201
|
Sheokand N, Malhotra H, Chauhan AS, Kumar M, Chaudhary S, Patidar A, Boradia VM, Raje CI, Raje M. Reverse overshot water-wheel retroendocytosis of apotransferrin extrudes cellular iron. J Cell Sci 2016; 129:843-53. [PMID: 26743084 DOI: 10.1242/jcs.180356] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 12/29/2015] [Indexed: 01/17/2023] Open
Abstract
Iron (Fe), a vital micronutrient for all organisms, must be managed judiciously because both deficiency or excess can trigger severe pathology. Although cellular Fe import is well understood, its export is thought to be limited to transmembrane extrusion through ferroportin (also known as Slc40a1), the only known mammalian Fe exporter. Utilizing primary cells and cell lines (including those with no discernible expression of ferroportin on their surface), we demonstrate that upon Fe loading, the multifunctional enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH), which is recruited to the cell surface, 'treadmills' apotransferrin in and out of the cell. Kinetic analysis utilizing labeled ligand, GAPDH-knockdown cells, (55)Fe-labeled cells and pharmacological inhibitors of endocytosis confirmed GAPDH-dependent apotransferrin internalization as a prerequisite for cellular Fe export. These studies define an unusual rapid recycling process of retroendocytosis for cellular Fe extrusion, a process mirroring receptor mediated internalization that has never before been considered for maintenance of cellular cationic homeostasis. Modulation of this unusual pathway could provide insights for management of Fe overload disorders.
Collapse
Affiliation(s)
- Navdeep Sheokand
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh 160036, India
| | - Himanshu Malhotra
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh 160036, India
| | - Anoop Singh Chauhan
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh 160036, India
| | - Manoj Kumar
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh 160036, India
| | - Surbhi Chaudhary
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh 160036, India
| | - Anil Patidar
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh 160036, India
| | - Vishant Mahendra Boradia
- National Institute of Pharmaceutical Education & Research, Phase X, Sector 67, SAS Nagar, Punjab 160062, India
| | - Chaaya Iyengar Raje
- National Institute of Pharmaceutical Education & Research, Phase X, Sector 67, SAS Nagar, Punjab 160062, India
| | - Manoj Raje
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh 160036, India
| |
Collapse
|
202
|
Skerratt SE, Humphreys S, Ferreira R, Jorgensen C, Warmus J, Zhao L, Tong X, Nickolls SA. Identification of a novel BODIPY minihepcidin tool for the high content analysis of ferroportin (SLC40A1) pharmacology. MEDCHEMCOMM 2016. [DOI: 10.1039/c6md00260a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Herein, we describe the design and synthesis of a novel BODIPY-labelled minihepcidin peptide to enable the high content analysis of ferroportin (SLC40A1) pharmacology.
Collapse
Affiliation(s)
| | | | | | | | - Joe Warmus
- Pfizer Worldwide Research & Development
- Groton
- USA
| | | | | | | |
Collapse
|
203
|
Expression of Iron-Related Proteins at the Neurovascular Unit Supports Reduction and Reoxidation of Iron for Transport Through the Blood-Brain Barrier. Mol Neurobiol 2015; 53:7237-7253. [DOI: 10.1007/s12035-015-9582-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/29/2015] [Indexed: 11/25/2022]
|
204
|
Dysregulation of ferroportin gene expression in β0-thalassemia/Hb E disease. Ann Hematol 2015; 95:387-96. [DOI: 10.1007/s00277-015-2572-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 12/02/2015] [Indexed: 12/13/2022]
|
205
|
Bonaccorsi di Patti MC, Polticelli F, Tortosa V, Furbetta PA, Musci G. A bacterial homologue of the human iron exporter ferroportin. FEBS Lett 2015; 589:3829-35. [PMID: 26608034 DOI: 10.1016/j.febslet.2015.11.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/13/2015] [Accepted: 11/16/2015] [Indexed: 10/22/2022]
Abstract
A bacterial homologue of the human iron exporter ferroportin found in the predatory Gram-negative bacterium Bdellovibrio bacteriovorus has been investigated. Molecular modelling, expression in recombinant form and iron binding and transport assays demonstrate that B. bacteriovorus ferroportin (bdFpn) is indeed an orthologue of human ferroportin. Key residues corresponding to those essential for iron binding and transport in human ferroportin are conserved in the bacterial homologue and are predicted to be correctly clustered in the central cavity of the protein. Mutation of these residues grossly affects the iron binding and transport ability of bdFpn.
Collapse
Affiliation(s)
| | - Fabio Polticelli
- Department of Sciences, Roma Tre University, Rome, Italy; National Institute of Nuclear Physics, Roma Tre Section, Rome, Italy.
| | | | | | - Giovanni Musci
- Department of Biosciences and Territory, University of Molise, Pesche, Italy.
| |
Collapse
|
206
|
Shanmugam NKN, Chen K, Cherayil BJ. Commensal Bacteria-induced Interleukin 1β (IL-1β) Secreted by Macrophages Up-regulates Hepcidin Expression in Hepatocytes by Activating the Bone Morphogenetic Protein Signaling Pathway. J Biol Chem 2015; 290:30637-47. [PMID: 26515063 DOI: 10.1074/jbc.m115.689190] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Indexed: 12/19/2022] Open
Abstract
The liver hormone hepcidin is the central regulator of systemic iron metabolism. Its increased expression in inflammatory states leads to hypoferremia and anemia. Elucidation of the mechanisms that up-regulate hepcidin during inflammation is essential for developing rational therapies for this anemia. Using mouse models of inflammatory bowel disease, we have shown previously that colitis-associated hepcidin induction is influenced by intestinal microbiota composition. Here we investigate how two commensal bacteria, Bifidobacterium longum and Bacteroides fragilis, representative members of the gut microbiota, affect hepcidin expression. We found that supernatants of a human macrophage cell line infected with either of the bacteria up-regulated hepcidin when added to a human hepatocyte cell line. This activity was abrogated by neutralization of IL-1β. Moreover, purified IL-1β increased hepcidin expression when added to the hepatocyte line or primary human hepatocytes and when injected into mice. IL-1β activated the bone morphogenetic protein (BMP) signaling pathway in hepatocytes and in mouse liver, as indicated by increased phosphorylation of small mothers against decapentaplegic proteins. Activation of BMP signaling correlated with IL-1β-induced expression of BMP2 in human hepatocytes and activin B in mouse liver. Treatment of hepatocytes with two different chemical inhibitors of BMP signaling or with a neutralizing antibody to BMP2 prevented IL-1β-induced up-regulation of hepcidin. Our results clarify how commensal bacteria affect hepcidin expression and reveal a novel connection between IL-1β and activation of BMP signaling. They also suggest that there may be differences between mice and humans with respect to the mechanism by which IL-1β up-regulates hepcidin.
Collapse
Affiliation(s)
- Nanda Kumar N Shanmugam
- From the Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129
| | - Kejie Chen
- From the Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129
| | - Bobby J Cherayil
- From the Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129
| |
Collapse
|
207
|
Giorgi G, D'Anna MC, Roque ME. Iron homeostasis and its disruption in mouse lung in iron deficiency and overload. Exp Physiol 2015; 100:1199-216. [DOI: 10.1113/ep085166] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 05/29/2015] [Indexed: 12/20/2022]
Affiliation(s)
- Gisela Giorgi
- Laboratory of Human Physiology; Department of Biology, Biochemistry and Pharmacy, Instituto de Investigaciones Biológicas y Biomédicas del Sur (INBIOSUR-CONICET); San Juan 670, Universidad Nacional del Sur; Bahía Blanca Argentina
| | - María Cecilia D'Anna
- Laboratory of Human Physiology; Department of Biology, Biochemistry and Pharmacy, Instituto de Investigaciones Biológicas y Biomédicas del Sur (INBIOSUR-CONICET); San Juan 670, Universidad Nacional del Sur; Bahía Blanca Argentina
| | - Marta Elena Roque
- Laboratory of Human Physiology; Department of Biology, Biochemistry and Pharmacy, Instituto de Investigaciones Biológicas y Biomédicas del Sur (INBIOSUR-CONICET); San Juan 670, Universidad Nacional del Sur; Bahía Blanca Argentina
| |
Collapse
|
208
|
Tripathi AK, Haldar S, Qian J, Beserra A, Suda S, Singh A, Hopfer U, Chen SG, Garrick MD, Turner JR, Knutson MD, Singh N. Prion protein functions as a ferrireductase partner for ZIP14 and DMT1. Free Radic Biol Med 2015; 84:322-330. [PMID: 25862412 PMCID: PMC4476631 DOI: 10.1016/j.freeradbiomed.2015.03.037] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 03/30/2015] [Accepted: 03/30/2015] [Indexed: 12/21/2022]
Abstract
Excess circulating iron is stored in the liver, and requires reduction of non-Tf-bound iron (NTBI) and transferrin (Tf) iron at the plasma membrane and endosomes, respectively, by ferrireductase (FR) proteins for transport across biological membranes through divalent metal transporters. Here, we report that prion protein (PrP(C)), a ubiquitously expressed glycoprotein most abundant on neuronal cells, functions as a FR partner for divalent-metal transporter-1 (DMT1) and ZIP14. Thus, absence of PrP(C) in PrP-knock-out (PrP(-/-)) mice resulted in markedly reduced liver iron stores, a deficiency that was not corrected by chronic or acute administration of iron by the oral or intraperitoneal routes. Likewise, preferential radiolabeling of circulating NTBI with (59)Fe revealed significantly reduced uptake and storage of NTBI by the liver of PrP(-/-) mice relative to matched PrP(+/+) controls. However, uptake, storage, and utilization of ferritin-bound iron that does not require reduction for uptake were increased in PrP(-/-) mice, indicating a compensatory response to the iron deficiency. Expression of exogenous PrP(C) in HepG2 cells increased uptake and storage of ferric iron (Fe(3+)), not ferrous iron (Fe(2+)), from the medium, supporting the function of PrP(C) as a plasma membrane FR. Coexpression of PrP(C) with ZIP14 and DMT1 in HepG2 cells increased uptake of Fe(3+) significantly, and surprisingly, increased the ratio of N-terminally truncated PrP(C) forms lacking the FR domain relative to full-length PrP(C). Together, these observations indicate that PrP(C) promotes, and possibly regulates, the uptake of NTBI through DMT1 and Zip14 via its FR activity. Implications of these observations for neuronal iron homeostasis under physiological and pathological conditions are discussed.
Collapse
Affiliation(s)
- Ajai K. Tripathi
- Department of Pathology, Case Western Reserve University/University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | - Swati Haldar
- Department of Pathology, Case Western Reserve University/University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | - Juan Qian
- Department of Pathology, Case Western Reserve University/University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | - Amber Beserra
- School of Arts and Sciences, Case Western Reserve University/University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | - Srinivas Suda
- Department of Pathology, Case Western Reserve University/University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | - Ajay Singh
- Department of Pathology, Case Western Reserve University/University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | - Ulrich Hopfer
- Department of physiology and Biophysics, Case Western Reserve University/University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | - Shu G. Chen
- Department of Pathology, Case Western Reserve University/University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | | | | | | | - Neena Singh
- Department of Pathology, Case Western Reserve University/University Hospitals Case Medical Center, Cleveland, Ohio, USA
- To whom correspondence should be addressed: Neena Singh, Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, Ohio 44106, USA. Tel: 216-368-2617;
| |
Collapse
|
209
|
Nikonorov AA, Skalnaya MG, Tinkov AA, Skalny AV. Mutual interaction between iron homeostasis and obesity pathogenesis. J Trace Elem Med Biol 2015; 30:207-14. [PMID: 24916791 DOI: 10.1016/j.jtemb.2014.05.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 04/15/2014] [Accepted: 05/14/2014] [Indexed: 02/08/2023]
Abstract
Obesity is identified as an important medical problem. One of the pathologic conditions observed in obesity is systemic iron deficiency and hypoferremia. Along with a large number of studies indicating disturbed iron homeostasis in obesity, recent data indicate a cause-effect relationship between iron status and obesity-related pathologies. The primary objective of the article is to consider two aspects of the iron-obesity interplay: (1) the mechanisms leading to impaired iron balance, and (2) the pathways of iron participation in obesity-related pathogenesis. While considering disturbance of iron homeostasis in obesity, a number of potential mechanisms of hypoferremia are proposed. At the same time, the inflammation of obesity and obesity-related hepcidin and lipocalin 2 hyperproduction seem to be the most probable reasons of obesity-related hypoferremia. Oversecretion of these proteins leads to iron sequestration in reticuloendothelial system cells. The latter also leads to increased adipose tissue iron content, thus producing preconditions for adverse effects of local iron overload. Being a redox-active metal, iron is capable of inducing oxidative stress as well as endoplasmic reticulum stress, inflammation and adipose tissue endocrine dysfunction. Iron-mediated mechanisms of toxicity may influence aspects of obesity pathogenesis possibly even leading to obesity aggravation. Thus, a mutual interaction between disturbance in iron homeostasis and obesity pathogenesis is proposed. All sides of this interaction should be considered to design new therapeutic approaches to the treatment of disturbed iron homeostasis in obesity.
Collapse
Affiliation(s)
- Alexandr A Nikonorov
- Department of Biochemistry, Orenburg State Medical Academy, Sovetskaya Street 6, Orenburg 460000, Russia.
| | - Margarita G Skalnaya
- Russian Society of Trace Elements in Medicine, Zemlyanoy Val Street 46, Moscow 105064, Russia
| | - Alexey A Tinkov
- Department of Biochemistry, Orenburg State Medical Academy, Sovetskaya Street 6, Orenburg 460000, Russia
| | - Anatoly V Skalny
- Russian Society of Trace Elements in Medicine, Zemlyanoy Val Street 46, Moscow 105064, Russia; Institute of Bioelementology (Russian Satellite Centre of Trace Element - Institute for UNESCO), Orenburg State University, Pobedy Avenue 13, Orenburg 460352, Russia
| |
Collapse
|
210
|
Duodenal cytochrome b (DCYTB) in iron metabolism: an update on function and regulation. Nutrients 2015; 7:2274-96. [PMID: 25835049 PMCID: PMC4425144 DOI: 10.3390/nu7042274] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 03/03/2015] [Accepted: 03/05/2015] [Indexed: 01/01/2023] Open
Abstract
Iron and ascorbate are vital cellular constituents in mammalian systems. The bulk-requirement for iron is during erythropoiesis leading to the generation of hemoglobin-containing erythrocytes. Additionally, both iron and ascorbate are required as co-factors in numerous metabolic reactions. Iron homeostasis is controlled at the level of uptake, rather than excretion. Accumulating evidence strongly suggests that in addition to the known ability of dietary ascorbate to enhance non-heme iron absorption in the gut, ascorbate regulates iron homeostasis. The involvement of ascorbate in dietary iron absorption extends beyond the direct chemical reduction of non-heme iron by dietary ascorbate. Among other activities, intra-enterocyte ascorbate appears to be involved in the provision of electrons to a family of trans-membrane redox enzymes, namely those of the cytochrome b561 class. These hemoproteins oxidize a pool of ascorbate on one side of the membrane in order to reduce an electron acceptor (e.g., non-heme iron) on the opposite side of the membrane. One member of this family, duodenal cytochrome b (DCYTB), may play an important role in ascorbate-dependent reduction of non-heme iron in the gut prior to uptake by ferrous-iron transporters. This review discusses the emerging relationship between cellular iron homeostasis, the emergent “IRP1-HIF2α axis”, DCYTB and ascorbate in relation to iron metabolism.
Collapse
|
211
|
Gammella E, Buratti P, Cairo G, Recalcati S. Macrophages: central regulators of iron balance. Metallomics 2015; 6:1336-45. [PMID: 24905850 DOI: 10.1039/c4mt00104d] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Macrophages are important to immune function and also actively participate in iron homeostasis. The involvement of splenic and liver macrophages in the processing of effete erythrocytes and the subsequent return of iron to the circulation is well established, and the molecular details of iron recycling have been characterized recently. Another important aspect regarding iron handling by macrophages is their capacity to act as immune cells, which involves the inflammatory response, as well as other pathological conditions in which macrophages are central. This review discusses the latest advances in macrophage iron trafficking and the pathophysiological consequences of altered iron homeostasis in these cells.
Collapse
Affiliation(s)
- Elena Gammella
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milano, Italy.
| | | | | | | |
Collapse
|
212
|
Dubick MA, Barr JL, Keen CL, Atkins JL. Ceruloplasmin and Hypoferremia: Studies in Burn and Non-Burn Trauma Patients. Antioxidants (Basel) 2015; 4:153-69. [PMID: 26785343 PMCID: PMC4665565 DOI: 10.3390/antiox4010153] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 02/25/2015] [Accepted: 02/28/2015] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE Normal iron handling appears to be disrupted in critically ill patients leading to hypoferremia that may contribute to systemic inflammation. Ceruloplasmin (Cp), an acute phase reactant protein that can convert ferrous iron to its less reactive ferric form facilitating binding to ferritin, has ferroxidase activity that is important to iron handling. Genetic absence of Cp decreases iron export resulting in iron accumulation in many organs. The objective of this study was to characterize iron metabolism and Cp activity in burn and non-burn trauma patients to determine if changes in Cp activity are a potential contributor to the observed hypoferremia. MATERIAL AND METHODS Under Brooke Army Medical Center Institutional Review Board approved protocols, serum or plasma was collected from burn and non-burn trauma patients on admission to the ICU and at times up to 14 days and measured for indices of iron status, Cp protein and oxidase activity and cytokines. RESULTS Burn patients showed evidence of anemia and normal or elevated ferritin levels. Plasma Cp oxidase activity in burn and trauma patients were markedly lower than controls on admission and increased to control levels by day 3, particularly in burn patients. Plasma cytokines were elevated throughout the 14 days study along with evidence of an oxidative stress. No significant differences in soluble transferrin receptor were noted among groups on admission, but levels in burn patients were lower than controls for the first 5 days after injury. CONCLUSION This study further established the hypoferremia and inflammation associated with burns and trauma. To our knowledge, this is the first study to show an early decrease in Cp oxidase activity in burn and non-burn trauma patients. The results support the hypothesis that transient loss of Cp activity contributes to hypoferremia and inflammation. Further studies are warranted to determine if decreased Cp activity increases the risk of iron-induced injury following therapeutic interventions such as transfusions with blood that has undergone prolonged storage in trauma resuscitation.
Collapse
Affiliation(s)
- Michael A Dubick
- Army Institute of Surgical Research, 3698 Chambers Pass, JBSA Fort Sam Houston, TX 78234, USA.
| | - Johnny L Barr
- Army Institute of Surgical Research, 3698 Chambers Pass, JBSA Fort Sam Houston, TX 78234, USA.
| | - Carl L Keen
- Departments of Nutrition and Internal Medicine, University of California, Davis, CA 95616, USA.
| | - James L Atkins
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| |
Collapse
|
213
|
Sudden sensorineural hearing loss and polymorphisms in iron homeostasis genes: new insights from a case-control study. BIOMED RESEARCH INTERNATIONAL 2015; 2015:834736. [PMID: 25789325 PMCID: PMC4348611 DOI: 10.1155/2015/834736] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 12/15/2014] [Accepted: 01/06/2015] [Indexed: 12/19/2022]
Abstract
Background. Even if various pathophysiological events have been proposed as explanations, the putative cause of sudden hearing loss remains unclear. Objectives. To investigate and to reveal associations (if any) between the main iron-related gene variants and idiopathic sudden sensorineural hearing loss. Study Design. Case-control study. Materials and Methods. A total of 200 sudden sensorineural hearing loss patients (median age 63.65 years; range 10-92) were compared with 400 healthy control subjects. The following genetic variants were investigated: the polymorphism c.-8CG in the promoter of the ferroportin gene (FPN1; SLC40A1), the two isoforms C1 and C2 (p.P570S) of the transferrin protein (TF), the amino acidic substitutions p.H63D and p.C282Y in the hereditary hemochromatosis protein (HFE), and the polymorphism c.-582AG in the promoter of the HEPC gene, which encodes the protein hepcidin (HAMP). Results. The homozygous genotype c.-8GG of the SLC40A1 gene revealed an OR for ISSNHL risk of 4.27 (CI 95%, 2.65-6.89; P = 0.001), being overrepresented among cases. Conclusions. Our study indicates that the homozygous genotype FPN1 -8GG was significantly associated with increased risk of developing sudden hearing loss. These findings suggest new research should be conducted in the field of iron homeostasis in the inner ear.
Collapse
|
214
|
Rogler G, Vavricka S. Anemia in inflammatory bowel disease: an under-estimated problem? Front Med (Lausanne) 2015; 1:58. [PMID: 25646159 PMCID: PMC4298217 DOI: 10.3389/fmed.2014.00058] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 12/18/2014] [Indexed: 12/16/2022] Open
Abstract
Anemia is one of the most frequent complications and/or extraintestinal manifestations of inflammatory bowel disease (IBD). Iron deficiency is the most important cause of anemia in Crohn’s disease and ulcerative colitis patients. Iron deficiency even without anemia may impact the quality of life of our IBD patients. In the last 10 years, the understanding of the pathology of iron-deficiency anemia and “anemia of chronic diseases” has increased; new diagnostic tools have been developed and new therapeutic strategies have been discussed. Hepcidin has been identified to be a central regulator of iron absorption from the intestine and of iron plasma levels. Hepcidin is regulated by iron deficiency but also as an acute phase protein by pro-inflammatory mediators such as interleukin-6. Innovative diagnostic tools have not been introduced in clinical routine or are not available for routine diagnostics. As iron substitution therapy is easy these days with a preference for intravenous substitution, the impact of differential diagnosis of anemia in IBD patients is underestimated.
Collapse
Affiliation(s)
- Gerhard Rogler
- Division of Gastroenterology and Hepatology, University Hospital Zürich , Zürich , Switzerland
| | - Stephan Vavricka
- Division of Gastroenterology and Hepatology, University Hospital Zürich , Zürich , Switzerland ; Division of Gastroenterology, Triemlispital , Zürich , Switzerland
| |
Collapse
|
215
|
van Beers EJ, Yang Y, Raghavachari N, Tian X, Allen DT, Nichols JS, Mendelsohn L, Nekhai S, Gordeuk VR, Taylor JG, Kato GJ. Iron, inflammation, and early death in adults with sickle cell disease. Circ Res 2015; 116:298-306. [PMID: 25378535 PMCID: PMC4297524 DOI: 10.1161/circresaha.116.304577] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 11/06/2014] [Indexed: 01/19/2023]
Abstract
RATIONALE Patients with sickle cell disease (SCD) have markers of chronic inflammation, but the mechanism of inflammation and its relevance to patient survival are unknown. OBJECTIVE To assess the relationship between iron, inflammation, and early death in SCD. METHODS AND RESULTS Using peripheral blood mononuclear cell transcriptome profile hierarchical clustering, we classified 24 patients and 10 controls in clusters with significantly different expression of genes known to be regulated by iron. Subsequent gene set enrichment analysis showed that many genes associated with the high iron cluster were involved in the toll-like receptor system (TLR4, TLR7, and TLR8) and inflammasome complex pathway (NLRP3, NLRC4, and CASP1). Quantitative PCR confirmed this classification and showed that ferritin light chain, TLR4, and interleukin-6 expression were >100-fold higher in patients than in controls (P<0.001). Further linking intracellular iron and inflammation, 14 SCD patients with a ferroportin Q248H variant that causes intracellular iron accumulation had significantly higher levels of interleukin-6 and C-reactive protein compared with 14 matched SCD patients with the wild-type allele (P<0.05). Finally, in a cohort of 412 patients followed for a median period of 47 months (interquartile range, 24-82), C-reactive protein was strongly and independently associated with early death (hazard ratio, 3.0; 95% confidence interval, 1.7-5.2; P<0.001). CONCLUSIONS Gene expression markers of high intracellular iron in patients with SCD are associated with markers of inflammation and mortality. The results support a model in which intracellular iron promotes inflammatory pathways, such as the TLR system and the inflammasome, identifying important new pathways for additional investigation.
Collapse
Affiliation(s)
- Eduard J van Beers
- From the Hematology Branch (E.J.v.B., D.T.A., J.S.N., L.M., J.G.T., G.J.K.), Genomics Core Facility (Y.Y., N.R.), and Office of Biostatistics Research (X.T.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Center for Sickle Cell Disease, Department of Medicine, Howard University, Washington, DC (S.N.); Comprehensive Sickle Cell Center, Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago (V.R.G.); and Division of Hematology-Oncology, Department of Medicine and the Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA (G.J.K.). Current address: Van Creveldkliniek, University Medical Center Utrecht, Utrecht, The Netherlands (E.J.v.B.)
| | - Yanqin Yang
- From the Hematology Branch (E.J.v.B., D.T.A., J.S.N., L.M., J.G.T., G.J.K.), Genomics Core Facility (Y.Y., N.R.), and Office of Biostatistics Research (X.T.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Center for Sickle Cell Disease, Department of Medicine, Howard University, Washington, DC (S.N.); Comprehensive Sickle Cell Center, Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago (V.R.G.); and Division of Hematology-Oncology, Department of Medicine and the Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA (G.J.K.). Current address: Van Creveldkliniek, University Medical Center Utrecht, Utrecht, The Netherlands (E.J.v.B.)
| | - Nalini Raghavachari
- From the Hematology Branch (E.J.v.B., D.T.A., J.S.N., L.M., J.G.T., G.J.K.), Genomics Core Facility (Y.Y., N.R.), and Office of Biostatistics Research (X.T.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Center for Sickle Cell Disease, Department of Medicine, Howard University, Washington, DC (S.N.); Comprehensive Sickle Cell Center, Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago (V.R.G.); and Division of Hematology-Oncology, Department of Medicine and the Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA (G.J.K.). Current address: Van Creveldkliniek, University Medical Center Utrecht, Utrecht, The Netherlands (E.J.v.B.)
| | - Xin Tian
- From the Hematology Branch (E.J.v.B., D.T.A., J.S.N., L.M., J.G.T., G.J.K.), Genomics Core Facility (Y.Y., N.R.), and Office of Biostatistics Research (X.T.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Center for Sickle Cell Disease, Department of Medicine, Howard University, Washington, DC (S.N.); Comprehensive Sickle Cell Center, Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago (V.R.G.); and Division of Hematology-Oncology, Department of Medicine and the Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA (G.J.K.). Current address: Van Creveldkliniek, University Medical Center Utrecht, Utrecht, The Netherlands (E.J.v.B.)
| | - Darlene T Allen
- From the Hematology Branch (E.J.v.B., D.T.A., J.S.N., L.M., J.G.T., G.J.K.), Genomics Core Facility (Y.Y., N.R.), and Office of Biostatistics Research (X.T.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Center for Sickle Cell Disease, Department of Medicine, Howard University, Washington, DC (S.N.); Comprehensive Sickle Cell Center, Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago (V.R.G.); and Division of Hematology-Oncology, Department of Medicine and the Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA (G.J.K.). Current address: Van Creveldkliniek, University Medical Center Utrecht, Utrecht, The Netherlands (E.J.v.B.)
| | - James S Nichols
- From the Hematology Branch (E.J.v.B., D.T.A., J.S.N., L.M., J.G.T., G.J.K.), Genomics Core Facility (Y.Y., N.R.), and Office of Biostatistics Research (X.T.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Center for Sickle Cell Disease, Department of Medicine, Howard University, Washington, DC (S.N.); Comprehensive Sickle Cell Center, Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago (V.R.G.); and Division of Hematology-Oncology, Department of Medicine and the Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA (G.J.K.). Current address: Van Creveldkliniek, University Medical Center Utrecht, Utrecht, The Netherlands (E.J.v.B.)
| | - Laurel Mendelsohn
- From the Hematology Branch (E.J.v.B., D.T.A., J.S.N., L.M., J.G.T., G.J.K.), Genomics Core Facility (Y.Y., N.R.), and Office of Biostatistics Research (X.T.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Center for Sickle Cell Disease, Department of Medicine, Howard University, Washington, DC (S.N.); Comprehensive Sickle Cell Center, Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago (V.R.G.); and Division of Hematology-Oncology, Department of Medicine and the Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA (G.J.K.). Current address: Van Creveldkliniek, University Medical Center Utrecht, Utrecht, The Netherlands (E.J.v.B.)
| | - Sergei Nekhai
- From the Hematology Branch (E.J.v.B., D.T.A., J.S.N., L.M., J.G.T., G.J.K.), Genomics Core Facility (Y.Y., N.R.), and Office of Biostatistics Research (X.T.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Center for Sickle Cell Disease, Department of Medicine, Howard University, Washington, DC (S.N.); Comprehensive Sickle Cell Center, Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago (V.R.G.); and Division of Hematology-Oncology, Department of Medicine and the Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA (G.J.K.). Current address: Van Creveldkliniek, University Medical Center Utrecht, Utrecht, The Netherlands (E.J.v.B.)
| | - Victor R Gordeuk
- From the Hematology Branch (E.J.v.B., D.T.A., J.S.N., L.M., J.G.T., G.J.K.), Genomics Core Facility (Y.Y., N.R.), and Office of Biostatistics Research (X.T.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Center for Sickle Cell Disease, Department of Medicine, Howard University, Washington, DC (S.N.); Comprehensive Sickle Cell Center, Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago (V.R.G.); and Division of Hematology-Oncology, Department of Medicine and the Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA (G.J.K.). Current address: Van Creveldkliniek, University Medical Center Utrecht, Utrecht, The Netherlands (E.J.v.B.)
| | - James G Taylor
- From the Hematology Branch (E.J.v.B., D.T.A., J.S.N., L.M., J.G.T., G.J.K.), Genomics Core Facility (Y.Y., N.R.), and Office of Biostatistics Research (X.T.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Center for Sickle Cell Disease, Department of Medicine, Howard University, Washington, DC (S.N.); Comprehensive Sickle Cell Center, Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago (V.R.G.); and Division of Hematology-Oncology, Department of Medicine and the Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA (G.J.K.). Current address: Van Creveldkliniek, University Medical Center Utrecht, Utrecht, The Netherlands (E.J.v.B.)
| | - Gregory J Kato
- From the Hematology Branch (E.J.v.B., D.T.A., J.S.N., L.M., J.G.T., G.J.K.), Genomics Core Facility (Y.Y., N.R.), and Office of Biostatistics Research (X.T.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; Center for Sickle Cell Disease, Department of Medicine, Howard University, Washington, DC (S.N.); Comprehensive Sickle Cell Center, Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago (V.R.G.); and Division of Hematology-Oncology, Department of Medicine and the Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA (G.J.K.). Current address: Van Creveldkliniek, University Medical Center Utrecht, Utrecht, The Netherlands (E.J.v.B.).
| |
Collapse
|
216
|
Chanprasert S, Scaglia F. Adult liver disorders caused by inborn errors of metabolism: review and update. Mol Genet Metab 2015; 114:1-10. [PMID: 25467056 DOI: 10.1016/j.ymgme.2014.10.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 10/23/2014] [Accepted: 10/23/2014] [Indexed: 12/22/2022]
Abstract
Inborn errors of metabolism (IEMs) are a group of genetic diseases that have protean clinical manifestations and can involve several organ systems. The age of onset is highly variable but IEMs afflict mostly the pediatric population. However, in the past decades, the advancement in management and new therapeutic approaches have led to the improvement in IEM patient care. As a result, many patients with IEMs are surviving into adulthood and developing their own set of complications. In addition, some IEMs will present in adulthood. It is important for internists to have the knowledge and be familiar with these conditions because it is predicted that more and more adult patients with IEMs will need continuity of care in the near future. The review will focus on Wilson disease, alpha-1 antitrypsin deficiency, citrin deficiency, and HFE-associated hemochromatosis which are typically found in the adult population. Clinical manifestations and pathophysiology, particularly those that relate to hepatic disease as well as diagnosis and management will be discussed in detail.
Collapse
Affiliation(s)
- Sirisak Chanprasert
- Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children Hospital, Houston, TX, USA
| | - Fernando Scaglia
- Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children Hospital, Houston, TX, USA.
| |
Collapse
|
217
|
Zhou F, Chen Y, Fan G, Feng C, Du G, Zhu G, Li Y, Jiao H, Guan L, Wang Z. Lead-induced iron overload and attenuated effects of ferroportin 1 overexpression in PC12 cells. Toxicol In Vitro 2014; 28:1339-48. [DOI: 10.1016/j.tiv.2014.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 05/12/2014] [Accepted: 07/11/2014] [Indexed: 01/21/2023]
|
218
|
Chen Y, Zhang S, Wang X, Guo W, Wang L, Zhang D, Yuan L, Zhang Z, Xu Y, Liu S. Disordered signaling governing ferroportin transcription favors breast cancer growth. Cell Signal 2014; 27:168-76. [PMID: 25451081 DOI: 10.1016/j.cellsig.2014.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 10/23/2014] [Accepted: 11/03/2014] [Indexed: 12/17/2022]
Abstract
Iron is a necessary chemical element needed by all organisms. Iron metabolism is finely tuned in mammals, and the hepcidin-ferroportin (FPN) axis is the central signaling in governing systemic iron homeostasis. Deregulation of this signaling would lead to iron disorders and likely other diseases including cancers. Reduced FPN was previously found to correlate with poor prognosis in breast cancer patients. Nonetheless, the biological effects of abnormal FPN expression in tumor cells remain largely unexplored, and the mechanisms underlying misregulated expression of FPN in cancers keep elusive. In the current study, we scrutinized the effects of abnormal FPN on tumor growth and the molecular mechanisms of diminished tumor FPN. Downregulation of FPN significantly promoted breast cancer growth, whereas FPN upregulation impeded tumor growth. We demonstrated that the transcription factors Nrf2 (nuclear factor erythroid 2-like 2) and MZF-1 (myeloid zinc finger-1) synergistically transactivated FPN expression in breast cancer cells. Moreover, CpG island methylation at the FPN promoter was the reason of attenuated FPN expression. Downregulation of Nrf2 and MZF-1 and hypermethylation of the FPN promoter were concurrently associated with decreased FPN concentration in breast tumors. Taken together, our study highlighted the contribution of disordered iron metabolism to breast cancer growth, and also signified an oncogenic effect of misregulated ferroportin in breast cancers. This work represents a promising starting point to the possibility of restraining breast cancer through targeting FPN or its upstream regulatory factors.
Collapse
Affiliation(s)
- Yue Chen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; Department of Urology, the Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China
| | - Shuping Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Xiaoyan Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Wenli Guo
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Lei Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Daoqiang Zhang
- Weifang Medical College, Wendeng Central Hospital, Weihai 264400, China
| | - Lin Yuan
- Weifang Medical College, Wendeng Central Hospital, Weihai 264400, China
| | - Zhihong Zhang
- Department of Urology, the Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China
| | - Yong Xu
- Department of Urology, the Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China.
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| |
Collapse
|
219
|
Iron transport across the blood-brain barrier: development, neurovascular regulation and cerebral amyloid angiopathy. Cell Mol Life Sci 2014; 72:709-27. [PMID: 25355056 DOI: 10.1007/s00018-014-1771-4] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 10/10/2014] [Accepted: 10/23/2014] [Indexed: 12/14/2022]
Abstract
There are two barriers for iron entry into the brain: (1) the brain-cerebrospinal fluid (CSF) barrier and (2) the blood-brain barrier (BBB). Here, we review the literature on developmental iron accumulation by the brain, focusing on the transport of iron through the brain microvascular endothelial cells (BMVEC) of the BBB. We review the iron trafficking proteins which may be involved in the iron flux across BMVEC and discuss the plausible mechanisms of BMVEC iron uptake and efflux. We suggest a model for how BMVEC iron uptake and efflux are regulated and a mechanism by which the majority of iron is trafficked across the developing BBB under the direct guidance of neighboring astrocytes. Thus, we place brain iron uptake in the context of the neurovascular unit of the adult brain. Last, we propose that BMVEC iron is involved in the aggregation of amyloid-β peptides leading to the progression of cerebral amyloid angiopathy which often occurs prior to dementia and the onset of Alzheimer's disease.
Collapse
|
220
|
Abstract
Metal ion assimilation is essential for all forms of life. However, organisms must properly control the availability of these nutrients within the cell to avoid inactivating proteins by mismetallation. To safeguard against an imbalance between supply and demand in eukaryotes, intracellular compartments contain metal transporters that load and unload metals. Although the vacuoles of Saccharomyces cerevisiae and Arabidopsis thaliana are well established locales for the storage of copper, zinc, iron, and manganese, related compartments are emerging as important mediators of metal homeostasis. Here we describe these compartments and review their metal transporter complement.
Collapse
Affiliation(s)
| | - Sabeeha S Merchant
- From the Department of Chemistry and Biochemistry and the Institute for Genomics and Proteomics, UCLA, Los Angeles, California 90095
| |
Collapse
|
221
|
Chen Y, Zhang Z, Yang K, Du J, Xu Y, Liu S. Myeloid zinc-finger 1 (MZF-1) suppresses prostate tumor growth through enforcing ferroportin-conducted iron egress. Oncogene 2014; 34:3839-47. [PMID: 25284586 DOI: 10.1038/onc.2014.310] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 07/09/2014] [Accepted: 08/13/2014] [Indexed: 12/24/2022]
Abstract
Although previous studies suggest that myeloid zinc-finger 1 (MZF-1) is a multifaceted transcription factor that may function as either an oncogene or a tumor suppressor, the molecular bases determining its different traits remain elusive. Increasing evidence suggests that disorders in iron metabolism affect tumorigenesis and tumor behaviors, and that excess tumor iron stimulates tumor progression through various mechanisms such as enhancing DNA replication and energy metabolism. Ferroportin (FPN) is the only known iron exporter in mammalian cells, and it determines global iron egress out of cells. FPN reduction leads to decreased iron efflux and increased intracellular iron that consequentially aggravates the oncogenic effects of iron. MZF-1 was recently identified as a transcription factor that regulates FPN expression. Thus far, however, the molecular mechanisms underlying the MZF-1-FPN signaling in cancers are largely unknown. Here, we found a significant reduction of FPN levels in prostate tumors relative to adjacent tissues, and demonstrated a crucial role of FPN in tumor growth through controlling tumor iron concentration. Inhibition of MZF-1 expression led to reduced FPN concentration, coupled with resultant intracellular iron retention, increased iron-related cellular activities and enhanced tumor cell growth. In contrast, increase of MZF-1 expression restrained tumor cell growth by promoting FPN-driven iron egress. Importantly, we demonstrated that AP4 and c-Myb jointly modulated MZF-1 transcription, and that miR-492 was also directly involved in regulating MZF-1 concentration through binding to the 3' untranslated regions of its mRNA. These results correlate with reduced AP4 and c-Myb expression and elevated miR-492 expression found in prostate tumors as compared with adjacent tissues that resulted in diminished MZF-1 and FPN. Moreover, we demonstrated that alterations of AP4, c-Myb and miR-492 levels significantly affected tumor cell growth. Targeting molecules within the MZF-1-FPN signaling thus appears to be a promising approach to restrain prostate cancer.
Collapse
Affiliation(s)
- Y Chen
- 1] Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin, China [2] State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
| | - Z Zhang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin, China
| | - K Yang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin, China
| | - J Du
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Y Xu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin, China
| | - S Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
222
|
Theil EC. IRE mRNA riboregulators use metabolic iron (Fe(2+)) to control mRNA activity and iron chemistry in animals. Metallomics 2014; 7:15-24. [PMID: 25209685 DOI: 10.1039/c4mt00136b] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
A family of noncoding RNAs bind Fe(2+) to increase protein synthesis. The structures occur in messenger RNAs encoding animal proteins for iron metabolism. Each mRNA regulatory sequence, ∼30 ribonucleotides long, is called an IRE (Iron Responsive Element), and folds into a bent, A-RNA helix with a terminal loop. Riboregulatory RNAs, like t-RNAs, r-RNAs micro-RNAs, etc. contrast with DNA, since single-stranded RNA can fold into a variety of complex, three-dimensional structures. IRE-RNAs bind two types of proteins: (1) IRPs which are protein repressors, sequence-related to mitochondrial aconitases. (2) eIF-4F, which bind ribosomes and enhances general protein biosynthesis. The competition between IRP and eIF-4F binding to IRE-RNA is controlled by Fe(2+)-induced changes in the IRE-RNA conformation. Mn(2+), which also binds to IRE-RNA in solution, is a convenient experimental proxy for air-sensitive Fe(2+) studies of in vitro protein biosynthesis and protein binding. However, only Fe(2+) has physiological effects on protein biosynthesis directed by IRE-mRNAs. The structures of the IRE-RNA riboregulators is known indirectly from effects of base substitutions on function, from solution NMR of the free RNA, and of X-ray crystallography of the IRE-RNA-IRP repressor complex. However, the inability to date, to crystallize the free IRE-RNA, and the dissociation of the IRE-RNA-IRP complex when metal binds, have hampered direct identification and characterization of the RNA-metal binding sites. The high conservation of the primary sequence in IRE-mRNA control elements has facilitated their identification and analysis of metal-assisted riboregulator function. Expansion of RNA search analyses beyond primary will likely reveal other, metal-dependent families of mRNA riboregulators.
Collapse
Affiliation(s)
- Elizabeth C Theil
- The Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA.
| |
Collapse
|
223
|
Abstract
SIGNIFICANCE Iron is the most abundant transition metal in biology and an essential cofactor for many cellular enzymes. Iron homeostasis impairment is also a component of peripheral neuropathies. RECENT ADVANCES During the past years, much effort has been paid to understand the molecular mechanism involved in maintaining systemic iron homeostasis in mammals. This has been stimulated by the evidence that iron dyshomeostasis is an initial cause of several disorders, including genetic and sporadic neurodegenerative disorders. CRITICAL ISSUES However, very little has been done to investigate the physiological role of iron in peripheral nervous system (PNS), despite the development of suitable cellular and animal models. FUTURE DIRECTIONS To stimulate research on iron metabolism and peripheral neuropathy, we provide a summary of the knowledge on iron homeostasis in the PNS, on its transport across the blood-nerve barrier, its involvement in myelination, and we identify unresolved questions. Furthermore, we comment on the role of iron in iron-related disorder with peripheral component, in demyelinating and metabolic peripheral neuropathies.
Collapse
Affiliation(s)
- Sonia Levi
- 1 University Vita-Salute San Raffaele , Milan, Italy
| | | |
Collapse
|
224
|
Abstract
Iron is one of the most important nonorganic substances that make life possible. Iron plays major roles in oxygen transport (eg, hemoglobin; -67% of total body iron [TBI]), short-term oxygen storage (eg, myoglobin; -3.5% of TBI), and energy generation (eg, cytochromes; -3% of TBI). Iron also serves vital roles in various nonheme-containing enzymes (-2% of TBI). Figure 1 lists heme-containing and nonheme iron-containing proteins. TBI is controlled by the rate of iron absorption; there are no physiologic mechanisms to excrete excess iron. Iron deficiency has many adverse consequences, including anemia, and in children, behavioral and learning disorders. Iron excess is toxic to the body, harming the heart, liver, skin, pancreatic islet beta cells, bones, joints, and pituitary gland. Maintaining proper iron balance is essential for maintaining homeostasis and health. TBI in adults normally ranges between 3.5 and 5.0 g. A total of 75% of TBI is functional, and 25% is stored within cells as ferritin or hemosiderin. Ferritin contains 24 subunits of light chains (L chains; 19.7 kDa) and heavy chains (H chains; 21.1 kDa). The L chains are encoded on chromosome 19q13.33 and are 175 amino acids long. The H chains are encoded on chromosome 11q1 and are 183 amino acids long. Each ferritin molecule can contain as many as approximately 4500 ferric ions. Because the major role of iron is in hemoglobin synthesis, this review will focus on iron, iron transport, and hematopoiesis.
Collapse
|
225
|
Abstract
Iron deficiency anemia is a common global problem whose etiology is typically attributed to acquired inadequate dietary intake and/or chronic blood loss. However, in several kindreds multiple family members are affected with iron deficiency anemia that is unresponsive to oral iron supplementation and only partially responsive to parenteral iron therapy. The discovery that many of these cases harbor mutations in the TMPRSS6 gene led to the recognition that they represent a single clinical entity: iron-refractory iron deficiency anemia (IRIDA). This article reviews clinical features of IRIDA, recent genetic studies, and insights this disorder provides into the regulation of systemic iron homeostasis.
Collapse
|
226
|
McCarthy RC, Park YH, Kosman DJ. sAPP modulates iron efflux from brain microvascular endothelial cells by stabilizing the ferrous iron exporter ferroportin. EMBO Rep 2014; 15:809-15. [PMID: 24867889 DOI: 10.15252/embr.201338064] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
A sequence within the E2 domain of soluble amyloid precursor protein (sAPP) stimulates iron efflux. This activity has been attributed to a ferroxidase activity suggested for this motif. We demonstrate that the stimulation of efflux supported by this peptide and by sAPPα is due to their stabilization of the ferrous iron exporter, ferroportin (Fpn), in the plasma membrane of human brain microvascular endothelial cells (hBMVEC). The peptide does not bind ferric iron explaining why it does not and thermodynamically cannot promote ferrous iron autoxidation. This peptide specifically pulls Fpn down from the plasma membrane of hBMVEC; based on these results, FTP, for ferroportin-targeting peptide, correctly identifies the function of this peptide. The data suggest that in stabilizing Fpn via the targeting due to the FTP sequence, sAPP will increase the flux of iron into the cerebral interstitium. This inference correlates with the observation of significant iron deposition in the amyloid plaques characteristic of Alzheimer's disease.
Collapse
Affiliation(s)
- Ryan C McCarthy
- Department of Biochemistry, School of Medicine and Biomedical Sciences University at Buffalo, Buffalo, NY, USA
| | - Yun-Hee Park
- Department of Biochemistry, School of Medicine and Biomedical Sciences University at Buffalo, Buffalo, NY, USA
| | - Daniel J Kosman
- Department of Biochemistry, School of Medicine and Biomedical Sciences University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
227
|
Bonaccorsi di Patti MC, Polticelli F, Cece G, Cutone A, Felici F, Persichini T, Musci G. A structural model of human ferroportin and of its iron binding site. FEBS J 2014; 281:2851-60. [PMID: 24767627 DOI: 10.1111/febs.12825] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 03/17/2014] [Accepted: 04/24/2014] [Indexed: 11/29/2022]
Abstract
A structural model of human ferroportin has been built using two Escherichia coli proteins belonging to the major facilitator superfamily of transporters. A potential iron binding site was identified in the inward-open conformation of the model, and its relevance was tested through measurement of iron export of HEK293T cells expressing wild-type or mutated ferroportin. Aspartates 39 and 181 were found to be essential for the transport ability of the protein. Noteworthy, the D181V mutation is naturally found in type 4 hemochromatosis with reticuloendothelial system iron retention phenotype. The outward-open conformation of ferroportin was also predicted, and showed that significant conformational changes must occur in the inward- to outward-open transition of ferroportin. In particular, putative iron ligands move several ångströms away from each other, leading to the logical conclusion that the iron binding site is not occupied by the metal in the outward-open conformation of ferroportin.
Collapse
|
228
|
Eleftheriadis T, Pissas G, Antoniadi G, Filippidis G, Golfinopoulos S, Spanoulis A, Liakopoulos V, Stefanidis I. Serum copper and ferroportin in monocytes of hemodialysis patients are both decreased but unassociated. Int Urol Nephrol 2014; 46:1825-31. [PMID: 24806658 DOI: 10.1007/s11255-014-0725-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 04/21/2014] [Indexed: 11/25/2022]
Abstract
PURPOSE Disturbed iron homeostasis contributes to resistance to recombinant human erythropoietin (rHuEpo) in hemodialysis (HD) patients. Although increased hepcidin, which downregulates the iron exporter ferroportin, had been incriminated, such an association has not been confirmed. Albeit not universally accepted, it has been supported that in case of copper deficiency, decreased activity of multicopper oxidases induces endocytosis and degradation of ferroportin. Ferroportin in monocytes, serum copper, ceruloplasmin and markers of iron status were measured, and associations with rHuEpo resistance index (ERI) were evaluated. METHODS After a 4-week washout period from iron treatment, 34 HD patients and 20 healthy volunteers enrolled in the study. Ferroportin was assessed by means of Western blotting, copper colorimetrically, whereas ceruloplasmin with enzyme-linked immunosorbent assay. Hemoglobin, serum iron, ferritin and transferrin saturation (TSAT) were also measured. RESULTS Ferroportin in monocytes of HD patients was decreased. Serum copper, ceruloplasmin, iron and TSAT were decreased. No correlation between copper or ceruloplasmin and ferroportin was detected. ERI was negatively correlated with ferroportin and all the markers of iron adequacy, but not with copper or ceruloplasmin. CONCLUSION Although copper deficiency and decreased ferroportin are common in HD patients, copper might not play role in ferroportin level in monocytes and in iron metabolism in this population.
Collapse
Affiliation(s)
- Theodoros Eleftheriadis
- Department of Nephrology, Medical School, University of Thessaly, Neo Ktirio, Mezourlo Hill, 411 10, Larissa, Greece,
| | | | | | | | | | | | | | | |
Collapse
|
229
|
Cutone A, Frioni A, Berlutti F, Valenti P, Musci G, Bonaccorsi di Patti MC. Lactoferrin prevents LPS-induced decrease of the iron exporter ferroportin in human monocytes/macrophages. Biometals 2014; 27:807-13. [DOI: 10.1007/s10534-014-9742-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Accepted: 04/18/2014] [Indexed: 01/01/2023]
|
230
|
Merlot S, Hannibal L, Martins S, Martinelli L, Amir H, Lebrun M, Thomine S. The metal transporter PgIREG1 from the hyperaccumulator Psychotria gabriellae is a candidate gene for nickel tolerance and accumulation. JOURNAL OF EXPERIMENTAL BOTANY 2014; 65:1551-64. [PMID: 24510940 DOI: 10.1093/jxb/eru025] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Nickel is an economically important metal and phytotechnologies are being developed to limit the impact of nickel mining on the environment. More than 300 plant species are known to hyperaccumulate nickel. However, our knowledge of the mechanisms involved in nickel accumulation in plants is very limited because it has not yet been possible to study these hyperaccumulators at the genomic level. Here, we used next-generation sequencing technologies to sequence the transcriptome of the nickel hyperaccumulator Psychotria gabriellae of the Rubiaceae family, and used yeast and Arabidopsis as heterologous systems to study the activity of identified metal transporters. We characterized the activity of three metal transporters from the NRAMP and IREG/FPN families. In particular, we showed that PgIREG1 is able to confer nickel tolerance when expressed in yeast and in transgenic plants, where it localizes in the tonoplast. In addition, PgIREG1 shows higher expression in P. gabriellae than in the related non-accumulator species Psychotria semperflorens. Our results designate PgIREG1 as a candidate gene for nickel tolerance and hyperaccumulation in P. gabriellae. These results also show how next-generation sequencing technologies can be used to access the transcriptome of non-model nickel hyperaccumulators to identify the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Sylvain Merlot
- CNRS, Institut des Sciences du Végétal, Labex SPS, Avenue de la terrasse, 91198 Gif-sur-Yvette cedex, France
| | | | | | | | | | | | | |
Collapse
|
231
|
|
232
|
Frazer DM, Anderson GJ. The regulation of iron transport. Biofactors 2014; 40:206-14. [PMID: 24132807 DOI: 10.1002/biof.1148] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 09/02/2013] [Accepted: 09/06/2013] [Indexed: 01/01/2023]
Abstract
Iron is an essential nutrient, but its concentration and distribution in the body must be tightly controlled due to its inherent toxicity and insolubility in aqueous solution. Living systems have successfully overcome these potential limitations by evolving a range of iron binding proteins and transport systems that effectively maintain iron in a nontoxic and soluble form for much, if not all, of its time within the body. In the circulation, iron is transported to target organs bound to the serum iron binding protein transferrin. Individual cells modulate their uptake of transferrin-bound iron depending on their iron requirements, using both transferrin receptor 1-dependent and independent pathways. Once inside the cell, iron can be chaperoned to sites of need or, if in excess, stored within ferritin. Iron is released from cells by the iron export protein ferroportin1, which requires the ferroxidase activity of ceruloplasmin or hephestin to load iron safely onto transferrin. The regulation of iron export is controlled predominantly at the systemic level by the master regulator of iron homeostasis hepcidin. Hepcidin, in turn, responds to changes in body iron demand, making use of a range of regulatory mechanisms that center on the bone morphogenetic protein signaling pathway. This review provides an overview of recent advances in the field of iron metabolism and outlines the key components of the iron transport and regulation systems.
Collapse
Affiliation(s)
- David M Frazer
- Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Qld, Australia
| | | |
Collapse
|
233
|
Chin D, Huebbe P, Frank J, Rimbach G, Pallauf K. Curcumin may impair iron status when fed to mice for six months. Redox Biol 2014; 2:563-9. [PMID: 24634837 PMCID: PMC3953957 DOI: 10.1016/j.redox.2014.01.018] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 01/14/2014] [Accepted: 01/18/2014] [Indexed: 11/16/2022] Open
Abstract
Curcumin has been shown to have many potentially health beneficial properties in vitro and in animal models with clinical studies on the toxicity of curcumin reporting no major side effects. However, curcumin may chelate dietary trace elements and could thus potentially exert adverse effects. Here, we investigated the effects of a 6 month dietary supplementation with 0.2% curcumin on iron, zinc, and copper status in C57BL/6J mice. Compared to non-supplemented control mice, we observed a significant reduction in iron, but not zinc and copper stores, in the liver and the spleen, as well as strongly suppressed liver hepcidin and ferritin expression in the curcumin-supplemented mice. The expression of the iron-importing transport proteins divalent metal transporter 1 and transferrin receptor 1 was induced, while hepatic and splenic inflammatory markers were not affected in the curcumin-fed mice. The mRNA expression of other putative target genes of curcumin, including the nuclear factor (erythroid-derived 2)-like 2 and haem oxygenase 1 did not differ between the groups. Most of the published animal trials with curcumin-feeding have not reported adverse effects on iron status or the spleen. However, it is possible that long-term curcumin supplementation and a Western-type diet may aggravate iron deficiency. Therefore, our findings show that further studies are needed to evaluate the effect of curcumin supplementation on iron status. 0.2% dietary curcumin for 6 months reduced iron stores in murine liver and spleen. Curcumin chelated iron but not zinc and copper in vivo. Liver hepcidin and ferritin expression was strongly suppressed in curcumin-fed mice. Curcumin induced expression of hepatic iron transporters DMT1 and TfR1. Curcumin did not affect hepatic and splenic inflammatory and oxidative markers.
Collapse
Key Words
- Curcumin
- DMT1, divalent metal transporter 1
- Enlarged spleen
- FPN, ferroportin
- HO1, haem oxygenase
- IL, interleukin
- Iron store
- Liver minerals
- NQO1, NAD(P)H quinone oxidoreductase
- NRF2, nuclear factor (erythroid-derived 2)-like 2
- Safety
- TBS, tris buffered saline
- TNFα, tumour necrosis factor α
- TfR1, transferrin receptor 1
- Toxicity
- qRT-PCR, quantitative real-time polymerase chain reaction
- γ-GCS, γ-glutamyl cysteine synthetase
Collapse
Affiliation(s)
- Dawn Chin
- Institute of Human Nutrition and Food Science, Christian-Albrechts-University Kiel, Hermann-Rodewald-Strasse 6-8, D-24118 Kiel, Germany
| | - Patricia Huebbe
- Institute of Human Nutrition and Food Science, Christian-Albrechts-University Kiel, Hermann-Rodewald-Strasse 6-8, D-24118 Kiel, Germany
| | - Jan Frank
- Institute of Biological Chemistry and Nutrition, University of Hohenheim, Garbenstrasse 28, D-70599 Stuttgart, Germany
| | - Gerald Rimbach
- Institute of Human Nutrition and Food Science, Christian-Albrechts-University Kiel, Hermann-Rodewald-Strasse 6-8, D-24118 Kiel, Germany
| | - Kathrin Pallauf
- Institute of Human Nutrition and Food Science, Christian-Albrechts-University Kiel, Hermann-Rodewald-Strasse 6-8, D-24118 Kiel, Germany
| |
Collapse
|
234
|
McCarthy RC, Kosman DJ. Glial cell ceruloplasmin and hepcidin differentially regulate iron efflux from brain microvascular endothelial cells. PLoS One 2014; 9:e89003. [PMID: 24533165 PMCID: PMC3923066 DOI: 10.1371/journal.pone.0089003] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 01/13/2014] [Indexed: 11/18/2022] Open
Abstract
We have used an in vitro model system to probe the iron transport pathway across the brain microvascular endothelial cells (BMVEC) of the blood-brain barrier (BBB). This model consists of human BMVEC (hBMVEC) and C6 glioma cells (as an astrocytic cell line) grown in a transwell, a cell culture system commonly used to quantify metabolite flux across a cell-derived barrier. We found that iron efflux from hBMVEC through the ferrous iron permease ferroportin (Fpn) was stimulated by secretion of the soluble form of the multi-copper ferroxidase, ceruloplasmin (sCp) from the co-cultured C6 cells. Reciprocally, expression of sCp mRNA in the C6 cells was increased by neighboring hBMVEC. In addition, data indicate that C6 cell-secreted hepcidin stimulates internalization of hBMVEC Fpn but only when the end-feet projections characteristic of this glia-derived cell line are proximal to the endothelial cells. This hepcidin-dependent loss of Fpn correlated with knock-down of iron efflux from the hBMVEC; this result was consistent with the mechanism by which hepcidin regulates iron efflux in mammalian cells. In summary, the data support a model of iron trafficking across the BBB in which the capillary endothelium induce the underlying astrocytes to produce the ferroxidase activity needed to support Fpn-mediated iron efflux. Reciprocally, astrocyte proximity modulates the effective concentration of hepcidin at the endothelial cell membrane and thus the surface expression of hBMVEC Fpn. These results are independent of the source of hBMVEC iron (transferrin or non-transferrin bound) indicating that the model developed here is broadly applicable to brain iron homeostasis.
Collapse
Affiliation(s)
- Ryan C McCarthy
- Department of Biochemistry, University at Buffalo, School of Medicine and Biomedical Scienes, Buffalo, New York, United States of America
| | - Daniel J Kosman
- Department of Biochemistry, University at Buffalo, School of Medicine and Biomedical Scienes, Buffalo, New York, United States of America
| |
Collapse
|
235
|
Ben-Othman R, Flannery AR, Miguel DC, Ward DM, Kaplan J, Andrews NW. Leishmania-mediated inhibition of iron export promotes parasite replication in macrophages. PLoS Pathog 2014; 10:e1003901. [PMID: 24497831 PMCID: PMC3907422 DOI: 10.1371/journal.ppat.1003901] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 12/10/2013] [Indexed: 12/20/2022] Open
Abstract
Leishmania parasites infect macrophages, cells that play an important role in organismal iron homeostasis. By expressing ferroportin, a membrane protein specialized in iron export, macrophages release iron stored intracellularly into the circulation. Iron is essential for the intracellular replication of Leishmania, but how the parasites compete with the iron export function of their host cell is unknown. Here, we show that infection with Leishmania amazonensis inhibits ferroportin expression in macrophages. In a TLR4-dependent manner, infected macrophages upregulated transcription of hepcidin, a peptide hormone that triggers ferroportin degradation. Parasite replication was inhibited in hepcidin-deficient macrophages and in wild type macrophages overexpressing mutant ferroportin that is resistant to hepcidin-induced degradation. Conversely, intracellular growth was enhanced by exogenously added hepcidin, or by expression of dominant-negative ferroportin. Importantly, dominant-negative ferroportin and macrophages from flatiron mice, a mouse model for human type IV hereditary hemochromatosis, restored the infectivity of mutant parasite strains defective in iron acquisition. Thus, inhibition of ferroportin expression is a specific strategy used by L. amazonensis to inhibit iron export and promote their own intracellular growth.
Collapse
Affiliation(s)
- Rym Ben-Othman
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Andrew R. Flannery
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Danilo C. Miguel
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Diane M. Ward
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Jerry Kaplan
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Norma W. Andrews
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
- * E-mail:
| |
Collapse
|
236
|
Ruiz JC, Bruick RK. F-box and leucine-rich repeat protein 5 (FBXL5): sensing intracellular iron and oxygen. J Inorg Biochem 2014; 133:73-7. [PMID: 24508277 DOI: 10.1016/j.jinorgbio.2014.01.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 01/15/2014] [Accepted: 01/16/2014] [Indexed: 12/25/2022]
Abstract
Though essential for many vital biological processes, excess iron results in the formation of damaging reactive oxygen species (ROS). Therefore, iron metabolism must be tightly regulated. F-box and leucine-rich repeat protein 5 (FBXL5), an E3 ubiquitin ligase subunit, regulates cellular and systemic iron homeostasis by facilitating iron regulatory protein 2 (IRP2) degradation. FBXL5 possesses an N-terminal hemerythrin (Hr)-like domain that mediates its own differential stability by switching between two different conformations to communicate cellular iron availability. In addition, the FBXL5-Hr domain also senses O2 availability, albeit by a distinct mechanism. Mice lacking FBXL5 fail to sense intracellular iron levels and die in utero due to iron overload and exposure to damaging levels of oxidative stress. By closely monitoring intracellular levels of iron and oxygen, FBLX5 prevents the formation of conditions that favor ROS formation. These findings suggest that FBXL5 is essential for the maintenance of iron homeostasis and is a key sensor of bioavailable iron. Here, we describe the iron and oxygen sensing mechanisms of the FBXL5 Hr-like domain and its role in mediating ROS biology.
Collapse
Affiliation(s)
- Julio C Ruiz
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9038, United States
| | - Richard K Bruick
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9038, United States.
| |
Collapse
|
237
|
Mallikarjun V, Sriram A, Scialo F, Sanz A. The interplay between mitochondrial protein and iron homeostasis and its possible role in ageing. Exp Gerontol 2014; 56:123-34. [PMID: 24394155 DOI: 10.1016/j.exger.2013.12.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/23/2013] [Accepted: 12/27/2013] [Indexed: 01/18/2023]
Abstract
Free (labile or chelatable) iron is extremely redox-active and only represents a small fraction of the total mitochondrial iron population. Several studies have shown that the proportion of free iron increases with age, leading to increased Fenton chemistry in later life. It is not clear why free iron accumulates in mitochondria, but it does so in parallel with an inability to degrade and recycle damaged proteins that causes loss of mitochondrial protein homeostasis (proteostasis). The increase in oxidative damage that has been shown to occur with age might be explained by these two processes. While this accumulation of oxidative damage has often been cited as causative to ageing there are examples of model organisms that possess high levels of oxidative damage throughout their lives with no effect on lifespan. Interestingly, these same animals are characterised by an outstanding ability to maintain correct proteostasis during their entire life. ROS can damage critical components of the iron homeostasis machinery, while the efficacy of mitochondrial quality control mechanisms will determine how detrimental that damage is. Here we review the interplay between iron and organellar quality control in mitochondrial dysfunction and we suggest that a decline in mitochondrial proteostasis with age leaves iron homeostasis (where several key stages are thought to be dependent on proteostasis machinery) vulnerable to oxidative damage and other age-related stress factors. This will have severe consequences for the electron transport chain and TCA cycle (among other processes) where several components are acutely dependent on correct assembly, insertion and maintenance of iron-sulphur clusters, leading to energetic crisis and death.
Collapse
Affiliation(s)
- Venkatesh Mallikarjun
- Institute of Biomedical Technology and BioMediTech, University of Tampere, FI-33014 Tampere, Finland.
| | - Ashwin Sriram
- Institute of Biomedical Technology and BioMediTech, University of Tampere, FI-33014 Tampere, Finland
| | - Filippo Scialo
- Institute of Biomedical Technology and BioMediTech, University of Tampere, FI-33014 Tampere, Finland
| | - Alberto Sanz
- Institute of Biomedical Technology and BioMediTech, University of Tampere, FI-33014 Tampere, Finland.
| |
Collapse
|
238
|
Argüello G, Martinez P, Peña J, Chen O, Platt F, Zanlungo S, González M. Hepatic metabolic response to restricted copper intake in a Niemann–Pick C murine model. Metallomics 2014; 6:1527-39. [DOI: 10.1039/c4mt00056k] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Niemann–Pick C disease (NPC) is a vesicular trafficking disorder primarily caused by mutations in theNpc1gene and characterized by liver dysfunction and neuropathology.
Collapse
Affiliation(s)
- Graciela Argüello
- INTA
- Laboratorio de Bioinformática y Expresión Génica
- Universidad de Chile
- Santiago, Chile
- FONDAP-Center of Genome Regulation (CGR)
| | - Pablo Martinez
- Departamento de Gastroenterología
- Facultad de Medicina
- Pontificia Universidad Católica de Chile
- Santiago, Chile
| | - Juan Peña
- INTA
- Laboratorio de Bioinformática y Expresión Génica
- Universidad de Chile
- Santiago, Chile
| | - Oscar Chen
- Department of Pharmacology
- University of Oxford
- Oxford OX1 3QT, UK
| | - Frances Platt
- Department of Pharmacology
- University of Oxford
- Oxford OX1 3QT, UK
| | - Silvana Zanlungo
- FONDAP-Center of Genome Regulation (CGR)
- Santiago, Chile
- Departamento de Gastroenterología
- Facultad de Medicina
- Pontificia Universidad Católica de Chile
| | - Mauricio González
- INTA
- Laboratorio de Bioinformática y Expresión Génica
- Universidad de Chile
- Santiago, Chile
- FONDAP-Center of Genome Regulation (CGR)
| |
Collapse
|
239
|
Díaz V, Gammella E, Recalcati S, Santambrogio P, Naldi AM, Vogel J, Gassmann M, Cairo G. Liver iron modulates hepcidin expression during chronically elevated erythropoiesis in mice. Hepatology 2013; 58:2122-32. [PMID: 23744538 DOI: 10.1002/hep.26550] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 05/22/2013] [Indexed: 01/01/2023]
Abstract
UNLABELLED The liver-derived peptide hepcidin controls the balance between iron demand and iron supply. By inhibiting the iron export activity of ferroportin, hepcidin modulates iron absorption and delivery from the body's stores. The regulation of hepcidin, however, is not completely understood and includes a variety of different signals. We studied iron metabolism and hepcidin expression in mice constitutively overexpressing erythropoietin (Epo) (Tg6 mice), which leads to excessive erythropoiesis. We observed a very strong down-regulation of hepcidin in Tg6 mice that was accompanied by a strong increase in duodenal expression of ferroportin and divalent metal tranporter-1, as well as enhanced duodenal iron absorption. Despite these compensatory mechanisms, Tg6 mice displayed marked circulating iron deficiency and low levels of iron in liver, spleen, and muscle. To elucidate the primary signal affecting hepcidin expression during chronically elevated erythropoiesis, we increased iron availability by either providing iron (thus further increasing the hematocrit) or reducing erythropoiesis-dependent iron consumption by means of splenectomy. Both treatments increased liver iron and up-regulated hepcidin expression and the BMP6/SMAD pathway despite continuously high plasma Epo levels and sustained erythropoiesis. This suggests that hepcidin expression is not controlled by erythropoietic signals directly in this setting. Rather, these results indicate that iron consumption for erythropoiesis modulates liver iron content, and ultimately BMP6 and hepcidin. Analysis of the BMP6/SMAD pathway targets showed that inhibitor of DNA binding 1 (ID1) and SMAD7, but not transmembrane serine protease 6 (TMPRSS6), were up-regulated by increased iron availability and thus may be involved in setting the upper limit of hepcidin. CONCLUSION We provide evidence that under conditions of excessive and effective erythropoiesis, liver iron regulates hepcidin expression through the BMP6/SMAD pathway.
Collapse
Affiliation(s)
- Víctor Díaz
- Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology (ZIHP), and University of Zurich, Switzerland; Department of Health and Human Performance, Faculty of Sports Science, INEF, Technical University of Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
240
|
Shanmugam NKN, Cherayil BJ. Serum-induced up-regulation of hepcidin expression involves the bone morphogenetic protein signaling pathway. Biochem Biophys Res Commun 2013; 441:383-6. [PMID: 24157792 DOI: 10.1016/j.bbrc.2013.10.065] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 10/13/2013] [Indexed: 12/20/2022]
Abstract
Hepcidin is a peptide hormone that is secreted by the liver and that functions as the central regulator of systemic iron metabolism in mammals. Its expression is regulated at the transcriptional level by changes in iron status and iron requirements, and by inflammatory cues. There is considerable interest in understanding the mechanisms that influence hepcidin expression because dysregulation of hepcidin production is associated with a number of disease states and can lead to iron overload or iron-restricted anemia. In order to shed light on the factors that alter hepcidin expression, we carried out experiments with HepG2 and HuH7, human hepatoma cell lines that are widely used for this purpose. We found that the addition of heat-inactivated fetal calf serum to these cells resulted in a significant dose- and time-dependent up-regulation of hepcidin expression. Serum also activated signaling events known to be downstream of bone morphogenetic proteins (BMPs), a group of molecules that have been implicated previously in hepcidin regulation. Inhibition of these signals with dorsomorphin significantly suppressed serum-induced hepcidin up-regulation. Our results indicate that a BMP or BMP-like molecule present in serum may play an important role in regulating hepcidin expression.
Collapse
Affiliation(s)
- Nanda Kumar N Shanmugam
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA 02114, USA
| | | |
Collapse
|
241
|
Linder MC. Mobilization of stored iron in mammals: a review. Nutrients 2013; 5:4022-50. [PMID: 24152745 PMCID: PMC3820057 DOI: 10.3390/nu5104022] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 09/04/2013] [Accepted: 09/12/2013] [Indexed: 12/16/2022] Open
Abstract
From the nutritional standpoint, several aspects of the biochemistry and physiology of iron are unique. In stark contrast to most other elements, most of the iron in mammals is in the blood attached to red blood cell hemoglobin and transporting oxygen to cells for oxidative phosphorylation and other purposes. Controlled and uncontrolled blood loss thus has a major impact on iron availability. Also, in contrast to most other nutrients, iron is poorly absorbed and poorly excreted. Moreover, amounts absorbed (~1 mg/day in adults) are much less than the total iron (~20 mg/day) cycling into and out of hemoglobin, involving bone marrow erythropoiesis and reticuloendothelial cell degradation of aged red cells. In the face of uncertainties in iron bioavailability, the mammalian organism has evolved a complex system to retain and store iron not immediately in use, and to make that iron available when and where it is needed. Iron is stored innocuously in the large hollow protein, ferritin, particularly in cells of the liver, spleen and bone marrow. Our current understanding of the molecular, cellular and physiological mechanisms by which this stored iron in ferritin is mobilized and distributed-within the cell or to other organs-is the subject of this review.
Collapse
Affiliation(s)
- Maria C Linder
- Department of Chemistry and Biochemistry, California State University, Fullerton, CA 92834-6866, USA.
| |
Collapse
|
242
|
Abstract
PURPOSE OF REVIEW The circulating peptide hepcidin modulates systemic iron balance by limiting the absorption of dietary iron and the release of iron from macrophage stores. Recent studies conducted in humans, animal models, and tissue culture systems have enhanced our understanding of the molecular mechanisms by which hepcidin levels are altered in response to iron stores, inflammation, and erythropoietic activity. RECENT FINDINGS The bone morphogenetic protein (BMP) type I receptors ALK2 and ALK3 play key, nonredundant roles in mediating hepcidin synthesis through the BMP signaling pathway. Actions of the hereditary hemochromatosis proteins HFE and transferrin receptor 2 may intersect with the BMP pathway. Hepcidin induction in response to inflammation requires cooperative BMP signaling. A variety of innate immune and infectious stimuli induce hepcidin expression. The hypoxia inducible factor pathway appears to suppress hepcidin indirectly through the capacity of erythropoietin to stimulate erythropoiesis. SUMMARY Study of the molecular mechanisms underlying the regulation of hepcidin synthesis has revealed complex biology. Improved understanding of the signaling pathways involved in hepcidin regulation may contribute to improved therapeutic outcomes for patients with genetic and acquired disorders that impact systemic iron balance.
Collapse
|
243
|
Zarjou A, Bolisetty S, Joseph R, Traylor A, Apostolov EO, Arosio P, Balla J, Verlander J, Darshan D, Kuhn LC, Agarwal A. Proximal tubule H-ferritin mediates iron trafficking in acute kidney injury. J Clin Invest 2013; 123:4423-34. [PMID: 24018561 DOI: 10.1172/jci67867] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 07/18/2013] [Indexed: 12/30/2022] Open
Abstract
Ferritin plays a central role in iron metabolism and is made of 24 subunits of 2 types: heavy chain and light chain. The ferritin heavy chain (FtH) has ferroxidase activity that is required for iron incorporation and limiting toxicity. The purpose of this study was to investigate the role of FtH in acute kidney injury (AKI) and renal iron handling by using proximal tubule-specific FtH-knockout mice (FtH(PT-/-) mice). FtH(PT-/-) mice had significant mortality, worse structural and functional renal injury, and increased levels of apoptosis in rhabdomyolysis and cisplatin-induced AKI, despite significantly higher expression of heme oxygenase-1, an antioxidant and cytoprotective enzyme. While expression of divalent metal transporter-1 was unaffected, expression of ferroportin (FPN) was significantly lower under both basal and rhabdomyolysis-induced AKI in FtH(PT-/-) mice. Apical localization of FPN was disrupted after AKI to a diffuse cytosolic and basolateral pattern. FtH, regardless of iron content and ferroxidase activity, induced FPN. Interestingly, urinary levels of the iron acceptor proteins neutrophil gelatinase-associated lipocalin, hemopexin, and transferrin were increased in FtH(PT-/-) mice after AKI. These results underscore the protective role of FtH and reveal the critical role of proximal tubule FtH in iron trafficking in AKI.
Collapse
|
244
|
Ferroportin in monocytes of hemodialysis patients and its associations with hepcidin, inflammation, markers of iron status and resistance to erythropoietin. Int Urol Nephrol 2013; 46:161-7. [PMID: 23860963 DOI: 10.1007/s11255-013-0497-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 06/17/2013] [Indexed: 12/17/2022]
Abstract
PURPOSE Disturbed iron homeostasis contributes to resistance to recombinant human erythropoietin (rHuEpo) in hemodialysis (HD) patients. Increased hepcidin, which downregulates the iron exporter ferroportin, has been incriminated. However, other factors also control ferroportin expression in mononuclear phagocyte system. Ferroportin in monocytes, as well as serum hepcidin, interleukin-6 (IL-6) and common markers of iron status were measured and correlations with rHuEpo resistance index (ERI) were evaluated. METHODS After a 4-week washout period from iron treatment, 34 HD patients and 20 healthy volunteers enrolled in the study. Ferroportin was assessed by means of western blotting, whereas hepcidin and IL-6 with enzyme-linked immunosorbent assay. Hemoglobin, serum iron, ferritin and transferrin saturation (TSAT) were also measured. RESULTS Ferroportin in monocytes of HD patients was decreased. Serum hepcidin and IL-6 were increased, whereas serum iron and TSAT were decreased. ERI was negatively correlated with ferroportin and all the markers of iron adequacy, but not with hepcidin. CONCLUSION Decreased ferroportin in monocytes of HD patients accompanies increased hepcidin, inflammation, decreased iron availability and is correlated with resistance to rHuEpo treatment.
Collapse
|
245
|
Mandilaras K, Pathmanathan T, Missirlis F. Iron absorption in Drosophila melanogaster. Nutrients 2013; 5:1622-47. [PMID: 23686013 PMCID: PMC3708341 DOI: 10.3390/nu5051622] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 05/03/2013] [Accepted: 05/07/2013] [Indexed: 12/20/2022] Open
Abstract
The way in which Drosophila melanogaster acquires iron from the diet remains poorly understood despite iron absorption being of vital significance for larval growth. To describe the process of organismal iron absorption, consideration needs to be given to cellular iron import, storage, export and how intestinal epithelial cells sense and respond to iron availability. Here we review studies on the Divalent Metal Transporter-1 homolog Malvolio (iron import), the recent discovery that Multicopper Oxidase-1 has ferroxidase activity (iron export) and the role of ferritin in the process of iron acquisition (iron storage). We also describe what is known about iron regulation in insect cells. We then draw upon knowledge from mammalian iron homeostasis to identify candidate genes in flies. Questions arise from the lack of conservation in Drosophila for key mammalian players, such as ferroportin, hepcidin and all the components of the hemochromatosis-related pathway. Drosophila and other insects also lack erythropoiesis. Thus, systemic iron regulation is likely to be conveyed by different signaling pathways and tissue requirements. The significance of regulating intestinal iron uptake is inferred from reports linking Drosophila developmental, immune, heat-shock and behavioral responses to iron sequestration.
Collapse
Affiliation(s)
- Konstantinos Mandilaras
- School of Biological and Chemical Sciences, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK; E-Mail:
| | - Tharse Pathmanathan
- Department of Physiology, Biophysics and Neuroscience, CINVESTAV-IPN, IPN Avenue 2508, Zacatenco, 07360, Mexico City, Mexico; E-Mail:
| | - Fanis Missirlis
- Department of Physiology, Biophysics and Neuroscience, CINVESTAV-IPN, IPN Avenue 2508, Zacatenco, 07360, Mexico City, Mexico; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +52-55-5747-3963; Fax: +52-55-5747-5713
| |
Collapse
|
246
|
Abstract
Iron is an essential nutrient that facilitates cell proliferation and growth. However, iron also has the capacity to engage in redox cycling and free radical formation. Therefore, iron can contribute to both tumour initiation and tumour growth; recent work has also shown that iron has a role in the tumour microenvironment and in metastasis. Pathways of iron acquisition, efflux, storage and regulation are all perturbed in cancer, suggesting that reprogramming of iron metabolism is a central aspect of tumour cell survival. Signalling through hypoxia-inducible factor (HIF) and WNT pathways may contribute to altered iron metabolism in cancer. Targeting iron metabolic pathways may provide new tools for cancer prognosis and therapy.
Collapse
Affiliation(s)
- Suzy V Torti
- Departments of Molecular, Microbial and Structural Biology, University of Connecticut Health Center, Farmington, Connecticut 06030, USA.
| | | |
Collapse
|
247
|
DeWitt MR, Chen P, Aschner M. Manganese efflux in Parkinsonism: insights from newly characterized SLC30A10 mutations. Biochem Biophys Res Commun 2013; 432:1-4. [PMID: 23357421 PMCID: PMC3594538 DOI: 10.1016/j.bbrc.2013.01.058] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 01/16/2013] [Indexed: 01/02/2023]
Abstract
Although manganese (Mn) is required for normal cellular function, overexposure to this metal may cause an extrapyramidal syndrome resembling Parkinson's disease (PD). Notably, high whole-blood Mn levels have been reported in patients with idiopathic PD. Because Mn is both essential at low dose and toxic at higher dose; its transport and homeostasis are tightly regulated. Previously, the only protein known to be operant in cellular Mn export was the iron-regulating transporter, ferroportin (Fpn). The causal role for Mn in PD has yet to be fully understood, but evidence of a familial predisposition to PD associated with Mn toxicity is mounting. A recently discovered mutation in SLC30A10 identified its gene product as putatively involved in Mn efflux. Patients with the SLC30A10 mutation display Parkinsonian-like gate disturbances and hypermanganesemia. This review will address Mn transport proteins, the newly discovered SLC30A10 mutations and their implications to Parkinsonism and Mn regulation.
Collapse
Affiliation(s)
- Margaret R. DeWitt
- Vanderbilt Center for Molecular Toxicology, Nashville, TN 37232-8552, USA
- Vanderbilt Brain Institute, Nashville, TN 37232-8552, USA
| | - Pan Chen
- Vanderbilt University Medical Center, Department of Pediatrics, Nashville, TN 37232-8552, USA
| | - Michael Aschner
- Vanderbilt Center for Molecular Toxicology, Nashville, TN 37232-8552, USA
- Vanderbilt Brain Institute, Nashville, TN 37232-8552, USA
- Vanderbilt University Medical Center, Department of Pediatrics, Nashville, TN 37232-8552, USA
| |
Collapse
|
248
|
The effect of lead exposure on brain iron homeostasis and the expression of DMT1/FP1 in the brain in developing and aged rats. Toxicol Lett 2012; 216:108-23. [PMID: 23219683 DOI: 10.1016/j.toxlet.2012.11.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 11/26/2012] [Accepted: 11/27/2012] [Indexed: 11/21/2022]
Abstract
The relation between lead (Pb) and iron (Fe) becomes increasingly concerned because they are both divalent metals that are absorbed by the same intestinal mechanism, and Pb exposure and Fe deficiency in the developmental brain, as well as Fe overload in the aged brain, can cause cognitive deficits. However, the interaction between Pb exposure and Fe status in the brain has not been established. Therefore, in the current study, we examined the effects of maternal ingestion of Pb in drinking water during gestation and lactation on the Fe status and the expression of divalent metal transporter 1 (DMT1) and ferroportin 1 (FP1) in the brain of offspring. The offspring were followed through old age, with measurements taken at postnatal week 3 (PNW3), 41 (PNW41) and 70 (PNW70). Pb exposure increases the Fe content in the old-aged rats' brain, which might be not subjected to DMT1 mediating, but may be associated with the decrease expression of FP1. Furthermore, the effect of Pb on FP1 expression is regulated at transcriptional and posttranscriptional levels. The perturbation in Fe homeostasis may contribute to the neurotoxicology consequences induced by Pb exposure, and FP1 may play a role in Pb-induced Fe cumulation in the brain.
Collapse
|
249
|
Castoldi M, Muckenthaler MU. Regulation of iron homeostasis by microRNAs. Cell Mol Life Sci 2012; 69:3945-52. [PMID: 22678662 PMCID: PMC11114850 DOI: 10.1007/s00018-012-1031-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 05/04/2012] [Accepted: 05/07/2012] [Indexed: 02/07/2023]
Abstract
Iron homeostasis is maintained at the cellular and systemic levels to assure adequate iron supply while preventing iron overload. The identification of genes mutated in patients with iron-related disorders or animal models with imbalances of iron homeostasis gave insight into the molecular mechanisms underlying processes critical for balancing iron levels, such as iron uptake, storage, export, and monitoring of available iron. MicroRNAs control genes involved in some of these processes adding an additional level of complexity to the regulation of iron metabolism. This review summarizes recent advances how miRNAs regulate iron homeostasis.
Collapse
Affiliation(s)
- Mirco Castoldi
- Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, 69120, Heidelberg, Germany,
| | | |
Collapse
|
250
|
Abstract
Ferroportin (FPN) is the sole iron export membrane protein identified in mammals that is abundantly expressed on absorptive enterocytes and macrophages, and is essential for physiological regulation of cellular iron. The expression of FPN is positively induced by cellular iron and is suppressed by liver hepcidin in response to either increased systemic iron or inflammatory stimuli. Hepcidin binds to cell surface FPN inducing FPN internalization followed by lysosomal degradation of the protein and consequently iron efflux from macrophages is blocked and there is suboptimal iron absorption by duodenal enterocytes. Dozens of FPN gene mutations have been identified in different ethnic populations and some of the mutations are associated with autosomal dominant iron overload disorder described as FPN disease or hemochromatosis type 4 that is distinct from hereditary hemochromatosis due to HFE mutations. Clinical manifestations of iron overload FPN disease can be classified into two groups according to whether there is selective macrophage iron loading or parenchymal and reticuloendothelial iron accumulation. There is evidence suggesting that altered hepcidin-FPN interaction can modulate host's response to infection. Resistance to hepcidin promotes iron egress from cells and this inhibits growth of intracellular pathogens. Conversely, iron retention due to loss of iron export activity by mutated FPN results in intracellular iron accumulation and a permissive environment for intracellular pathogens.
Collapse
Affiliation(s)
- Ishmael Kasvosve
- Department of Medical Laboratory Sciences Faculty of Health Sciences, University of Botswana Private Bag UB 00712, Gaborone, Botswana.
| |
Collapse
|