1
|
Lu C, Tan C, Ouyang H, Chen Z, Yan Z, Zhang M. Ferroptosis in Intracerebral Hemorrhage: A Panoramic Perspective of the Metabolism, Mechanism and Theranostics. Aging Dis 2022; 13:1348-1364. [PMID: 36186133 PMCID: PMC9466971 DOI: 10.14336/ad.2022.01302] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/30/2022] [Indexed: 11/22/2022] Open
Abstract
Iron is one of the most crucial elements in the human body. In recent years, a kind of programmed, non-apoptotic cell death closely related to iron metabolism-called ferroptosis- has aroused much interest among many scientists. Ferroptosis also interacts with other pathways involved in cell death including iron abnormality, the cystine/glutamate antiporter and lipid peroxidation. Together these pathological pathways exert great impacts on intracerebral hemorrhage (ICH), a lethal cerebrovascular disease with a high incidence rate and mortality rate. Furthermore, the ferroptosis also affects different brain cells (neurons and neuroglial cells) and different organelles (mitochondria and endoplasmic reticulum). Clinical treatments for ferroptosis in ICH have been closely investigated recently. This perspective provides a comprehensive summary of ferroptosis mechanisms after ICH and its interaction with other cell death patterns. Understanding the role of ferroptosis in ICH will open new windows for the future treatments and preventions for ICH and other intracerebral diseases.
Collapse
Affiliation(s)
- Chenxiao Lu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Xiangya School of Medicine, Central South University, Changsha, 410031, China
| | - Changwu Tan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Xiangya School of Medicine, Central South University, Changsha, 410031, China
| | - Hongfei Ouyang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Xiangya School of Medicine, Central South University, Changsha, 410031, China
| | - Zhuohui Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.
| | - Zhouyi Yan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Mengqi Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Correspondence should be addressed to: Dr. Mengqi Zhang, Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China. ..
| |
Collapse
|
2
|
Silva AM, Moniz T, de Castro B, Rangel M. Human transferrin: An inorganic biochemistry perspective. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
3
|
Kosman DJ. A holistic view of mammalian (vertebrate) cellular iron uptake. Metallomics 2021; 12:1323-1334. [PMID: 32766655 DOI: 10.1039/d0mt00065e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cell iron uptake in mammals is commonly distinguished by whether the iron is presented to the cell as transferrin-bound or not: TBI or NTBI. This generic perspective conflates TBI with canonical transferrin receptor, endosomal iron uptake, and NTBI with uptake supported by a plasma membrane-localized divalent metal ion transporter, most often identified as DMT1. In fact, iron uptake by mammalian cells is far more nuanced than this somewhat proscribed view suggests. This view fails to accommodate the substantial role that ZIP8 and ZIP14 play in iron uptake, while adhering to the traditional premise that a relatively high endosomal [H+] is thermodynamically required for release of iron from holo-Tf. The canonical view of iron uptake also does not encompass the fact that plasma membrane electron transport - PMET - has long been linked to cell iron uptake. In fact, the known mammalian metallo-reductases - Dcytb and the STEAP proteins - are members of this cohort of cytochrome-dependent oxido-reductases that shuttle reducing equivalents across the plasma membrane. A not commonly appreciated fact is the reduction potential of ferric iron in holo-Tf is accessible to cytoplasmic reducing equivalents - reduced pyridine and flavin mono- and di-nucleotides and dihydroascorbic acid. This allows for the reductive release of Fe2+ at the extracellular surface of the PM and subsequent transport into the cytoplasm by a neutral pH transporter - a ZIP protein. What this perspective emphasizes is that there are two TfR-dependent uptake pathways, one which does and one which does not involve clathrin-dependent, endolysosomal trafficking. This raises the question as to the selective advantage of having two Tf, TfR-dependent routes of iron accumulation. This review of canonical and non-canonical iron uptake uses cerebral iron trafficking as a point of discussion, a focus that encourages inclusion also of the importance of ferritin as a circulating 'chaperone' of ferric iron.
Collapse
Affiliation(s)
- Daniel J Kosman
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, The University of Buffalo, Suite 4102, 995 Main St., Buffalo, NY 14203, USA.
| |
Collapse
|
4
|
Kulakova A, Indrakumar S, Sønderby P, Gentiluomo L, Streicher W, Roessner D, Frieß W, Peters GHJ, Harris P. Small angle X-ray scattering and molecular dynamic simulations provide molecular insight for stability of recombinant human transferrin. JOURNAL OF STRUCTURAL BIOLOGY-X 2020; 4:100017. [PMID: 32647821 PMCID: PMC7337065 DOI: 10.1016/j.yjsbx.2019.100017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/15/2019] [Accepted: 11/25/2019] [Indexed: 12/20/2022]
Abstract
Combination of SAXS and MD simulations can explain the molecular background for the stability studies. Destabilizing excipients in general induce opening of the N-lobe by binding to specific residues in the protein. The fully open conformation of transferrin is not seen without aggregation.
Transferrin is an attractive candidate for drug delivery due to its ability to cross the blood brain barrier. However, in order to be able to use it for therapeutic purposes, it is important to investigate how its stability depends on different formulation conditions. Combining high-throughput thermal and chemical denaturation studies with small angle X-ray scattering (SAXS) and molecular dynamics (MD) simulations, it was possible to connect the stability of transferrin with its conformational changes. Lowering pH induces opening of the transferrin N-lobe, which results in a negative effect on the stability. Presence of NaCl or arginine at low pH enhances the opening and has a negative impact on the overall protein stability. Statement of Significance Protein-based therapeutics have become an essential part of medical treatment. They are highly specific, have high affinity and fewer off-target effects. However, stabilization of proteins is critical, time-consuming, and expensive, and it is not yet possible to predict the behavior of proteins under different conditions. The current work is focused on a molecular understanding of the stability of human serum transferrin; a protein which is abundant in blood serum, may pass the blood brain barrier and therefore with high potential in drug delivery. Combination of high throughput unfolding techniques and structural studies, using small angle X-ray scattering and molecular dynamic simulations, allows us to understand the behavior of transferrin on a molecular level.
Collapse
Affiliation(s)
- Alina Kulakova
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800 Kgs. Lyngby, Denmark
| | - Sowmya Indrakumar
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800 Kgs. Lyngby, Denmark
| | - Pernille Sønderby
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800 Kgs. Lyngby, Denmark
| | - Lorenzo Gentiluomo
- Wyatt Technology Europe GmbH, Hochstrasse 18, 56307 Dernbach, Germany.,Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-University of Munich, Butenandtstrasse 5, 81377 Munich, Germany
| | | | - Dierk Roessner
- Wyatt Technology Europe GmbH, Hochstrasse 18, 56307 Dernbach, Germany
| | - Wolfgang Frieß
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-University of Munich, Butenandtstrasse 5, 81377 Munich, Germany
| | - Günther H J Peters
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800 Kgs. Lyngby, Denmark
| | - Pernille Harris
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800 Kgs. Lyngby, Denmark
| |
Collapse
|
5
|
Reilley DJ, Fuller JT, Nechay MR, Victor M, Li W, Ruberry JD, Mujika JI, Lopez X, Alexandrova AN. Toxic and Physiological Metal Uptake and Release by Human Serum Transferrin. Biophys J 2020; 118:2979-2988. [PMID: 32497515 PMCID: PMC7300305 DOI: 10.1016/j.bpj.2020.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/03/2020] [Accepted: 04/29/2020] [Indexed: 10/24/2022] Open
Abstract
An atomistic understanding of metal transport in the human body is critical to anticipate the side effects of metal-based therapeutics and holds promise for new drugs and drug delivery designs. Human serum transferrin (hTF) is a central part of the transport processes because of its ubiquitous ferrying of physiological Fe(III) and other transition metals to tightly controlled parts of the body. There is an atomistic mechanism for the uptake process with Fe(III), but not for the release process, or for other metals. This study provides initial insight into these processes for a range of transition metals-Ti(IV), Co(III), Fe(III), Ga(III), Cr(III), Fe(II), Zn(II)-through fully atomistic, extensive quantum mechanical/discrete molecular dynamics sampling and provides, to our knowledge, a new technique we developed to calculate relative binding affinities between metal cations and the protein. It identifies protonation of Tyr188 as a trigger for metal release rather than protonation of Lys206 or Lys296. The study identifies the difficulty of metal release from hTF as potentially related to cytotoxicity. Simulations identify a few critical interactions that stabilize the metal binding site in a flexible, nuanced manner.
Collapse
Affiliation(s)
- David J Reilley
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California
| | - Jack T Fuller
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California
| | - Michael R Nechay
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California
| | - Marie Victor
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California; Institut Lumire Matire, Villeurbanne, France
| | - Wei Li
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts
| | - Josiah D Ruberry
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California
| | - Jon I Mujika
- Kimika Fakultatea, Euskal Herriko Unibertsitatea (UPV/EHU) and Donostia, International Physics Center, Donostia, Euskadi, Spain
| | - Xabier Lopez
- Kimika Fakultatea, Euskal Herriko Unibertsitatea (UPV/EHU) and Donostia, International Physics Center, Donostia, Euskadi, Spain
| | - Anastassia N Alexandrova
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
6
|
Wehrstedt S, Kubis J, Zimmermann A, Bruns H, Mayer D, Grieshober M, Stenger S. The tyrosine kinase inhibitor dasatinib reduces the growth of intracellular Mycobacterium tuberculosis despite impairing T-cell function. Eur J Immunol 2018; 48:1892-1903. [PMID: 30242834 DOI: 10.1002/eji.201847656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/10/2018] [Accepted: 09/20/2018] [Indexed: 12/31/2022]
Abstract
Tyrosine kinases are checkpoints for multiple cellular pathways and dysregulation induces malignancies, most notably chronic myeloid leukemia (CML). Inhibition of Abl-tyrosine kinases has evolved as a new concept for the treatment of CML and other malignant diseases. Due to the multiple immune-modulatory pathways controlled by tyrosine kinases, treatment with tyrosine kinase inhibitors (TKIs) will not only affect the biology of malignant cells but also modulate physiological immune functions. To understand the effects of TKIs on host defense against intracellular bacteria, we investigated the immunological impact of the dual Abl/Src TKI dasatinib on the cellular immune response to Mycobacterium tuberculosis (Mtb). Our results demonstrate that dasatinib impaired proliferation, cytokine release (IFN-γ, TNF-α, GM-CSF), expression of granulysin and degranulation of cytotoxic effector molecules of human Mtb-specific T-lymphocytes by inhibition of lymphocyte-specific protein tyrosine kinase (Lck) phosphorylation. Despite this profound inhibition of T-cell function, dasatinib suppressed growth of virulent Mtb in human macrophages co-cultured with autologous Mtb-specific T-cells (49±15%). Functional analysis suggested that growth inhibition is due to dasatinib-triggered lysosomal acidification in Mtb-infected macrophages. These results highlight the significance of innate immune responses, i.e. acidification of lysosomes, which control the multiplication of intracellular bacteria despite the lack of efficient T-cell support.
Collapse
Affiliation(s)
- Stephanie Wehrstedt
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Germany
| | - Jan Kubis
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Germany
| | - Andreas Zimmermann
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Germany
| | - Heiko Bruns
- Department of Internal Medicine 5, Hematology/Oncology, University Hospital Erlangen, Germany
| | - Daniel Mayer
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Germany
| | - Mark Grieshober
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Germany
| | - Steffen Stenger
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Germany
| |
Collapse
|
7
|
Harris MT, Hussain SS, Inouye CM, Castle AM, Castle JD. Reinterpretation of the localization of the ATP binding cassette transporter ABCG1 in insulin-secreting cells and insights regarding its trafficking and function. PLoS One 2018; 13:e0198383. [PMID: 30235209 PMCID: PMC6147399 DOI: 10.1371/journal.pone.0198383] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/04/2018] [Indexed: 01/08/2023] Open
Abstract
The ABC transporter ABCG1 contributes to the regulation of cholesterol efflux from cells and to the distribution of cholesterol within cells. We showed previously that ABCG1 deficiency inhibits insulin secretion by pancreatic beta cells and, based on its immunolocalization to insulin granules, proposed its essential role in forming granule membranes that are enriched in cholesterol. While we confirm elsewhere that ABCG1, alongside ABCA1 and oxysterol binding protein OSBP, supports insulin granule formation, the aim here is to clarify the localization of ABCG1 within insulin-secreting cells and to provide added insight regarding ABCG1's trafficking and sites of function. We show that stably expressed GFP-tagged ABCG1 closely mimics the distribution of endogenous ABCG1 in pancreatic INS1 cells and accumulates in the trans-Golgi network (TGN), endosomal recycling compartment (ERC) and on the cell surface but not on insulin granules, early or late endosomes. Notably, ABCG1 is short-lived, and proteasomal and lysosomal inhibitors both decrease its degradation. Following blockade of protein synthesis, GFP-tagged ABCG1 first disappears from the ER and TGN and later from the ERC and plasma membrane. In addition to aiding granule formation, our findings raise the prospect that ABCG1 may act beyond the TGN to regulate activities involving the endocytic pathway, especially as the amount of transferrin receptor is increased in ABCG1-deficient cells. Thus, ABCG1 may function at multiple intracellular sites and the plasma membrane as a roving sensor and modulator of cholesterol distribution, membrane trafficking and cholesterol efflux.
Collapse
Affiliation(s)
- Megan T. Harris
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Syed Saad Hussain
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Candice M. Inouye
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Anna M. Castle
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - J. David Castle
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| |
Collapse
|
8
|
Ciarlone GE, Dean JB. Acute hypercapnic hyperoxia stimulates reactive species production in the caudal solitary complex of rat brain slices but does not induce oxidative stress. Am J Physiol Cell Physiol 2016; 311:C1027-C1039. [DOI: 10.1152/ajpcell.00161.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/05/2016] [Indexed: 12/28/2022]
Abstract
Central CO2 chemoreceptive neurons in the caudal solitary complex (cSC) are stimulated by hyperoxia via a free radical mechanism. Hyperoxia has been shown to increase superoxide and nitric oxide in the cSC, but it remains unknown how changes in Pco2 during hyperoxia affect the production of O2-dependent reactive oxygen and nitrogen species (RONS) downstream that can lead to increased levels of oxidative and nitrosative stress, cellular excitability, and, potentially, dysfunction. We used real-time fluorescence microscopy in rat brain slices to determine how hyperoxia and hypercapnic acidosis (HA) modulate one another in the production of key RONS, as well as colorimetric assays to measure levels of oxidized and nitrated lipids and proteins. We also examined the effects of CO2 narcosis and hypoxia before euthanasia and brain slice harvesting, as these neurons are CO2 sensitive and hypothesized to employ CO2/H+ mechanisms that exacerbate RONS production and potentially oxidative stress. Our findings show that hyperoxia ± HA increases the production of peroxynitrite and its derivatives, whereas increases in Fenton chemistry are most prominent during hyperoxia + HA. Using CO2 narcosis before euthanasia modulates cellular sensitivity to HA postmortem and enhances the magnitude of the peroxynitrite pathway, but blunts the activity of Fenton chemistry. Overall, hyperoxia and HA do not result in increased production of markers of oxidative and nitrosative stress as expected. We postulate this is due to antioxidant and proteosomal removal of damaged lipids and proteins to maintain cell viability and avoid death during protracted hyperoxia.
Collapse
Affiliation(s)
- Geoffrey E. Ciarlone
- Department of Molecular Pharmacology and Physiology, Hyperbaric Biomedical Research Laboratory, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Jay B. Dean
- Department of Molecular Pharmacology and Physiology, Hyperbaric Biomedical Research Laboratory, Morsani College of Medicine, University of South Florida, Tampa, Florida
| |
Collapse
|
9
|
Khan P, Shandilya A, Jayaram B, Islam A, Ahmad F, Hassan MI. Effect of pH on the stability of hemochromatosis factor E: a combined spectroscopic and molecular dynamics simulation-based study. J Biomol Struct Dyn 2016; 35:1582-1598. [PMID: 27174123 DOI: 10.1080/07391102.2016.1189359] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Hereditary hemochromatosis is an iron overburden condition, which is mainly governed by hereditary hemochromatosis factor E (HFE), a member of major histocompatibility complex class I. To understand the effect of pH on the structure and stability of HFE, we have cloned, expressed, and purified the HFE in the bacterial system and performed circular dichroism, fluorescence, and absorbance measurements at a wide pH range (pH 3.0-11.0). We found that HFE remains stable in the pH range 7.5-11.0 and gets completely acid denatured at low pH values. In this work, we also analyzed the contribution of salt bridges to the stability of HFE. We further performed molecular dynamics simulations for 80 ns at different pH values. An excellent agreement was observed between results from biophysical and MD simulation studies. At lower pH, HFE undergoes denaturation and may be driven toward a degradation pathway, such as ubiquitination. Hence, HFE is not available to bind again with transferrin receptor1 to negatively regulate iron homeostasis. Further we postulated that, might be low pH of cancerous cells helps them to meet their high iron requirement.
Collapse
Affiliation(s)
- Parvez Khan
- a Centre for Interdisciplinary Research in Basic Sciences , Jamia Millia Islamia , New Delhi 110025 , India
| | - Ashutosh Shandilya
- b Department of Chemistry , Indian Institute of Technology Delhi , New Delhi 110016 , India
| | - B Jayaram
- b Department of Chemistry , Indian Institute of Technology Delhi , New Delhi 110016 , India
| | - Asimul Islam
- a Centre for Interdisciplinary Research in Basic Sciences , Jamia Millia Islamia , New Delhi 110025 , India
| | - Faizan Ahmad
- a Centre for Interdisciplinary Research in Basic Sciences , Jamia Millia Islamia , New Delhi 110025 , India
| | - Md Imtaiyaz Hassan
- a Centre for Interdisciplinary Research in Basic Sciences , Jamia Millia Islamia , New Delhi 110025 , India
| |
Collapse
|
10
|
Mirshafiey A, Simhag A, El Rouby NMM, Azizi G. T-helper 22 cells as a new player in chronic inflammatory skin disorders. Int J Dermatol 2015; 54:880-8. [PMID: 26183243 DOI: 10.1111/ijd.12883] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 09/01/2014] [Accepted: 10/19/2014] [Indexed: 12/30/2022]
Abstract
T-helper 22 (Th22) cell is a new subset of CD4+ T cells that secrets interleukin (IL)-22 but not IL-17 or interferon-γ. Th22 is distinct from Th17 and other known CD4+ T-cell subsets with distinguished gene expression and function. Th22 subsets have chemokine receptors CCR6+ CCR4+ CCR10+ phenotype and aryl hydrocarbon receptor as the key transcription factor. This T-helper subset, by producing cytokines such as IL-22, IL-13, and tumor necrosis factor-α, is implicated in the pathogenesis of inflammatory skin disorder. This review discusses the role of Th22 and its cytokine IL-22 in the immunopathogenesis of inflammatory skin disorders such as psoriasis and atopic dermatitis.
Collapse
Affiliation(s)
- Abbas Mirshafiey
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Anita Simhag
- Karolinska Institutet Science Park AB, Huddinge, Sweden
| | | | - Gholamreza Azizi
- Imam Hassan Mojtaba Hospital, Alborz University of Medical Sciences, Karaj, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Ali S. Questions and answers. THE EGYPTIAN JOURNAL OF INTERNAL MEDICINE 2015. [DOI: 10.4103/1110-7782.159479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
12
|
Increased plasma catalytic iron in patients may mediate acute kidney injury and death following cardiac surgery. Kidney Int 2015; 87:1046-54. [PMID: 25565307 DOI: 10.1038/ki.2014.374] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 09/10/2014] [Accepted: 09/18/2014] [Indexed: 02/03/2023]
Abstract
Catalytic iron, the chemical form of iron capable of participating in redox cycling, is a key mediator of acute kidney injury (AKI) in multiple animal models, but its role in human AKI has not been studied. Here we tested in a prospective cohort of 250 patients undergoing cardiac surgery whether plasma catalytic iron levels are elevated and associated with the composite outcome of AKI requiring renal replacement therapy or in-hospital mortality. Plasma catalytic iron, free hemoglobin, and other iron parameters were measured preoperatively, at the end of cardiopulmonary bypass, and on postoperative days 1 and 3. Plasma catalytic iron levels, but not other iron parameters, rose significantly at the end of cardiopulmonary bypass and were directly associated with bypass time and number of packed red blood cell transfusions. In multivariate analyses adjusting for age and preoperative eGFR, patients in the highest compared with the lowest quartile of catalytic iron on postoperative day 1 had a 6.71 greater odds of experiencing the primary outcome, and also had greater odds of AKI, hospital mortality, and postoperative myocardial injury. Thus, our data are consistent with and expand on findings from animal models demonstrating a pathologic role of catalytic iron in mediating adverse postoperative outcomes. Interventions aimed at reducing plasma catalytic iron levels as a strategy for preventing AKI in humans are warranted.
Collapse
|
13
|
Iron homeostasis in breast cancer. Cancer Lett 2014; 347:1-14. [DOI: 10.1016/j.canlet.2014.01.029] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/16/2013] [Accepted: 01/24/2014] [Indexed: 02/08/2023]
|
14
|
The herpes virus Fc receptor gE-gI mediates antibody bipolar bridging to clear viral antigens from the cell surface. PLoS Pathog 2014; 10:e1003961. [PMID: 24604090 PMCID: PMC3946383 DOI: 10.1371/journal.ppat.1003961] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 01/16/2014] [Indexed: 11/19/2022] Open
Abstract
The Herpes Simplex Virus 1 (HSV-1) glycoprotein gE-gI is a transmembrane Fc receptor found on the surface of infected cells and virions that binds human immunoglobulin G (hIgG). gE-gI can also participate in antibody bipolar bridging (ABB), a process by which the antigen-binding fragments (Fabs) of the IgG bind a viral antigen while the Fc binds to gE-gI. IgG Fc binds gE-gI at basic, but not acidic, pH, suggesting that IgG bound at extracellular pH by cell surface gE-gI would dissociate and be degraded in acidic endosomes/lysosomes if endocytosed. The fate of viral antigens associated with gE-gI-bound IgG had been unknown: they could remain at the cell surface or be endocytosed with IgG. Here, we developed an in vitro model system for ABB and investigated the trafficking of ABB complexes using 4-D confocal fluorescence imaging of ABB complexes with transferrin or epidermal growth factor, well-characterized intracellular trafficking markers. Our data showed that cells expressing gE-gI and the viral antigen HSV-1 gD endocytosed anti-gD IgG and gD in a gE-gI-dependent process, resulting in lysosomal localization. These results suggest that gE-gI can mediate clearance of infected cell surfaces of anti-viral host IgG and viral antigens to evade IgG-mediated responses, representing a general mechanism for viral Fc receptors in immune evasion and viral pathogenesis.
Collapse
|
15
|
Dumas Z, Ross-Gillespie A, Kümmerli R. Switching between apparently redundant iron-uptake mechanisms benefits bacteria in changeable environments. Proc Biol Sci 2013; 280:20131055. [PMID: 23760867 DOI: 10.1098/rspb.2013.1055] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Bacteria often possess multiple siderophore-based iron uptake systems for scavenging this vital resource from their environment. However, some siderophores seem redundant, because they have limited iron-binding efficiency and are seldom expressed under iron limitation. Here, we investigate the conundrum of why selection does not eliminate this apparent redundancy. We focus on Pseudomonas aeruginosa, a bacterium that can produce two siderophores-the highly efficient but metabolically expensive pyoverdine, and the inefficient but metabolically cheap pyochelin. We found that the bacteria possess molecular mechanisms to phenotypically switch from mainly producing pyoverdine under severe iron limitation to mainly producing pyochelin when iron is only moderately limited. We further show that strains exclusively producing pyochelin grew significantly better than strains exclusively producing pyoverdine under moderate iron limitation, whereas the inverse was seen under severe iron limitation. This suggests that pyochelin is not redundant, but that switching between siderophore strategies might be beneficial to trade off efficiencies versus costs of siderophores. Indeed, simulations parameterized from our data confirmed that strains retaining the capacity to switch between siderophores significantly outcompeted strains defective for one or the other siderophore under fluctuating iron availabilities. Finally, we discuss how siderophore switching can be viewed as a form of collective decision-making, whereby a coordinated shift in behaviour at the group level emerges as a result of positive and negative feedback loops operating among individuals at the local scale.
Collapse
Affiliation(s)
- Zoé Dumas
- Department of Ecology and Evolution, University of Lausanne, , Biophore Building, 1015 Lausanne, Switzerland
| | | | | |
Collapse
|
16
|
Iron-sulphur clusters, their biosynthesis, and biological functions in protozoan parasites. ADVANCES IN PARASITOLOGY 2013; 83:1-92. [PMID: 23876871 DOI: 10.1016/b978-0-12-407705-8.00001-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Fe-S clusters are ensembles of sulphide-linked di-, tri-, and tetra-iron centres of a variety of metalloproteins that play important roles in reduction and oxidation of mitochondrial electron transport, energy metabolism, regulation of gene expression, cell survival, nitrogen fixation, and numerous other metabolic pathways. The Fe-S clusters are assembled by one of four distinct systems: NIF, SUF, ISC, and CIA machineries. The ISC machinery is a house-keeping system conserved widely from prokaryotes to higher eukaryotes, while the other systems are present in a limited range of organisms and play supplementary roles under certain conditions such as stress. Fe-S cluster-containing proteins and the components required for Fe-S cluster biosynthesis are modulated under stress conditions, drug resistance, and developmental stages. It is also known that a defect in Fe-S proteins and Fe-S cluster biogenesis leads to many genetic disorders in humans, which indicates the importance of the systems. In this review, we describe the biological and physiological significance of Fe-S cluster-containing proteins and their biosynthesis in parasitic protozoa including Plasmodium, Trypanosoma, Leishmania, Giardia, Trichomonas, Entamoeba, Cryptosporidium, Blastocystis, and microsporidia. We also discuss the roles of Fe-S cluster biosynthesis in proliferation, differentiation, and stress response in protozoan parasites. The heterogeneity of the systems and the compartmentalization of Fe-S cluster biogenesis in the protozoan parasites likely reflect divergent evolution under highly diverse environmental niches, and influence their parasitic lifestyle and pathogenesis. Finally, both Fe-S cluster-containing proteins and their biosynthetic machinery in protozoan parasites are remarkably different from those in their mammalian hosts. Thus, they represent a rational target for the development of novel chemotherapeutic and prophylactic agents against protozoan infections.
Collapse
|
17
|
Chen C, Paw BH. Cellular and mitochondrial iron homeostasis in vertebrates. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:1459-67. [PMID: 22285816 DOI: 10.1016/j.bbamcr.2012.01.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 01/07/2012] [Accepted: 01/07/2012] [Indexed: 02/08/2023]
Abstract
Iron plays an essential role in cellular metabolism and biological processes. However, due to its intrinsic redox activity, free iron is a potentially toxic molecule in cellular biochemistry. Thus, organisms have developed sophisticated ways to import, sequester, and utilize iron. The transferrin cycle is a well-studied iron uptake pathway that is important for most vertebrate cells. Circulating iron can also be imported into cells by mechanisms that are independent of transferrin. Once imported into erythroid cells, iron is predominantly consumed by the mitochondria for the biosynthesis of heme and iron sulfur clusters. This review focuses on canonical transferrin-mediated and the newly discovered, non-transferrin mediated iron uptake pathways, as well as, mitochondrial iron homeostasis in higher eukaryotes. This article is part of a Special Issue entitled: Cell Biology of Metals.
Collapse
Affiliation(s)
- Caiyong Chen
- Department of Medicine, Hematology Division, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
18
|
Iron transport machinery of human cells: players and their interactions. CURRENT TOPICS IN MEMBRANES 2012; 69:67-93. [PMID: 23046647 DOI: 10.1016/b978-0-12-394390-3.00003-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Organisms, like cells, maintain tight control of iron. In humans as well as other mammals, control is achieved through the regulation of iron uptake into the body rather than through the excretion of iron. The mechanisms by which humans and mice regulate both iron uptake and the distribution of iron within the body and cells are reviewed. Special emphasis is given to the iron transporters involved in this process.
Collapse
|
19
|
Leitner DF, Connor JR. Functional roles of transferrin in the brain. Biochim Biophys Acta Gen Subj 2011; 1820:393-402. [PMID: 22138408 DOI: 10.1016/j.bbagen.2011.10.016] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 10/13/2011] [Accepted: 10/24/2011] [Indexed: 12/11/2022]
Abstract
BACKGROUND Transferrin is synthesized in the brain by choroid plexus and oligodendrocytes, but only that in the choroid plexus is secreted. Transferrin is a major iron delivery protein to the brain, but the amount transcytosed across the brain microvasculature is minimal. Transferrin is the major source of iron delivery to neurons. It may deliver iron to immature oligodendrocytes but this trophic effect declines over time while iron requirements for maintaining myelination continue. Finally, transferrin may play an important role in neurodegenerative diseases through its ability to mobilize iron. SCOPE OF REVIEW The role of transferrin in maintaining brain iron homeostasis and the mechanism by which it enters the brain and delivers iron will be discussed. Its relevance to neurological disorders will also be addressed. MAJOR CONCLUSIONS Transferrin is the major iron delivery protein for neurons and the microvasculature, but has a limited role for glial cells. The main source of transferrin in the brain is likely from the choroid plexus although the concentration of transferrin at any given time in the brain includes that synthesized in oligodendrocytes. Little is known about brain iron egress or the role of transferrin in this process. GENERAL SIGNIFICANCE Neuron survival requires iron, which is predominantly delivered by transferrin. The concentration of transferrin in the cerebrospinal fluid is reflective of brain iron availability and can function as a biomarker in disease. Accumulation of iron in the brain contributes to neurodegenerative processes, thus an understanding of the role that transferrin plays in regulating brain iron homeostasis is essential. This article is part of a Special Issue entitled Transferrins: Molecular mechanisms of iron transport and disorders.
Collapse
Affiliation(s)
- Dominique F Leitner
- Department of Neurosurgery, Penn State University, M.S. Hershey Medical Center, 500 University Dr., Hershey, PA 17033-0850, USA
| | | |
Collapse
|
20
|
Regulation of brain iron and copper homeostasis by brain barrier systems: implication in neurodegenerative diseases. Pharmacol Ther 2011; 133:177-88. [PMID: 22115751 DOI: 10.1016/j.pharmthera.2011.10.006] [Citation(s) in RCA: 215] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 10/27/2011] [Indexed: 12/21/2022]
Abstract
Iron (Fe) and copper (Cu) are essential to neuronal function; excess or deficiency of either is known to underlie the pathoetiology of several commonly known neurodegenerative disorders. This delicate balance of Fe and Cu in the central milieu is maintained by the brain barrier systems, i.e., the blood-brain barrier (BBB) between the blood and brain interstitial fluid and the blood-cerebrospinal fluid barrier (BCB) between the blood and cerebrospinal fluid (CSF). This review provides a concise description on the structural and functional characteristics of the brain barrier systems. Current understanding of Fe and Cu transport across the brain barriers is thoroughly examined, with major focuses on whether the BBB and BCB coordinate the direction of Fe and Cu fluxes between the blood and brain/CSF. In particular, the mechanism by which pertinent metal transporters in the barriers, such as the transferrin receptor (TfR), divalent metal transporter (DMT1), copper transporter (CTR1), ATP7A/B, and ferroportin (FPN), regulate metal movement across the barriers is explored. Finally, the detrimental consequences of dysfunctional metal transport by brain barriers, as a result of endogenous disorders or exogenous insults, are discussed. Understanding the regulation of Fe and Cu homeostasis in the central nervous system aids in the design of new drugs targeted on the regulatory proteins at the brain barriers for the treatment of metal's deficiency or overload-related neurological diseases.
Collapse
|
21
|
Chen J, Enns CA. Hereditary hemochromatosis and transferrin receptor 2. Biochim Biophys Acta Gen Subj 2011; 1820:256-63. [PMID: 21864651 DOI: 10.1016/j.bbagen.2011.07.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 07/26/2011] [Accepted: 07/29/2011] [Indexed: 02/08/2023]
Abstract
BACKGROUND Multicellular organisms regulate the uptake of calories, trace elements, and other nutrients by complex feedback mechanisms. In the case of iron, the body senses internal iron stores, iron requirements for hematopoiesis, and inflammatory status, and regulates iron uptake by modulating the uptake of dietary iron from the intestine. Both the liver and the intestine participate in the coordination of iron uptake and distribution in the body. The liver senses inflammatory signals and iron status of the organism and secretes a peptide hormone, hepcidin. Under high iron or inflammatory conditions hepcidin levels increase. Hepcidin binds to the iron transport protein, ferroportin (FPN), promoting FPN internalization and degradation. Decreased FPN levels reduce iron efflux out of intestinal epithelial cells and macrophages into the circulation. Derangements in iron metabolism result in either the abnormal accumulation of iron in the body, or in anemias. The identification of the mutations that cause the iron overload disease, hereditary hemochromatosis (HH), or iron-refractory iron-deficiency anemia has revealed many of the proteins used to regulate iron uptake. SCOPE OF THE REVIEW In this review we discuss recent data concerning the regulation of iron homeostasis in the body by the liver and how transferrin receptor 2 (TfR2) affects this process. MAJOR CONCLUSIONS TfR2 plays a key role in regulating iron homeostasis in the body. GENERAL SIGNIFICANCE The regulation of iron homeostasis is important. One third of the people in the world are anemic. HH is the most common inherited disease in people of Northern European origin and can lead to severe health complications if left untreated. This article is part of a Special Issue entitled Transferrins: Molecular mechanisms of iron transport and disorders.
Collapse
Affiliation(s)
- Juxing Chen
- Department of Cell and Developmental Biology L215, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, USA
| | | |
Collapse
|
22
|
Ciudin A, Hernández C, Simó R. Iron overload in diabetic retinopathy: a cause or a consequence of impaired mechanisms? EXPERIMENTAL DIABETES RESEARCH 2010; 2010:714108. [PMID: 20827392 PMCID: PMC2935195 DOI: 10.1155/2010/714108] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Accepted: 06/29/2010] [Indexed: 11/17/2022]
Abstract
Iron is an essential ion for life, playing a central role in many metabolic processes. The most important property of free iron is its capacity to be reversibly oxidized and reduced, but at same time this make it highly pro-oxidant molecule. In this regard, iron is able to generate powerful reactive oxygen species (ROS). For this reason, careful control on iron availability is central to the maintenance of normal cell function in the retina. In the diabetic eye there is an impairment of iron homeostasis, thus leading to iron overload. The mechanisms involved in this process include: (1) Destruction of heme molecules induced by hyperglycemia (2) Intraretinal and vitreal hemorrhages (3) Overexpression of the renin-angiotensin system. The main consequences of iron overload are the following: (1) Retinal neurodegeneration due to the increase of oxidative stress (2) Increase of AGE-RAGE binding (3) Defective phagocytosis of retinal pigment epithelium, which generates the accumulation of autoantigens and the synthesis of proinflammatory cytokines. Further studies addressed to explore not only the role of iron in the pathogenesis of diabetic retinopathy, but also to design novel therapeutic strategies based on the regulation of iron homeostasis are needed.
Collapse
Affiliation(s)
- Andreea Ciudin
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Unidad de Diabetes y Metabolismo, Instituto de Investigación Hospital Universitario Vall d'Hebron, Paseo Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Cristina Hernández
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Unidad de Diabetes y Metabolismo, Instituto de Investigación Hospital Universitario Vall d'Hebron, Paseo Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Rafael Simó
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Unidad de Diabetes y Metabolismo, Instituto de Investigación Hospital Universitario Vall d'Hebron, Paseo Vall d'Hebron 119-129, 08035 Barcelona, Spain
| |
Collapse
|
23
|
Dean JB. Hypercapnia causes cellular oxidation and nitrosation in addition to acidosis: implications for CO2 chemoreceptor function and dysfunction. J Appl Physiol (1985) 2010; 108:1786-95. [PMID: 20150563 PMCID: PMC2886689 DOI: 10.1152/japplphysiol.01337.2009] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Accepted: 02/08/2010] [Indexed: 12/22/2022] Open
Abstract
Cellular mechanisms of CO2 chemoreception are discussed and debated in terms of the stimuli produced during hypercapnic acidosis and their molecular targets: protons generated by the hydration of CO2 and dissociation of carbonic acid, which target membrane-bound proteins and lipids in brain stem neurons. The CO2 hydration reaction, however, is not the only reaction that CO2 undergoes that generates molecules capable of modifying proteins and lipids. Molecular CO2 also reacts with peroxynitrite (ONOO-), a reactive nitrogen species (RNS), which is produced from nitric oxide (*NO) and superoxide (*O2-). The CO2/ONOO- reaction, in turn, produces additional nitrosative and oxidative reactive intermediates. Furthermore, protons facilitate additional redox reactions that generate other reactive oxygen species (ROS). ROS/RNS generated by these redox reactions may act as additional stimuli of CO2 chemoreceptors since neurons in chemosensitive areas produce both *NO and *O2- and, therefore, ONOO-. Perturbing *NO, *O2-, and ONOO- activities in chemosensitive areas modulates cardiorespiration. Moreover, neurons in at least one chemosensitive area, the solitary complex, are stimulated by cellular oxidation. Together, these data raise the following two questions: 1) do pH and ROS/RNS work in tandem to stimulate CO2 chemoreceptors during hypercapnic acidosis; and 2) does nitrosative stress and oxidative stress contribute to CO2 chemoreceptor dysfunction? To begin considering these two issues and their implications for central chemoreception, this minireview has the following three goals: 1) summarize the nitrosative and oxidative reactions that occur during hypercapnic acidosis and isocapnic acidosis; 2) review the evidence that redox signaling occurs in chemosensitive areas; and 3) review the evidence that neurons in the solitary complex are stimulated by cellular oxidation.
Collapse
Affiliation(s)
- Jay B Dean
- Department of Molecular Pharmacology and Physiology, Hyperbaric Biomedical Research Laboratory, University of South Florida, College of Medicine, MDC 8, 12901 Bruce B. Downs Blvd., Tampa, Florida 33612, USA.
| |
Collapse
|
24
|
Chen J, Wang J, Meyers KR, Enns CA. Transferrin-directed internalization and cycling of transferrin receptor 2. Traffic 2009; 10:1488-501. [PMID: 19682329 DOI: 10.1111/j.1600-0854.2009.00961.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Transferrin receptor 2 (TfR2) is a homologue of transferrin receptor 1 (TfR1) but has distinct functions from TfR1 in iron homeostasis. In keeping with its proposed role in iron sensing, previous studies showed that TfR2 has a short half-life and that holo-Tf stabilizes TfR2 by redirecting it from a degradative pathway to a recycling pathway. In this study, we characterized how the endocytosis, recycling and degradation of TfR2 relates to its function and differs from TfR1. TfR2 endocytosis was adaptor protein-2 (AP-2) dependent. Flow cytometry analysis showed that TfR1 and TfR2 utilized the same endocytic pathway only in the presence of holo-Tf, indicating that holo-Tf alters the interaction of TfR2 with the endocytic machinery. Unlike TfR1, phosphofurin acidic cluster sorting protein 1 (PACS-1) binds to the cytoplasmic domain of TfR2 and data suggest that PACS-1 is involved in the TfR2 recycling. Depletion of TSG101 by siRNA or expression of a dominant negative Vps4 inhibited TfR2 degradation, indicating that TfR2 degradation occurs through a multivesicular body (MVB) pathway. TfR2 degradation is not mediated through ubiquitination on the single lysine (K31) in the cytoplasmic domain or on the amino terminal residue. No ubiquitination of TfR2 by HA-ubiquitin was detected, indicating a lack of direct TfR2 ubiquitination involvement in its degradation.
Collapse
Affiliation(s)
- Juxing Chen
- Department of Cell and Developmental Biology L215, Oregon Health & Science University, Portland, OR 97239, USA
| | | | | | | |
Collapse
|
25
|
Nashida T, Yoshie S, Imai A, Shimomura H. Transferrin secretory pathways in rat parotid acinar cells. Arch Biochem Biophys 2009; 487:131-8. [DOI: 10.1016/j.abb.2009.05.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Revised: 05/14/2009] [Accepted: 05/16/2009] [Indexed: 11/16/2022]
|
26
|
Hassan W, Ibrahim M, Nogueira CW, Braga AL, Deobald AM, MohammadZai IU, Rocha JBT. Influence of pH on the reactivity of diphenyl ditelluride with thiols and anti-oxidant potential in rat brain. Chem Biol Interact 2009; 180:47-53. [DOI: 10.1016/j.cbi.2008.12.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Revised: 12/18/2008] [Accepted: 12/19/2008] [Indexed: 10/21/2022]
|
27
|
Garrick MD, Garrick LM. Cellular iron transport. Biochim Biophys Acta Gen Subj 2009; 1790:309-25. [DOI: 10.1016/j.bbagen.2009.03.018] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2008] [Revised: 03/23/2009] [Accepted: 03/23/2009] [Indexed: 01/24/2023]
|
28
|
Hower V, Mendes P, Torti FM, Laubenbacher R, Akman S, Shulaev V, Torti SV. A general map of iron metabolism and tissue-specific subnetworks. MOLECULAR BIOSYSTEMS 2009; 5:422-43. [PMID: 19381358 PMCID: PMC2680238 DOI: 10.1039/b816714c] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Iron is required for survival of mammalian cells. Recently, understanding of iron metabolism and trafficking has increased dramatically, revealing a complex, interacting network largely unknown just a few years ago. This provides an excellent model for systems biology development and analysis. The first step in such an analysis is the construction of a structural network of iron metabolism, which we present here. This network was created using CellDesigner version 3.5.2 and includes reactions occurring in mammalian cells of numerous tissue types. The iron metabolic network contains 151 chemical species and 107 reactions and transport steps. Starting from this general model, we construct iron networks for specific tissues and cells that are fundamental to maintaining body iron homeostasis. We include subnetworks for cells of the intestine and liver, tissues important in iron uptake and storage, respectively, as well as the reticulocyte and macrophage, key cells in iron utilization and recycling. The addition of kinetic information to our structural network will permit the simulation of iron metabolism in different tissues as well as in health and disease.
Collapse
Affiliation(s)
- Valerie Hower
- Department of Cancer Biology, Wake Forest University School of Medicine, Medical Center Blvd, Winston Salem, NC 27157, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
pH-Dependent Fe (II) pathophysiology and protective effect of an organoselenium compound. FEBS Lett 2009; 583:1011-6. [DOI: 10.1016/j.febslet.2009.02.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2008] [Revised: 02/08/2009] [Accepted: 02/11/2009] [Indexed: 10/21/2022]
|
30
|
Hassan W, Ibrahim M, Nogueira CW, Braga AL, Mohammadzai IU, Taube PS, Rocha JBT. Enhancement of iron-catalyzed lipid peroxidation by acidosis in brain homogenate: comparative effect of diphenyl diselenide and ebselen. Brain Res 2008; 1258:71-7. [PMID: 19135432 DOI: 10.1016/j.brainres.2008.12.046] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Revised: 12/12/2008] [Accepted: 12/17/2008] [Indexed: 11/26/2022]
Abstract
Iron is more soluble at lower pH values; therefore we hypothesized that decreasing the environmental pH would lead to increased iron-mediated lipid peroxidation. Diphenyl diselenide and ebselen are potential candidates as neuroprotective agent, particularly in situations involving overproduction of free radicals and involving cellular pH fall. The aim of the present study was (a) to investigate the relationship between lipid peroxidation and acidosis in brain homogenate and (b) to test the influence of pH on the antioxidant properties of diphenyl diselenide and ebselen. For the purpose rat brain homogenate was incubated at different pH ranging from physiological to acidic values and extent of lipid peroxidation was measured. Thiobarbituric acid-reactive species (TBARS) production significantly increased when homogenate was incubated in the pH (5.4-6.8) medium both in the absence and presence of Fe (II) as compared with physiological pH (7.4). These data indicate that lipid peroxidation processes, mediated by iron, are enhanced with decreasing extracellular pH. The iron mobilized may come from reserves where it is weakly bound. Diphenyl diselenide significantly protected TBARS production at all studied pH values while ebselen offered only a small statistically non-significant protection. However, calculated IC(50) for TBARS inhibition indicated that pH did not change anti-oxidant activities of the tested compounds. This study provides in-vitro evidence for acidosis induced oxidative stress in brain homogenate and anti-oxidant action of diphenyl diselenide.
Collapse
Affiliation(s)
- Waseem Hassan
- Departamento de Química, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, CEP 97105-900, RS, Brazil.
| | | | | | | | | | | | | |
Collapse
|
31
|
Zhang AS, Enns CA. Iron homeostasis: recently identified proteins provide insight into novel control mechanisms. J Biol Chem 2008; 284:711-5. [PMID: 18757363 DOI: 10.1074/jbc.r800017200] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Iron is an essential nutrient required for a variety of biochemical processes. It is a vital component of the heme in hemoglobin, myoglobin, and cytochromes and is also an essential cofactor for non-heme enzymes such as ribonucleotide reductase, the limiting enzyme for DNA synthesis. When in excess, iron is toxic because it generates superoxide anions and hydroxyl radicals that react readily with biological molecules, including proteins, lipids, and DNA. As a result, humans possess elegant control mechanisms to maintain iron homeostasis by coordinately regulating iron absorption, iron recycling, and mobilization of stored iron. Disruption of these processes causes either iron-deficient anemia or iron overload disorders. In this minireview, we focus on the roles of recently identified proteins in the regulation of iron homeostasis.
Collapse
Affiliation(s)
- An-Sheng Zhang
- Department of Cell and Developmental Biology, Oregon Health & Science University, Portland, Oregon 97239, USA
| | | |
Collapse
|
32
|
Giannetti AM, Halbrooks PJ, Mason AB, Vogt TM, Enns CA, Björkman PJ. The molecular mechanism for receptor-stimulated iron release from the plasma iron transport protein transferrin. Structure 2008; 13:1613-23. [PMID: 16271884 DOI: 10.1016/j.str.2005.07.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2005] [Revised: 07/19/2005] [Accepted: 07/23/2005] [Indexed: 01/08/2023]
Abstract
Human transferrin receptor 1 (TfR) binds iron-loaded transferrin (Fe-Tf) and transports it to acidic endosomes where iron is released in a TfR-facilitated process. Consistent with our hypothesis that TfR binding stimulates iron release from Fe-Tf at acidic pH by stabilizing the apo-Tf conformation, a TfR mutant (W641A/F760A-TfR) that binds Fe-Tf, but not apo-Tf, cannot stimulate iron release from Fe-Tf, and less iron is released from Fe-Tf inside cells expressing W641A/F760A-TfR than cells expressing wild-type TfR (wtTfR). Electron paramagnetic resonance spectroscopy shows that binding at acidic pH to wtTfR, but not W641A/F760A-TfR, changes the Tf iron binding site > or =30 A from the TfR W641/F760 patch. Mutation of Tf histidine residues predicted to interact with the W641/F760 patch eliminates TfR-dependent acceleration of iron release. Identification of TfR and Tf residues critical for TfR-facilitated iron release, yet distant from a Tf iron binding site, demonstrates that TfR transmits long-range conformational changes and stabilizes the conformation of apo-Tf to accelerate iron release from Fe-Tf.
Collapse
Affiliation(s)
- Anthony M Giannetti
- Division of Biology 114-96, California Institute of Technology, Pasadena, California 91125, USA
| | | | | | | | | | | |
Collapse
|
33
|
Intracellular localization and subsequent redistribution of metal transporters in a rat choroid plexus model following exposure to manganese or iron. Toxicol Appl Pharmacol 2008; 230:167-74. [PMID: 18420243 DOI: 10.1016/j.taap.2008.02.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Revised: 02/19/2008] [Accepted: 02/19/2008] [Indexed: 11/23/2022]
Abstract
Confocal microscopy was used to investigate the effects of manganese (Mn) and iron (Fe) exposure on the subcellular distribution of metal transporting proteins, i.e., divalent metal transporter 1 (DMT1), metal transporter protein 1 (MTP1), and transferrin receptor (TfR), in the rat intact choroid plexus which comprises the blood-cerebrospinal fluid barrier. In control tissue, DMT1 was concentrated below the apical epithelial membrane, MTP1 was diffuse within the cytosol, and TfR was distributed in vesicles around nuclei. Following Mn or Fe treatment (1 and 10 microM), the distribution of DMT1 was not affected. However, MTP1 and TfR moved markedly toward the apical pole of the cells. These shifts were abolished when microtubules were disrupted. Quantitative RT-PCR and Western blot analyses revealed a significant increase in mRNA and protein levels of TfR but not DMT1 and MTP1 after Mn exposure. These results suggest that early events in the tissue response to Mn or Fe exposure involve microtubule-dependent, intracellular trafficking of MTP1 and TfR. The intracellular trafficking of metal transporters in the choroid plexus following Mn exposure may partially contribute to Mn-induced disruption in Fe homeostasis in the cerebrospinal fluid (CSF) following Mn exposure.
Collapse
|
34
|
Haber A, Agadjanian H, Medina-Kauwe LK, Gross Z. Corroles that bind with high affinity to both apo and holo transferrin. J Inorg Biochem 2008; 102:446-57. [DOI: 10.1016/j.jinorgbio.2007.10.026] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2007] [Revised: 09/30/2007] [Accepted: 10/12/2007] [Indexed: 10/22/2022]
|
35
|
De Domenico I, McVey Ward D, Kaplan J. Regulation of iron acquisition and storage: consequences for iron-linked disorders. Nat Rev Mol Cell Biol 2008; 9:72-81. [PMID: 17987043 DOI: 10.1038/nrm2295] [Citation(s) in RCA: 317] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mammalian iron homeostasis must be meticulously regulated so that this essential element is available for use, but at the same time prevented from promoting the formation of toxic radicals. Controlling the entry of iron into blood plasma is the main mechanism by which iron stores in the body are physiologically manipulated and regulated. Defects in iron acquisition at the cellular and systemic levels lead to human disorders, which involve either iron overload or iron deficiency. Discoveries of iron transporters and insights into their regulation have provided important information about iron metabolism and genetic iron disorders.
Collapse
Affiliation(s)
- Ivana De Domenico
- Department of Pathology, School of Medicine, University of Utah, Salt Lake City, Utah 84132, USA
| | | | | |
Collapse
|
36
|
He X, Hahn P, Iacovelli J, Wong R, King C, Bhisitkul R, Massaro-Giordano M, Dunaief JL. Iron homeostasis and toxicity in retinal degeneration. Prog Retin Eye Res 2007; 26:649-73. [PMID: 17921041 DOI: 10.1016/j.preteyeres.2007.07.004] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Iron is essential for many metabolic processes but can also cause damage. As a potent generator of hydroxyl radical, the most reactive of the free radicals, iron can cause considerable oxidative stress. Since iron is absorbed through diet but not excreted except through menstruation, total body iron levels buildup with age. Macular iron levels increase with age, in both men and women. This iron has the potential to contribute to retinal degeneration. Here we present an overview of the evidence suggesting that iron may contribute to retinal degenerations. Intraocular iron foreign bodies cause retinal degeneration. Retinal iron buildup resulting from hereditary iron homeostasis disorders aceruloplasminemia, Friedreich's ataxia, and panthothenate kinase-associated neurodegeneration cause retinal degeneration. Mice with targeted mutation of the iron exporter ceruloplasmin have age-dependent retinal iron overload and a resulting retinal degeneration with features of age-related macular degeneration (AMD). Post mortem retinas from patients with AMD have more iron and the iron carrier transferrin than age-matched controls. Over the past 10 years much has been learned about the intricate network of proteins involved in iron handling. Many of these, including transferrin, transferrin receptor, divalent metal transporter-1, ferritin, ferroportin, ceruloplasmin, hephaestin, iron-regulatory protein, and histocompatibility leukocyte antigen class I-like protein involved in iron homeostasis (HFE) have been found in the retina. Some of these proteins have been found in the cornea and lens as well. Levels of the iron carrier transferrin are high in the aqueous and vitreous humors. The functions of these proteins in other tissues, combined with studies on cultured ocular tissues, genetically engineered mice, and eye exams on patients with hereditary iron diseases provide clues regarding their ocular functions. Iron may play a role in a broad range of ocular diseases, including glaucoma, cataract, AMD, and conditions causing intraocular hemorrhage. While iron deficiency must be prevented, the therapeutic potential of limiting iron-induced ocular oxidative damage is high. Systemic, local, or topical iron chelation with an expanding repertoire of drugs has clinical potential.
Collapse
Affiliation(s)
- Xining He
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, 305 Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Teh EM, Hewitt J, Ung KC, Griffiths TAM, Nguyen V, Briggs SK, Mason AB, MacGillivray RTA. Identification of the epitope of a monoclonal antibody that disrupts binding of human transferrin to the human transferrin receptor. FEBS J 2006; 272:6344-53. [PMID: 16336271 DOI: 10.1111/j.1742-4658.2005.05028.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The molecular basis of the transferrin (TF)-transferrin receptor (TFR) interaction is not known. The C-lobe of TF is required to facilitate binding to the TFR and both the N- and C-lobes are necessary for maximal binding. Several mAb have been raised against human transferrin (hTF). One of these, designated F11, is specific to the C-lobe of hTF and does not recognize mouse or pig TF. Furthermore, mAb F11 inhibits the binding of TF to TFR on HeLa cells. To map the epitope for mAb F11, constructs spanning various regions of hTF were expressed as glutathione S-transferase (GST) fusion proteins in Escherichia coli. The recombinant fusion proteins were analysed in an iterative fashion by immunoblotting using mAb F11 as the probe. This process resulted in the localization of the F11 epitope to the C1 domain (residues 365-401) of hTF. Subsequent computer modelling suggested that the epitope is probably restricted to a surface patch of hTF consisting of residues 365-385. Mutagenesis of the F11 epitope of hTF to the sequence of either mouse or pig TF confirmed the identity of the epitope as immunoreactivity was diminished or lost. In agreement with other studies, these epitope mapping studies support a role for residues in the C1 domain of hTF in receptor binding.
Collapse
Affiliation(s)
- Evelyn M Teh
- Department of Biochemistry and Molecular Biology and Centre for Blood Research, University of British Columbia, Vancouver, Canada
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Heeney MM, Andrews NC. Iron homeostasis and inherited iron overload disorders: an overview. Hematol Oncol Clin North Am 2004; 18:1379-403, ix. [PMID: 15511621 DOI: 10.1016/j.hoc.2004.06.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Iron is an ubiquitous metal of vital importance to the normal physiologic processes of many organisms. Recent discoveries of mutations in genes that lead to inherited iron overload diseases have advanced the understanding of iron homeostasis in humans. This article provides an overview of the human iron cycle, regulation of iron homeostasis, how perturbations in this homeostasis lead to iron overload disease in adults and children, and strategies for diagnosis of inherited iron overload.
Collapse
|
39
|
Giannetti AM, Björkman PJ. HFE and transferrin directly compete for transferrin receptor in solution and at the cell surface. J Biol Chem 2004; 279:25866-75. [PMID: 15056661 DOI: 10.1074/jbc.m401467200] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Transferrin receptor (TfR) is a dimeric cell surface protein that binds both the serum iron transport protein transferrin (Fe-Tf) and HFE, the protein mutated in patients with the iron overload disorder hereditary hemochromatosis. HFE and Fe-Tf can bind simultaneously to TfR to form a ternary complex, but HFE binding to TfR lowers the apparent affinity of the Fe-Tf/TfR interaction. This apparent affinity reduction could result from direct competition between HFE and Fe-Tf for their overlapping binding sites on each TfR polypeptide chain, from negative cooperativity, or from a combination of both. To explore the mechanism of the affinity reduction, we constructed a heterodimeric TfR that contains mutations such that one TfR chain binds only HFE and the other binds only Fe-Tf. Binding studies using a heterodimeric form of soluble TfR demonstrate that TfR does not exhibit cooperativity in heterotropic ligand binding, suggesting that some or all of the effects of HFE on iron homeostasis result from competition with Fe-Tf for TfR binding. Experiments using transfected cell lines demonstrate a physiological role for this competition in altering HFE trafficking patterns.
Collapse
Affiliation(s)
- Anthony M Giannetti
- Graduate Option in Biochemistry and Molecular Biophysics, California Institute of Technology, Pasadena, CA 91125, USA
| | | |
Collapse
|
40
|
Deane R, Zheng W, Zlokovic BV. Brain capillary endothelium and choroid plexus epithelium regulate transport of transferrin-bound and free iron into the rat brain. J Neurochem 2004; 88:813-20. [PMID: 14756801 PMCID: PMC3980859 DOI: 10.1046/j.1471-4159.2003.02221.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Iron transport into the CNS is still not completely understood. Using a brain perfusion technique in rats, we have shown a significant brain capillary uptake of circulating transferrin (Tf)-bound and free 59Fe (1 nm) at rates of 136 +/- 26 and 182 +/- 23 microL/g/min, respectively, while their respective transport rates into brain parenchyma were 1.68 +/- 0.56 and 1.52 +/- 0.48 microL/g/min. Regional Tf receptor density (Bmax) in brain endothelium determined with 125I-holo-Tf correlated well with 59Fe-Tf regional brain uptake rates reflecting significant vascular association of iron. Tf-bound and free circulating 59Fe were sequestered by the choroid plexus and transported into the CSF at low rates of 0.17 +/- 0.01 and 0.09 +/- 0.02 microL/min/g, respectively, consistent with a 10-fold brain-CSF concentration gradient for 59Fe, Tf-bound or free. We conclude that transport of circulating Tf-bound and free iron could be equally important for its delivery to the CNS. Moreover, data suggest that entry of Tf-bound and free iron into the CNS is determined by (i) its initial sequestration by brain capillaries and choroid plexus, and (ii) subsequent controlled and slow release from vascular structures into brain interstitial fluid and CSF.
Collapse
Affiliation(s)
- Rashid Deane
- Frank P. Smith Neurosurgical Research Laboratory, Department of Neurosurgery, University of Rochester Medical Center, Rochester, New York 14642, USA.
| | | | | |
Collapse
|
41
|
Abstract
Aluminum, a trivalent cation unable to undergo redox reactions, is shown to faciliate iron-initiated DOPA oxidation in the melanin pathway under acidic condition of pH 5.5, which is a favored medium for aluminum facilitation of iron-induced lipid peroxidation. In the process of oxidation of DOPA to melanin in the presence of the metal ions, Fe3+ and H2O2 oxidize DOPA to dopachrome (DC), then Al3+ catalyzes the conversion of DC to 5,6-dihydroxyindole (DHI) and finally Fe3+ oxidizes DHI to indole-5,6-quinone (IQ), which polymerizes immediately to melanochrome and melanin. The reactions involve the intermediate complexes of metal ions and DOPA or its derivative. The present results indicate that aluminum can enhance the oxidative stress on iron-mediated DOPA oxidation in melanin pathway under acidic condition through the cooperation of iron and aluminum ions.
Collapse
Affiliation(s)
- Junwei Di
- Department of Chemistry, State Key Laboratory of Coordination Chemistry of China, Nanjing University, Nanjing 210093, P R China
| | | |
Collapse
|
42
|
Giannetti AM, Snow PM, Zak O, Björkman PJ. Mechanism for multiple ligand recognition by the human transferrin receptor. PLoS Biol 2003; 1:E51. [PMID: 14691533 PMCID: PMC300677 DOI: 10.1371/journal.pbio.0000051] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2003] [Accepted: 09/10/2003] [Indexed: 11/18/2022] Open
Abstract
Transferrin receptor 1 (TfR) plays a critical role in cellular iron import for most higher organisms. Cell surface TfR binds to circulating iron-loaded transferrin (Fe-Tf) and transports it to acidic endosomes, where low pH promotes iron to dissociate from transferrin (Tf) in a TfR-assisted process. The iron-free form of Tf (apo-Tf) remains bound to TfR and is recycled to the cell surface, where the complex dissociates upon exposure to the slightly basic pH of the blood. Fe-Tf competes for binding to TfR with HFE, the protein mutated in the iron-overload disease hereditary hemochromatosis. We used a quantitative surface plasmon resonance assay to determine the binding affinities of an extensive set of site-directed TfR mutants to HFE and Fe-Tf at pH 7.4 and to apo-Tf at pH 6.3. These results confirm the previous finding that Fe-Tf and HFE compete for the receptor by binding to an overlapping site on the TfR helical domain. Spatially distant mutations in the TfR protease-like domain affect binding of Fe-Tf, but not iron-loaded Tf C-lobe, apo-Tf, or HFE, and mutations at the edge of the TfR helical domain affect binding of apo-Tf, but not Fe-Tf or HFE. The binding data presented here reveal the binding footprints on TfR for Fe-Tf and apo-Tf. These data support a model in which the Tf C-lobe contacts the TfR helical domain and the Tf N-lobe contacts the base of the TfR protease-like domain. The differential effects of some TfR mutations on binding to Fe-Tf and apo-Tf suggest differences in the contact points between TfR and the two forms of Tf that could be caused by pH-dependent conformational changes in Tf, TfR, or both. From these data, we propose a structure-based model for the mechanism of TfR-assisted iron release from Fe-Tf. Differences in the contact points between the transferrin receptor and the two forms of transferrin (with or without iron) are consistent with pH-dependent conformational changes in transferrin, its receptor, or both
Collapse
Affiliation(s)
- Anthony M Giannetti
- 1Graduate Option in Biochemistry and Molecular Biophysics, California Institute of TechnologyPasadena, CaliforniaUnited States of America
| | - Peter M Snow
- 2Caltech Protein Expression Center, Division of Biology California Institute of TechnologyPasadena, CaliforniaUnited States of America
| | - Olga Zak
- 3Department of Physiology and Biophysics, Albert Einstein College of MedicineBronx, New YorkUnited States of America
| | - Pamela J Björkman
- 4Division of Biology and Howard Hughes Medical Institute, California Institute of TechnologyPasadena, CaliforniaUnited States of America
| |
Collapse
|
43
|
Pfanzagl B, Tribl F, Koller E, Möslinger T. Homocysteine strongly enhances metal-catalyzed LDL oxidation in the presence of cystine and cysteine. Atherosclerosis 2003; 168:39-48. [PMID: 12732385 DOI: 10.1016/s0021-9150(03)00057-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Here we show that homocysteine stimulates low density lipoprotein (LDL) oxidation at copper(II) concentrations causing only a slight oxidation of LDL lipids. LDL oxidation by homocysteine and copper(II) is further enhanced in the presence of cystine, although cystine alone does not stimulate LDL oxidation with copper(II). Similarly, a combination of cysteine with homocysteine provoked a more than additive increase of oxidation. Simultaneous presence of cysteine and homocystine also resulted in a more than additive oxidative effect which was not statistically significant, however. Stimulation of LDL oxidation in the presence of homocysteine by cystine was also observed with iron(III) at acidic pH and when LDL oxidation was initiated by azo-compound generated peroxyl radicals. At pH 7.4 histidine is able to prevent LDL oxidation by copper(II) in a thiol mixture similar to the one found in human plasma if present in tenfold excess over homocysteine, but loses its inhibitory effect at higher homocysteine concentrations. The synergistic effect on metal-catalyzed LDL oxidation observed with mixtures of homocysteine and cystine or cysteine sustains the hypothesis that the epidemiological association between raised homocysteine levels and risk of cardiovascular disease is caused by an increase in oxidative stress.
Collapse
Affiliation(s)
- Beatrix Pfanzagl
- Institute of Physiology, University of Wien, Schwarzspanierstrasse 17, Austria.
| | | | | | | |
Collapse
|
44
|
Abstract
To acquire iron, all species have had to overcome the problems of iron insolubility and toxicity. This review surveys the approaches taken in solubilizing and transporting iron by species in different kingdoms. In some instances, iron uptake systems are novel and are restricted to a few species or to a biologic kingdom. In other instances, it is clear that all organisms use homologous genes. With rare exceptions, ferrous iron (Fe(2+)) transport systems are not specific for iron and will effect uptake of other transition metals. Ferric iron (Fe(3+)) transport systems are highly specific for iron, and in vertebrates are used to target iron transport to specific tissues.
Collapse
Affiliation(s)
- Jerry Kaplan
- Department of Pathology, School of Medicine, University of Utah, Salt Lake City, UT 84132-2408, USA
| |
Collapse
|
45
|
|
46
|
Aberkane H, Stoltz JF, Galteau MM, Wellman M. Erythrocytes as targets for gamma-glutamyltranspeptidase initiated pro-oxidant reaction. Eur J Haematol 2002; 68:262-71. [PMID: 12144532 DOI: 10.1034/j.1600-0609.2002.01636.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Gamma-glutamyltranspeptidase (GGT) is a well known cell plasma membrane and serum circulating enzyme. In clinical chemistry, GGT is used as a marker of alcohol consumption and drug uptake. Serum GGT activity varies in hepatobiliary diseases and cancer. This enzyme is involved in glutathione (GSH) metabolism, which is generally associated with antioxidant properties. However, in recent years, findings from our group and from others showed that GGT-catalysed extracellular metabolism of GSH leads, in the presence of iron, to the generation of reactive oxygen species (ROS). It was demonstrated that those highly reactive species oxidise lipids, cell surface protein thiols or activate transcriptional factors such as Nuclear Factor kappaB (NFkappaB). The objective of the present work is to determine whether the red blood cells are targets for plasma GGT-initiated pro-oxidant reaction. The results obtained demonstrate that the GGT/GSH/iron system oxidises isolated erythrocyte membranes. A significant release of haemoglobin and a decrease of erythrocyte deformability are also observed. In addition, in vivo studies showed a relationship between plasma GGT activity and erythrocyte deformability in 20 studied subjects. In conclusion, GGT-mediated ROS production is able to oxidise erythrocytes and thus disturbs their functions.
Collapse
Affiliation(s)
- Hayet Aberkane
- Thiols et Fonctions Cellulaires, Faculty of Pharmacy, University Henri Poincaré, Nancy 1, 30 rue Lionnois, 54000 Nancy, France
| | | | | | | |
Collapse
|
47
|
Walker BL, Tiong JW, Jefferies WA. Iron metabolism in mammalian cells. INTERNATIONAL REVIEW OF CYTOLOGY 2002; 211:241-78. [PMID: 11597005 DOI: 10.1016/s0074-7696(01)11020-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Most living things require iron to exist. Iron has many functions within cells but is rarely found unbound because of its propensity to catalyze the formation of toxic free radicals. Thus the regulation of iron requirements by cells and the acquisition and uptake of iron into tissues in multicellular organisms is tightly regulated. In humans, understanding iron transport and utility has recently been advanced by a "great conjunction" of molecular genetics in simple organisms, identifying genes involved in genetic diseases of metal metabolism and by the application of traditional cell physiology approaches. We are now able to approach a rudimentary understanding of the "iron cycle" within mammals. In the future, this information will be applied toward modulating the outcome of therapies designed to overcome diseases involving metals.
Collapse
Affiliation(s)
- B L Walker
- Biomedical Research Centre, and Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | | | | |
Collapse
|
48
|
Abstract
Iron is an essential cofactor in a variety of cellular processes. Except for a few unusual bacterial species, iron is indispensable for living organisms. However, free iron is toxic because of its propensity to induce the formation of dangerous free radicals. Consequently, iron balance is tightly regulated. Disorders of iron homeostasis are among the most common afflictions of humans. This review discusses inherited iron deficiency and iron overload disorders and recent insights into their pathophysiology.
Collapse
Affiliation(s)
- N C Andrews
- Howard Hughes Medical Institute, Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA.
| |
Collapse
|
49
|
Enns CA. Pumping iron: the strange partnership of the hemochromatosis protein, a class I MHC homolog, with the transferrin receptor. Traffic 2001; 2:167-74. [PMID: 11260522 DOI: 10.1034/j.1600-0854.2001.020303.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
People suffering from hereditary hemochromatosis (HH) can not regulate the uptake of iron properly and gradually accumulate iron in their body over their lifetime. The protein involved in HH, HFE, has been recently identified as a class I major histocompatibility complex (MHC) homolog. The wild-type HFE associates and co-traffics with the transferrin receptor (TfR). The mutation responsible for 83% of HH (C260Y) results in the failure of HFE to form a critical disulfide bond, bind beta2 microglobulin, bind TfR, and traffic to the cell surface. In non-polarized cells, the partnership of HFE and TfR results in decreased iron uptake into cells. The mechanism whereby a class I MHC homolog modifies the function of a membrane receptor and how this dynamic complex of molecules regulates iron transport across intestinal epithelial cells is the subject of this review.
Collapse
Affiliation(s)
- C A Enns
- Department of Cell and Developmental Biology, L-215, Oregon Health Sciences University, Portland, OR 97201, USA.
| |
Collapse
|
50
|
Abstract
Iron homeostasis is maintained by regulating its absorption: Under conditions of deficiency, assimilation is enhanced but iron uptake is otherwise limited to prevent toxicity due to overload. Iron deficiency remains the most important micronutrient deficiency worldwide, but increasing awareness of the genetic basis for iron-loading diseases points to iron overload as a major public health issue as well. Recent identification of mutant alleles causing iron uptake disorders in mice and humans provides new insights into the mechanisms involved in iron transport and its regulation. This article summarizes these discoveries and discusses their impact on our current understanding of iron transport and its regulation.
Collapse
Affiliation(s)
- M Wessling-Resnick
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts 02115, USA.
| |
Collapse
|