Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
1.
J Fish Dis ; 47(8): e13953, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38616496

ABSTRACT

Ferritin, transferrin, and transferrin receptors I and II play a vital role in iron metabolism, health, and indication of iron deficiency anaemia in fish. To evaluate the use of high-iron diets to prevent or reverse channel catfish (Ictalurus punctatus) anaemia of unknown causes, we investigated the expression of these iron-regulatory genes and proteins in channel catfish fed plant-based diets. Catfish fingerlings were fed five diets supplemented with 0 (basal), 125, and 250 mg/kg of either inorganic iron or organic iron for 2 weeks. Ferritin, transferrin, and transferrin receptor I and II mRNA and protein expression levels in fish tissues (liver, intestine, trunk kidney, and head kidney) and plasma were determined. Transferrin (iron transporter) and TfR (I and II) genes were generally highly expressed in fish fed the basal diet compared to those fed the iron-supplemented diets. In contrast, ferritin (iron storage) genes were more expressed in the trunk kidney of fish fed the iron-supplemented diets than in those fed the basal diet. Our results demonstrate that supplementing channel catfish plant-based diets with iron from either organic or inorganic iron sources affected the expression of the iron-regulatory genes and increased body iron status in the fish.


Subject(s)
Animal Feed , Diet , Ferritins , Ictaluridae , Iron , Receptors, Transferrin , Transferrin , Animals , Ictaluridae/genetics , Ferritins/genetics , Ferritins/metabolism , Ferritins/blood , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Transferrin/metabolism , Transferrin/genetics , Diet/veterinary , Animal Feed/analysis , Iron/metabolism , Dietary Supplements/analysis , Gene Expression Regulation/drug effects , Fish Diseases , Iron, Dietary/administration & dosage , Iron, Dietary/metabolism , Gene Expression/drug effects
2.
J Tradit Chin Med ; 44(2): 345-352, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38504540

ABSTRACT

OBJECTIVE: To explore the effect of acupuncture treatment on cerebral ischaemia-reperfusion injury (CIRI) and reveal the underlying mechanism of the effect based on nuclear receptor coactivator 4 (NCOA4) mediated ferritinophagy. METHODS: Sprague-Dawley male rats were divided into four groups: the sham group, model group, acupuncture group, and sham acupuncture group. After 2 h of middle cerebral artery occlusion (MCAO), reperfusion was performed for 24 h to induce CIRI. The rats were treated with acupuncture at the Neiguan (PC6) and Shuigou (GV26) acupoints. Their neurological function was evaluated by taking their Bederson scores at 2 h after ischaemia and 24 h after reperfusion. Triphenyltetrazolium chloride staining was applied to assess the cerebral infarct volume at 24 h after reperfusion. The malondialdehyde (MDA) and ferrous iron (Fe2+) levels were observed after 24 h of reperfusion using an assay kit. Western blotting was performed to detect the expression of NCOA4 and ferritin heavy chain 1 (FTH1) at 24 h after reperfusion. Moreover, the colocalization of ferritin with neurons, NCOA4 with microtubule-associated protein 1 light chain 3 (LC3), and NCOA4 with ferritin was visualized using immunofluorescence staining. RESULTS: Acupuncture significantly improved neurological function and decreased cerebral infarct volume in the acupuncture group. Following CIRI, the expression of NCOA4, LC3 and FTH1 was increased, which enhanced ferritinophagy and induced an inappropriate accumulation of Fe2+ and MDA in the ischaemic brain. However, acupuncture dramatically downregulated the expression of NCOA4, LC3 and FTH1, inhibited the overactivation of ferritinophagy, and decreased the levels of MDA and Fe2+. CONCLUSIONS: Acupuncture can inhibit NCOA4-mediated ferritinophagy and protect neurons against CIRI in a rat model.


Subject(s)
Acupuncture Therapy , Brain Ischemia , Reperfusion Injury , Rats , Male , Animals , Rats, Sprague-Dawley , Brain Ischemia/genetics , Brain Ischemia/therapy , Brain Ischemia/metabolism , Cerebral Infarction , Reperfusion Injury/genetics , Reperfusion Injury/therapy , Reperfusion Injury/metabolism , Ferritins/genetics , Nuclear Receptor Coactivators/metabolism
3.
BMC Biol ; 21(1): 279, 2023 12 05.
Article in English | MEDLINE | ID: mdl-38049852

ABSTRACT

BACKGROUND: The rapid reproduction of malaria parasites requires proper iron uptake. However, the process of iron absorption by parasites is rarely studied. Divalent metal transporter (DMT1) is a critical iron transporter responsible for uptaking iron. A homolog of human DMT1 exists in the malaria parasite genome, which in Plasmodium yoelii is hereafter named PyDMT1. RESULTS: PyDMT1 knockout appears to be lethal. Surprisingly, despite dwelling in an iron-rich environment, the parasite cannot afford to lose even partial expression of PyDMT1; PyDMT1 hypomorphs were associated with severe growth defects and quick loss of pathogenicity. Iron supplementation could completely suppress the defect of the PyDMT1 hypomorph during in vitro culturing. Genetic manipulation through host ferritin (Fth1) knockout to increase intracellular iron levels enforced significant growth inhibition in vivo on the normal parasites but not the mutant. In vitro culturing with isolated ferritin knockout mouse erythrocytes completely rescued PyDMT1-hypomorph parasites. CONCLUSION: A critical iron requirement of malaria parasites at the blood stage as mediated by this newly identified iron importer PyDMT1, and the iron homeostasis in malarial parasites is finely tuned. Tipping the iron balance between the parasite and host will efficiently kill the pathogenicity of the parasite. Lastly, PyDMT1 hypomorph parasites were less sensitive to the action of artemisinin.


Subject(s)
Malaria , Plasmodium yoelii , Animals , Mice , Humans , Iron/metabolism , Ferritins/genetics , Ferritins/metabolism , Biological Transport , Membrane Transport Proteins/metabolism , Erythrocytes/parasitology
4.
Stem Cell Res Ther ; 14(1): 330, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37964388

ABSTRACT

BACKGROUND: A non-invasive imaging technology that can monitor cell viability, retention, distribution, and interaction with host tissue after transplantation is needed for optimizing and translating stem cell-based therapies. Current cell imaging approaches are limited in sensitivity or specificity, or both, for in vivo cell tracking. The objective of this study was to apply a novel ferritin-based magnetic resonance imaging (MRI) platform to longitudinal tracking of human embryonic stem cells (hESCs) in vivo. METHODS: Human embryonic stem cells (hESCs) were genetically modified to stably overexpress ferritin using the CRISPR-Cas9 system. Cellular toxicity associated with ferritin overexpression and manganese (Mn) supplementation were assessed based on cell viability, proliferation, and metabolic activity. Ferritin-overexpressing hESCs were characterized based on stem cell pluripotency and cardiac-lineage differentiation capability. Cells were supplemented with Mn and imaged in vitro as cell pellets on a preclinical 3 T MR scanner. T1-weighted images and T1 relaxation times were analyzed to assess contrast. For in vivo study, three million cells were injected into the leg muscle of non-obese diabetic severe combined immunodeficiency (NOD SCID) mice. Mn was administrated subcutaneously. T1-weighted sequences and T1 mapping were used to image the animals for longitudinal in vivo cell tracking. Cell survival, proliferation, and teratoma formation were non-invasively monitored by MRI. Histological analysis was used to validate MRI results. RESULTS: Ferritin-overexpressing hESCs labeled with 0.1 mM MnCl2 provided significant T1-induced bright contrast on in vitro MRI, with no adverse effect on cell viability, proliferation, pluripotency, and differentiation into cardiomyocytes. Transplanted hESCs displayed significant bright contrast on MRI 24 h after Mn administration, with contrast persisting for 5 days. Bright contrast was recalled at 4-6 weeks with early teratoma outgrowth. CONCLUSIONS: The bright-ferritin platform provides the first demonstration of longitudinal cell tracking with signal recall, opening a window on the massive cell death that hESCs undergo in the weeks following transplantation before the surviving cell fraction proliferates to form teratomas.


Subject(s)
Human Embryonic Stem Cells , Teratoma , Mice , Animals , Humans , Human Embryonic Stem Cells/pathology , Ferritins/genetics , Mice, SCID , Magnetic Resonance Imaging/methods , Embryonic Stem Cells
5.
Front Biosci (Elite Ed) ; 14(3): 17, 2022 07 04.
Article in English | MEDLINE | ID: mdl-36137989

ABSTRACT

BACKGROUND: Eukaryotic initiation factor (eIF) 4G plays an important role in assembling the initiation complex required for ribosome binding to mRNA and promote translation. Translation of ferritin IRE mRNAs is regulated by iron through iron responsive elements (IREs) and iron regulatory protein (IRP). The noncoding IRE stem-loop (30-nt) structure control synthesis of proteins in iron trafficking, cell cycling, and nervous system function. High cellular iron concentrations promote IRE RNA binding to ribosome and initiation factors, and allow synthesis of ferritin. METHODS: In vitro translation assay was performed in depleted wheat germ lysate with supplementation of initiation factors. Fluorescence spectroscopy was used to characterize eIF4F/IRE binding. RESULTS: Eukaryotic initiation factor eIF4G increases the translation of ferritin through binding to stem loop structure of iron responsive elements mRNA in the 5'-untranslated region. Our translation experiment demonstrated that exogenous addition of eIF4G selectively enhanced the translation of ferritin IRE RNA in depleted WG lysate. However, eIF4G facilitates capped IRE RNA translation significantly higher than uncapped IRE RNA translation. Addition of iron with eIF4G to depleted WG lysate significantly enhanced translation for both IRE mRNA (capped and uncapped), confirming the contribution of eIF4G and iron as a potent enhancer of ferritin IRE mRNA translation. Fluorescence data revealed that ferritin IRE strongly interacts to eIF4G (Kd = 63 nM), but not eIF4E. Further equilibrium studies showed that iron enhanced (~4-fold) the ferritin IRE binding to eIF4G. The equilibrium binding effects of iron on ferritin IRE RNA/eIFs interaction and the temperature dependence of this reaction were measured and compared. The Kd values for the IRE binding to eIF4G ranging from 18.2 nM to 63.0 nM as temperature elevated from 5 °C to 25 °C, while the presence of iron showed much stronger affinity over the same range of temperatures. Thermodynamic parameter revealed that IRE RNA binds to eIF4G with ΔH = -42.6 ± 3.3 kJ. mole-1, ΔS = -11.5 ± 0.4 J. mole-1K-1, and ΔG = -39.2 ± 2.7 kJ. mole-1, respectively. Furthermore, addition of iron significantly changed the values of thermodynamic parameters, favoring stable complex formation, thus favoring efficient protein synthesis. This study first time demonstrate the participation of eIF4G in ferritin IRE mRNA translation. CONCLUSIONS: eIF4G specifically interacts with ferritin IRE RNA and promotes eIF4G-dependent translation.


Subject(s)
Eukaryotic Initiation Factor-4F , Eukaryotic Initiation Factor-4G , Eukaryotic Initiation Factor-4F/genetics , Eukaryotic Initiation Factor-4F/metabolism , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-4G/metabolism , Ferritins/genetics , Iron/metabolism , Iron-Regulatory Proteins/genetics , Iron-Regulatory Proteins/metabolism , RNA Caps/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Untranslated Regions
6.
Vet Parasitol ; 304: 109684, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35259632

ABSTRACT

Ferritins are iron-binding proteins that play critical functions in iron metabolism. Tick ferritins are essential in blood feeding, reproduction, iron transport, and protection of ticks from the iron-mediated oxidative stress during blood feeding and digestion. In ixodids, ferritin 2 (Fer2) is responsible for iron transport into peripheral tissues, it is critically involved in tick reproduction and has been identified as a good candidate antigen to be included in anti-tick vaccines. In argasids, information on the molecular and functional characteristics of ferritins is almost nonexistent. Given the potential of ixodid Fer2 as a vaccine target, the aim of the current study was to characterise the Fer2 orthologues in Ornithodoros erraticus (OEFer2) and O. moubata (OMFer2), including functional analyses by RNAi gene knockdown and the assessment of the protective efficacy of recombinant Fer2 protein in an animal vaccination trials. Characterisation and analysis of the OMFer2 and OEFer2 amino acid sequences showed high similarity to each other, and high similarity to the Fer2 sequences of ixodid species as well, confirming that Fer2 is highly conserved between both tick families and suggesting a similar function in the physiology of both argasid and ixodid ticks. Fer2 gene knockdown in O. moubata reduced egg hatchability rate and the subsequent number of emerging nymphs-1 up to 71%. Conversely, Fer2 gene knockdown in O. erraticus did not affect the treated ticks even though the Fer2 mRNA expression level was reduced by 90%. The recombinant form of O. moubata Fer2 (tOMFer2) was highly immunogenic and induced strong humoral responses when administered to rabbits formulated with Montanide adjuvant. The protective effect of the anti-tOMFer2 response was limited. While in O. erraticus, we did not observe any protective effect, in O. moubata it induced a significant reduction in oviposition without affecting the other parameters analysed. Accordingly, Fer2 seems to be involved in O. moubata embryogenesis. This study provides the first data on the molecular and functional characterisation of Fer2 in soft tick species and paves the way for further studies aimed at unveiling the functional aspects of Fer2 in soft ticks and confirming its potential as a vaccine candidate antigen.


Subject(s)
Ornithodoros , Vaccines , Animals , Antigens , Arthropod Proteins/metabolism , Female , Ferritins/genetics , Humans , Iron/metabolism , Mineral Oil , Rabbits , Recombinant Proteins
7.
Microb Biotechnol ; 15(5): 1525-1541, 2022 05.
Article in English | MEDLINE | ID: mdl-34644442

ABSTRACT

Ferritin proteins have an enormous capacity to store iron in cells. In search for the best conditions to accumulate and store bioavailable iron, we made use of a double mutant null for the monothiol glutaredoxins GRX3 and GRX4. The strain grx3grx4 accumulates high iron concentrations in the cytoplasm, making the metal easily available for ferritin chelation. Here, we perform a comparative study between human (L and H) and soya bean ferritins (H1 and H2) function in the eukaryotic system Saccharomyces cerevisiae. We demonstrate that the four human and soya bean ferritin chains are successfully expressed in our model system. Upon coexpression of either both human or soya bean ferritin chains, respiratory conditions along with iron supplementation led us to obtain the maximum yields of iron stored in yeast described to date. Human and soya bean ferritin chains are functional and present equivalent properties as promoters of cell survival in iron overload conditions. The best system revealed that the four human and soya bean ferritins possess a novel function as anti-ageing proteins in conditions of iron excess. In this respect, both ferritin chains with oxidoreductase capacity (human-H and soya bean-H2) bear the highest capacity to extend life suggesting the possibility of an evolutionary conservation.


Subject(s)
Fabaceae , Saccharomyces cerevisiae Proteins , Saccharomycetales , Ferritins/genetics , Ferritins/metabolism , Humans , Iron/metabolism , Oxidoreductases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
8.
Int J Biol Macromol ; 191: 152-160, 2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34547309

ABSTRACT

The favorable physicochemical properties are essential for the application of protein-based nanovehicles in the field of biomaterials. Herein, we found that the thermal stability of Marsupenaeus japonicus ferritin (MjFer) (Tm = 109.1 ± 0.4 °C) is markedly higher than human H-chain ferritin (HuHF) (Tm = 87.7 ± 0.3 °C), although they share a high structural similarity. Multiple results indicated that the promoted thermal stability of MjFer is mainly derived from the salt bridges located at the C3 interface. Consequently, MjFer exhibits strong protective effects on encapsulated curcumin upon exposure at high temperatures. In contrast, most of the curcumin loaded HuHF composites precipitated rapidly under the same conditions. These findings elucidated the molecular mechanism of the hyperthermostability of MjFer and illustrated that MjFer could act as a robust insulation nanocarrier for bioactive compounds against various thermal treatments.


Subject(s)
Dietary Supplements , Ferritins/chemistry , Nanoparticles/chemistry , Pharmaceutical Vehicles/chemistry , Animals , Curcumin/administration & dosage , Ferritins/genetics , Mutation , Penaeidae/chemistry , Protein Domains , Protein Stability
9.
Int J Mol Sci ; 22(13)2021 Jun 29.
Article in English | MEDLINE | ID: mdl-34209892

ABSTRACT

Ferritin naturally exists in most organisms and can specifically recognize the transferrin 1 receptor (TfR1), which is generally highly expressed on various types of tumor cells. The pH dependent reversible assembling and disassembling property of ferritin renders it as a suitable candidate for encapsulating a variety of anticancer drugs and imaging probes. Ferritins external surface is chemically and genetically modifiable which can serve as attachment site for tumor specific targeting peptides or moieties. Moreover, the biological origin of these protein cages makes it a biocompatible nanocarrier that stabilizes and protects the enclosed particles from the external environment without provoking any toxic or immunogenic responses. Recent studies, further establish ferritin as a multifunctional nanocarrier for targeted cancer chemotherapy and phototherapy. In this review, we introduce the favorable characteristics of ferritin drug carriers, the specific targeted surface modification and a multifunctional nanocarriers combined chemotherapy with phototherapy for tumor treatment. Taken together, ferritin is a potential ideal base of engineered nanoparticles for tumor therapy and still needs to explore more on its way.


Subject(s)
Antigens, CD/metabolism , Bioengineering/methods , Ferritins/metabolism , Neoplasms/metabolism , Receptors, Transferrin/metabolism , Animals , Drug Carriers , Drug Compounding , Drug Delivery Systems , Ferritins/genetics , Humans , Hydrogen-Ion Concentration , Nanoparticles , Neoplasms/drug therapy
10.
Fish Shellfish Immunol ; 115: 1-6, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34051346

ABSTRACT

Iron sequestration through ferritin forms a major part of innate immune response in molluscs and detailed understanding of ferritin gene and its functions can be directly applied in infection and disease management studies. Accordingly, identification and detailed molecular characterization of a ferritin subunit gene from a commercially significant marine mussel Perna viridis was targeted. Molecular screening using degenerate primers in total mantle RNA resulted in the amplification of a novel ferritin gene fragment having <87% identity to the reported ferritin gene sequences. Rapid amplification of cDNA ends-PCR was followed to generate complete cDNA sequence of P.viridis ferritin (PvFer). The complete cDNA was found to be 798 bp, containing an open reading frame of 522 bp, 5' untranslated region (UTR) of 112 bp and 3' UTR of 165 bp. The 5' UTR and 3' UTR were shown to contain an iron response element (IRE) and a polyadenylation signal (767AATAAA772) with poly (A) tail, respectively. Prediction of stem loop structure revealed that, PvFer-IRE can be folded into a typical secondary stem loop structure, having 5-CAGUGA-3' loop, proximal stem of five paired bases followed by a bulged cysteine, and six nucleotide bottom stem, indicating that expression of PvFer is regulated by iron at the translational level. ORF was found to encode 175 amino acid protein with calculated molecular mass of 19.97 kDa and isoelectric point of 4.97. Examination for signal peptide and phylogenetic analysis confirmed that PvFer belonged to cytosolic ferritins of molluscs. Conserved domain analysis showed that PvFer contained both ferroxidase diiron center and ferrihydrite nucleation center, analogous to ferritin M subunit of bony fishes and amphibians. However, amino acid sequence and glycosylation site showed more homology to vertebrate ferritin H subunits. Predicted 3D models of PvFer resembled the typical spatial features of ferritin proteins. The study forms the first comprehensive identification of a ferritin subunit gene in a true/common mussel (Order: Mytilida). Further, the detailed molecular phylogeny conducted through the present study revealed certain thought provoking insights on ferritin genes of the phylum Mollusca.


Subject(s)
Ferritins/genetics , Ferritins/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Perna/genetics , Perna/immunology , Animals , Base Sequence , DNA, Complementary/analysis , Ferritins/chemistry , Phylogeny , Sequence Alignment , Sequence Analysis, DNA
11.
Int J Mol Sci ; 21(18)2020 Sep 05.
Article in English | MEDLINE | ID: mdl-32899447

ABSTRACT

Physical training and antioxidant supplementation may influence iron metabolism through reduced oxidative stress and subsequent lowering of mRNA levels of genes that are easily induced by this stress, including those responsible for iron homeostasis. Fifteen elderly women participated in our 12-week experiment, involving six weeks of training without supplementation and six weeks of training supported by oral supplementation of 1000 mg of vitamin C daily. The participants were divided into two groups (n = 7 in group 1 and n = 8 in group 2). In group 1, we applied vitamin C supplementation in the first six weeks of training, while in group 2 during the remaining six weeks of training. In both phases, the health-related training occurred three times per week. Training accompanied by vitamin C supplementation did not affect prooxidative/antioxidative balance but significantly decreased ferritin heavy chain (FTH) and ferritin light chain (FTL) mRNA in leukocytes (for FTH mRNA from 2^64.24 to 2^11.06, p = 0.03 in group 1 and from 2^60.54 to 2^16.03, p = 0.01 in group 2, for FTL mRNA from 2^20.22 to 2^4.53, p = 0.01 in group 2). We concluded that vitamin C supplementation might have caused a decrease in gene expression of two important antioxidative genes (FTH, FTL) and had no effect on plasma prooxidative/antioxidative balance.


Subject(s)
Ascorbic Acid/pharmacology , Exercise/physiology , Ferritins/metabolism , Aged , Antioxidants/pharmacology , Apoferritins/genetics , Ascorbic Acid/metabolism , Dietary Supplements , Female , Ferritins/drug effects , Ferritins/genetics , Humans , Iron/metabolism , Leukocytes/drug effects , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , RNA, Messenger/metabolism
12.
Biochemistry ; 57(38): 5533-5543, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30183257

ABSTRACT

Mobilization of iron from bacterioferritin (BfrB) requires specific interactions with a [2Fe-2S] ferredoxin (Bfd). Blocking the BfrB:Bfd interaction results in irreversible iron accumulation in BfrB and iron deficiency in the cytosol [Eshelman, K., et al. (2017) Metallomics 9, 646-659]. The only known Bfd structure, which was obtained in complex with BfrB (Protein Data Bank entry 4E6K ), indicated a new fold and suggested that the stability of Bfd is aided by an anion binding site consisting of R26, R29, and K46. We investigated the Bfd fold using site-directed mutagenesis, X-ray crystallography, and biochemistry in solution. The X-ray structure, which is nearly identical to that of Bfd in the BfrB:Bfd complex, shows that the [2Fe-2S] cluster preorganizes residues at the BfrB:Bfd interface into a structure complementary to the Bfd binding site on BfrB. Studies in solution showed rapid loss of the [2Fe-2S] cluster at a low ionic strength but higher stability with an increasing ionic strength, thus supporting a structural anion binding site. Structures of the R26E and R26E/K46Y mutants are nearly identical to that of Bfd, except for a new network of hydrogen bonds stabilizing the region encompassing the former anion binding site. The stability of the R26E and R26E/K46Y mutants, which is weakly and completely independent of solution ionic strength, respectively, corroborates that Bfd requires an anion binding site. The mutations, which caused only small changes to the strength of the BfrB:Bfd interaction and mobilization of iron from BfrB, indicate that the anion binding site in Bfd serves primarily a structural role.


Subject(s)
Anions/metabolism , Bacterial Proteins/metabolism , Cytochrome b Group/metabolism , Ferritins/metabolism , Homeostasis , Iron-Sulfur Proteins/metabolism , Iron/metabolism , Pseudomonas aeruginosa/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Catalysis , Crystallography, X-Ray , Cytochrome b Group/chemistry , Cytochrome b Group/genetics , Ferredoxins/metabolism , Ferritins/chemistry , Ferritins/genetics , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/genetics , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Protein Conformation , Protein Domains
13.
Dev Comp Immunol ; 82: 104-112, 2018 05.
Article in English | MEDLINE | ID: mdl-29341872

ABSTRACT

It is well known that iron is an essential element for all living organism. The intracellular iron availability is also important for the host's innate immune response to various pathogens, in which the iron homeostasis can be regulated by ferritin due to its iron storage property. In this study, a full-length cDNA sequence of ferritin (named as CqFerritin) was identified with 1410 bp from red claw crayfish Cherax quadricarinatus, which contained an open reading frame of 513 bp, encoding 170 amino acids with a conserved ferritin domain. Tissue distribution analysis demonstrated that CqFerritin was widely expressed in various tissues with high presence in haemocyte, haematopoietic tissue (Hpt) and heart, while lowest expression in hepatopancreas. In addition, loss-of-function of CqFerritin by gene silencing resulted in significantly higher expression of an envelope protein VP28 of white spot syndrome virus (WSSV) in red claw crayfish Hpt cell cultures, indicating the potential antiviral response of CqFerritin. To further explore the effect on WSSV replication by CqFerritin, recombinant CqFerritin protein (rCqFerritin) was transfected into Hpt cells followed by WSSV infection. Importantly, the replication of WSSV was obviously decreased in Hpt cells if transfected with rCqFerritin protein, suggesting that CqFerritin had clearly negative effect on WSSV infection. Furthermore, intracellular accumulation of iron ions was found to promote the WSSV replication in a dose-dependent manner, illustrating that the iron level regulated by CqFerritin was likely to be vital for WSSV infection in red claw crayfish. Taken together, these data suggest that CqFerritin plays an important role in immune defense against WSSV infection in a crustacean C. quadricarinatus.


Subject(s)
Arthropod Proteins/metabolism , Astacoidea/immunology , DNA Virus Infections/immunology , Ferritins/metabolism , Hematopoietic System/metabolism , Iron/metabolism , White spot syndrome virus 1/physiology , Animals , Arthropod Proteins/genetics , Astacoidea/virology , Cells, Cultured , Cloning, Molecular , DNA, Complementary/genetics , Ferritins/genetics , Immunity, Innate , Ion Transport , Myocardium/metabolism , Virus Replication
14.
PLoS One ; 12(11): e0188933, 2017.
Article in English | MEDLINE | ID: mdl-29190821

ABSTRACT

Iron is an indispensable element for plant growth and defense and hence it is essential to improve the plant's ability to accumulate iron. Besides, it is also an important aspect for human health. In view of this, we attempted to increase the iron content in banana cultivar Rasthali using MusaFer1 as a candidate gene. Initially, the expression of all five genes of the MusaFer family (MusaFer1-5) was quantified under iron-excess and -deficient conditions. The supplementation of 250 and 350 µM iron enhanced expression of all MusaFer genes; however, MusaFer1 was increased maximally by 2- and 4- fold in leaves and roots respectively. Under iron deficient condition, all five MusaFer genes were downregulated, indicating their iron dependent regulation. In MusaFer1 overexpressing lines, iron content was increased by 2- and 3-fold in leaves and roots respectively, as compared with that of untransformed lines. The increased iron was mainly localized in the epidermal regions of petiole. The analysis of MusaFer1 promoter indicated that it might control the expression of iron metabolism related genes and also other genes of MusaFer family. MusaFer1 overexpression led to downregulated expression of MusaFer3, MusaFer4 and MusaFer5 in transgenic leaves which might be associated with the plant's compensatory mechanism in response to iron flux. Other iron metabolism genes like Ferric reductase (FRO), transporters (IRT, VIT and YSL) and chelators (NAS, DMAS and NAAT) were also differentially expressed in transgenic leaf and root, suggesting the multifaceted impact of MusaFer1 towards iron uptake and organ distribution. Additionally, MusaFer1 overexpression increased plant tolerance against methyl viologen and excess iron which was quantified in terms of photosynthetic efficiency and malondialdehyde content. Thus, the study not only broadens our understanding about iron metabolism but also highlights MusaFer1 as a suitable candidate gene for iron fortification in banana.


Subject(s)
Ferritins/genetics , Iron/metabolism , Musa/genetics , Oxidative Stress , Plants, Genetically Modified/genetics , Reverse Transcriptase Polymerase Chain Reaction
15.
Clin Biochem ; 50(18): 1087-1092, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28941592

ABSTRACT

OBJECTIVE: Parkinson's disease (PD) is a common neurodegenerative disease. Oxidative stress is considered as a key modulator in the development of PD. This study aimed to investigate associations between serum NOX1 (NADPH oxidase1), ferritin, selenium (Se), and uric acid (UA) levels and clinical parameters in patients with PD. DESIGN AND METHODS: Serum levels of NOX1, ferritin, Se, and UA were measured in 40 PD patients and 40 healthy individuals. Receiver operating characteristic (ROC) analysis was performed to investigate incremental diagnostic value of each factor in the study groups. RESULTS: Mean serum NOX1 levels were markedly higher in patient group (22.36±5.80ng/mL) versus healthy individuals (8.89±2.37ng/mL) (p<0.001). Significant differences were also observed in the serum concentrations of ferritin (p=0.005) and Se (p=0.001) between patients with PD and healthy individuals. However, the serum concentrations of UA were not statistically significant between the study groups (p=0.560). ROC analysis revealed a diagnostic ability of serum NOX1 and ferritin levels for PD with an area under ROC curve of ≥0.7 (p<0.05) and relatively high sensitivity and specificity. Combination of serum NOX1 and Se along with ferritin and UA levels increased the sensitivity up to 85%, specificity up to 97% and area under the ROC curve up to 0.94 (95% confidence interval (95% CI): 0.89 to 0.99, p<0.001). CONCLUSION: Our findings indicated that serum concentrations of NOX1, ferritin, and Se are significantly higher in the patients with PD. Therefore, these factors can be considered as potential diagnostic biomarkers for diagnosis and monitoring of PD patients. Further studies are required with larger sample size to provide more detailed information about the cognitive profile of participants and the outcome measures.


Subject(s)
Ferritins/analysis , NADPH Oxidases/analysis , Parkinson Disease/metabolism , Aged , Case-Control Studies , Female , Ferritins/blood , Ferritins/genetics , Humans , Iran , Male , Middle Aged , NADP/metabolism , NADPH Oxidases/blood , NADPH Oxidases/genetics , Oxidative Stress/genetics , Oxidative Stress/physiology , ROC Curve , Selenium/analysis , Selenium/blood , Selenium/metabolism , Sensitivity and Specificity , Uric Acid/analysis , Uric Acid/blood
16.
Cell Rep ; 20(10): 2341-2356, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28877469

ABSTRACT

Iron is vital for many homeostatic processes, and its liberation from ferritin nanocages occurs in the lysosome. Studies indicate that ferritin and its binding partner nuclear receptor coactivator-4 (NCOA4) are targeted to lysosomes by a form of selective autophagy. By using genome-scale functional screening, we identify an alternative lysosomal transport pathway for ferritin that requires FIP200, ATG9A, VPS34, and TAX1BP1 but lacks involvement of the ATG8 lipidation machinery that constitutes classical macroautophagy. TAX1BP1 binds directly to NCOA4 and is required for lysosomal trafficking of ferritin under basal and iron-depleted conditions. Under basal conditions ULK1/2-FIP200 controls ferritin turnover, but its deletion leads to TAX1BP1-dependent activation of TBK1 that regulates redistribution of ATG9A to the Golgi enabling continued trafficking of ferritin. Cells expressing an amyotrophic lateral sclerosis (ALS)-associated TBK1 allele are incapable of degrading ferritin suggesting a molecular mechanism that explains the presence of iron deposits in patient brain biopsies.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Proteins/metabolism , Autophagy/physiology , DNA, Complementary/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomes/metabolism , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Vesicular Transport Proteins/metabolism , Autophagy/genetics , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Proteins/genetics , Cell Line , Cell Line, Tumor , Ferritins/genetics , Ferritins/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Vesicular Transport Proteins/genetics
17.
Fish Shellfish Immunol ; 66: 411-422, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28535971

ABSTRACT

Iron is an essential microelement for almost all living organisms, while an excess of iron is toxic, thus maintenance of iron homeostasis is vital. As iron storage protein, ferritin plays an important role in iron metabolism. In the present study, we cloned and characterized the ferritin H subunit from Megalobrama amblycephala, termed as MamFerH. An iron-responsive element (IRE) was predicted in the 5' untranslated region (UTR) of MamFerH, while its bulge structural was different from that of the reported ferritin M subunit (MamFerM). The MamFerH and MamFerM genes exhibited similar expression patterns during early development with specifically high expression post hatching, whereas their tissue expression patterns were different. Specifically, MamFerM was highly expressed in the spleen, liver and kidney, while MamFerH was predominantly expressed in the blood and brain, indicating their different functions. In addition, the expression of the two genes was induced upon Aeromonas hydrophila infection at both transcriptional and translational levels, and MamFerH was more efficient. Immunohistochemistry and immunofluorescence analysis confirmed their significant changes at protein level and distribution in the liver post infection, indicating their participation in host immune response. Furthermore, bacteriostatic experiment revealed that recombinant MamFerH displayed more significant inhibitory effect on the growth of A. hydrophila.


Subject(s)
Cyprinidae , Ferritins/genetics , Fish Proteins/genetics , Fish Proteins/metabolism , Gene Expression Regulation , Aeromonas hydrophila/drug effects , Aeromonas hydrophila/physiology , Animals , Apoferritins/chemistry , Apoferritins/genetics , Apoferritins/metabolism , Apoferritins/pharmacology , Base Sequence , Cloning, Molecular , Cyprinidae/embryology , DNA, Complementary/genetics , DNA, Complementary/metabolism , Ferritins/chemistry , Ferritins/metabolism , Ferritins/pharmacology , Fish Diseases/genetics , Fish Diseases/metabolism , Fish Diseases/microbiology , Fish Proteins/chemistry , Fish Proteins/pharmacology , Gram-Negative Bacterial Infections/genetics , Gram-Negative Bacterial Infections/metabolism , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/veterinary , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Sequence Alignment/veterinary
18.
Fish Shellfish Immunol ; 63: 297-303, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28232280

ABSTRACT

Ferritin plays important roles in iron storage, detoxification, and immune response. Here, a ferritin gene (PcFer) was identified in Procambarus clarkii, an economically important freshwater crayfish. Full-length PcFer cDNA was 1022-bp, including a 135-bp 5'-untranslated region (UTR) with a typical iron responsive element, a 374-bp 3'-UTR, and a 513-bp open reading frame encoding a polypeptide of 170 amino acids which contained the Ferritin domain. PcFer has ion binding sites, a ferrihydrite nucleation center, and an iron ion channel. PcFer is phylogenetically closely-related to Pacifastacus leniusculus and Eriocheir sinensis ferritins. Real-time quantitative reverse-transcription PCR analysis showed that PcFer was expressed in all tested P. clarkii tissues, and expressed most in hepatopancreas. After challenge with various heavy metals and lipopolysaccharide, respectively, the hepatopancreatic expression levels of PcFer were markedly upregulated. These results suggest that expression of PcFer might be involved in immune defense and protection of P. clarkii against heavy metal stress.


Subject(s)
Arthropod Proteins/genetics , Astacoidea/genetics , Ferritins/genetics , Lipopolysaccharides/pharmacology , Metals, Heavy/toxicity , Water Pollutants, Chemical/toxicity , Amino Acid Sequence , Animals , Arthropod Proteins/chemistry , Arthropod Proteins/metabolism , Astacoidea/immunology , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/metabolism , Ferritins/chemistry , Ferritins/metabolism , Immunity, Innate , Phylogeny , Polymerase Chain Reaction/veterinary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment , Tissue Distribution
19.
Kardiol Pol ; 75(3): 267-275, 2017.
Article in English | MEDLINE | ID: mdl-27747853

ABSTRACT

BACKGROUND: Iron is presumed to play an important role in the functioning of cardiomyocytes and skeletal myocytes. There is scarcity of direct data characterising the cells functioning when exposed to iron depletion or iron overload in a cellular environment. There is some clinical evidence demonstrating that iron deficiency has serious negative prognostic consequences in heart failure (HF) patients and its correction brought clinical benefit. AIM: The viability of the cells upon unfavourable iron concentration in the cell culture medium and the presence of the molecular system of proteins involved in intracellular iron metabolism in these cells have been studied. METHODS: H9C2 rat adult cardiomyocytes and L6G8C5 rat adult skeletal myocytes were cultured for 24 h in optimal vs. reduced vs. increased iron concentrations. Intracellular iron content was measured by flame atomic absorption spectroscopy (FAAS). We analysed the mRNA expression of: ferritin heavy and light chains (FTH and FTL; iron storage proteins), myoglobin (MB, oxygen storage protein) ferroportin type 1 (FPN1; iron exporter), transferrin receptor type 1 (TfR1; iron importer), hepcidin (HAMP; iron metabolism regulator) using qPCR, the level of respective proteins using Western Blot (WB), and the viability of the cells using flow cytometry and cell viability tetrazolium reduction assay (MTS). RESULTS: Cardiomyocytes exposed to gradually reduced iron concentrations in the medium demonstrated a decrease in the mRNA expression of FTH, FTL, FPN1, MB, and HAMP (all R = -0.75, p < 0.05), indicating depleted iron status in the cells. As a consequence, the expression of TfR1 (R = 0.7, p < 0.05) was increased, reflecting a facilitated entrance of iron to the cells. The inverse changes occurred in H9C2 cells exposed to increased iron concentrations in the medium in comparison to control cells. The same pattern of changes in the mRNA expressions was observed in myocytes, and there was a strong correlation between analogous genes in both cell lines (all R > 0.9, p < 0.0001). WB analysis revealed the analogous pattern of changes in protein expression as an mRNA profile. Both iron depletion and iron excess impaired viability of cardiomyocytes and skeletal myocytes. CONCLUSIONS: Both rat cardiomyocytes and myocyte cells contain the set of genes involved in the intracellular iron metabolism, and both types of investigated cells respond to changing iron concentrations in the cultured environment. Both iron deficiency (ID) and iron overload is detrimental for the cells. This data may explain the beneficial effects of iron supplementation in patients with ID in HF.


Subject(s)
Iron/physiology , Muscle Cells/physiology , Nutritional Status , Anemia, Iron-Deficiency , Animals , Antigens, CD/genetics , Cation Transport Proteins/genetics , Cell Line , Cell Survival , Ferritins/genetics , Gene Expression Regulation , Hepcidins/genetics , Iron/analysis , Iron/metabolism , Iron Overload , Muscle Cells/metabolism , Rats , Receptors, Transferrin/genetics
20.
Fish Shellfish Immunol ; 57: 87-95, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27539708

ABSTRACT

Ferritins are conserved iron storage proteins that exist in most living organisms and play an essential role in iron homeostasis. In this study, we reported the identification and analysis of a ferritin middle-chain (M) subunit, MaFerM, from blunt snout bream, Megalobrama amblycephala. The full length cDNA of MaFerM contains a 5'-untranslated region (UTR) of 152 bp, an open reading frame (ORF) of 522 bp and a 3'-UTR of 270 bp. The ORF encodes a putative protein of 174 amino acids, which shares extensive sequence identities with the M ferritins of several fish species. In silico analysis identified both the ferroxidase center of mammalian heavy-chain (H) ferritins and the iron nucleation site of mammalian light-chain (L) ferritins in MaFerM. Quantitative real-time reverse transcription polymerase chain reaction analysis indicated that MaFerM expression was highest in the liver and lowest in the heart and responded positively to experimental challenges with Aeromonas hydrophila. The exposure of cultured M. amblycephala to treatment with stress inducers (iron and H2O2) significantly up-regulated the expression of MaFerM in a dose-dependent manner. Iron chelation analysis showed that recombinant MaFerM purified from Escherichia coli exhibited apparent iron binding activity. These results suggest that MaFerM is a functional M ferritin and is likely to play a role in iron sequestration and protection against oxidative stress and immune stimulus.


Subject(s)
Cyprinidae , Ferritins/genetics , Fish Diseases/genetics , Fish Proteins/genetics , Gene Expression Regulation , Gram-Negative Bacterial Infections/veterinary , Oxidative Stress , Aeromonas hydrophila/physiology , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/metabolism , Ferritins/chemistry , Ferritins/metabolism , Fish Diseases/immunology , Fish Diseases/microbiology , Fish Proteins/chemistry , Fish Proteins/metabolism , Gram-Negative Bacterial Infections/genetics , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/microbiology , Immunity, Innate , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment/veterinary , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL