Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Nutrients ; 12(5)2020 May 08.
Article in English | MEDLINE | ID: mdl-32397086

ABSTRACT

Despite the crucial role of the liver as the central regulator of iron homeostasis, no studies have directly tested the modulation of liver gene and protein expression patterns during iron deficiency instauration and recovery with fermented milks. Fermented goat milk consumption improves the key proteins of intestinal iron metabolism during iron deficiency recovery, enhancing the digestive and metabolic utilization of iron. The aim of this study was to assess the influence of fermented goat or cow milk consumption on liver iron homeostasis during iron-deficiency anemia recovery with normal or iron-overload diets. Analysis included iron status biomarkers, gene and protein expression in hepatocytes. In general, fermented goat milk consumption either with normal or high iron content up-regulated liver DMT1, FPN1 and FTL1 gene expression and DMT1 and FPN1 protein expression. However, HAMP mRNA expression was lower in all groups of animals fed fermented goat milk. Additionally, hepcidin protein expression decreased in control and anemic animals fed fermented goat milk with normal iron content. In conclusion, fermented goat milk potentiates the up-regulation of key genes coding for proteins involved in iron metabolism, such as DMT1, and FPN1, FTL1 and down-regulation of HAMP, playing a key role in enhanced iron repletion during anemia recovery, inducing a physiological adaptation of the liver key genes and proteins coordinated with the fluctuation of the cellular iron levels, favoring whole-body iron homeostasis.


Subject(s)
Anemia, Iron-Deficiency/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Eating/physiology , Fermentation , Gene Expression , Hepcidins/genetics , Hepcidins/metabolism , Homeostasis/genetics , Iron/metabolism , Liver/metabolism , Milk , Animals , Apoferritins/genetics , Apoferritins/metabolism , Apoferritins/physiology , Cation Transport Proteins/physiology , Cattle , Goats , Hepcidins/physiology , Humans , Intestinal Mucosa/metabolism , Rats, Wistar
2.
PLoS Genet ; 15(9): e1008396, 2019 09.
Article in English | MEDLINE | ID: mdl-31568497

ABSTRACT

The interplay between signalling pathways and metabolism is crucial for tissue growth. Yet, it remains poorly understood. Here, we studied the consequences of modulating iron metabolism on the growth of Drosophila imaginal discs. We find that reducing the levels of the ferritin heavy chain in the larval wing discs leads to drastic growth defects, whereas light chain depletion causes only minor defects. Mutant cell clones for the heavy chain lack the ability to compete against Minute mutant cells. Reactive oxygen species (ROS) accumulate in wing discs with reduced heavy chain levels, causing severe mitochondrial defects and ferroptosis. Preventing ROS accumulation alleviates some of the growth defects. We propose that the increased expression of ferritin in hippo mutant cells may protect against ROS accumulation.


Subject(s)
Apoferritins/metabolism , Iron/metabolism , Wings, Animal/metabolism , Animals , Apoferritins/physiology , Cell Death , Cells, Cultured , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/metabolism , Ferroptosis/physiology , Imaginal Discs/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Wings, Animal/growth & development
3.
Arterioscler Thromb Vasc Biol ; 39(3): 413-431, 2019 03.
Article in English | MEDLINE | ID: mdl-30700131

ABSTRACT

Objective- Calcific aortic valve disease is a prominent finding in elderly and in patients with chronic kidney disease. We investigated the potential role of iron metabolism in the pathogenesis of calcific aortic valve disease. Approach and Results- Cultured valvular interstitial cells of stenotic aortic valve with calcification from patients undergoing valve replacement exhibited significant susceptibility to mineralization/osteoblastic transdifferentiation in response to phosphate. This process was abrogated by iron via induction of H-ferritin as reflected by lowering ALP and osteocalcin secretion and preventing extracellular calcium deposition. Cellular phosphate uptake and accumulation of lysosomal phosphate were decreased. Accordingly, expression of phosphate transporters Pit1 and Pit2 were repressed. Translocation of ferritin into lysosomes occurred with high phosphate-binding capacity. Importantly, ferritin reduced nuclear accumulation of RUNX2 (Runt-related transcription factor 2), and as a reciprocal effect, it enhanced nuclear localization of transcription factor Sox9 (SRY [sex-determining region Y]-box 9). Pyrophosphate generation was also increased via upregulation of ENPP2 (ectonucleotide pyrophosphatase/phosphodiesterase-2). 3H-1, 2-dithiole-3-thione mimicked these beneficial effects in valvular interstitial cell via induction of H-ferritin. Ferroxidase activity of H-ferritin was essential for this function, as ceruloplasmin exhibited similar inhibitory functions. Histological analysis of stenotic aortic valve revealed high expression of H-ferritin without iron accumulation and its relative dominance over ALP in noncalcified regions. Increased expression of H-ferritin accompanied by elevation of TNF-α (tumor necrosis factor-α) and IL-1ß (interleukin-1ß) levels, inducers of H-ferritin, corroborates the essential role of ferritin/ferroxidase via attenuating inflammation in calcific aortic valve disease. Conclusions- Our results indicate that H-ferritin is a stratagem in mitigating valvular mineralization/osteoblastic differentiation. Utilization of 3H-1, 2-dithiole-3-thione to induce ferritin expression may prove a novel therapeutic potential in valvular mineralization.


Subject(s)
Aortic Valve Stenosis/metabolism , Apoferritins/physiology , Vascular Calcification/metabolism , Aortic Valve/metabolism , Aortic Valve/pathology , Aortic Valve Stenosis/pathology , Apoferritins/antagonists & inhibitors , Apoferritins/pharmacology , Biological Transport , Cell Nucleus/metabolism , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/biosynthesis , Core Binding Factor Alpha 1 Subunit/genetics , Endothelial Cells/metabolism , Gene Expression Regulation , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Ion Channels/biosynthesis , Iron/pharmacology , Lysosomes/metabolism , Phosphates/metabolism , Phosphoric Diester Hydrolases/biosynthesis , Phosphoric Diester Hydrolases/genetics , SOX9 Transcription Factor/metabolism , Thiones/pharmacology , Thiophenes/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Vascular Calcification/pathology
4.
Int J Nanomedicine ; 12: 4371-4395, 2017.
Article in English | MEDLINE | ID: mdl-28652739

ABSTRACT

The discovery of biogenic magnetic nanoparticles (BMNPs) in the human brain gives a strong impulse to study and understand their origin. Although knowledge of the subject is increasing continuously, much remains to be done for further development to help our society fight a number of pathologies related to BMNPs. This review provides an insight into the puzzle of the physiological origin of BMNPs in organisms of all three domains of life: prokaryotes, archaea, and eukaryotes, including humans. Predictions based on comparative genomic studies are presented along with experimental data obtained by physical methods. State-of-the-art understanding of the genetic control of biomineralization of BMNPs and their properties are discussed in detail. We present data on the differences in BMNP levels in health and disease (cancer, neurodegenerative disorders, and atherosclerosis), and discuss the existing hypotheses on the biological functions of BMNPs, with special attention paid to the role of the ferritin core and apoferritin.


Subject(s)
Bacteria/chemistry , Ferritins/physiology , Magnetite Nanoparticles , Apoferritins/chemistry , Apoferritins/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Ferritins/chemistry , Humans
5.
Dev Comp Immunol ; 73: 88-96, 2017 08.
Article in English | MEDLINE | ID: mdl-28336188

ABSTRACT

Ferritin is a major iron storage protein essential not only in the infectious process, but also in any circumstance generating oxidative stress. In this study, the cDNA coding sequence of ferritin-H was obtained from the sub-Antarctic Notothenioid fish Eleginops maclovinus through transcriptomic analysis of the head kidney. This sequence contained a 534 bp open reading frame that coded for a 177 amino acid protein with a molecular weight of 20,786.2 Da and a theoretical pI of 5.56. The protein displayed a region of iron putative response elements in the 5'UTR, two putative ferritin iron-binding region signatures, and seven characteristic amino acids with ferroxidase functions. Phylogenetic analysis related this sequence to ferritin-H sequences of other Antarctic Notothenioid fish, sharing 96.61% similarity. Constitutive gene expression analysis in different organs revealed increased ferritin-H gene expression in the gills, spleen, muscle, and liver. After infection with two bacterial strains of Piscirickettsia salmonis (LF-89 and Austral-005), ferritin-H was differentially expressed depending on bacterial strain and tissue. This study provides relevant information towards understanding the iron metabolism of a sub-Antarctic Notothenioid fish.


Subject(s)
Apoferritins/physiology , Fish Diseases/immunology , Fishes/physiology , Animals , Fish Diseases/metabolism , Iron/metabolism , Piscirickettsia , Piscirickettsiaceae Infections/veterinary , Transcriptome
6.
Biochim Biophys Acta ; 1850(12): 2506-17, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26423448

ABSTRACT

The phenomenon that heme oxygenase-1 (HO-1) protects cell from injury yet its enzymatic product, iron, may facilitate generation of free radical has been long puzzling. Here we establish a functional connection between ferritin heavy chain (FHC) and HO-1. In human lupus nephritis HO-1 and FHC are colocalized within the glomeruli. In rodent anti-Thy1 (thymocyte antigen 1) induced glomerulonephritis, heme oxygenase blockade lowers the expression of FHC and accelerates mesangial cell death. Stimulation of heme oxygenase in cultured rat mesangial cell enhances its resistance to hydrogen peroxide, whereas FHC knockdown by RNA interference compromises this salutary effect. RNA interference of HO-1 makes the cell more susceptible to hydrogen peroxide, which can be rescued by forced expression of wild-type FHC but not mutants that lose the capacity of iron storage and ferroxidase activity. Phosphorylation of JunD was not sustained in these cells. Microarray analysis identifies four candidate transcriptional factors that may regulate the HO-1-induced transcription of FHC. Our results support the role of FHC in neutralizing the iron toxicity as well as mediating the protective effect of HO-1 in response to oxidative stress.


Subject(s)
Apoferritins/physiology , Heme Oxygenase-1/physiology , Oxidative Stress , Animals , Glomerular Mesangium/cytology , Glomerular Mesangium/metabolism , Rats
7.
Exp Hematol ; 42(1): 59-69, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24141093

ABSTRACT

The objective was to explore how ferritin-H deletion influences (59)Fe-distribution and excretion-kinetics in mice. Kinetics of (59)Fe-release from organs, whole-body excretion, and distribution-kinetics of intravenously injected (59)Fe trace amounts were compared in iron-deficient and iron-replete mice with (Fth(Δ/Δ)) and without (Fth(lox/lox)) conditional Mx-Cre-induced ferritin-H deletion. (59)Fe was released from spleen and liver beginning on day 2 and day 5 after ferritin-H deletion, respectively, but was not excreted from the body. Plasma-(59)Fe was cleared significantly faster in iron-deficient Fth(Δ/Δ)-mice than in iron-adequate Fth(lox/lox)-controls. (59)Fe-distribution showed a transient peak (e.g., in heart, kidney, muscle) in Fth(lox/lox) control mice, but not in ferritin-H-deleted Fth(Δ/Δ) mice 24 hours after (59)Fe injection. (59)Fe uptake into the liver and spleen was significantly lower in iron-deficient Fth(Δ/Δ) than in Fth(lox/lox) mice 24 hours and 7 days after injection, respectively, and rapidly appeared in circulating erythrocytes instead. The rate of (59)Fe release after ferritin-H deletion supports earlier data on ferritin turnover in mammals; released (59)Fe is not excreted from the body. Instead, (59)Fe is channeled into erythropoiesis and circulating erythrocytes significantly more extensively and faster. Along with a lack of transient interim (59)Fe storage (e.g., in the heart and kidney), this finding is evidence for ferritin-related iron storage-capacity affecting rate and extent of iron utilization.


Subject(s)
Apoferritins/physiology , Iron Radioisotopes/pharmacokinetics , Animals , Iron/metabolism , Mice , Mice, Inbred C57BL
8.
Antioxid Redox Signal ; 20(11): 1754-69, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24124891

ABSTRACT

SIGNIFICANCE: Inflammation and immunity can be associated with varying degrees of heme release from hemoproteins, eventually leading to cellular and tissue iron (Fe) overload, oxidative stress, and tissue damage. Presumably, these deleterious effects contribute to the pathogenesis of systemic infections. RECENT ADVANCES: Heme release from hemoglobin sensitizes parenchyma cells to undergo programmed cell death in response to proinflammatory cytokines, such as tumor necrosis factor. This cytotoxic effect is driven by a mechanism involving intracellular accumulation of free radicals, which sustain the activation of the c-Jun N-terminal kinase (JNK) signaling transduction pathway. While heme catabolism by heme oxygenase-1 (HO-1) prevents programmed cell death, this cytoprotective effect requires the co-expression of ferritin H (heart/heavy) chain (FTH), which controls the pro-oxidant effect of labile Fe released from the protoporphyrin IX ring of heme. This antioxidant effect of FTH restrains JNK activation, whereas JNK activation inhibits FTH expression, a cross talk that controls metabolic adaptation to cellular Fe overload associated with systemic infections. CRITICAL ISSUES AND FUTURE DIRECTIONS: Identification and characterization of the mechanisms via which FTH provides metabolic adaptation to tissue Fe overload should provide valuable information to our current understanding of the pathogenesis of systemic infections as well as other immune-mediated inflammatory diseases.


Subject(s)
Apoferritins/physiology , Heme/metabolism , Iron/metabolism , Animals , Heme Oxygenase-1/physiology , Homeostasis , Humans , Immunity, Innate
9.
Biochem Biophys Res Commun ; 426(4): 643-8, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22975354

ABSTRACT

This study investigated the expression and functions of ferritin, which is involved in osteoblastogenesis, in the periodontal ligament (PDL). The PDL is one of the most important tissues for maintaining the homeostasis of teeth and tooth-supporting tissues. Real-time PCR analyses of the human PDL revealed abundant expression of ferritin light polypeptide (FTL) and ferritin heavy polypeptide (FTH), which encode the highly-conserved iron storage protein, ferritin. Immunohistochemical staining demonstrated predominant expression of FTL and FTH in mouse PDL tissues in vivo. In in vitro-maintained mouse PDL cells, FTL and FTH expressions were upregulated at both the mRNA and protein levels during the course of cytodifferentiation and mineralization. Interestingly, stimulation of PDL cells with exogenous apoferritin (iron-free ferritin) increased calcified nodule formation and alkaline phosphatase activity as well as the mRNA expressions of mineralization-related genes during the course of cytodifferentiation. On the other hand, RNA interference of FTH inhibited the mineralized nodule formation of PDL cells. This is the first report to demonstrate that ferritin is predominantly expressed in PDL tissues and positively regulates the cytodifferentiation and mineralization of PDL cells.


Subject(s)
Apoferritins/physiology , Calcification, Physiologic , Cell Differentiation , Periodontal Ligament/cytology , Animals , Apoferritins/biosynthesis , Apoferritins/genetics , Cell Line , Humans , Mice , Mice, Inbred C57BL , Periodontal Ligament/metabolism , RNA Interference
10.
Carcinogenesis ; 33(11): 2269-75, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22843505

ABSTRACT

H-ferritin (HF) is a core subunit of the iron storage protein ferritin and is related to the pathogenesis of malignant diseases. HF overexpression is present in human hematologic malignancies, suggesting that HF overexpression may contribute to the development of hematologic cancers. However, in vivo evidence that HF is directly linked to hematologic tumorigenesis has not yet been shown. In this study, we show that transgenic (tg) mice overexpressing the human HF gene (hHF-tg) developed aggressive radiation-induced thymic lymphoma/leukemia (TL) compared with wild-type (WT) mice, providing evidence that HF overexpression promotes leukemia/lymphomagenesis. Fractionated X-irradiation of hHF-tg mice caused a higher incidence and earlier onset of TL compared with WT mice. Immunological and pathological features of TLs were similar in both groups. However, proliferative activity of hHF-tg lymphoma cells was higher than that of WT lymphoma cells, and microarray analyses revealed that some leukemia/lymphoma-related genes were differentially expressed in hHF-tg TLs compared with WT TLs. To investigate whether cell damage induced by irradiation is related to leukemia/lymphomagenesis, we evaluated apoptotic levels in the thymus and bone marrow (BM) of hHF-tg and WT groups after fractionated X-irradiation. Apoptosis was augmented in the hHF-tg BM, but not in the thymus, compared with the WT BM, suggesting a possible linkage between increased BM apoptosis by HF overexpression and accelerated radiation-induced TL development. Our findings indicate that HF overexpression is closely related to the development of leukemia/lymphoma, which could have implications for the prevention of malignant hematologic diseases.


Subject(s)
Apoferritins/physiology , Apoptosis/radiation effects , Leukemia, Radiation-Induced/etiology , Lymphoma/etiology , X-Rays/adverse effects , 8-Hydroxy-2'-Deoxyguanosine , Animals , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Flow Cytometry , Humans , Leukemia, Radiation-Induced/mortality , Leukemia, Radiation-Induced/pathology , Lymphoma/mortality , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Survival Rate
11.
Free Radic Biol Med ; 52(9): 1692-7, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22348978

ABSTRACT

Intracellular inclusion bodies (IBs) containing ferritin and iron are hallmarks of hereditary ferritinopathy (HF). This neurodegenerative disease is caused by mutations in the coding sequence of the ferritin light chain (FTL) gene that generate FTL polypeptides with a C-terminus that is altered in amino acid sequence and length. Previous studies of ferritin formed with p.Phe167SerfsX26 mutant FTL (Mt-FTL) subunits found disordered 4-fold pores, iron mishandling, and proaggregative behavior, as well as a general increase in cellular oxidative stress when expressed in vivo. Herein, we demonstrate that Mt-FTL is also a target of iron-catalyzed oxidative damage in vitro and in vivo. Incubation of recombinant Mt-FTL ferritin with physiological concentrations of iron and ascorbate resulted in shell structural disruption and polypeptide cleavage not seen with the wild type, as well as a 2.5-fold increase in carbonyl group formation. However, Mt-FTL shell disruption and polypeptide cleavage were completely inhibited by the addition of the radical trap 5,5-dimethyl-1-pyrroline N-oxide. These results indicate an enhanced propensity of Mt-FTL toward free radical-induced oxidative damage in vitro. We also found evidence of extensive carbonylation in IBs from a patient with HF together with isolation of a C-terminal Mt-FTL fragment, which are both indicative of oxidative ferritin damage in vivo. Our data demonstrate an enhanced propensity of mutant ferritin to undergo iron-catalyzed oxidative damage and support this as a mechanism causing disruption of ferritin structure and iron mishandling that contribute to the pathology of HF.


Subject(s)
Apoferritins/physiology , Neurodegenerative Diseases/physiopathology , Oxidative Stress , Apoferritins/genetics , Blotting, Western , Brain/pathology , Electrophoresis, Polyacrylamide Gel , Humans , Neurodegenerative Diseases/pathology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
12.
Transplantation ; 93(4): 390-7, 2012 Feb 27.
Article in English | MEDLINE | ID: mdl-22222784

ABSTRACT

BACKGROUND: Recent development of immunosuppressive therapy has provided a platform for clinical human leukocyte antigen (HLA)- and ABO-incompatible kidney transplantation. However, the prognosis seems to be different between the two. Accommodation, the condition of no injury even in the presence of antidonor antibody, is one of the key factors for successful transplantation with antidonor antibody. The purpose of this study was to compare signal transduction between anti-A/B and anti-HLA antibody reaction and to elucidate the mechanisms underlying accommodation. METHODS: Blood type A- or B-transferase gene was transfected into human EA.hy926 endothelial cells. After cell sorting, A- or B-expressing cells at high levels were obtained. The effects of anti-HLA and anti-A/B antibody binding on complement-mediated cytotoxicity and signal transduction were examined. RESULTS: Preincubation with anti-HLA antibodies only at low levels (<10% of saturation level) or anti-A/B antibodies at high levels (even at near saturation levels) for 24 hr resulted in resistance to complement-mediated cytotoxicity. Anti-A/B antibody ligation inactivated ERK1/2 pathway and increased complement regulatory proteins such as CD55 and CD59, whereas anti-HLA ligation activated PI3K/AKT pathway and increased cytoprotective genes such as hemeoxygenase-1 and ferritin H. CONCLUSION: Complement inhibition by upregulation of CD55 and CD59 through ERK1/2 inactivation might play a substantial role in accommodation after ABO-incompatible transplantation, which could also explain the intriguing finding of C4d deposition in the graft without rejection.


Subject(s)
ABO Blood-Group System/immunology , Antibodies, Anti-Idiotypic/immunology , Endothelial Cells/physiology , Graft Rejection/prevention & control , HLA Antigens/immunology , Organ Transplantation/physiology , Signal Transduction/physiology , Antibodies, Anti-Idiotypic/pharmacology , Apoferritins/physiology , CD55 Antigens/physiology , CD59 Antigens/physiology , Cells, Cultured , Complement System Proteins/physiology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Graft Rejection/immunology , Heme Oxygenase-1/physiology , Humans , MAP Kinase Signaling System/physiology , Oncogene Protein v-akt/physiology , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/immunology
13.
Glia ; 59(6): 927-35, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21446040

ABSTRACT

There is a critical relationship between oligodendrocyte development, myelin production, and iron bioavailability. Iron deficiency leads to hypomyelination both in humans and animal models, and the neurological sequelae of hypomyelination are significant. Therefore, understanding molecular mechanisms of iron import into oligodendrocytes is necessary for devising effective strategies for iron supplementation. Although transferrin has been considered as an essential component of oligodendrocyte media in culture, oligodendrocytes in vivo lack transferrin receptors. We have established that receptors for H-ferritin (HF) exist on cells of oligodendroglial lineage and that uptake of extracellular HF by oligodendrocyte progenitors is via receptor mediated endocytosis. These data strongly argue that ferritin is a major source of iron for oligodendrocytes. In this study, we demonstrate that media deficient in transferrin results in loss of viability of oligodendrocyte progenitors in culture. Cell loss could be prevented by supplementing the media with HF. Moreover, the addition of extracellular HF stimulates development of oligodendrocyte progenitor cells (OPCs) by increasing expression of myelin basic protein (MBP) and olig2 proteins without increasing their proliferation. The effect of HF on the OPCs could be mimicked by addition of membrane permeable 3,5,5-trimethylhexanoyl ferrocene (TMH-Fe) as an iron source to the media, but not membrane-impermeable ferric ammonium citrate. Overall, therefore, our results demonstrate the importance of iron for OPCs viability and differentiation and identify extracellular HF as a critical source of iron for oligodendrocytes. Given that ferritin receptors, but not transferrin receptors can be demonstrated on oligodendrocytes in vivo, the delivery of iron to oligodendrocytes via ferritin may be the more biological relevant delivery system.


Subject(s)
Apoferritins/chemistry , Iron/physiology , Oligodendroglia/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Apoferritins/physiology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Culture Media/pharmacology , Iron/chemistry , Iron-Binding Proteins/drug effects , Iron-Binding Proteins/physiology , Oligodendroglia/chemistry , Oligodendroglia/cytology , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/drug effects , Receptors, Cell Surface/physiology , Stem Cells/chemistry , Stem Cells/cytology , Transferrin/deficiency , Transferrin/genetics
14.
J Neuroimmunol ; 224(1-2): 66-71, 2010 Jul 27.
Article in English | MEDLINE | ID: mdl-20627326

ABSTRACT

The chemokine CXCL12 and its receptor, CXCR4, regulate neuronal migration, differentiation, and survival. Alterations of CXCL12/CXCR4 signaling are implicated in different neuropathologies, including the neurological complications of HIV infection. Opiates are important co-factors for progression to neuroAIDS and can disrupt the CXCL12/CXCR4 axis in vitro and in vivo. This paper will review recently identified mechanisms of opiate-induced CXCR4 impairment in neurons and introduce results from pilot studies in human brain tissue, which highlight the role of the protein ferritin heavy chain in HIV neuropathology in patients with history of drug abuse.


Subject(s)
AIDS Dementia Complex/etiology , AIDS Dementia Complex/immunology , Apoferritins/adverse effects , Neurons/immunology , Neurons/pathology , Opioid-Related Disorders/immunology , Opioid-Related Disorders/metabolism , Receptors, CXCR4/metabolism , AIDS Dementia Complex/pathology , Animals , Apoferritins/physiology , Chemokine CXCL12/metabolism , Chemokine CXCL12/physiology , Comorbidity , Humans , Neurons/metabolism , Opioid-Related Disorders/pathology , Receptors, CXCR4/physiology
15.
Biochem Pharmacol ; 80(3): 316-24, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20385107

ABSTRACT

Because both iron deficiency and iron excess are deleterious to normal cell function, the intracellular level of iron must be tightly controlled. Ferritin, an iron binding protein, regulates iron balance by storing iron in a bioavailable but nontoxic form. Ferritin protein comprises two subunits: ferritin H, which contains ferroxidase activity, and ferritin L. Here we demonstrate that ferritin H mRNA and protein are induced by histone deacetylase inhibitors (HDAC inhibitors), a promising class of anti-cancer drugs, in cultured human cancer cells. Deletion analysis and EMSA assays reveal that the induction of ferritin H occurs at a transcriptional level via Sp1 and NF-Y binding sites near the transcriptional start site of the human ferritin H promoter. Classically, HDAC inhibitors modulate gene expression by increasing histone acetylation. However, ChIP assays demonstrate that HDAC inhibitors induce ferritin H transcription by increasing NF-Y binding to the ferritin H promoter without changes in histone acetylation. These results identify ferritin H as a new target of HDAC inhibitors, and recruitment of NF-Y as a novel mechanism of action of HDAC inhibitors.


Subject(s)
Apoferritins/biosynthesis , Histone Deacetylase Inhibitors/pharmacology , Apoferritins/genetics , Apoferritins/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , HeLa Cells , Humans , Protein Binding/drug effects , Protein Binding/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tumor Cells, Cultured
16.
J Neurochem ; 109(4): 1067-78, 2009 May.
Article in English | MEDLINE | ID: mdl-19519778

ABSTRACT

Insertional mutations in exon 4 of the ferritin light chain (FTL) gene are associated with hereditary ferritinopathy (HF) or neuroferritinopathy, an autosomal dominant neurodegenerative disease characterized by progressive impairment of motor and cognitive functions. To determine the pathogenic mechanisms by which mutations in FTL lead to neurodegeneration, we investigated iron metabolism and markers of oxidative stress in the brain of transgenic (Tg) mice that express the mutant human FTL498-499InsTC cDNA. Compared with wild-type mice, brain extracts from Tg (FTL-Tg) mice showed an increase in the cytoplasmic levels of both FTL and ferritin heavy chain polypeptides, a decrease in the protein and mRNA levels of transferrin receptor-1, and a significant increase in iron levels. Transgenic mice also showed the presence of markers for lipid peroxidation, protein carbonyls, and nitrone-protein adducts in the brain. However, gene expression analysis of iron management proteins in the liver of Tg mice indicates that the FTL-Tg mouse liver is iron deficient. Our data suggest that disruption of iron metabolism in the brain has a primary role in the process of neurodegeneration in HF and that the pathogenesis of HF is likely to result from a combination of reduction in iron storage function and enhanced toxicity associated with iron-induced ferritin aggregates in the brain.


Subject(s)
Apoferritins/genetics , Apoferritins/physiology , Iron Metabolism Disorders/genetics , Iron Metabolism Disorders/metabolism , Iron/metabolism , Oxidative Stress/genetics , Animals , Brain Chemistry/physiology , Electrophoretic Mobility Shift Assay , Exons/genetics , Homeostasis/genetics , Homeostasis/physiology , Immunohistochemistry , Lipid Peroxidation/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nonheme Iron Proteins/metabolism
17.
Cancer Res ; 69(13): 5340-8, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19531652

ABSTRACT

The epithelial-mesenchymal transition (EMT) plays a critical role in tumor progression. To obtain a broad view of the molecules involved in EMT, we carried out a comparative proteomic analysis of transforming growth factor-beta1 (TGF-beta1)-induced EMT in AML-12 murine hepatocytes. A total of 36 proteins with significant alterations in abundance were identified. Among these proteins, ferritin heavy chain (FHC), a cellular iron storage protein, was characterized as a novel modulator in TGF-beta1-induced EMT. In response to TGF-beta1, there was a dramatic decrease in the FHC levels, which caused iron release from FHC and, therefore, increased the intracellular labile iron pool (LIP). Abolishing the increase in LIP blocked TGF-beta1-induced EMT. In addition, increased LIP levels promoted the production of reactive oxygen species (ROS), which in turn activated p38 mitogen-activated protein kinase. The elimination of ROS inhibited EMT, whereas H2O2 treatment rescued TGF-beta1-induced EMT in cells in which the LIP increase was abrogated. Overexpression of exogenous FHC attenuated the increases in LIP and ROS production, leading to a suppression of EMT. We also showed that TGF-beta1-mediated down-regulation of FHC occurs via 3' untranslated region-dependent repression of the translation of FHC mRNA. Moreover, we found that FHC down-regulation is an event that occurs between the early and highly invasive advanced stages in esophageal adenocarcinoma and that depletion of LIP or ROS suppresses the migration of tumor cells. Our data show that cellular iron homeostasis regulated by FHC plays a critical role in TGF-beta1-induced EMT.


Subject(s)
Apoferritins/physiology , Cell Differentiation/physiology , Epithelial Cells/cytology , Hepatocytes/cytology , Iron/metabolism , Mesoderm/cytology , Reactive Oxygen Species/metabolism , Adenocarcinoma/pathology , Animals , Apoferritins/genetics , Cell Line, Tumor , Cell Movement/physiology , Epithelial Cells/physiology , Esophageal Neoplasms/pathology , Hepatocytes/drug effects , Hepatocytes/physiology , Homeostasis , Humans , Iron Deficiencies , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mesoderm/physiology , Mice , Neoplasms/pathology , Proteome , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta1/physiology
18.
Biochim Biophys Acta ; 1790(7): 606-14, 2009 Jul.
Article in English | MEDLINE | ID: mdl-18778755

ABSTRACT

BACKGROUND: Iron status is higher in the substantia nigra than in other brain regions but can fluctuate as function of diet and genetics and disease. Of particular note is the compartmentalization of the iron-enrichment in this region; the pars reticulata contains higher levels of stainable iron as compared to the pars compacta. The latter area is where the dopaminergic neurons reside. How this compartmentalization impacts the interpretation of data that iron contributes to cell death as in Parkinson's disease or iron deficiency contributes to altered dopaminergic activity is unknown. Nonetheless, that iron can influence neuronal cell death and dopamine function is clear. METHODS: The mechanisms by which iron may be managed in the substantia nigra, particularly in the neuromelanin cells where minimal levels of ferritin the iron storage protein have been detected are addressed. The current approaches to detect iron in the substantia nigra are also reviewed. In addition, the potential mechanisms by which iron enrichment may occur in the substantia nigra are explored. GENERAL SIGNIFICANCE: This review attempts to provide a critical evaluation of the many avenues of exploration into the role of iron in one of the most iron-enriched and clinically investigated areas of the brain, the substantia nigra.


Subject(s)
Iron/metabolism , Substantia Nigra/metabolism , Aging , Animals , Apoferritins/physiology , Disease Models, Animal , Ferritins/metabolism , Hemochromatosis Protein , Histocompatibility Antigens Class I/genetics , Membrane Proteins/genetics , Microglia/physiology , Parkinson Disease/physiopathology , Restless Legs Syndrome/physiopathology
19.
J Biol Chem ; 282(41): 29927-35, 2007 Oct 12.
Article in English | MEDLINE | ID: mdl-17702748

ABSTRACT

Cytoplasmic serine hydroxymethyltransferase (cSHMT) enzyme levels are elevated by the expression of the heavy chain ferritin (H ferritin) cDNA in cultured cells without corresponding changes in mRNA levels, resulting in enhanced folate-dependent de novo thymidylate biosynthesis and impaired homocysteine remethylation. In this study, the mechanism whereby H ferritin regulates cSHMT expression was determined. cSHMT translation is shown to be regulated by an H ferritin-responsive internal ribosome entry site (IRES) located within the cSHMT mRNA 5'-untranslated region (5'-UTR). The cSHMT 5'-UTR exhibited IRES activity during in vitro translation of bicistronic mRNA templates, and in MCF-7 and HeLa cells transfected with bicistronic mRNAs. IRES activity was depressed in H ferritin-deficient mouse embryonic fibroblasts and elevated in cells expressing the H ferritin cDNA. H ferritin was shown to interact with the mRNA-binding protein CUGBP1, a protein known to interact with the alpha and beta subunits of eukaryotic initiation factor eIF2. Small interference RNA-mediated depletion of CUGBP1 decreased IRES activity from bicistronic templates that included the cSHMT 3'-UTR in the bicistronic construct. The identification of this H ferritin-responsive IRES represents a mechanism that accounts for previous observations that H ferritin regulates folate metabolism.


Subject(s)
Apoferritins/physiology , Folic Acid/metabolism , Ribosomes/metabolism , 5' Untranslated Regions , Apoferritins/metabolism , CELF1 Protein , Cell Line, Tumor , DNA, Complementary/metabolism , Gene Deletion , Genes , HeLa Cells , Homocysteine/chemistry , Humans , Models, Biological , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Two-Hybrid System Techniques
20.
Carcinogenesis ; 28(9): 2047-52, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17434931

ABSTRACT

Exposure to asbestos is a known etiological factor in malignant mesothelioma (MM). However, in vitro cell culture studies have provided paradoxical evidence that asbestos exposure to mesothelial cells causes cytotoxicity or apoptosis rather than malignant transformation. Although it has been shown that the iron associated with asbestos participates in the cell toxicity and probably MM pathogenesis via generation of reactive oxygen species (ROS), the molecular mechanisms largely remain unknown. Here, we demonstrate that ferritin heavy chain (FHC), a core subunit of iron-binding protein ferritin, works as an anti-apoptotic protein against toxic asbestos and oxidative stress in human mesothelial cells and MM cells. We found that FHC was induced in asbestos-exposed MeT-5A human mesothelial cells. The mesothelial cell line stably expressing FHC generated less amount of hydrogen peroxide (H2O2), one of the main ROS, after asbestos exposure and was more resistant to apoptosis induced by H2O2 compared with the cells transfected with the empty vector. Next, we investigated biological roles of FHC in human MM cell. We found that NCI-H2052, a human MM cell line, had a higher expression of endogenous FHC than MeT-5A and used the cell to address FHC function in MM. NCI-H2052 showed reduced H2O2 production and an apoptosis-resistant phenotype compared with MeT-5A. Suppression of the over-expressed FHC by using FHC small interfering RNA rendered the MM cells sensitive to apoptosis, suggesting the contribution of FHC to apoptosis resistance of the MM cells. Our findings highlight the potential role of FHC in the pathogenesis of asbestos-induced mesothelioma.


Subject(s)
Apoferritins/physiology , Apoptosis/physiology , Asbestos/toxicity , Mesothelioma/physiopathology , Oxidative Stress/physiology , Pleura/cytology , Apoptosis/drug effects , Cell Line , Epithelium/pathology , Humans , Hydrogen Peroxide/metabolism , Mesothelioma/chemically induced , Mesothelioma/pathology , Pleura/drug effects , Pleura/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...