeka, Rijeka, Croatia
| | - Edita Devcic
- Department of Gastroenterology, UHC Rijeka, Rijeka, Croatia
| | | | - Davor Stimac
- Department of Gastroenterology, UHC Rijeka, Rijeka, Croatia
| | - Miljenko Kapovic
- Department of Biology and Medical Genetics, Faculty of Medicine, Rijeka, Croatia
| | - Smiljana Ristic
- Department of Biology and Medical Genetics, Faculty of Medicine, Rijeka, Croatia
| |
Collapse
|
19
|
Sebastiani G, Wilkinson N, Pantopoulos K. Pharmacological Targeting of the Hepcidin/Ferroportin Axis. Front Pharmacol 2016; 7:160. [PMID: 27445804 PMCID: PMC4914558 DOI: 10.3389/fphar.2016.00160] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 05/31/2016] [Indexed: 12/22/2022] Open
Abstract
The iron regulatory hormone hepcidin limits iron fluxes to the bloodstream by promoting degradation of the iron exporter ferroportin in target cells. Hepcidin insufficiency causes hyperabsorption of dietary iron, hyperferremia and tissue iron overload, which are hallmarks of hereditary hemochromatosis. Similar responses are also observed in iron-loading anemias due to ineffective erythropoiesis (such as thalassemias, dyserythropoietic anemias and myelodysplastic syndromes) and in chronic liver diseases. On the other hand, excessive hepcidin expression inhibits dietary iron absorption and leads to hypoferremia and iron retention within tissue macrophages. This reduces iron availability for erythroblasts and contributes to the development of anemias with iron-restricted erythropoiesis (such as anemia of chronic disease and iron-refractory iron-deficiency anemia). Pharmacological targeting of the hepcidin/ferroportin axis may offer considerable therapeutic benefits by correcting iron traffic. This review summarizes the principles underlying the development of hepcidin-based therapies for the treatment of iron-related disorders, and discusses the emerging strategies for manipulating hepcidin pathways.
Collapse
Affiliation(s)
- Giada Sebastiani
- Department of Medicine, McGill UniversityMontreal, QC, Canada; Division of Gastroenterology, Royal Victoria HospitalMontreal, QC, Canada
| | - Nicole Wilkinson
- Lady Davis Institute for Medical Research, Jewish General Hospital Montreal, QC, Canada
| | - Kostas Pantopoulos
- Department of Medicine, McGill UniversityMontreal, QC, Canada; Lady Davis Institute for Medical Research, Jewish General HospitalMontreal, QC, Canada
| |
Collapse
|
20
|
Kontoghiorghe CN, Kontoghiorghes GJ. New developments and controversies in iron metabolism and iron chelation therapy. World J Methodol 2016; 6:1-19. [PMID: 27019793 PMCID: PMC4804243 DOI: 10.5662/wjm.v6.i1.1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 11/17/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023] Open
Abstract
Iron is essential for all organisms including microbial, cancer and human cells. More than a quarter of the human population is affected by abnormalities of iron metabolism, mainly from iron deficiency and iron overload. Iron also plays an important role in free radical pathology and oxidative damage which is observed in almost all major diseases, cancer and ageing. New developments include the complete treatment of iron overload and reduction of morbidity and mortality in thalassaemia using deferiprone and selected deferiprone/deferoxamine combinations and also the use of the maltol iron complex in the treatment of iron deficiency anaemia. There is also a prospect of using deferiprone as a universal antioxidant in non iron overloaded diseases such as neurodegenerative, cardiovascular, renal, infectious diseases and cancer. New regulatory molecules of iron metabolism such as endogenous and dietary chelating molecules, hepcidin, mitochondrial ferritin and their role in health and disease is under evaluation. Similarly, new mechanisms of iron deposition, removal, distribution and toxicity have been identified using new techniques such as magnetic resonance imaging increasing our understanding of iron metabolic processes and the targeted treatment of related diseases. The uniform distribution of iron in iron overload between organs and within each organ is no longer valid. Several other controversies such as the toxicity impact of non transferrin bound iron vs injected iron, the excess levels of iron in tissues causing toxicity and the role of chelation on iron absorption need further investigation. Commercial interests of pharmaceutical companies and connections to leading journals are playing a crucial role in shaping worldwide medical opinion on drug sales and use but also patients' therapeutic outcome and safety. Major controversies include the selection criteria and risk/benefit assessment in the use of deferasirox in thalassaemia and more so in idiopathic haemochromatosis, thalassaemia intermedia and ex-thalassaemia transplanted patients who are safely treated with venesection. Iron chelating drugs can override normal regulatory pathways, correct iron imbalance and minimise iron toxicity. The use of iron chelating drugs as main, alternative or adjuvant therapy is in progress in many conditions, especially those with non established or effective therapies.
Collapse
|
21
|
Dual Role of ROS as Signal and Stress Agents: Iron Tips the Balance in favor of Toxic Effects. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8629024. [PMID: 27006749 PMCID: PMC4783558 DOI: 10.1155/2016/8629024] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 01/28/2016] [Indexed: 01/01/2023]
Abstract
Iron is essential for life, while also being potentially harmful. Therefore, its level is strictly monitored and complex pathways have evolved to keep iron safely bound to transport or storage proteins, thereby maintaining homeostasis at the cellular and systemic levels. These sequestration mechanisms ensure that mildly reactive oxygen species like anion superoxide and hydrogen peroxide, which are continuously generated in cells living under aerobic conditions, keep their physiologic role in cell signaling while escaping iron-catalyzed transformation in the highly toxic hydroxyl radical. In this review, we describe the multifaceted systems regulating cellular and body iron homeostasis and discuss how altered iron balance may lead to oxidative damage in some pathophysiological settings.
Collapse
|
22
|
Fujikura Y, Krijt J, Povýšil C, Mělková Z, Přikryl P, Vokurka M, Nečas E. Iron Overload Causes Alterations of E-Cadherin in the Liver. Folia Biol (Praha) 2016; 62:95-102. [PMID: 27516188 DOI: 10.14712/fb2016062030095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Iron overload causes tissue damage in the liver, but its initial effects at the molecular and cellular level are not well understood. Epithelial cadherin (E-cad) is a major adhesion protein in adherens junctions and is associated with several signal transduction pathways. Dysfunction of E-cad causes instability of adherens junctions, which leads to cell invasion, cell migration, and carcinogenesis. We found in liver samples from iron-overloaded mice that the apparent molecular mass of E-cad was reduced from 125 to 115 kDa in sodium dodecyl sulphate polyacrylamide gel electrophoresis under reducing conditions and immunoblotting, and that the cellular expression of E-cad was decreased in immunohistochemistry. The mRNA level of E-cad, however, did not change significantly, suggesting that the alterations are posttranslational. Interestingly, incubation of control liver extracts with Fe2+ alone also produced the same mobility shift. Neither an oxidant nor an antioxidant influenced this shift in vitro, suggesting that reactive oxygen species, which are generated by iron and known to cause damage to macromolecules, are not involved. Treatment of the 115 kDa E-cad with deferoxamine, an iron chelator, thus removing Fe2+, shifted the molecular mass back to 125 kDa, demonstrating that the shift is reversible. The observation also implies that the alteration that causes the mobility shift is not due to transcriptional control, deglycosylation, and proteolysis. This reversible mobility shift of E-cad has not been previously known. The alteration of E-cad that causes the mobility shift might be an initial step to liver diseases by iron overload.
Collapse
Affiliation(s)
- Y Fujikura
- Institute of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Czech Republic
| | - J Krijt
- Institute of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Czech Republic
| | - C Povýšil
- Institute of Pathology, First Faculty of Medicine, Charles University in Prague, Czech Republic
| | - Z Mělková
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University in Prague, Czech Republic
| | - P Přikryl
- Institute of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Czech Republic
| | - M Vokurka
- Institute of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Czech Republic
| | - E Nečas
- Institute of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Czech Republic
| |
Collapse
|
23
|
Iron-induced damage in cardiomyopathy: oxidative-dependent and independent mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:230182. [PMID: 25878762 PMCID: PMC4387903 DOI: 10.1155/2015/230182] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 03/06/2015] [Accepted: 03/15/2015] [Indexed: 02/08/2023]
Abstract
The high incidence of cardiomyopathy in patients with hemosiderosis, particularly in transfusional iron overload, strongly indicates that iron accumulation in the heart plays a major role in the process leading to heart failure. In this context, iron-mediated generation of noxious reactive oxygen species is believed to be the most important pathogenetic mechanism determining cardiomyocyte damage, the initiating event of a pathologic progression involving apoptosis, fibrosis, and ultimately cardiac dysfunction. However, recent findings suggest that additional mechanisms involving subcellular organelles and inflammatory mediators are important factors in the development of this disease. Moreover, excess iron can amplify the cardiotoxic effect of other agents or events. Finally, subcellular misdistribution of iron within cardiomyocytes may represent an additional pathway leading to cardiac injury. Recent advances in imaging techniques and chelators development remarkably improved cardiac iron overload detection and treatment, respectively. However, increased understanding of the pathogenic mechanisms of iron overload cardiomyopathy is needed to pave the way for the development of improved therapeutic strategies.
Collapse
|
24
|
Kitsati N, Liakos D, Ermeidi E, Mantzaris MD, Vasakos S, Kyratzopoulou E, Eliadis P, Andrikos E, Kokkolou E, Sferopoulos G, Mamalaki A, Siamopoulos K, Galaris D. Rapid elevation of transferrin saturation and serum hepcidin concentration in hemodialysis patients after intravenous iron infusion. Haematologica 2015; 100:e80-3. [PMID: 25425685 PMCID: PMC4349282 DOI: 10.3324/haematol.2014.116806] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Natalia Kitsati
- Laboratory of Biological Chemistry, University of Ioannina Medical School, Ioannina
| | | | - Eleni Ermeidi
- Department of Nephrology, University of Ioannina Medical School, Ioannina
| | - Michalis D Mantzaris
- Laboratory of Biological Chemistry, University of Ioannina Medical School, Ioannina
| | - Spyros Vasakos
- Biochemical Laboratory, Hatzikosta Regional Hospital, Ioannina
| | - Eleni Kyratzopoulou
- Laboratory of Molecular Biology and Immunobiotechnology, Hellenic Pasteur Institute, Athens, Greece
| | - Petros Eliadis
- Laboratory of Molecular Biology and Immunobiotechnology, Hellenic Pasteur Institute, Athens, Greece
| | | | | | | | - Avgi Mamalaki
- Laboratory of Molecular Biology and Immunobiotechnology, Hellenic Pasteur Institute, Athens, Greece
| | | | - Dimitrios Galaris
- Laboratory of Biological Chemistry, University of Ioannina Medical School, Ioannina
| |
Collapse
|
25
|
Padda RS, Gkouvatsos K, Guido M, Mui J, Vali H, Pantopoulos K. A high-fat diet modulates iron metabolism but does not promote liver fibrosis in hemochromatotic Hjv⁻/⁻ mice. Am J Physiol Gastrointest Liver Physiol 2015; 308:G251-61. [PMID: 25501544 DOI: 10.1152/ajpgi.00137.2014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hemojuvelin (Hjv) is a membrane protein that controls body iron metabolism by enhancing signaling to hepcidin. Hjv mutations cause juvenile hemochromatosis, a disease of systemic iron overload. Excessive iron accumulation in the liver progressively leads to inflammation and disease, such as fibrosis, cirrhosis, or hepatocellular cancer. Fatty liver (steatosis) may also progress to inflammation (steatohepatitis) and liver disease, and iron is considered as pathogenic cofactor. The aim of this study was to investigate the pathological implications of parenchymal iron overload due to Hjv ablation in the fatty liver. Wild-type (WT) and Hjv(-/-) mice on C57BL/6 background were fed a standard chow, a high-fat diet (HFD), or a HFD supplemented with 2% carbonyl iron (HFD+Fe) for 12 wk. The animals were analyzed for iron and lipid metabolism. As expected, all Hjv(-/-) mice manifested higher serum and hepatic iron and diminished hepcidin levels compared with WT controls. The HFD reduced iron indexes and promoted liver steatosis in both WT and Hjv(-/-) mice. Notably, steatosis was attenuated in Hjv(-/-) mice on the HFD+Fe regimen. Hjv(-/-) animals gained less body weight and exhibited reduced serum glucose and cholesterol levels. Histological and ultrastructural analysis revealed absence of iron-induced inflammation or liver fibrosis despite early signs of liver injury (expression of α-smooth muscle actin). We conclude that parenchymal hepatic iron overload does not suffice to trigger progression of liver steatosis to steatohepatitis or fibrosis in C57BL/6 mice.
Collapse
Affiliation(s)
- Ranjit Singh Padda
- Lady Davis Institute for Medical Research, Jewish General Hospital, and Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Konstantinos Gkouvatsos
- Lady Davis Institute for Medical Research, Jewish General Hospital, and Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Maria Guido
- Department of Diagnostic Sciences and Special Therapies, University of Padova, Padova, Italy; and
| | - Jeannie Mui
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | - Hojatollah Vali
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research, Jewish General Hospital, and Department of Medicine, McGill University, Montreal, Quebec, Canada;
| |
Collapse
|
26
|
Abstract
Non-transferrin-bound iron (NTBI) emerges in plasma of patients with systemic iron overload, but has also been documented in further pathological conditions. Quantification of NTBI can be useful for diagnosis and management of these disorders. However, currently available detection methods are tedious and often inaccurate, hampering wide applicability. In this issue of the Biochemical Journal, Ma et al. report the development of a novel assay for NTBI measurement, based on an iron-sensitive fluorescent probe that is linked to magnetic beads. The approach offers several advantages over existing technology and may bring NTBI assessment closer to the clinic.
Collapse
|
27
|
Fillebeen C, Wilkinson N, Pantopoulos K. Electrophoretic mobility shift assay (EMSA) for the study of RNA-protein interactions: the IRE/IRP example. J Vis Exp 2014:52230. [PMID: 25548934 PMCID: PMC4396942 DOI: 10.3791/52230] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
RNA/protein interactions are critical for post-transcriptional regulatory pathways. Among the best-characterized cytosolic RNA-binding proteins are iron regulatory proteins, IRP1 and IRP2. They bind to iron responsive elements (IREs) within the untranslated regions (UTRs) of several target mRNAs, thereby controlling the mRNAs translation or stability. IRE/IRP interactions have been widely studied by EMSA. Here, we describe the EMSA protocol for analyzing the IRE-binding activity of IRP1 and IRP2, which can be generalized to assess the activity of other RNA-binding proteins as well. A crude protein lysate containing an RNA-binding protein, or a purified preparation of this protein, is incubated with an excess of(32) P-labeled RNA probe, allowing for complex formation. Heparin is added to preclude non-specific protein to probe binding. Subsequently, the mixture is analyzed by non-denaturing electrophoresis on a polyacrylamide gel. The free probe migrates fast, while the RNA/protein complex exhibits retarded mobility; hence, the procedure is also called "gel retardation" or "bandshift" assay. After completion of the electrophoresis, the gel is dried and RNA/protein complexes, as well as free probe, are detected by autoradiography. The overall goal of the protocol is to detect and quantify IRE/IRP and other RNA/protein interactions. Moreover, EMSA can also be used to determine specificity, binding affinity, and stoichiometry of the RNA/protein interaction under investigation.
Collapse
Affiliation(s)
- Carine Fillebeen
- Lady Davis Institute for Medical Research, Jewish General Hospital; Department of Medicine, McGill University
| | - Nicole Wilkinson
- Lady Davis Institute for Medical Research, Jewish General Hospital; Department of Medicine, McGill University
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research, Jewish General Hospital; Department of Medicine, McGill University;
| |
Collapse
|
28
|
Chaturvedi R, Banerjee S, Chattopadhyay P, Bhattacharjee CR, Raul P, Borah K. High iron accumulation in hair and nail of people living in iron affected areas of Assam, India. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2014; 110:216-20. [PMID: 25261608 DOI: 10.1016/j.ecoenv.2014.08.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 08/21/2014] [Accepted: 08/23/2014] [Indexed: 05/12/2023]
Abstract
Human populace of Assam, India repeatedly exposed to high concentration of iron in groundwater results in adverse health effects like hemochromatosis, liver cirrhosis and siderosis. In the present study, human hair and nail analysis were carried out to establish a possible relationship between iron toxicity and its deposition among the residents. Nail and hair iron concentrations ranged from 28.2 to 1046μgg(-1) (n=114) and 26.5-838 (n=108)μgg(-1) levels, respectively, among all the study participants. The iron content of the groundwater (421-5340μgL(-1)) (n=64) used for drinking purposes was positively correlated with both nail (r=0.788, p<0.0001) and hair (r=0.709, p<0.0001) iron concentrations. Age- and sex-matched controls corresponding to each group were selected from population residing in other parts of the country where groundwater does not have excess iron. All the study groups included population drinking iron-contaminated water above the WHO/BIS limit (>300µgL(-1)) for 5 years (Group 1), for more than 5-10 years (Group 2) and for more than 10 years (Group 3). Results suggested that the participants consuming groundwater exceeding the WHO limit of iron had significantly more iron accumulation than those using groundwater containing ≤300μgL(-1) iron (p<0.01). There was statistically higher concentration of iron in the nail samples than the hair samples in all the study groups (p<0.01). There was a positive correlation in iron concentration and the residence time of the participants (p<0.01). Iron levels in the male participants were significantly higher than the female participants in the present study (p<0.01). The current findings are sufficiently compelling to warrant more extensive study of iron exposure through drinking water and adverse effects to the human in the areas where iron concentration is high.
Collapse
Affiliation(s)
- Richa Chaturvedi
- Department of Chemistry, Defence Research Laboratory, Tezpur-784001, Assam; Department of Chemistry, Assam University, Silchar-788001, Assam
| | - Saumen Banerjee
- Department of Chemistry, Defence Research Laboratory, Tezpur-784001, Assam.
| | | | | | - Prasanta Raul
- Department of Chemistry, Defence Research Laboratory, Tezpur-784001, Assam
| | - Kusum Borah
- Department of Chemistry, Defence Research Laboratory, Tezpur-784001, Assam
| |
Collapse
|
29
|
Janaway BM, Simpson JE, Hoggard N, Highley JR, Forster G, Drew D, Gebril OH, Matthews FE, Brayne C, Wharton SB, Ince PG. Brain haemosiderin in older people: pathological evidence for an ischaemic origin of magnetic resonance imaging (MRI) microbleeds. Neuropathol Appl Neurobiol 2014; 40:258-69. [PMID: 23678850 PMCID: PMC4282337 DOI: 10.1111/nan.12062] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 05/10/2013] [Indexed: 01/03/2023]
Abstract
Introduction Magnetic resonance imaging (MRI) cerebral microbleeds (CMB) arise from ferromagnetic haemosiderin iron assumed to derive from extravasation of erythrocytes. Light microscopy of ageing brain frequently reveals foci of haemosiderin from single crystalloids to larger, predominantly perivascular, aggregates. The pathological and radiological relationship between these findings is not resolved. Methods Haemosiderin deposition and vascular pathology in the putamen were quantified in 200 brains donated to the population-representative Medical Research Council Cognitive Function and Ageing Study. Molecular markers of gliosis and tissue integrity were assessed by immunohistochemistry in brains with highest (n = 20) and lowest (n = 20) levels of putamen haemosiderin. The association between haemosiderin counts and degenerative and vascular brain pathology, clinical data, and the haemochromatosis (HFE) gene H63D genotype were analysed. The frequency of MRI CMB in 10 cases with highest and lowest burden of putamen haemosiderin, was compared using post mortem 3T MRI. Results Greater putamen haemosiderin was significantly associated with putaminal indices of small vessel ischaemia (microinfarcts, P < 0.05; arteriolosclerosis, P < 0.05; perivascular attenuation, P < 0.001) and with lacunes in any brain region (P < 0.023) but not large vessel disease, or whole brain measures of neurodegenerative pathology. Higher levels of putamen haemosiderin correlated with more CMB (P < 0.003). Conclusions The MRI-CMB concept should take account of brain iron homeostasis, and small vessel ischaemic change in later life, rather than only as a marker for minor episodes of cerebrovascular extravasation. These data are of clinical relevance, suggesting that basal ganglia MRI microbleeds may be a surrogate for ischaemic small vessel disease rather than exclusively a haemorrhagic diathesis.
Collapse
Affiliation(s)
- B M Janaway
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Goswami D, Machini MT, Silvestre DM, Nomura CS, Esposito BP. Cell penetrating peptide (CPP)-conjugated desferrioxamine for enhanced neuroprotection: synthesis and in vitro evaluation. Bioconjug Chem 2014; 25:2067-80. [PMID: 25299707 DOI: 10.1021/bc5004197] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Iron overload causes progressive and sometimes irreversible damage due to accelerated production of reactive oxygen species. Desferrioxamine (DFO), a siderophore, has been used clinically to remove excess iron. However, the applications of DFO are limited because of its inability to access intracellular labile iron. Cell penetrating peptides (CPPs) have become an efficient delivery vector for the enhanced internalization of drugs into the cytosol. We describe, herein, an efficient method for covalently conjugating DFO to the CPPs TAT(47-57) and Penetratin. Both conjugates suppressed the redox activity of labile plasma iron in buffered solutions and in iron-overloaded sera. Enhanced access to intracellular labile iron compared to the parent siderophore was achieved in HeLa and RBE4 (a model of blood-brain-barrier) cell lines. Iron complexes of both conjugates also had better permeability in both cell models. DFO antioxidant and iron binding properties were preserved and its bioavailability was increased upon CPP conjugation, which opens new therapeutic possibilities for neurodegenerative processes associated with brain iron overload.
Collapse
Affiliation(s)
- Dibakar Goswami
- Departamentos de Química Fundamental e de ‡Bioquímica, Instituto de Química, Universidade de São Paulo , Av. Lineu Prestes 748, 05508-000, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
31
|
Wilkinson N, Pantopoulos K. The IRP/IRE system in vivo: insights from mouse models. Front Pharmacol 2014; 5:176. [PMID: 25120486 PMCID: PMC4112806 DOI: 10.3389/fphar.2014.00176] [Citation(s) in RCA: 245] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 07/07/2014] [Indexed: 12/16/2022] Open
Abstract
Iron regulatory proteins 1 and 2 (IRP1 and IRP2) post-transcriptionally control the expression of several mRNAs encoding proteins of iron, oxygen and energy metabolism. The mechanism involves their binding to iron responsive elements (IREs) in the untranslated regions of target mRNAs, thereby controlling mRNA translation or stability. Whereas IRP2 functions solely as an RNA-binding protein, IRP1 operates as either an RNA-binding protein or a cytosolic aconitase. Early experiments in cultured cells established a crucial role of IRPs in regulation of cellular iron metabolism. More recently, studies in mouse models with global or localized Irp1 and/or Irp2 deficiencies uncovered new physiological functions of IRPs in the context of systemic iron homeostasis. Thus, IRP1 emerged as a key regulator of erythropoiesis and iron absorption by controlling hypoxia inducible factor 2α (HIF2α) mRNA translation, while IRP2 appears to dominate the control of iron uptake and heme biosynthesis in erythroid progenitor cells by regulating the expression of transferrin receptor 1 (TfR1) and 5-aminolevulinic acid synthase 2 (ALAS2) mRNAs, respectively. Targeted disruption of either Irp1 or Irp2 in mice is associated with distinct phenotypic abnormalities. Thus, Irp1(-/-) mice develop polycythemia and pulmonary hypertension, while Irp2(-/-) mice present with microcytic anemia, iron overload in the intestine and the liver, and neurologic defects. Combined disruption of both Irp1 and Irp2 is incombatible with life and leads to early embryonic lethality. Mice with intestinal- or liver-specific disruption of both Irps are viable at birth but die later on due to malabsorption or liver failure, respectively. Adult mice lacking both Irps in the intestine exhibit a profound defect in dietary iron absorption due to a "mucosal block" that is caused by the de-repression of ferritin mRNA translation. Herein, we discuss the physiological function of the IRE/IRP regulatory system.
Collapse
Affiliation(s)
- Nicole Wilkinson
- Lady Davis Institute for Medical Research, Jewish General Hospital, and Department of Medicine, McGill University Montreal, QC, Canada
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research, Jewish General Hospital, and Department of Medicine, McGill University Montreal, QC, Canada
| |
Collapse
|
32
|
Gkouvatsos K, Fillebeen C, Daba A, Wagner J, Sebastiani G, Pantopoulos K. Iron-dependent regulation of hepcidin in Hjv-/- mice: evidence that hemojuvelin is dispensable for sensing body iron levels. PLoS One 2014; 9:e85530. [PMID: 24409331 PMCID: PMC3883712 DOI: 10.1371/journal.pone.0085530] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 11/27/2013] [Indexed: 12/20/2022] Open
Abstract
Hemojuvelin (Hjv) is a bone morphogenetic protein (BMP) co-receptor involved in the control of systemic iron homeostasis. Functional inactivation of Hjv leads to severe iron overload in humans and mice due to marked suppression of the iron-regulatory hormone hepcidin. To investigate the role of Hjv in body iron sensing, Hjv−/− mice and isogenic wild type controls were placed on a moderately low, a standard or a high iron diet for four weeks. Hjv−/− mice developed systemic iron overload under all regimens. Transferrin (Tf) was highly saturated regardless of the dietary iron content, while liver iron deposition was proportional to it. Hepcidin mRNA expression responded to fluctuations in dietary iron intake, despite the absence of Hjv. Nevertheless, iron-dependent upregulation of hepcidin was more than an order of magnitude lower compared to that seen in wild type controls. Likewise, iron signaling via the BMP/Smad pathway was preserved but substantially attenuated. These findings suggest that Hjv is not required for sensing of body iron levels and merely functions as an enhancer for iron signaling to hepcidin.
Collapse
Affiliation(s)
- Konstantinos Gkouvatsos
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Carine Fillebeen
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Alina Daba
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - John Wagner
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Giada Sebastiani
- Division of Gastroenterology, Royal Victoria Hospital, McGill University Health Center, Montreal, Quebec, Canada ; Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada ; Department of Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
33
|
Fillebeen C, Pantopoulos K. Hepatitis C virus infection causes iron deficiency in Huh7.5.1 cells. PLoS One 2013; 8:e83307. [PMID: 24349485 PMCID: PMC3862679 DOI: 10.1371/journal.pone.0083307] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 11/05/2013] [Indexed: 02/04/2023] Open
Abstract
Patients with chronic hepatitis C virus (HCV) infection frequently develop systemic iron overload, which exacerbates morbidity. Nevertheless, iron inhibits HCV replication in cell culture models and thereby exerts antiviral activity. We hypothesized that the cellular iron status is crucial for the establishment of HCV infection. We show that HCV infection of permissive Huh7.5.1 hepatoma cells promotes an iron deficient phenotype. Thus, HCV leads to increased iron regulatory protein (IRP) activity, accumulation of IRP2 and suppression of transferrin receptor 1 (TfR1) and divalent metal transporter 1 (DMT1) in the host. These data suggest that HCV regulates cellular iron levels to bypass iron-mediated inhibition in viral replication.
Collapse
Affiliation(s)
- Carine Fillebeen
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
34
|
Koskenkorva-Frank TS, Weiss G, Koppenol WH, Burckhardt S. The complex interplay of iron metabolism, reactive oxygen species, and reactive nitrogen species: insights into the potential of various iron therapies to induce oxidative and nitrosative stress. Free Radic Biol Med 2013; 65:1174-1194. [PMID: 24036104 DOI: 10.1016/j.freeradbiomed.2013.09.001] [Citation(s) in RCA: 301] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 09/05/2013] [Accepted: 09/05/2013] [Indexed: 02/07/2023]
Abstract
Production of minute concentrations of superoxide (O2(*-)) and nitrogen monoxide (nitric oxide, NO*) plays important roles in several aspects of cellular signaling and metabolic regulation. However, in an inflammatory environment, the concentrations of these radicals can drastically increase and the antioxidant defenses may become overwhelmed. Thus, biological damage may occur owing to redox imbalance-a condition called oxidative and/or nitrosative stress. A complex interplay exists between iron metabolism, O2(*-), hydrogen peroxide (H2O2), and NO*. Iron is involved in both the formation and the scavenging of these species. Iron deficiency (anemia) (ID(A)) is associated with oxidative stress, but its role in the induction of nitrosative stress is largely unclear. Moreover, oral as well as intravenous (iv) iron preparations used for the treatment of ID(A) may also induce oxidative and/or nitrosative stress. Oral administration of ferrous salts may lead to high transferrin saturation levels and, thus, formation of non-transferrin-bound iron, a potentially toxic form of iron with a propensity to induce oxidative stress. One of the factors that determine the likelihood of oxidative and nitrosative stress induced upon administration of an iv iron complex is the amount of labile (or weakly-bound) iron present in the complex. Stable dextran-based iron complexes used for iv therapy, although they contain only negligible amounts of labile iron, can induce oxidative and/or nitrosative stress through so far unknown mechanisms. In this review, after summarizing the main features of iron metabolism and its complex interplay with O2(*-), H2O2, NO*, and other more reactive compounds derived from these species, the potential of various iron therapies to induce oxidative and nitrosative stress is discussed and possible underlying mechanisms are proposed. Understanding the mechanisms, by which various iron formulations may induce oxidative and nitrosative stress, will help us develop better tolerated and more efficient therapies for various dysfunctions of iron metabolism.
Collapse
Affiliation(s)
- Taija S Koskenkorva-Frank
- Chemical and Preclinical Research and Development, Vifor (International) Ltd., CH-9001 St. Gallen, Switzerland
| | - Günter Weiss
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Willem H Koppenol
- Institute of Inorganic Chemistry, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Susanna Burckhardt
- Chemical and Preclinical Research and Development, Vifor (International) Ltd., CH-9001 St. Gallen, Switzerland; Institute of Inorganic Chemistry, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
35
|
Non-Alcoholic Fatty Liver Disease: Diagnosis and Evaluation of Disease Severity. ACTA ACUST UNITED AC 2013. [DOI: 10.5812/thrita.11795] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
36
|
Ugarte M, Osborne NN, Brown LA, Bishop PN. Iron, zinc, and copper in retinal physiology and disease. Surv Ophthalmol 2013; 58:585-609. [DOI: 10.1016/j.survophthal.2012.12.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 12/09/2012] [Accepted: 12/11/2012] [Indexed: 12/26/2022]
|
37
|
Abstract
Iron is a redox active metal which is abundant in the Earth's crust. It has played a key role in the evolution of living systems and as such is an essential element in a wide range of biological phenomena, being critical for the function of an enormous array of enzymes, energy transduction mechanisms, and oxygen carriers. The redox nature of iron renders the metal toxic in excess and consequently all biological organisms carefully control iron levels. In this overview the mechanisms adopted by man to control body iron levels are described.Low body iron levels are related to anemia which can be treated by various forms of iron fortification and supplementation. Elevated iron levels can occur systemically or locally, each giving rise to specific symptoms. Systemic iron overload results from either the hyperabsorption of iron or regular blood transfusion and can be treated by the use of a selection of iron chelating molecules. The symptoms of many forms of neurodegeneration are associated with elevated levels of iron in certain regions of the brain and iron chelation therapy is beginning to find an application in the treatment of such diseases. Iron chelators have also been widely investigated for the treatment of cancer, tuberculosis, and malaria. In these latter studies, selective removal of iron from key enzymes or iron binding proteins is sought. Sufficient selectivity between the invading organism and the host has yet to be established for such chelators to find application in the clinic.Iron chelation for systemic iron overload and iron supplementation therapy for the treatment of various forms of anemia are now established procedures in clinical medicine. Chelation therapy may find an important role in the treatment of various neurodegenerative diseases in the near future.
Collapse
|
38
|
Wysokinski D, Danisz K, Blasiak J, Dorecka M, Romaniuk D, Szaflik J, Szaflik JP. An association of transferrin gene polymorphism and serum transferrin levels with age-related macular degeneration. Exp Eye Res 2013; 106:14-23. [DOI: 10.1016/j.exer.2012.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Revised: 09/18/2012] [Accepted: 10/08/2012] [Indexed: 12/16/2022]
|
39
|
Farina M, Avila DS, da Rocha JBT, Aschner M. Metals, oxidative stress and neurodegeneration: a focus on iron, manganese and mercury. Neurochem Int 2012; 62:575-94. [PMID: 23266600 DOI: 10.1016/j.neuint.2012.12.006] [Citation(s) in RCA: 357] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Revised: 12/07/2012] [Accepted: 12/10/2012] [Indexed: 02/08/2023]
Abstract
Essential metals are crucial for the maintenance of cell homeostasis. Among the 23 elements that have known physiological functions in humans, 12 are metals, including iron (Fe) and manganese (Mn). Nevertheless, excessive exposure to these metals may lead to pathological conditions, including neurodegeneration. Similarly, exposure to metals that do not have known biological functions, such as mercury (Hg), also present great health concerns. This review focuses on the neurodegenerative mechanisms and effects of Fe, Mn and Hg. Oxidative stress (OS), particularly in mitochondria, is a common feature of Fe, Mn and Hg toxicity. However, the primary molecular targets triggering OS are distinct. Free cationic iron is a potent pro-oxidant and can initiate a set of reactions that form extremely reactive products, such as OH. Mn can oxidize dopamine (DA), generating reactive species and also affect mitochondrial function, leading to accumulation of metabolites and culminating with OS. Cationic Hg forms have strong affinity for nucleophiles, such as -SH and -SeH. Therefore, they target critical thiol- and selenol-molecules with antioxidant properties. Finally, we address the main sources of exposure to these metals, their transport mechanisms into the brain, and therapeutic modalities to mitigate their neurotoxic effects.
Collapse
Affiliation(s)
- Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, SC, Brazil
| | | | | | | |
Collapse
|
40
|
MRI Assessment of Hepatic Iron Clearance Rates After USPIO Administration in Healthy Adults. Invest Radiol 2012; 47:717-24. [DOI: 10.1097/rli.0b013e31826dc151] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
41
|
Daba A, Gkouvatsos K, Sebastiani G, Pantopoulos K. Differences in activation of mouse hepcidin by dietary iron and parenterally administered iron dextran: compartmentalization is critical for iron sensing. J Mol Med (Berl) 2012; 91:95-102. [DOI: 10.1007/s00109-012-0937-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 07/06/2012] [Accepted: 07/18/2012] [Indexed: 10/28/2022]
|
42
|
Pantopoulos K, Porwal SK, Tartakoff A, Devireddy L. Mechanisms of mammalian iron homeostasis. Biochemistry 2012; 51:5705-24. [PMID: 22703180 DOI: 10.1021/bi300752r] [Citation(s) in RCA: 419] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Iron is vital for almost all organisms because of its ability to donate and accept electrons with relative ease. It serves as a cofactor for many proteins and enzymes necessary for oxygen and energy metabolism, as well as for several other essential processes. Mammalian cells utilize multiple mechanisms to acquire iron. Disruption of iron homeostasis is associated with various human diseases: iron deficiency resulting from defects in the acquisition or distribution of the metal causes anemia, whereas iron surfeit resulting from excessive iron absorption or defective utilization causes abnormal tissue iron deposition, leading to oxidative damage. Mammals utilize distinct mechanisms to regulate iron homeostasis at the systemic and cellular levels. These involve the hormone hepcidin and iron regulatory proteins, which collectively ensure iron balance. This review outlines recent advances in iron regulatory pathways as well as in mechanisms underlying intracellular iron trafficking, an important but less studied area of mammalian iron homeostasis.
Collapse
Affiliation(s)
- Kostas Pantopoulos
- Lady Davis Institute for Medical Research, Jewish General Hospital and Department of Medicine, McGill University, Montreal, QC, Canada
| | | | | | | |
Collapse
|
43
|
Yoon HH, Huh SJ, Lee JH, Lee S, Kim SH, Kwon HC, Kim HJ. Should we still use Camitta's criteria for severe aplastic anemia? THE KOREAN JOURNAL OF HEMATOLOGY 2012; 47:126-30. [PMID: 22783359 PMCID: PMC3389061 DOI: 10.5045/kjh.2012.47.2.126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 05/17/2012] [Accepted: 05/18/2012] [Indexed: 12/16/2022]
Abstract
Background The criteria by Camitta for diagnosis in severe aplastic anemia (SAA) has been used since 1976. However, there has been no attempt to verify the Camitta's criteria, that the survival in patients with SAA may differ by absolute neutrophil count (ANC), platelet count (PLT), and corrected reticulocyte count (CRC), which are components of the Camitta's criteria. Methods 117 SAA patients diagnosed by the Camitta's criteria were analyzed, retrospectively. Univariate and multivariate analyses were used to evaluate the factors affecting overall survival (OS). Results Response by immunosuppressive therapy (IST) or stem cell transplantation (SCT) significantly affected OS (P=0.001). Therefore, we excluded treatment responders for analysis. Finally, 92 SAA patients including treatment non-responders by IST or SCT and conservative care group were analyzed by using univariate and multivariate analyses. The median age of analyzed patients was 54.5 years. Male to female ratio was 1:1. The median follow-up duration was 74.23 months (range, 54.71-93.74 months). The median ANC, PLT, and CRC were 394/µL, 12,000/µL, and 0.39%, respectively. In multivariate analyses, ANC <500/µL or ≥500/µL (P=0.015, HR 2.694, 95% CI: 1.20-6.01) and age (P=0.015, HR 1.022, 95% CI: 1.00-1.04) were the significant factors for OS. Conclusion ANC could be an essential, not an optional criterion for diagnosing SAA. This study suggests the possibility that the Camitta's criteria be modified. Studies in large cohorts are needed to transform the Camitta's criteria.
Collapse
Affiliation(s)
- Hyun Hwa Yoon
- Department of Internal Medicine, Dong-A University College of Medicine, Busan, Korea
| | | | | | | | | | | | | |
Collapse
|
44
|
Regulation of iron transport and the role of transferrin. Biochim Biophys Acta Gen Subj 2012; 1820:188-202. [DOI: 10.1016/j.bbagen.2011.10.013] [Citation(s) in RCA: 303] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2011] [Revised: 10/27/2011] [Accepted: 10/30/2011] [Indexed: 12/15/2022]
|
45
|
Nanaki SG, Pantopoulos K, Bikiaris DN. Synthesis of biocompatible poly(ɛ-caprolactone)- block-poly(propylene adipate) copolymers appropriate for drug nanoencapsulation in the form of core-shell nanoparticles. Int J Nanomedicine 2011; 6:2981-95. [PMID: 22162656 PMCID: PMC3230566 DOI: 10.2147/ijn.s26568] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Poly(propylene adipate)-block-poly(ɛ-caprolactone) copolymers were synthesized using a combination of polycondensation and ring-opening polymerization of ɛ-caprolactone in the presence of poly(propylene adipate). Gel permeation chromatography was used for molecular weight determination, whereas hydrogen-1 nuclear magnetic resonance and carbon-13 nuclear magnetic resonance spectroscopy were employed for copolymer characterization and composition evaluation. The copolymers were found to be block while their composition was similar to the feeding ratio. They formed semicrystalline structures, while only poly(ɛ-caprolactone) formed crystals, as shown by wide angle X-ray diffraction. Differential scanning calorimetry data suggest that the melting point and heat of fusion of copolymers decreased by increasing the poly(propylene adipate) amount. The synthesized polymers exhibited low cytotoxicity and were used to encapsulate desferrioxamine, an iron-chelating drug. The desferrioxamine nanoparticles were self-assembled into core shell structures, had mean particle size <250 nm, and the drug remained in crystalline form. Further studies revealed that the dissolution rate was mainly related to the melting temperature, as well as to the degree of crystallinity of copolymers.
Collapse
Affiliation(s)
- Stavroula G Nanaki
- Laboratory of Polymer Chemistry and Technology, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | | | | |
Collapse
|