101
|
Volani C, Doerrier C, Demetz E, Haschka D, Paglia G, Lavdas AA, Gnaiger E, Weiss G. Dietary iron loading negatively affects liver mitochondrial function. Metallomics 2018; 9:1634-1644. [PMID: 29026901 DOI: 10.1039/c7mt00177k] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Iron is an essential co-factor for several metabolic processes, including mitochondrial respiration, and mitochondria are the major sites of iron-utilization. Cellular iron homeostasis must be tightly regulated, as intracellular iron deficiency can lead to insufficient energy production, whereas iron overload triggers ROS (reactive oxygen species) formation via the Fenton reaction. So far little is known on how iron imbalances affect mitochondrial function in vivo and the impact of the genotype on that, we studied the effects of dietary iron loading on mitochondrial respiratory capacity in liver by comparing two genetically divergent mouse strains, namely C57BL/6N and FVB mice. Both mouse strains differed in their basal iron levels and their metabolic responses to iron loading as determined by expression of iron trafficking proteins (ferritin was increased in livers of animals receiving high iron diet) as well as tissue iron content (2-fold increase, FVB p = 0.0013; C57BL/6N p = 0.0022). Dietary iron exposure caused a significant impairment of mitochondrial oxidative phosphorylation, especially regarding OXPHOS capacity (FVB p = 0.0006; C57BL/6N p = 0.0087) and S-ETS capacity (FVB p = 0.0281; C57BL/6N p = 0.0159). These effects were more pronounced in C57BL/6N than in FVB mice and were paralleled by an iron mediated induction of oxidative stress in mitochondria. The increased susceptibility of C57BL6/N mice to iron loading may be due to reduced expression of anti-oxidant defense mechanisms and altered iron trafficking upon dietary challenge pointing to a role of genetic modifiers for cellular and mitochondrial iron trafficking. Finally, iron-mediated induction of mitochondrial oxidative stress and reduction of oxidative phosphorylation may underlie fatigue in subjects with iron loading diseases.
Collapse
Affiliation(s)
- Chiara Volani
- Department of Internal Medicine II (Infectious Diseases, Immunology, Rheumatology and Pneumology), Medical University of Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
102
|
Stokes BA, Sabatino JA, Zohn IE. High levels of iron supplementation prevents neural tube defects in the Fpn1 ffe mouse model. Birth Defects Res 2018; 109:81-91. [PMID: 28008752 DOI: 10.1002/bdra.23542] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/11/2016] [Accepted: 06/05/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND Periconception maternal nutrition and folate in particular are important factors influencing the incidence of neural tube defects (NTDs). Many but not all NTDs are prevented by folic acid supplementation and there is a pressing need for additional strategies to prevent these birth defects. Other micronutrients such as iron are potential candidates, yet a clear role for iron deficiency in contributing to NTDs is lacking. Our previous studies with the flatiron (ffe) mouse model of Ferroportin1 (Fpn1) deficiency suggest that iron is required for neural tube closure and forebrain development raising the possibility that iron supplementation could prevent NTDs. METHODS We determined the effect of periconception iron and/or folic acid supplementation on the penetrance of NTDs in the Fpn1ffe mouse model. Concurrently, measurements of folate and iron were made to ensure supplementation had the intended effects. RESULTS High levels of iron supplementation significantly reduced the incidence of NTDs in Fpn1ffe mutants. Fpn1 deficiency resulted in reduced folate levels in both pregnant dams and embryos. Yet folic acid supplementation did not prevent NTDs in the Fpn1ffe model. Similarly, forebrain truncations were rescued with iron. Surprisingly, the high levels of iron supplementation used in this study caused folate deficiency in wild-type dams and embryos. CONCLUSION Our results demonstrate that iron supplementation can prevent NTDs and forebrain truncations in the Fpn1ffe model. Surprisingly, high levels of iron supplementation and iron overload can cause folate deficiency. If iron is essential for neural tube closure, it is possible that iron deficiency might contribute to NTDs. Birth Defects Research 109:81-91, 2017. © 2016 The Authors Birth Defects Research Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bethany A Stokes
- Department of Biology, The George Washington University, Washington, DC.,Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC
| | - Julia A Sabatino
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC
| | - Irene E Zohn
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC
| |
Collapse
|
103
|
Damasceno FC, Condeles AL, Lopes AKB, Facci RR, Linares E, Truzzi DR, Augusto O, Toledo JC. The labile iron pool attenuates peroxynitrite-dependent damage and can no longer be considered solely a pro-oxidative cellular iron source. J Biol Chem 2018; 293:8530-8542. [PMID: 29661935 DOI: 10.1074/jbc.ra117.000883] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 04/12/2018] [Indexed: 01/01/2023] Open
Abstract
The ubiquitous cellular labile iron pool (LIP) is often associated with the production of the highly reactive hydroxyl radical, which forms through a redox reaction with hydrogen peroxide. Peroxynitrite is a biologically relevant peroxide produced by the recombination of nitric oxide and superoxide. It is a strong oxidant that may be involved in multiple pathological conditions, but whether and how it interacts with the LIP are unclear. Here, using fluorescence spectroscopy, we investigated the interaction between the LIP and peroxynitrite by monitoring peroxynitrite-dependent accumulation of nitrosated and oxidized fluorescent intracellular indicators. We found that, in murine macrophages, removal of the LIP with membrane-permeable iron chelators sustainably accelerates the peroxynitrite-dependent oxidation and nitrosation of these indicators. These observations could not be reproduced in cell-free assays, indicating that the chelator-enhancing effect on peroxynitrite-dependent modifications of the indicators depended on cell constituents, presumably including LIP, that react with these chelators. Moreover, neither free nor ferrous-complexed chelators stimulated intracellular or extracellular oxidative and nitrosative chemistries. On the basis of these results, LIP appears to be a relevant and competitive cellular target of peroxynitrite or its derived oxidants, and thereby it reduces oxidative processes, an observation that may change the conventional notion that the LIP is simply a cellular source of pro-oxidant iron.
Collapse
Affiliation(s)
- Fernando Cruvinel Damasceno
- From the Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, CEP 14040-901 and
| | - André Luis Condeles
- From the Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, CEP 14040-901 and
| | - Angélica Kodama Bueno Lopes
- From the Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, CEP 14040-901 and
| | - Rômulo Rodrigues Facci
- From the Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, CEP 14040-901 and
| | - Edlaine Linares
- the Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, CEP 05508-000, Brazil
| | - Daniela Ramos Truzzi
- the Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, CEP 05508-000, Brazil
| | - Ohara Augusto
- the Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, CEP 05508-000, Brazil
| | - José Carlos Toledo
- From the Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, CEP 14040-901 and
| |
Collapse
|
104
|
Gammella E, Buratti P, Cairo G, Recalcati S. The transferrin receptor: the cellular iron gate. Metallomics 2018; 9:1367-1375. [PMID: 28671201 DOI: 10.1039/c7mt00143f] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The transferrin receptor (TfR1), which mediates cellular iron uptake through clathrin-dependent endocytosis of iron-loaded transferrin, plays a key role in iron homeostasis. Since the number of TfR1 molecules at the cell surface is the rate-limiting step for iron entry into cells and is essential to prevent iron overload, TfR1 expression is precisely controlled at multiple levels. In this review, we have discussed the latest advances in the molecular regulation of TfR1 expression and we have considered current understanding of TfR1 function beyond its canonical role in providing iron for erythroid precursors and rapidly proliferating cells.
Collapse
Affiliation(s)
- Elena Gammella
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milano, Italy.
| | | | | | | |
Collapse
|
105
|
Abstract
Brain iron is tightly regulated by a multitude of proteins to ensure homeostasis. Iron dyshomeostasis has become a molecular signature associated with aging which is accompanied by progressive decline in cognitive processes. A common theme in neurodegenerative diseases where age is the major risk factor, iron dyshomeostasis coincides with neuroinflammation, abnormal protein aggregation, neurodegeneration, and neurobehavioral deficits. There is a great need to determine the mechanisms governing perturbations in iron metabolism, in particular to distinguish between physiological and pathological aging to generate fruitful therapeutic targets for neurodegenerative diseases. The aim of the present review is to focus on the age-related alterations in brain iron metabolism from a cellular and molecular biology perspective, alongside genetics, and neuroimaging aspects in man and rodent models, with respect to normal aging and neurodegeneration. In particular, the relationship between iron dyshomeostasis and neuroinflammation will be evaluated, as well as the effects of systemic iron overload on the brain. Based on the evidence discussed here, we suggest a synergistic use of iron-chelators and anti-inflammatories as putative anti-brain aging therapies to counteract pathological aging in neurodegenerative diseases.
Collapse
Affiliation(s)
- Azhaar Ashraf
- Institute of Psychiatry, Psychology and Neuroscience, Department of Neuroimaging, King's College London, London, United Kingdom
| | - Maryam Clark
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Po-Wah So
- Institute of Psychiatry, Psychology and Neuroscience, Department of Neuroimaging, King's College London, London, United Kingdom
| |
Collapse
|
106
|
Viaud M, Ivanov S, Vujic N, Duta-Mare M, Aira LE, Barouillet T, Garcia E, Orange F, Dugail I, Hainault I, Stehlik C, Marchetti S, Boyer L, Guinamard R, Foufelle F, Bochem A, Hovingh KG, Thorp EB, Gautier EL, Kratky D, Dasilva-Jardine P, Yvan-Charvet L. Lysosomal Cholesterol Hydrolysis Couples Efferocytosis to Anti-Inflammatory Oxysterol Production. Circ Res 2018. [PMID: 29523554 DOI: 10.1161/circresaha.117.312333] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Macrophages face a substantial amount of cholesterol after the ingestion of apoptotic cells, and the LIPA (lysosomal acid lipase) has a major role in hydrolyzing cholesteryl esters in the endocytic compartment. OBJECTIVE Here, we directly investigated the role of LIPA-mediated clearance of apoptotic cells both in vitro and in vivo. METHODS AND RESULTS We show that LIPA inhibition causes a defective efferocytic response because of impaired generation of 25-hydroxycholesterol and 27-hydroxycholesterol. Reduced synthesis of 25-hydroxycholesterol after LIPA inhibition contributed to defective mitochondria-associated membrane leading to mitochondrial oxidative stress-induced NLRP3 (NOD-like receptor family, pyrin domain containing) inflammasome activation and caspase-1-dependent Rac1 (Ras-related C3 botulinum toxin substrate 1) degradation. A secondary event consisting of failure to appropriately activate liver X receptor-mediated pathways led to mitigation of cholesterol efflux and apoptotic cell clearance. In mice, LIPA inhibition caused defective clearance of apoptotic lymphocytes and stressed erythrocytes by hepatic and splenic macrophages, culminating in splenomegaly and splenic iron accumulation under hypercholesterolemia. CONCLUSIONS Our findings position lysosomal cholesterol hydrolysis as a critical process that prevents metabolic inflammation by enabling efficient macrophage apoptotic cell clearance.
Collapse
Affiliation(s)
- Manon Viaud
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Stoyan Ivanov
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Nemanja Vujic
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria (N.V., M.D.-M., D.K.)
| | - Madalina Duta-Mare
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria (N.V., M.D.-M., D.K.)
| | - Lazaro-Emilio Aira
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | | | - Elsa Garcia
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Francois Orange
- UFR Sciences, Faculté des Sciences de l'Université de Nice-Sophia Antipolis, France (F.O.)
| | - Isabelle Dugail
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1166, Pierre & Marie Curie University, ICAN Institute of Cardiometabolism & Nutrition, Hôpital de la Pitié, Boulevard de l'Hôpital, Paris, France (I.D., E.L.G.)
| | - Isabelle Hainault
- Institut National de la Santé et de la Recherche Médicale (Inserm) UMRS 1138, Centre de Recherche des Cordeliers, Paris, France (I.H., F.F.)
| | - Christian Stehlik
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL (C.S., E.B.T.)
| | - Sandrine Marchetti
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Laurent Boyer
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Rodolphe Guinamard
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Fabienne Foufelle
- Institut National de la Santé et de la Recherche Médicale (Inserm) UMRS 1138, Centre de Recherche des Cordeliers, Paris, France (I.H., F.F.)
| | | | | | - Edward B Thorp
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL (C.S., E.B.T.)
| | - Emmanuel L Gautier
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1166, Pierre & Marie Curie University, ICAN Institute of Cardiometabolism & Nutrition, Hôpital de la Pitié, Boulevard de l'Hôpital, Paris, France (I.D., E.L.G.)
| | - Dagmar Kratky
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria (N.V., M.D.-M., D.K.)
| | - Paul Dasilva-Jardine
- Academic Medical Center, Amsterdam, The Netherlands; and Staten Biotechnology, Nijmegen, The Netherlands (P.D.-J.)
| | - Laurent Yvan-Charvet
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| |
Collapse
|
107
|
Unraveling the Burden of Iron in Neurodegeneration: Intersections with Amyloid Beta Peptide Pathology. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2850341. [PMID: 29581821 PMCID: PMC5831758 DOI: 10.1155/2018/2850341] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 12/17/2017] [Indexed: 12/14/2022]
Abstract
Iron overload is a hallmark of many neurodegenerative processes such as Alzheimer's, Parkinson's, and Huntington's diseases. Unbound iron accumulated as a consequence of brain aging is highly reactive with water and oxygen and produces reactive oxygen species (ROS) or free radicals. ROS are toxic compounds able to damage cell membranes, DNA, and mitochondria. Which are the mechanisms involved in neuronal iron homeostasis and in neuronal response to iron-induced oxidative stress constitutes a cutting-edge topic in metalloneurobiology. Increasing our knowledge about the underlying mechanisms that operate in iron accumulation and their consequences would shed light on the comprehension of the molecular events that participate in the pathophysiology of the abovementioned neurodegenerative diseases. In this review, current evidences about iron accumulation in the brain, the signaling mechanisms triggered by metal overload, as well as the interaction between amyloid β (Aβ) and iron, will be summarized.
Collapse
|
108
|
Sedgwick AC, Wu L, Han HH, Bull SD, He XP, James TD, Sessler JL, Tang BZ, Tian H, Yoon J. Excited-state intramolecular proton-transfer (ESIPT) based fluorescence sensors and imaging agents. Chem Soc Rev 2018; 47:8842-8880. [DOI: 10.1039/c8cs00185e] [Citation(s) in RCA: 690] [Impact Index Per Article: 98.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
We review recent advances in the design and application of excited-state intramolecular proton-transfer (ESIPT) based fluorescent probes. These sensors and imaging agents (probes) are important in biology, physiology, pharmacology, and environmental science.
Collapse
Affiliation(s)
- Adam C. Sedgwick
- Department of Chemistry
- University of Bath
- Bath
- UK
- Department of Chemistry
| | - Luling Wu
- Department of Chemistry
- University of Bath
- Bath
- UK
| | - Hai-Hao Han
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering
- Feringa Nobel Prize Scientist Joint Research Center
- School of Chemistry and Molecular Engineering
- East China University of Science and Technology
- Shanghai 200237
| | | | - Xiao-Peng He
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering
- Feringa Nobel Prize Scientist Joint Research Center
- School of Chemistry and Molecular Engineering
- East China University of Science and Technology
- Shanghai 200237
| | - Tony D. James
- Department of Chemistry
- University of Bath
- Bath
- UK
- Department of Materials and Life Sciences
| | | | - Ben Zhong Tang
- Department of Chemistry
- The Hong Kong University of Science & Technology (HKUST)
- Clear Water Bay
- Kowloon
- China
| | - He Tian
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering
- Feringa Nobel Prize Scientist Joint Research Center
- School of Chemistry and Molecular Engineering
- East China University of Science and Technology
- Shanghai 200237
| | - Juyoung Yoon
- Department of Chemistry and Nano Science
- Ewha Womans University
- Seoul 120-750
- Korea
| |
Collapse
|
109
|
Yoon S, Cho B, Shin M, Koranteng F, Cha N, Shim J. Iron Homeostasis Controls Myeloid Blood Cell Differentiation in Drosophila. Mol Cells 2017; 40:976-985. [PMID: 29237257 PMCID: PMC5750716 DOI: 10.14348/molcells.2017.0287] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 11/12/2017] [Indexed: 02/04/2023] Open
Abstract
Iron is an essential divalent ion for aerobic life. Life has evolved to maintain iron homeostasis for normal cellular and physiological functions and therefore imbalances in iron levels exert a wide range of consequences. Responses to iron dysregulation in blood development, however, remain elusive. Here, we found that iron homeostasis is critical for differentiation of Drosophila blood cells in the larval hematopoietic organ, called the lymph gland. Supplementation of an iron chelator, bathophenanthroline disulfate (BPS) results in an excessive differentiation of the crystal cell in the lymph gland. This phenotype is recapitulated by loss of Fer1HCH in the intestine, indicating that reduced levels of systemic iron enhances crystal cell differentiation. Detailed analysis of Fer1HCH-tagged-GFP revealed that Fer1HCH is also expressed in the hematopoietic systems. Lastly, blocking Fer1HCH expression in the mature blood cells showed marked increase in the blood differentiation of both crystal cells and plasmatocytes. Thus, our work suggests a relevance of systemic and local iron homeostasis in blood differentiation, prompting further investigation of molecular mechanisms underlying iron regulation and cell fate determination in the hematopoietic system.
Collapse
Affiliation(s)
- Sunggyu Yoon
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul 04763,
Korea
| | - Bumsik Cho
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul 04763,
Korea
| | - Mingyu Shin
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul 04763,
Korea
| | - Ferdinand Koranteng
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul 04763,
Korea
| | - Nuri Cha
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul 04763,
Korea
| | - Jiwon Shim
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul 04763,
Korea
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul 04763,
Korea
- Research Institute for Natural Science, Hanyang University, Seoul 04763,
Korea
| |
Collapse
|
110
|
Nakashige TG, Nolan EM. Human calprotectin affects the redox speciation of iron. Metallomics 2017; 9:1086-1095. [PMID: 28561859 DOI: 10.1039/c7mt00044h] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We report that the metal-sequestering human host-defense protein calprotectin (CP, S100A8/S100A9 oligomer) affects the redox speciation of iron (Fe) in bacterial growth media and buffered aqueous solution. Under aerobic conditions and in the absence of an exogenous reducing agent, CP-Ser (S100A8(C42S)/S100A9(C3S) oligomer) depletes Fe from three different bacterial growth media preparations over a 48 h timeframe (T = 30 °C). The presence of the reducing agent β-mercaptoethanol accelerates this process and allows CP-Ser to deplete Fe over a ≈1 h timeframe. Fe-depletion assays performed with metal-binding-site variants of CP-Ser show that the hexahistidine (His6) site, which coordinates Fe(ii) with high affinity, is required for Fe depletion. An analysis of Fe redox speciation in buffer containing Fe(iii) citrate performed under aerobic conditions demonstrates that CP-Ser causes a time-dependent increase in the [Fe(ii)]/[Fe(iii)] ratio. Taken together, these results indicate that the hexahistidine site of CP stabilizes Fe(ii) and thereby shifts the redox equilibrium of Fe to the reduced ferrous state under aerobic conditions. We also report that the presence of bacterial metabolites affects the Fe-depleting activity of CP-Ser. Supplementation of bacterial growth media with an Fe(iii)-scavenging siderophore (enterobactin, staphyloferrin B, or desferrioxamine B) attenuates the Fe-depleting activity of CP-Ser. This result indicates that formation of Fe(iii)-siderophore complexes blocks CP-mediated reduction of Fe(iii) and hence the ability of CP to coordinate Fe(ii). In contrast, the presence of pyocyanin (PYO), a redox-cycling phenazine produced by Pseudomonas aeruginosa that reduces Fe(iii) to Fe(ii), accelerates Fe depletion by CP-Ser under aerobic conditions. These findings indicate that the presence of microbial metabolites that contribute to metal homeostasis at the host/pathogen interface can affect the metal-sequestering function of CP.
Collapse
Affiliation(s)
- Toshiki G Nakashige
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | | |
Collapse
|
111
|
Camarena V, Sant DW, Huff TC, Mustafi S, Muir RK, Aron AT, Chang CJ, Renslo AR, Monje PV, Wang G. cAMP signaling regulates DNA hydroxymethylation by augmenting the intracellular labile ferrous iron pool. eLife 2017; 6:29750. [PMID: 29239726 PMCID: PMC5745079 DOI: 10.7554/elife.29750] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 12/13/2017] [Indexed: 12/11/2022] Open
Abstract
It is widely accepted that cAMP regulates gene transcription principally by activating the protein kinase A (PKA)-targeted transcription factors. Here, we show that cAMP enhances the generation of 5-hydroxymethylcytosine (5hmC) in multiple cell types. 5hmC is converted from 5-methylcytosine (5mC) by Tet methylcytosine dioxygenases, for which Fe(II) is an essential cofactor. The promotion of 5hmC was mediated by a prompt increase of the intracellular labile Fe(II) pool (LIP). cAMP enhanced the acidification of endosomes for Fe(II) release to the LIP likely through RapGEF2. The effect of cAMP on Fe(II) and 5hmC was confirmed by adenylate cyclase activators, phosphodiesterase inhibitors, and most notably by stimulation of G protein-coupled receptors (GPCR). The transcriptomic changes caused by cAMP occurred in concert with 5hmC elevation in differentially transcribed genes. Collectively, these data show a previously unrecognized regulation of gene transcription by GPCR-cAMP signaling through augmentation of the intracellular labile Fe(II) pool and DNA hydroxymethylation.
Collapse
Affiliation(s)
- Vladimir Camarena
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, United States.,Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, United States
| | - David W Sant
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, United States.,Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, United States
| | - Tyler C Huff
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, United States.,Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, United States
| | - Sushmita Mustafi
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, United States.,Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, United States
| | - Ryan K Muir
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Allegra T Aron
- Department of Chemistry, University of California, Berkeley, Berkeley, United States
| | - Christopher J Chang
- Department of Chemistry, University of California, Berkeley, Berkeley, United States.,Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Adam R Renslo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Paula V Monje
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, United States.,Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, United States
| | - Gaofeng Wang
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, United States.,Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, United States.,Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, United States.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, United States
| |
Collapse
|
112
|
Iron-loaded transferrin (Tf) is detrimental whereas iron-free Tf confers protection against brain ischemia by modifying blood Tf saturation and subsequent neuronal damage. Redox Biol 2017; 15:143-158. [PMID: 29248829 PMCID: PMC5975212 DOI: 10.1016/j.redox.2017.11.026] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/28/2017] [Accepted: 11/29/2017] [Indexed: 01/18/2023] Open
Abstract
Despite transferrin being the main circulating carrier of iron in body fluids, and iron overload conditions being known to worsen stroke outcome through reactive oxygen species (ROS)-induced damage, the contribution of blood transferrin saturation (TSAT) to stroke brain damage is unknown. The objective of this study was to obtain evidence on whether TSAT determines the impact of experimental ischemic stroke on brain damage and whether iron-free transferrin (apotransferrin, ATf)-induced reduction of TSAT is neuroprotective. We found that experimental ischemic stroke promoted an early extravasation of circulating iron-loaded transferrin (holotransferrin, HTf) to the ischemic brain parenchyma. In vitro, HTf was found to boost ROS production and to be harmful to primary neuronal cultures exposed to oxygen and glucose deprivation. In stroked rats, whereas increasing TSAT with exogenous HTf was detrimental, administration of exogenous ATf and the subsequent reduction of TSAT was neuroprotective. Mechanistically, ATf did not prevent extravasation of HTf to the brain parenchyma in rats exposed to ischemic stroke. However, ATf in vitro reduced NMDA-induced neuronal uptake of HTf and also both the NMDA-mediated lipid peroxidation derived 4-HNE and the resulting neuronal death without altering Ca2+-calcineurin signaling downstream the NMDA receptor. Removal of transferrin from the culture media or blockade of transferrin receptors reduced neuronal death. Together, our data establish that blood TSAT exerts a critical role in experimental stroke-induced brain damage. In addition, our findings suggest that the protective effect of ATf at the neuronal level resides in preventing NMDA-induced HTf uptake and ROS production, which in turn reduces neuronal damage. Blood TSAT is pivotal to determine neuronal fate in rat models of stroke During ischemia blood transferrin extravasates and accumulates in ischemic neurons. Increasing TSAT with holotransferrin (HTf) is detrimental in rat models of stroke. Decreasing TSAT with apotransferrin (ATf) is beneficial in rat models of stroke HTf promotes and ATf reduces ROS-, iron- and NMDAR-initiated ischemic neuronal death.
Collapse
|
113
|
Discovery of DS28120313 as a potent orally active hepcidin production inhibitor: Design and optimization of novel 4,6-disubstituted indazole derivatives. Bioorg Med Chem Lett 2017; 27:5252-5257. [DOI: 10.1016/j.bmcl.2017.10.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/13/2017] [Accepted: 10/15/2017] [Indexed: 11/19/2022]
|
114
|
Rosa L, Cutone A, Lepanto MS, Paesano R, Valenti P. Lactoferrin: A Natural Glycoprotein Involved in Iron and Inflammatory Homeostasis. Int J Mol Sci 2017; 18:1985. [PMID: 28914813 PMCID: PMC5618634 DOI: 10.3390/ijms18091985&n948647=v984776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Human lactoferrin (hLf), an iron-binding multifunctional cationic glycoprotein secreted by exocrine glands and by neutrophils, is a key element of host defenses. HLf and bovine Lf (bLf), possessing high sequence homology and identical functions, inhibit bacterial growth and biofilm dependently from iron binding ability while, independently, bacterial adhesion to and the entry into cells. In infected/inflamed host cells, bLf exerts an anti-inflammatory activity against interleukin-6 (IL-6), thus up-regulating ferroportin (Fpn) and transferrin receptor 1 (TfR1) and down-regulating ferritin (Ftn), pivotal actors of iron and inflammatory homeostasis (IIH). Consequently, bLf inhibits intracellular iron overload, an unsafe condition enhancing in vivo susceptibility to infections, as well as anemia of inflammation (AI), re-establishing IIH. In pregnant women, affected by AI, bLf oral administration decreases IL-6 and increases hematological parameters. This surprising effect is unrelated to iron supplementation by bLf (80 μg instead of 1-2 mg/day), but to its role on IIH. AI is unrelated to the lack of iron, but to iron delocalization: cellular/tissue overload and blood deficiency. BLf cures AI by restoring iron from cells to blood through Fpn up-expression. Indeed, anti-inflammatory activity of oral and intravaginal bLf prevents preterm delivery. Promising bLf treatments can prevent/cure transitory inflammation/anemia/oral pathologies in athletes.
Collapse
Affiliation(s)
- Luigi Rosa
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Antimo Cutone
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Maria Stefania Lepanto
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Rosalba Paesano
- Department of Gynecological-Obstetric and Urological Sciences, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Piera Valenti
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| |
Collapse
|
115
|
Rosa L, Cutone A, Lepanto MS, Paesano R, Valenti P. Lactoferrin: A Natural Glycoprotein Involved in Iron and Inflammatory Homeostasis. Int J Mol Sci 2017; 18:E1985. [PMID: 28914813 PMCID: PMC5618634 DOI: 10.3390/ijms18091985] [Citation(s) in RCA: 242] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/11/2017] [Accepted: 09/12/2017] [Indexed: 02/07/2023] Open
Abstract
Human lactoferrin (hLf), an iron-binding multifunctional cationic glycoprotein secreted by exocrine glands and by neutrophils, is a key element of host defenses. HLf and bovine Lf (bLf), possessing high sequence homology and identical functions, inhibit bacterial growth and biofilm dependently from iron binding ability while, independently, bacterial adhesion to and the entry into cells. In infected/inflamed host cells, bLf exerts an anti-inflammatory activity against interleukin-6 (IL-6), thus up-regulating ferroportin (Fpn) and transferrin receptor 1 (TfR1) and down-regulating ferritin (Ftn), pivotal actors of iron and inflammatory homeostasis (IIH). Consequently, bLf inhibits intracellular iron overload, an unsafe condition enhancing in vivo susceptibility to infections, as well as anemia of inflammation (AI), re-establishing IIH. In pregnant women, affected by AI, bLf oral administration decreases IL-6 and increases hematological parameters. This surprising effect is unrelated to iron supplementation by bLf (80 μg instead of 1-2 mg/day), but to its role on IIH. AI is unrelated to the lack of iron, but to iron delocalization: cellular/tissue overload and blood deficiency. BLf cures AI by restoring iron from cells to blood through Fpn up-expression. Indeed, anti-inflammatory activity of oral and intravaginal bLf prevents preterm delivery. Promising bLf treatments can prevent/cure transitory inflammation/anemia/oral pathologies in athletes.
Collapse
Affiliation(s)
- Luigi Rosa
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Antimo Cutone
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Maria Stefania Lepanto
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Rosalba Paesano
- Department of Gynecological-Obstetric and Urological Sciences, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Piera Valenti
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| |
Collapse
|
116
|
Regenboog M, Bohte AE, Akkerman EM, Stoker J, Hollak CE. Iron storage in liver, bone marrow and splenic Gaucheroma reflects residual disease in type 1 Gaucher disease patients on treatment. Br J Haematol 2017; 179:635-647. [DOI: 10.1111/bjh.14915] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/18/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Martine Regenboog
- Department of Internal Medicine; Division of Endocrinology & Metabolism; Academic Medical Centre; University of Amsterdam; Amsterdam the Netherlands
- Department of Radiology; Academic Medical Centre; University of Amsterdam; Amsterdam the Netherlands
| | - Anneloes E. Bohte
- Department of Radiology; Academic Medical Centre; University of Amsterdam; Amsterdam the Netherlands
| | - Erik M. Akkerman
- Department of Radiology; Academic Medical Centre; University of Amsterdam; Amsterdam the Netherlands
| | - Jaap Stoker
- Department of Radiology; Academic Medical Centre; University of Amsterdam; Amsterdam the Netherlands
| | - Carla E.M. Hollak
- Department of Internal Medicine; Division of Endocrinology & Metabolism; Academic Medical Centre; University of Amsterdam; Amsterdam the Netherlands
| |
Collapse
|
117
|
Liu Z, Purro M, Qiao J, Xiong MP. Multifunctional Polymeric Micelles for Combining Chelation and Detection of Iron in Living Cells. Adv Healthc Mater 2017; 6:10.1002/adhm.201700162. [PMID: 28661064 PMCID: PMC5587393 DOI: 10.1002/adhm.201700162] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 05/24/2017] [Indexed: 01/13/2023]
Abstract
Multifunctional self-assembled micelles composed of Pluronics F127 polymer chains are developed and investigated for chelation and selective detection of iron(III) in vitro and in iron-overloaded cells. Tetraphenylethene (TPE) is encapsulated into the micelle core and the iron chelate drug deferoxamine (DFO) is conjugated to micelles to generate a fluorescence quenching detection system termed DFO-TFM for short, where T stands for TPE, F for F127, and M for micelle. The key to the successful formation of this fluorescence quenching system is due to the near-ideal overlap between the absorption spectrum of the DFO:iron(III) complex and fluorescence spectrum of TPE. DFO-TFM can retain the iron-chelation properties of DFO and exhibits negligible cytotoxicity compared to free DFO. Furthermore, this fluorescence "turn-off" system can be utilized to detect the presence of iron and to monitor the chelation process in an iron overload cell model. This study serves as an effective proof-of-concept model for designing future in vivo systems capable of combining the features of iron chelation with iron detection and efforts toward the development of such detection systems are currently underway.
Collapse
Affiliation(s)
- Zhi Liu
- Department of Pharmaceutical & Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, 30602-2352, USA
| | - Max Purro
- Department of Pharmaceutical & Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, 30602-2352, USA
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705-2222, USA
| | - Jing Qiao
- Department of Pharmaceutical & Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, 30602-2352, USA
| | - May P Xiong
- Department of Pharmaceutical & Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, 30602-2352, USA
| |
Collapse
|
118
|
Soto-Castro L, Plata-Guzmán LY, Figueroa-Angulo EE, Calla-Choque JS, Reyes-López M, de la Garza M, León-Sicairos N, Garzón-Tiznado JA, Arroyo R, León-Sicairos C. Iron responsive-like elements in the parasite Entamoeba histolytica. Microbiology (Reading) 2017; 163:1329-1342. [DOI: 10.1099/mic.0.000431] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Liliana Soto-Castro
- Programa Regional del Noroeste para el Posgrado en Biotecnología de la Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Av. de las Américas y Josefa Ortíz (Cd. Universitaria) Culiacán, Sinaloa 80030, Mexico
| | - Laura Yuliana Plata-Guzmán
- Programa Regional del Noroeste para el Posgrado en Biotecnología de la Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Av. de las Américas y Josefa Ortíz (Cd. Universitaria) Culiacán, Sinaloa 80030, Mexico
| | - Elisa Elvira Figueroa-Angulo
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Avenida Instituto Politécnico Nacional No. 2508, Colonia San Pedro Zacatenco, México, D.F. 07360, Mexico
| | - Jaeson Santos Calla-Choque
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Avenida Instituto Politécnico Nacional No. 2508, Colonia San Pedro Zacatenco, México, D.F. 07360, Mexico
| | - Magda Reyes-López
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Avenida Instituto Politécnico Nacional No. 2508, Colonia San Pedro Zacatenco, México D.F. 07360, Mexico
| | - Mireya de la Garza
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Avenida Instituto Politécnico Nacional No. 2508, Colonia San Pedro Zacatenco, México D.F. 07360, Mexico
| | - Nidia León-Sicairos
- Programa Regional del Noroeste para el Posgrado en Biotecnología de la Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Av. de las Américas y Josefa Ortíz (Cd. Universitaria) Culiacán, Sinaloa 80030, Mexico
| | - José Antonio Garzón-Tiznado
- Programa Regional del Noroeste para el Posgrado en Biotecnología de la Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Av. de las Américas y Josefa Ortíz (Cd. Universitaria) Culiacán, Sinaloa 80030, Mexico
| | - Rossana Arroyo
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Avenida Instituto Politécnico Nacional No. 2508, Colonia San Pedro Zacatenco, México, D.F. 07360, Mexico
| | - Claudia León-Sicairos
- Programa Regional del Noroeste para el Posgrado en Biotecnología de la Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Av. de las Américas y Josefa Ortíz (Cd. Universitaria) Culiacán, Sinaloa 80030, Mexico
| |
Collapse
|
119
|
Fukuda T, Goto R, Kiho T, Ueda K, Muramatsu S, Hashimoto M, Aki A, Watanabe K, Tanaka N. Discovery of DS79182026: A potent orally active hepcidin production inhibitor. Bioorg Med Chem Lett 2017; 27:3716-3722. [DOI: 10.1016/j.bmcl.2017.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/27/2017] [Accepted: 07/01/2017] [Indexed: 10/19/2022]
|
120
|
Visscher C, Middendorf L, Günther R, Engels A, Leibfacher C, Möhle H, Düngelhoef K, Weier S, Haider W, Radko D. Fat content, fatty acid pattern and iron content in livers of turkeys with hepatic lipidosis. Lipids Health Dis 2017; 16:98. [PMID: 28558775 PMCID: PMC5450264 DOI: 10.1186/s12944-017-0484-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/11/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The so-called "hepatic lipidosis" in turkeys is an acute progressive disease associated with a high mortality rate in a very short time. Dead animals show a massive fatty degeneration of the liver. The cause is still unclear. Previous findings suggest that there may be parallels to human non-alcoholic fatty liver disease. The object of the study was to examine the changes in the fat contents, the fatty acid composition and the iron content in livers of animals, which have died from hepatic lipidosis. METHODS The conspicuous livers (n = 85) were collected from 20 flocks where the phenomenon of massive increased animal losses accompanied by marked macroscopically visible pathological liver steatosis suddenly occurred. For comparison and as a reference, livers (n = 16) of two healthy flocks were taken. Healthy and diseased flocks were fed identical diets concerning official nutrient recommendations and were operating under standardized, comparable conventional conditions. RESULTS Compared to livers of healthy animals, in the livers of turkeys died from hepatic lipidosis there were found massively increased fat levels (130 ± 33.2 vs. 324 ± 101 g/kg dry matter-DM). In all fatty livers, different fatty acids concentrations were present in significantly increased concentrations compared to controls (palmitic acid: 104 g/kg DM, +345%; palmitoleic acid: 18.0 g/kg DM, + 570%; oleic acid: 115 g/kg DM, +437%). Fatty acids concentrations relevant for liver metabolism and inflammation were significantly reduced (arachidonic acid: 2.92 g/kg DM, -66.6%; eicosapentaenoic acid: 0.141 g/kg DM, -78.3%; docosahexaenoic acid: 0.227 g/kg DM, -90.4%). The ratio of certain fatty acids to one another between control and case livers changed analogously to liver diseases in humans (e.g.: C18:0/C16:0 - 0.913 against 0.311; C16:1n7/C16:0 - 0.090 against 0.165; C18:1/C18:0 - 0.938 against 4.03). The iron content in the liver tissue also increased massively (271 ± 51.5 vs 712 ± 214 mg/kg DM). CONCLUSION The hepatic lipidosis has a massive impact on the lipid content, the lipid composition and the iron content in the liver. The character of the metabolic disorder includes parallels to the non-alcoholic steatohepatitis in humans.
Collapse
Affiliation(s)
- Christian Visscher
- Institute for Animal Nutrition, University of Veterinary Medicine Hannover, Foundation, Bischofsholer Damm 15, D-30173, Hannover, Germany.
| | - Lea Middendorf
- Institute for Animal Nutrition, University of Veterinary Medicine Hannover, Foundation, Bischofsholer Damm 15, D-30173, Hannover, Germany
| | - Ronald Günther
- Heidemark GmbH, Veterinärlabor, Jakob-Uffrecht-Str. 20, D-39340, Haldensleben, Germany
| | - Alexandra Engels
- Tierarztpraxis Dr. A. Engels, Gut Hacheney 2-5, D-59199, Bönen-Lenningsen, Germany
| | - Christof Leibfacher
- Tierarztpraxis Dr. A. Engels, Gut Hacheney 2-5, D-59199, Bönen-Lenningsen, Germany
| | - Henrik Möhle
- Tierärztliche Gemeinschaftspraxis Dres. Windhaus & Hemme, An der Ohe 1, D-49377, Vechta, Germany
| | - Kristian Düngelhoef
- Tierarztpraxis an der Güterstraße, Güterstraße 7, D-46499, Hamminkeln, Germany
| | - Stefan Weier
- Praxis am Bergweg, Bergweg 20, D-49393, Lohne (Oldenburg), Germany
| | - Wolfram Haider
- Institut für Tierpathologie, Schönhauser Str. 62, D-13127, Berlin, Germany
| | - Dimitri Radko
- Elanco Animal Health GmbH, Werner-Reimers-Str. 2-4, Bad Homburg, D-61352, Germany
| |
Collapse
|
121
|
Carr CE, Musiani F, Huang HT, Chivers PT, Ciurli S, Maroney MJ. Glutamate Ligation in the Ni(II)- and Co(II)-Responsive Escherichia coli Transcriptional Regulator, RcnR. Inorg Chem 2017; 56:6459-6476. [PMID: 28517938 DOI: 10.1021/acs.inorgchem.7b00527] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Escherichia coli RcnR (resistance to cobalt and nickel regulator, EcRcnR) is a metal-responsive repressor of the genes encoding the Ni(II) and Co(II) exporter proteins RcnAB by binding to PRcnAB. The DNA binding affinity is weakened when the cognate ions Ni(II) and Co(II) bind to EcRcnR in a six-coordinate site that features a (N/O)5S ligand donor-atom set in distinct sites: while both metal ions are bound by the N terminus, Cys35, and His64, Co(II) is additionally bound by His3. On the other hand, the noncognate Zn(II) and Cu(I) ions feature a lower coordination number, have a solvent-accessible binding site, and coordinate protein ligands that do not include the N-terminal amine. A molecular model of apo-EcRcnR suggested potential roles for Glu34 and Glu63 in binding Ni(II) and Co(II) to EcRcnR. The roles of Glu34 and Glu63 in metal binding, metal selectivity, and function were therefore investigated using a structure/function approach. X-ray absorption spectroscopy was used to assess the structural changes in the Ni(II), Co(II), and Zn(II) binding sites of Glu → Ala and Glu → Cys variants at both positions. The effect of these structural alterations on the regulation of PrcnA by EcRcnR in response to metal binding was explored using LacZ reporter assays. These combined studies indicate that while Glu63 is a ligand for both metal ions, Glu34 is a ligand for Co(II) but possibly not for Ni(II). The Glu34 variants affect the structure of the cognate metal sites, but they have no effect on the transcriptional response. In contrast, the Glu63 variants affect both the structure and transcriptional response, although they do not completely abolish the function of EcRcnR. The structure of the Zn(II) site is not significantly perturbed by any of the glutamic acid variations. The spectroscopic and functional data obtained on the mutants were used to calculate models of the metal-site structures of EcRcnR bound to Ni(II), Co(II), and Zn(II). The results are interpreted in terms of a switch mechanism, in which a subset of the metal-binding ligands is responsible for the allosteric response required for DNA release.
Collapse
Affiliation(s)
- Carolyn E Carr
- Department of Chemistry, University of Massachusetts , Amherst, Massachusetts 01003, United States
| | - Francesco Musiani
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna , Bologna 40126, Italy
| | - Hsin-Ting Huang
- Department of Chemistry, University of Massachusetts , Amherst, Massachusetts 01003, United States
| | - Peter T Chivers
- Departments of Biosciences and Chemistry, Durham University , Durham DH1 3LE, United Kingdom
| | - Stefano Ciurli
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna , Bologna 40126, Italy
| | - Michael J Maroney
- Department of Chemistry, University of Massachusetts , Amherst, Massachusetts 01003, United States
| |
Collapse
|
122
|
Pino JMV, da Luz MHM, Antunes HKM, Giampá SQDC, Martins VR, Lee KS. Iron-Restricted Diet Affects Brain Ferritin Levels, Dopamine Metabolism and Cellular Prion Protein in a Region-Specific Manner. Front Mol Neurosci 2017; 10:145. [PMID: 28567002 PMCID: PMC5434142 DOI: 10.3389/fnmol.2017.00145] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/28/2017] [Indexed: 01/03/2023] Open
Abstract
Iron is an essential micronutrient for several physiological functions, including the regulation of dopaminergic neurotransmission. On the other hand, both iron, and dopamine can affect the folding and aggregation of proteins related with neurodegenerative diseases, such as cellular prion protein (PrPC) and α-synuclein, suggesting that deregulation of iron homeostasis and the consequential disturbance of dopamine metabolism can be a risk factor for conformational diseases. These proteins, in turn, are known to participate in the regulation of iron and dopamine metabolism. In this study, we evaluated the effects of dietary iron restriction on brain ferritin levels, dopamine metabolism, and the expression levels of PrPC and α-synuclein. To achieve this goal, C57BL/6 mice were fed with iron restricted diet (IR) or with normal diet (CTL) for 1 month. IR reduced iron and ferritin levels in liver. Ferritin reduction was also observed in the hippocampus. However, in the striatum of IR group, ferritin level was increased, suggesting that under iron-deficient condition, each brain area might acquire distinct capacity to store iron. Increased lipid peroxidation was observed only in hippocampus of IR group, where ferritin level was reduced. IR also generated discrete results regarding dopamine metabolism of distinct brain regions: in striatum, the level of dopamine metabolites (DOPAC and HVA) was reduced; in prefrontal cortex, only HVA was increased along with the enhanced MAO-A activity; in hippocampus, no alterations were observed. PrPC levels were increased only in the striatum of IR group, where ferritin level was also increased. PrPC is known to play roles in iron uptake. Thus, the increase of PrPC in striatum of IR group might be related to the increased ferritin level. α-synuclein was not altered in any regions. Abnormal accumulation of ferritin, increased MAO-A activity or lipid peroxidation are molecular features observed in several neurological disorders. Our findings show that nutritional iron deficiency produces these molecular alterations in a region-specific manner and provide new insight into the variety of molecular pathways that can lead to distinct neurological symptoms upon iron deficiency. Thus, adequate iron supplementation is essential for brain health and prevention of neurological diseases.
Collapse
Affiliation(s)
- Jessica M V Pino
- Departamento de Bioquímica, Universidade Federal de São PauloSão Paulo, Brazil
| | - Marcio H M da Luz
- Departamento de Bioquímica, Universidade Federal de São PauloSão Paulo, Brazil
| | - Hanna K M Antunes
- Departamento de Psicobiologia, Universidade Federal de São PauloSão Paulo, Brazil.,Departamento de Biociências, Universidade Federal de São PauloSão Paulo, Brazil
| | | | | | - Kil S Lee
- Departamento de Bioquímica, Universidade Federal de São PauloSão Paulo, Brazil
| |
Collapse
|
123
|
Fukuda T, Ueda K, Ishiyama T, Goto R, Muramatsu S, Hashimoto M, Watanabe K, Tanaka N. Synthesis and SAR studies of 3,6-disubstituted indazole derivatives as potent hepcidin production inhibitors. Bioorg Med Chem Lett 2017; 27:2148-2152. [DOI: 10.1016/j.bmcl.2017.03.056] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/09/2017] [Accepted: 03/22/2017] [Indexed: 10/19/2022]
|
124
|
FBXL5 Inactivation in Mouse Brain Induces Aberrant Proliferation of Neural Stem Progenitor Cells. Mol Cell Biol 2017; 37:MCB.00470-16. [PMID: 28069738 DOI: 10.1128/mcb.00470-16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 01/03/2017] [Indexed: 01/05/2023] Open
Abstract
FBXL5 is the substrate recognition subunit of an SCF-type ubiquitin ligase that serves as a master regulator of iron metabolism in mammalian cells. We previously showed that mice with systemic deficiency of FBXL5 fail to sense intracellular iron levels and die in utero at embryonic day 8.5 (E8.5) as a result of iron overload and subsequent oxidative stress. This early embryonic mortality has thus impeded study of the role of FBXL5 in brain development. We have now generated mice lacking FBXL5 specifically in nestin-expressing neural stem progenitor cells (NSPCs) in the brain. Unexpectedly, the mutant embryos manifested a progressive increase in the number of NSPCs and astroglia in the cerebral cortex. Stabilization of iron regulatory protein 2 (IRP2) as a result of FBXL5 deficiency led to accumulation of ferrous and ferric iron as well as to generation of reactive oxygen species. Pharmacological manipulation suggested that the phenotypes of FBXL5 deficiency are attributable to aberrant activation of mammalian target of rapamycin (mTOR) signaling. Our results thus show that FBXL5 contributes to regulation of NSPC proliferation during mammalian brain development.
Collapse
|
125
|
Lippitsch A, Chukovetskyi Y, Baal N, Bein G, Hackstein H. Unique high and homogenous surface expression of the transferrin receptor CD71 on murine plasmacytoid dendritic cells in different tissues. Cell Immunol 2017; 316:41-52. [PMID: 28372797 DOI: 10.1016/j.cellimm.2017.03.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 02/28/2017] [Accepted: 03/27/2017] [Indexed: 01/03/2023]
Abstract
Plasmacytoid dendritic cells (pDC) are of increasing interest in cancer vaccine development, but many functions of these highly specialized, multifaceted cells are poorly understood. The transferrin receptor CD71 has also been suggested to function as an antigen uptake receptor on professional antigen-presenting cells. In this study, we employed multiparameter flow cytometry to investigate CD71 expression on various leukocyte subsets, including DC subsets, granulocytes, macrophages, T and B lymphocytes, γδ T cells, and natural killer cells. Cells from various lymphoid and non-lymphoid murine tissues were analyzed using fluorochrome-conjugated monoclonal antibodies. High CD71 expression (90-100%) was observed, uniquely on pDC amongst the leukocyte populations examined, in both lymphoid and non-lymphoid tissues, including other DC subsets. In contrast, CD71 expression on non-tissue pDC, in the bone marrow and peripheral blood, was reduced. The cause and function of this high tissue pDC-selective CD71 expression remain to be examined.
Collapse
Affiliation(s)
- Anne Lippitsch
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig-University Giessen, Biomedical Research Center Seltersberg (BFS), Schubertstrasse 81, D-35392 Giessen, Germany
| | - Yuri Chukovetskyi
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig-University Giessen, Biomedical Research Center Seltersberg (BFS), Schubertstrasse 81, D-35392 Giessen, Germany
| | - Nelli Baal
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig-University Giessen, Biomedical Research Center Seltersberg (BFS), Schubertstrasse 81, D-35392 Giessen, Germany
| | - Gregor Bein
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig-University Giessen, Biomedical Research Center Seltersberg (BFS), Schubertstrasse 81, D-35392 Giessen, Germany
| | - Holger Hackstein
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig-University Giessen, Biomedical Research Center Seltersberg (BFS), Schubertstrasse 81, D-35392 Giessen, Germany.
| |
Collapse
|
126
|
Oliveira ALGD, Chaves AT, Menezes CAS, Guimarães NS, Bueno LL, Fujiwara RT, Rocha MODC. Vitamin D receptor expression and hepcidin levels in the protection or severity of leprosy: a systematic review. Microbes Infect 2017; 19:311-322. [PMID: 28323068 DOI: 10.1016/j.micinf.2017.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/06/2017] [Accepted: 03/03/2017] [Indexed: 12/20/2022]
Abstract
Leprosy is a chronic infectious disease whose disequilibrium in the host's genetic, immunological and clinical mechanisms leads to distinct manifestations defining the type of immunological response. This review focuses its attention on the influence of the Vitamin D Receptor and hepcidin expressions that can suggest the protection or severity of leprosy.
Collapse
Affiliation(s)
- Ana Laura Grossi de Oliveira
- Post-graduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil.
| | - Ana Thereza Chaves
- Post-graduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | | | - Nathalia Sernizon Guimarães
- Post-graduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | | | - Ricardo Toshio Fujiwara
- Post-graduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Manoel Otávio da Costa Rocha
- Post-graduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
127
|
Yang X, Park SH, Chang HC, Shapiro JS, Vassilopoulos A, Sawicki KT, Chen C, Shang M, Burridge PW, Epting CL, Wilsbacher LD, Jenkitkasemwong S, Knutson M, Gius D, Ardehali H. Sirtuin 2 regulates cellular iron homeostasis via deacetylation of transcription factor NRF2. J Clin Invest 2017; 127:1505-1516. [PMID: 28287409 DOI: 10.1172/jci88574] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 01/19/2017] [Indexed: 01/01/2023] Open
Abstract
SIRT2 is a cytoplasmic sirtuin that plays a role in various cellular processes, including tumorigenesis, metabolism, and inflammation. Since these processes require iron, we hypothesized that SIRT2 directly regulates cellular iron homeostasis. Here, we have demonstrated that SIRT2 depletion results in a decrease in cellular iron levels both in vitro and in vivo. Mechanistically, we determined that SIRT2 maintains cellular iron levels by binding to and deacetylating nuclear factor erythroid-derived 2-related factor 2 (NRF2) on lysines 506 and 508, leading to a reduction in total and nuclear NRF2 levels. The reduction in nuclear NRF2 leads to reduced ferroportin 1 (FPN1) expression, which in turn results in decreased cellular iron export. Finally, we observed that Sirt2 deletion reduced cell viability in response to iron deficiency. Moreover, livers from Sirt2-/- mice had decreased iron levels, while this effect was reversed in Sirt2-/- Nrf2-/- double-KO mice. Taken together, our results uncover a link between sirtuin proteins and direct control over cellular iron homeostasis via regulation of NRF2 deacetylation and stability.
Collapse
|
128
|
Iron Reduces M1 Macrophage Polarization in RAW264.7 Macrophages Associated with Inhibition of STAT1. Mediators Inflamm 2017; 2017:8570818. [PMID: 28286378 PMCID: PMC5327769 DOI: 10.1155/2017/8570818] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/18/2017] [Indexed: 01/22/2023] Open
Abstract
Iron metabolism in inflammation has been mostly characterized in macrophages exposed to pathogens or inflammatory conditions. The aim of this study is to investigate the cross-regulatory interactions between M1 macrophage polarization and iron metabolism. Firstly, we characterized the transcription of genes related to iron homeostasis in M1 RAW264.7 macrophages stimulated by IFN-γ. The molecular signature of M1 macrophages showed high levels of iron storage (ferritin), a low level of iron export (ferroportin), and changes of iron regulators (hepcidin and transferrin receptors), which favour iron sequestration in the reticuloendothelial system and are benefit for inflammatory disorders. Then, we evaluated the effect of iron on M1 macrophage polarization. Iron significantly reduced mRNA levels of IL-6, IL-1β, TNF-α, and iNOS produced by IFN-γ-polarized M1 macrophages. Immunofluorescence analysis showed that iron also reduced iNOS production. However, iron did not compromise but enhanced the ability of M1-polarized macrophages to phagocytose FITC-dextran. Moreover, we demonstrated that STAT1 inhibition was required for reduction of iNOS and M1-related cytokines production by the present of iron. Together, these findings indicated that iron decreased polarization of M1 macrophages and inhibited the production of the proinflammatory cytokines. The results expanded our knowledge about the role of iron in macrophage polarization.
Collapse
|
129
|
Lihi N, Godó AJ, Sciortino G, Garribba E, Várnagy K. Tridentate (O,N,O) ligands as potential chelator compounds for iron overload. Polyhedron 2017. [DOI: 10.1016/j.poly.2016.11.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
130
|
Milto IV, Suhodolo IV, Prokopieva VD, Klimenteva TK. Molecular and Cellular Bases of Iron Metabolism in Humans. BIOCHEMISTRY (MOSCOW) 2017; 81:549-64. [PMID: 27301283 DOI: 10.1134/s0006297916060018] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Iron is a microelement with the most completely studied biological functions. Its wide dissemination in nature and involvement in key metabolic pathways determine the great importance of this metal for uni- and multicellular organisms. The biological role of iron is characterized by its indispensability in cell respiration and various biochemical processes providing normal functioning of cells and organs of the human body. Iron also plays an important role in the generation of free radicals, which under different conditions can be useful or damaging to biomolecules and cells. In the literature, there are many reviews devoted to iron metabolism and its regulation in pro- and eukaryotes. Significant progress has been achieved recently in understanding molecular bases of iron metabolism. The purpose of this review is to systematize available data on mechanisms of iron assimilation, distribution, and elimination from the human body, as well as on its biological importance and on the major iron-containing proteins. The review summarizes recent ideas about iron metabolism. Special attention is paid to mechanisms of iron absorption in the small intestine and to interrelationships of cellular and extracellular pools of this metal in the human body.
Collapse
Affiliation(s)
- I V Milto
- Siberian State Medical University, Tomsk, 634050, Russia.
| | | | | | | |
Collapse
|
131
|
Does any drug to treat cancer target mTOR and iron hemostasis in neurodegenerative disorders? Biometals 2016; 30:1-16. [PMID: 27853903 DOI: 10.1007/s10534-016-9981-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 11/08/2016] [Indexed: 12/23/2022]
Abstract
The prevalence of neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease and Huntington's disease are increased by age. Alleviation of their symptoms and protection of normal neurons against degeneration are the main aspects of the research to establish novel therapeutic strategies. Iron as the one of most important cation not only play important role in the structure of electron transport chain proteins but also has pivotal duties in cellular activities. But disruption in iron hemostasis can make it toxin to neurons which causes lipid peroxidation, DNA damage and etc. In patients with Alzheimer and Parkinson misbalancing in iron homeostasis accelerate neurodegeneration and cause neuroinflmmation. mTOR as the common signaling pathway between cancer and neurodegenerative disorders controls iron uptake and it is in active form in both diseases. Anti-cancer drugs which target mTOR causes iron deficiency and dual effects of mTOR inhibitors can candidate them as a therapeutic strategy to alleviate neurodegeneration/inflammation because of iron overloading.
Collapse
|
132
|
Regenboog M, van Kuilenburg AB, Verheij J, Swinkels DW, Hollak CE. Hyperferritinemia and iron metabolism in Gaucher disease: Potential pathophysiological implications. Blood Rev 2016; 30:431-437. [DOI: 10.1016/j.blre.2016.05.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/13/2016] [Accepted: 05/24/2016] [Indexed: 01/10/2023]
|
133
|
Thévenod F, Wolff NA. Iron transport in the kidney: implications for physiology and cadmium nephrotoxicity. Metallomics 2016; 8:17-42. [PMID: 26485516 DOI: 10.1039/c5mt00215j] [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
The kidney has recently emerged as an organ with a significant role in systemic iron (Fe) homeostasis. Substantial amounts of Fe are filtered by the kidney, which have to be reabsorbed to prevent Fe deficiency. Accordingly Fe transporters and receptors for protein-bound Fe are expressed in the nephron that may also function as entry pathways for toxic metals, such as cadmium (Cd), by way of "ionic and molecular mimicry". Similarities, but also differences in handling of Cd by these transport routes offer rationales for the propensity of the kidney to develop Cd toxicity. This critical review provides a comprehensive update on Fe transport by the kidney and its relevance for physiology and Cd nephrotoxicity. Based on quantitative considerations, we have also estimated the in vivo relevance of the described transport pathways for physiology and toxicology. Under physiological conditions all segments of the kidney tubules are likely to utilize Fe for cellular Fe requiring processes for metabolic purposes and also to contribute to reabsorption of free and bound forms of Fe into the circulation. But Cd entering tubule cells disrupts metabolic pathways and is unable to exit. Furthermore, our quantitative analyses contest established models linking chronic Cd nephrotoxicity to proximal tubular uptake of metallothionein-bound Cd. Hence, Fe transport by the kidney may be beneficial by preventing losses from the body. But increased uptake of Fe or Cd that cannot exit tubule cells may lead to kidney injury, and Fe deficiency may facilitate renal Cd uptake.
Collapse
Affiliation(s)
- Frank Thévenod
- Institute of Physiology, Pathophysiology & Toxicology, Center for Biomedical Training and Research (ZBAF), University of Witten/Herdecke, Stockumer Str. 12, 58453 Witten, Germany.
| | - Natascha A Wolff
- Institute of Physiology, Pathophysiology & Toxicology, Center for Biomedical Training and Research (ZBAF), University of Witten/Herdecke, Stockumer Str. 12, 58453 Witten, Germany.
| |
Collapse
|
134
|
Liu Z, Lin TM, Purro M, Xiong MP. Enzymatically Biodegradable Polyrotaxane-Deferoxamine Conjugates for Iron Chelation. ACS APPLIED MATERIALS & INTERFACES 2016; 8:25788-25797. [PMID: 27623539 PMCID: PMC5560162 DOI: 10.1021/acsami.6b09077] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Chelation therapy is frequently used to help reduce excess iron in the body, but current chelators such as deferoxamine (DFO) are plagued by short blood circulation times, which necessitates infusions and can cause undesirable toxic side effects in patients. To address these issues, polyrotaxanes (PR) were synthesized by threading α-cyclodextrin (α-CD) onto poly(ethylene glycol) bis(amine) (PEG-BA, MW 3400 g/mol) capped with enzymatically cleavable bulky Z-L phenylalanine (Z-L Phe) moieties. The resulting PR was conjugated to DFO and hydroxypropylated to generate the final polyrotaxane-DFO (hPR-DFO). The iron chelating capability of hPR-DFO was verified by UV-vis absorption spectroscopy and the ability of materials to degrade into smaller CD-conjugated DFO fragments (hCD-DFO) in the presence of the protease was confirmed via gel permeation chromatography. In vitro studies in iron-overloaded macrophages reveal that hPR-DFO can significantly reduce the cytotoxicity of the drug while maintaining its chelation efficacy, and that it is more rapidly endocytosed and trafficked to lysosomes of iron-overloaded cells in comparison to non-iron-overloaded macrophages. In vivo studies indicate that iron-overloaded mice treated with hPR-DFO displayed lower serum ferritin levels (a measure of iron burden in the body) and could eliminate excess iron by both the renal and fecal routes. Moreover, there was no gross evidence of acute toxicological damage to the liver or spleen.
Collapse
Affiliation(s)
- Zhi Liu
- Department of Pharmaceutical & Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602-2352, USA
| | - Tien-min Lin
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin–Madison, Madison, WI 53705-2222, USA
| | - Max Purro
- Department of Pharmaceutical & Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602-2352, USA
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin–Madison, Madison, WI 53705-2222, USA
| | - May P. Xiong
- Department of Pharmaceutical & Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602-2352, USA
- Correspondence and requests for materials should be addressed to May P. Xiong.
| |
Collapse
|
135
|
Tian T, Blanco E, Smoukov SK, Velev OD, Velikov KP. Dissolution behaviour of ferric pyrophosphate and its mixtures with soluble pyrophosphates: Potential strategy for increasing iron bioavailability. Food Chem 2016; 208:97-102. [DOI: 10.1016/j.foodchem.2016.03.078] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 03/03/2016] [Accepted: 03/22/2016] [Indexed: 11/26/2022]
|
136
|
Hamdi A, Roshan TM, Kahawita TM, Mason AB, Sheftel AD, Ponka P. Erythroid cell mitochondria receive endosomal iron by a "kiss-and-run" mechanism. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2859-2867. [PMID: 27627839 DOI: 10.1016/j.bbamcr.2016.09.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 08/24/2016] [Accepted: 09/08/2016] [Indexed: 12/11/2022]
Abstract
In erythroid cells, more than 90% of transferrin-derived iron enters mitochondria where ferrochelatase inserts Fe2+ into protoporphyrin IX. However, the path of iron from endosomes to mitochondrial ferrochelatase remains elusive. The prevailing opinion is that, after its export from endosomes, the redox-active metal spreads into the cytosol and mysteriously finds its way into mitochondria through passive diffusion. In contrast, this study supports the hypothesis that the highly efficient transport of iron toward ferrochelatase in erythroid cells requires a direct interaction between transferrin-endosomes and mitochondria (the "kiss-and-run" hypothesis). Using a novel method (flow sub-cytometry), we analyze lysates of reticulocytes after labeling these organelles with different fluorophores. We have identified a double-labeled population definitively representing endosomes interacting with mitochondria, as demonstrated by confocal microscopy. Moreover, we conclude that this endosome-mitochondrion association is reversible, since a "chase" with unlabeled holotransferrin causes a time-dependent decrease in the size of the double-labeled population. Importantly, the dissociation of endosomes from mitochondria does not occur in the absence of holotransferrin. Additionally, mutated recombinant holotransferrin, that cannot release iron, significantly decreases the uptake of 59Fe by reticulocytes and diminishes 59Fe incorporation into heme. This suggests that endosomes, which are unable to provide iron to mitochondria, cause a "traffic jam" leading to decreased endocytosis of holotransferrin. Altogether, our results suggest that a molecular mechanism exists to coordinate the iron status of endosomal transferrin with its trafficking. Besides its contribution to the field of iron metabolism, this study provides evidence for a new intracellular trafficking pathway of organelles.
Collapse
Affiliation(s)
- Amel Hamdi
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada; Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Tariq M Roshan
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Tanya M Kahawita
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada; Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Anne B Mason
- Department of Biochemistry, University of Vermont, Burlington, VT, USA
| | - Alex D Sheftel
- Spartan Bioscience Inc., Ottawa, Ontario, Canada; High Impact Editing, Ottawa, Ontario, Canada
| | - Prem Ponka
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada; Department of Physiology, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
137
|
Zhang P, Yue Y, Pan D, Yang R, Xu Y, Wang L, Yan J, Li X, Yang M. Pharmacokinetics study of Zr-89-labeled melanin nanoparticle in iron-overload mice. Nucl Med Biol 2016; 43:529-533. [DOI: 10.1016/j.nucmedbio.2016.05.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 05/24/2016] [Accepted: 05/28/2016] [Indexed: 10/21/2022]
|
138
|
Oh CK, Park SH, Kim J, Moon Y. Non-mutagenic Suppression of Enterocyte Ferroportin 1 by Chemical Ribosomal Inactivation via p38 Mitogen-activated Protein Kinase (MAPK)-mediated Regulation: EVIDENCE FOR ENVIRONMENTAL HEMOCHROMATOSIS. J Biol Chem 2016; 291:19858-72. [PMID: 27445333 DOI: 10.1074/jbc.m116.722520] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Indexed: 01/09/2023] Open
Abstract
Iron transfer across the basolateral membrane of an enterocyte into the circulation is the rate-limiting step in iron absorption and is regulated by various pathophysiological factors. Ferroportin (FPN), the only known mammalian iron exporter, transports iron from the basolateral surface of enterocytes, macrophages, and hepatocytes into the blood. Patients with genetic mutations in FPN or repeated blood transfusion develop hemochromatosis. In this study, non-mutagenic ribosomal inactivation was assessed as an etiological factor of FPN-associated hemochromatosis in enterocytes. Non-mutagenic chemical ribosomal inactivation disrupted iron homeostasis by regulating expression of the iron exporter FPN-1, leading to intracellular accumulation in enterocytes. Mechanistically, a xenobiotic insult stimulated the intracellular sentinel p38 MAPK signaling pathway, which was positively involved in FPN-1 suppression by ribosomal dysfunction. Moreover, ribosomal inactivation-induced iron accumulation in Caenorhabditis elegans as a simplified in vivo model for gut nutrition uptake was dependent on SEK-1, a p38 kinase activator, leading to suppression of FPN-1.1 expression and iron accumulation. In terms of gene regulation, ribosomal stress-activated p38 signaling down-regulated NRF2 and NF-κB, both of which were positive transcriptional regulators of FPN-1 transcription. This study provides molecular evidence for the modulation of iron bioavailability by ribosomal dysfunction as a potent etiological factor of non-mutagenic environmental hemochromatosis in the gut-to-blood axis.
Collapse
Affiliation(s)
- Chang-Kyu Oh
- From the Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan 50612, South Korea and
| | - Seong-Hwan Park
- From the Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan 50612, South Korea and
| | - Juil Kim
- From the Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan 50612, South Korea and
| | - Yuseok Moon
- From the Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan 50612, South Korea and the Medical Research Institute, Pusan National University, Busan 46241, South Korea
| |
Collapse
|
139
|
Jiang Y, Yan Y, Wang X, Zhu G, Xu YJ. Hepcidin inhibition on the effect of osteogenesis in zebrafish. Biochem Biophys Res Commun 2016; 476:1-6. [PMID: 27233600 DOI: 10.1016/j.bbrc.2016.05.118] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 05/23/2016] [Indexed: 12/26/2022]
Abstract
Iron overload, as a risk factor for osteoporosis, can result in the up-regulation of Hepcidin, and Hepcidin knockout mice display defects in their bone microarchitecture. However, the molecular and genetic mechanisms underlying Hepcidin deficiency-derived bone loss remain unclear. Here, we show that hepcidin knockdown in zebrafish using morpholinos leads to iron overload. Furthermore, a mineralization delay is observed in osteoblast cells in hepcidin morphants, and these defects could be partially restored with microinjection of hepcidin mRNA. Quantitative real-time PCR analyses revealed the osteoblast-specific genes alp, runx2a, runx2b, and sp7 in morphants are down-regulated. Furthermore, we confirmed qRT-PCR results by in situ hybridization and found down-regulated genes related to osteoblast function in hepcidin morphants. Most importantly, we revealed that hepcidin was capable of removing whole-body iron which facilitated larval recovery from the reductions in bone formation and osteogenesis induced by iron overload.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou 215004, China; Department of Orthopedics, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi 214000, China; Osteoporosis Diagnosis and Treatment Technology, Institute of Soochow University, 1055 Sanxiang Road, Suzhou 215004, China
| | - Yilin Yan
- Institute of Neuroscience, University of Oregon, Eugene, OR, USA
| | - Xiao Wang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou 215004, China; Osteoporosis Diagnosis and Treatment Technology, Institute of Soochow University, 1055 Sanxiang Road, Suzhou 215004, China
| | - Guoxing Zhu
- Department of Orthopedics, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi 214000, China.
| | - You-Jia Xu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou 215004, China; Osteoporosis Diagnosis and Treatment Technology, Institute of Soochow University, 1055 Sanxiang Road, Suzhou 215004, China.
| |
Collapse
|
140
|
Cao H, Schroeder B, Chen J, Schott MB, McNiven MA. The Endocytic Fate of the Transferrin Receptor Is Regulated by c-Abl Kinase. J Biol Chem 2016; 291:16424-37. [PMID: 27226592 PMCID: PMC4974358 DOI: 10.1074/jbc.m116.724997] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Indexed: 12/19/2022] Open
Abstract
Clathrin-mediated endocytosis of transferrin (Tf) and its cognate receptor (TfR1) is a central pathway supporting the uptake of trophic iron. It has generally been assumed that this is a constitutive process. However, we have reported that the non-receptor tyrosine kinase, Src, is activated by Tf to facilitate the internalization of the Tf-TfR1 ligand-receptor complex. As an extension of these findings, we have tested whether subsequent trafficking steps might be regulated by additional kinase-dependent cascades, and we observed a significant endocytic block by inhibiting c-Abl kinase by a variety of methods. Importantly, Tf internalization was reduced significantly in all of these cell models and could be restored by re-expression of WT c-Abl. Surprisingly, this attenuated Tf-TfR1 endocytosis was due to a substantial drop in both the surface and total cellular receptor levels. Additional studies with the LDL receptor showed a similar effect. Surprisingly, immunofluorescence microscopy of imatinib-treated cells revealed a marked colocalization of internalized TfR1 with late endosomes/lysosomes, whereas attenuating the lysosome function with several inhibitors reduced this receptor loss. Importantly, inhibition of c-Abl resulted in a striking redistribution of the chaperone Hsc70 from a diffuse cytosolic localization to an association with the TfR1 at the late endosome-lysosome. Pharmacological inhibition of Hsc70 ATPase activity in cultured cells by the drug VER155008 prevents this chaperone-receptor interaction, resulting in an accumulation of the TfR1 in the early endosome. Thus, inhibition of c-Abl minimizes receptor recycling pathways and results in chaperone-dependent trafficking of the TfR1 to the lysosome for degradation. These findings implicate a novel role for c-Abl and Hsc70 as an unexpected regulator of Hsc70-mediated transport of trophic receptor cargo between the early and late endosomal compartments.
Collapse
Affiliation(s)
- Hong Cao
- From the Department of Biochemistry and Molecular Biology, Center for Basic Research in Digestive Diseases, and
| | - Barbara Schroeder
- Department of Experimental Pathology and Laboratory Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Jing Chen
- From the Department of Biochemistry and Molecular Biology, Center for Basic Research in Digestive Diseases, and
| | - Micah B Schott
- From the Department of Biochemistry and Molecular Biology, Center for Basic Research in Digestive Diseases, and
| | - Mark A McNiven
- From the Department of Biochemistry and Molecular Biology, Center for Basic Research in Digestive Diseases, and
| |
Collapse
|
141
|
Chang HC, Wu R, Shang M, Sato T, Chen C, Shapiro JS, Liu T, Thakur A, Sawicki KT, Prasad SVN, Ardehali H. Reduction in mitochondrial iron alleviates cardiac damage during injury. EMBO Mol Med 2016; 8:247-267. [PMID: 26896449 PMCID: PMC4772952 DOI: 10.15252/emmm.201505748] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 01/18/2016] [Accepted: 01/20/2016] [Indexed: 01/19/2023] Open
Abstract
Excess cellular iron increases reactive oxygen species (ROS) production and causes cellular damage. Mitochondria are the major site of iron metabolism and ROS production; however, few studies have investigated the role of mitochondrial iron in the development of cardiac disorders, such as ischemic heart disease or cardiomyopathy (CM). We observe increased mitochondrial iron in mice after ischemia/reperfusion (I/R) and in human hearts with ischemic CM, and hypothesize that decreasing mitochondrial iron protects against I/R damage and the development of CM. Reducing mitochondrial iron genetically through cardiac-specific overexpression of a mitochondrial iron export protein or pharmacologically using a mitochondria-permeable iron chelator protects mice against I/R injury. Furthermore, decreasing mitochondrial iron protects the murine hearts in a model of spontaneous CM with mitochondrial iron accumulation. Reduced mitochondrial ROS that is independent of alterations in the electron transport chain's ROS producing capacity contributes to the protective effects. Overall, our findings suggest that mitochondrial iron contributes to cardiac ischemic damage, and may be a novel therapeutic target against ischemic heart disease.
Collapse
Affiliation(s)
- Hsiang-Chun Chang
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rongxue Wu
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Meng Shang
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tatsuya Sato
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Chunlei Chen
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jason S Shapiro
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ting Liu
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Anita Thakur
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Konrad T Sawicki
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sathyamangla V N Prasad
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Hossein Ardehali
- Feinberg Cardiovascular Research Institute (FCVRI), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
142
|
Tabu K, Muramatsu N, Mangani C, Wu M, Zhang R, Kimura T, Terashima K, Bizen N, Kimura R, Wang W, Murota Y, Kokubu Y, Nobuhisa I, Kagawa T, Kitabayashi I, Bradley M, Taga T. A Synthetic Polymer Scaffold Reveals the Self-Maintenance Strategies of Rat Glioma Stem Cells by Organization of the Advantageous Niche. Stem Cells 2016; 34:1151-62. [DOI: 10.1002/stem.2299] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 11/18/2015] [Accepted: 12/02/2015] [Indexed: 01/27/2023]
Affiliation(s)
- Kouichi Tabu
- Department of Stem Cell Regulation; Medical Research Institute, Tokyo Medical and Dental University (TMDU); Tokyo Japan
| | - Nozomi Muramatsu
- Department of Stem Cell Regulation; Medical Research Institute, Tokyo Medical and Dental University (TMDU); Tokyo Japan
| | - Christian Mangani
- EaStChem, School of Chemistry; University of Edinburgh; Edinburgh London United Kingdom
| | - Mei Wu
- EaStChem, School of Chemistry; University of Edinburgh; Edinburgh London United Kingdom
| | - Rong Zhang
- EaStChem, School of Chemistry; University of Edinburgh; Edinburgh London United Kingdom
- School of Materials Science and Engineering; Changzhou University; Changzhou Jiangsu China
| | - Taichi Kimura
- Department of Pathology, Laboratory of Cancer Research; Hokkaido University Graduate School of Medicine; Sapporo Japan
| | - Kazuo Terashima
- Department of Stem Cell Regulation; Medical Research Institute, Tokyo Medical and Dental University (TMDU); Tokyo Japan
| | - Norihisa Bizen
- Department of Stem Cell Regulation; Medical Research Institute, Tokyo Medical and Dental University (TMDU); Tokyo Japan
| | - Ryosuke Kimura
- Department of Stem Cell Regulation; Medical Research Institute, Tokyo Medical and Dental University (TMDU); Tokyo Japan
| | - Wenqian Wang
- Department of Stem Cell Regulation; Medical Research Institute, Tokyo Medical and Dental University (TMDU); Tokyo Japan
| | - Yoshitaka Murota
- Department of Stem Cell Regulation; Medical Research Institute, Tokyo Medical and Dental University (TMDU); Tokyo Japan
| | - Yasuhiro Kokubu
- Department of Stem Cell Regulation; Medical Research Institute, Tokyo Medical and Dental University (TMDU); Tokyo Japan
| | - Ikuo Nobuhisa
- Department of Stem Cell Regulation; Medical Research Institute, Tokyo Medical and Dental University (TMDU); Tokyo Japan
| | - Tetsushi Kagawa
- Department of Stem Cell Regulation; Medical Research Institute, Tokyo Medical and Dental University (TMDU); Tokyo Japan
| | - Issay Kitabayashi
- Division of Hematological Malignancy; National Cancer Center Research Institute; Tokyo Japan
| | - Mark Bradley
- EaStChem, School of Chemistry; University of Edinburgh; Edinburgh London United Kingdom
| | - Tetsuya Taga
- Department of Stem Cell Regulation; Medical Research Institute, Tokyo Medical and Dental University (TMDU); Tokyo Japan
| |
Collapse
|
143
|
Liu Z, Wang Y, Purro M, Xiong MP. Oxidation-Induced Degradable Nanogels for Iron Chelation. Sci Rep 2016; 6:20923. [PMID: 26868174 PMCID: PMC4751432 DOI: 10.1038/srep20923] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 01/13/2016] [Indexed: 01/22/2023] Open
Abstract
Iron overload can increase cellular oxidative stress levels due to formation of reactive oxygen species (ROS); untreated, it can be extremely destructive to organs and fatal to patients. Since elevated oxidative stress levels are inherent to the condition in such patients, oxidation-induced degradable nanogels for iron chelation were rationally designed by simultaneously polymerizing oxidation-sensitive host-guest crosslinkers between β-cyclodextrin (β-CD) and ferrocene (Fc) and iron chelating moieties composed of deferoxamine (DFO) into the final gel scaffold in reverse emulsion reaction chambers. UV-Vis absorption and atomic absorption spectroscopy (AAS) was used to verify iron chelating capability of nanogels. These materials can degrade into smaller chelating fragments at rates proportional to the level of oxidative stress present. Conjugating DFO reduces the cytotoxicity of the chelator in the macrophage cells. Importantly, the nanogel can effectively reduce cellular ferritin expression in iron overloaded cells and regulate intracellular iron levels at the same time, which is important for maintaining a homeostatic level of this critical metal in cells.
Collapse
Affiliation(s)
- Zhi Liu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison 777 Highland Avenue, Madison, WI 53705-2222, USA
| | - Yan Wang
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison 777 Highland Avenue, Madison, WI 53705-2222, USA
| | - Max Purro
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison 777 Highland Avenue, Madison, WI 53705-2222, USA
| | - May P Xiong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison 777 Highland Avenue, Madison, WI 53705-2222, USA
| |
Collapse
|
144
|
Redox cycling metals: Pedaling their roles in metabolism and their use in the development of novel therapeutics. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:727-48. [PMID: 26844773 DOI: 10.1016/j.bbamcr.2016.01.026] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/29/2016] [Indexed: 12/12/2022]
Abstract
Essential metals, such as iron and copper, play a critical role in a plethora of cellular processes including cell growth and proliferation. However, concomitantly, excess of these metal ions in the body can have deleterious effects due to their ability to generate cytotoxic reactive oxygen species (ROS). Thus, the human body has evolved a very well-orchestrated metabolic system that keeps tight control on the levels of these metal ions. Considering their very high proliferation rate, cancer cells require a high abundance of these metals compared to their normal counterparts. Interestingly, new anti-cancer agents that take advantage of the sensitivity of cancer cells to metal sequestration and their susceptibility to ROS have been developed. These ligands can avidly bind metal ions to form redox active metal complexes, which lead to generation of cytotoxic ROS. Furthermore, these agents also act as potent metastasis suppressors due to their ability to up-regulate the metastasis suppressor gene, N-myc downstream regulated gene 1. This review discusses the importance of iron and copper in the metabolism and progression of cancer, how they can be exploited to target tumors and the clinical translation of novel anti-cancer chemotherapeutics.
Collapse
|
145
|
Wang L, Li R, Li L, Wang H, Liu J. Study on the interaction between Fe 3+and fibrinogen and its influence on the polymerization behavior of fibrin networks. RSC Adv 2016. [DOI: 10.1039/c6ra17661e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The interactions between fibrinogen molecule and Fe3+were studied and applied to explicate the polymerization behavior of fibrinogen mediated with Fe3+. Overloading Fe3+in the fibrinogen solution will accelerate the amorphous aggregation of fibrin.
Collapse
Affiliation(s)
- Lei Wang
- Department of Chemistry
- Liaocheng University
- Liaocheng 252000
- China
| | - Rui Li
- Department of Chemistry
- Liaocheng University
- Liaocheng 252000
- China
| | - Lianzhi Li
- Department of Chemistry
- Liaocheng University
- Liaocheng 252000
- China
| | - Huaisheng Wang
- Department of Chemistry
- Liaocheng University
- Liaocheng 252000
- China
| | - Jifeng Liu
- Key Laboratory of Food Nutrition and Safety
- Ministry of Education of China
- Tianjin University of Science and Technology
- Tianjin 300457
- China
| |
Collapse
|
146
|
da Silva BP, Matyelka JCDS, Moreira MEDC, Toledo RCL, Della Lucia CM, Pinheiro-Sant'Ana HM, Martino HSD. A high fat diet does not affect the iron bioavailability in Wistar rats fed with chia and increases gene expression of iron metabolism proteins. Food Funct 2016; 7:4861-4868. [DOI: 10.1039/c6fo00759g] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This study evaluated the effect of chia on the iron bioavailability and gene expression of proteins involved in iron metabolism in animals fed with a high fat diet and a standard diet.
Collapse
|
147
|
Li M, Koshi T, Emr SD. Membrane-anchored ubiquitin ligase complex is required for the turnover of lysosomal membrane proteins. J Cell Biol 2015; 211:639-52. [PMID: 26527740 PMCID: PMC4639871 DOI: 10.1083/jcb.201505062] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 09/25/2015] [Indexed: 12/02/2022] Open
Abstract
Cells must regulate the abundance and activity of numerous nutrient transporters in different organelle membranes to achieve nutrient homeostasis. As the recycling center and major storage organelle, lysosomes are essential for maintaining nutrient homeostasis. However, very little is known about mechanisms that govern the regulation of its membrane proteins. In this study, we demonstrated that changes of Zn(2+) levels trigger the downregulation of vacuolar Zn(2+) transporters. Low Zn(2+) levels cause the degradation of the influx transporter Cot1, whereas high Zn(2+) levels trigger the degradation of the efflux channel Zrt3. The degradation process depends on the vacuole membrane recycling and degradation pathway. Unexpectedly, we identified a RING domain-containing E3 ligase Tul1 and its interacting proteins in the Dsc complex that are important for the ubiquitination of Cot1 and partial ubiquitination of Zrt3. Our study demonstrated that the Dsc complex can function at the vacuole to regulate the composition and lifetime of vacuolar membrane proteins.
Collapse
Affiliation(s)
- Ming Li
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| | - Tatsuhiro Koshi
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| | - Scott D Emr
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| |
Collapse
|
148
|
Mouton M, Przybylowicz W, Mesjasz-Przybylowicz J, Postma F, Thornton M, Archer E, Botha A. Linking the occurrence of cutaneous opportunistic fungal invaders with elemental concentrations in false killer whale (Pseudorca crassidens) skin. ENVIRONMENTAL MICROBIOLOGY REPORTS 2015; 7:728-737. [PMID: 26034019 DOI: 10.1111/1758-2229.12302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 05/20/2015] [Indexed: 06/04/2023]
Abstract
Cetaceans, occupying the top levels in marine food chains, are vulnerable to elevated levels of potentially toxic trace elements, such as aluminium (Al), mercury (Hg) and nickel (Ni). Negative effects associated with these toxic metals include infection by opportunistic microbial invaders. To corroborate the link between the presence of cutaneous fungal invaders and trace element levels, skin samples from 40 stranded false killer whales (FKWs) were analysed using culture techniques and inductively coupled plasma-mass spectroscopy. Twenty-two skin samples yielded 18 clinically relevant fungal species. While evidence for bioaccumulation of Hg in the skin of the FKWs was observed, a strong link was found to exist between the occurrence of opportunistic fungal invaders and higher Al : Se and Al : Zn ratios. This study provides indications that elevated levels of some toxic metals, such as Al, contribute to immunotoxicity rendering FKWs susceptible to colonization by cutaneous opportunistic fungal invaders.
Collapse
Affiliation(s)
- Marnel Mouton
- Department of Microbiology, Stellenbosch University, Private Bag X1, Stellenbosch, 7600, South Africa
| | - Wojciech Przybylowicz
- Materials Research Department, iThemba LABS, National Research Foundation, P.O. Box 722, Somerset West, 7129, South Africa
- AGH University of Science and Technology, Faculty of Physics & Applied Computer Science, Al. A. Mickiewicza 30, Krakow, 30-059, Poland
| | - Jolanta Mesjasz-Przybylowicz
- Materials Research Department, iThemba LABS, National Research Foundation, P.O. Box 722, Somerset West, 7129, South Africa
| | - Ferdinand Postma
- Department of Microbiology, Stellenbosch University, Private Bag X1, Stellenbosch, 7600, South Africa
| | - Meredith Thornton
- Mammal Research Institute, University of Pretoria, c/o Iziko South African Museum, P.O. Box 61, Cape Town, 8000, South Africa
| | - Edward Archer
- Department of Botany & Zoology, Stellenbosch University, Stellenbosch, South Africa
| | - Alfred Botha
- Department of Microbiology, Stellenbosch University, Private Bag X1, Stellenbosch, 7600, South Africa
| |
Collapse
|
149
|
Handa P, Kowdley KV. Glyceronephosphate O-acyltransferase as a hemochromatosis modifier gene: Another iron in the fire? Hepatology 2015; 62:337-9. [PMID: 25820544 DOI: 10.1002/hep.27813] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 03/26/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Priya Handa
- Liver Care Network and Organ Care Research, Swedish Medical Center, Seattle, WA
| | - Kris V Kowdley
- Liver Care Network and Organ Care Research, Swedish Medical Center, Seattle, WA
| |
Collapse
|
150
|
Ferritin Is Required in Multiple Tissues during Drosophila melanogaster Development. PLoS One 2015; 10:e0133499. [PMID: 26192321 PMCID: PMC4508113 DOI: 10.1371/journal.pone.0133499] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/28/2015] [Indexed: 11/19/2022] Open
Abstract
In Drosophila melanogaster, iron is stored in the cellular endomembrane system inside a protein cage formed by 24 ferritin subunits of two types (Fer1HCH and Fer2LCH) in a 1:1 stoichiometry. In larvae, ferritin accumulates in the midgut, hemolymph, garland, pericardial cells and in the nervous system. Here we present analyses of embryonic phenotypes for mutations in Fer1HCH, Fer2LCH and in both genes simultaneously. Mutations in either gene or deletion of both genes results in a similar set of cuticular embryonic phenotypes, ranging from non-deposition of cuticle to defects associated with germ band retraction, dorsal closure and head involution. A fraction of ferritin mutants have embryonic nervous systems with ventral nerve cord disruptions, misguided axonal projections and brain malformations. Ferritin mutants die with ectopic apoptotic events. Furthermore, we show that ferritin maternal contribution, which varies reflecting the mother's iron stores, is used in early development. We also evaluated phenotypes arising from the blockage of COPII transport from the endoplasmic reticulum to the Golgi apparatus, feeding the secretory pathway, plus analysis of ectopically expressed and fluorescently marked Fer1HCH and Fer2LCH. Overall, our results are consistent with insect ferritin combining three functions: iron storage, intercellular iron transport, and protection from iron-induced oxidative stress. These functions are required in multiple tissues during Drosophila embryonic development.
Collapse
|