Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Am J Physiol Cell Physiol ; 323(6): C1791-C1806, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36342159

ABSTRACT

Iron absorption is a complex and highly controlled process where DMT1 transports nonheme iron through the brush-border membrane of enterocytes to the cytoplasm but does not transport alkaline-earth metals such as calcium. However, it has been proposed that high concentrations of calcium in the diet could reduce iron bioavailability. In this work, we investigate the effect of intracellular and extracellular calcium on iron uptake by Caco-2 cells, as determined by calcein fluorescence quenching. We found that extracellular calcium inhibits iron uptake by Caco-2 cells in a concentration-dependent manner. Chelation of intracellular calcium with BAPTA did not affect iron uptake, which indicates that the inhibitory effect of calcium is not exerted through intracellular calcium signaling. Kinetic studies performed, provided evidence that calcium acts as a reversible noncompetitive inhibitor of the iron transport activity of DMT1. Based on these experimental results, a mathematical model was developed that considers the dynamics of noncompetitive inhibition using a four-state mechanism to describe the inhibitory effect of calcium on the DMT1 iron transport process in intestinal cells. The model accurately predicts the calcein fluorescence quenching dynamics observed experimentally after an iron challenge. Therefore, the proposed model structure is capable of representing the inhibitory effect of extracellular calcium on DMT1-mediated iron entry into the cLIP of Caco-2 cells. Considering the range of calcium concentrations that can inhibit iron uptake, the possible inhibition of dietary calcium on intestinal iron uptake is discussed.


Subject(s)
Cation Transport Proteins , Iron , Humans , Iron/metabolism , Caco-2 Cells , Calcium , Calcium, Dietary , Cation Transport Proteins/metabolism , Kinetics , Intestinal Absorption , Models, Theoretical
2.
RSC Adv ; 8(55): 31261-31266, 2018 Sep 05.
Article in English | MEDLINE | ID: mdl-35548216

ABSTRACT

A new probe (E)-7-(diethylamino)-3-(3-(thiophen-2-yl)acryloyl)-2H-chromen-2-one (ChC16) was synthesized and studied as a turn-on fluorescent probe, based on a Michael addition mechanism for sensing SO2 derivatives, which is favored in the presence of cationic micellar media such as cetylpyridinium bromide (CPB). The probe showed high selectivity and sensitivity toward bisulfite over other anions and biothiols, including cysteine (Cys), homocysteine (Hcy) and glutathione (GSH), with a detection limit of 240 nM. Moreover, the probe showed great potential for its practical application in the detection of bisulfite in real samples, such as dry white wine, and in bioimaging.

3.
PLoS One ; 12(12): e0189043, 2017.
Article in English | MEDLINE | ID: mdl-29228015

ABSTRACT

Iron accumulation, oxidative stress and calcium signaling dysregulation are common pathognomonic signs of several neurodegenerative diseases, including Parkinson´s and Alzheimer's diseases, Friedreich ataxia and Huntington's disease. Given their therapeutic potential, the identification of multifunctional compounds that suppress these damaging features is highly desirable. Here, we report the synthesis and characterization of N-(1,3-dihydroxy-2-(hydroxymethyl)propan-2-yl)-2-(7-hydroxy-2-oxo-2H-chromen-4-yl)acetamide, named CT51, which exhibited potent free radical neutralizing activity both in vitro and in cells. CT51 bound Fe2+ with high selectivity and Fe3+ with somewhat lower affinity. Cyclic voltammetric analysis revealed irreversible binding of Fe3+ to CT51, an important finding since stopping Fe2+/Fe3+ cycling in cells should prevent hydroxyl radical production resulting from the Fenton-Haber-Weiss cycle. When added to human neuroblastoma cells, CT51 freely permeated the cell membrane and distributed to both mitochondria and cytoplasm. Intracellularly, CT51 bound iron reversibly and protected against lipid peroxidation. Treatment of primary hippocampal neurons with CT51 reduced the sustained calcium release induced by an agonist of ryanodine receptor-calcium channels. These protective properties of CT51 on cellular function highlight its possible therapeutic use in diseases with significant oxidative, iron and calcium dysregulation.


Subject(s)
Antioxidants/metabolism , Iron/metabolism , Neurons/physiology , Calcium Signaling , Cell Line, Tumor , Humans , Neurons/metabolism , Oxidative Stress
4.
Biochim Biophys Acta Mol Basis Dis ; 1863(9): 2202-2209, 2017 09.
Article in English | MEDLINE | ID: mdl-28502703

ABSTRACT

Mitochondrial dysfunction and oxidative damage, often accompanied by elevated intracellular iron levels, are pathophysiological features in a number of neurodegenerative processes. The question arises as to whether iron dyshomeostasis is a consequence of mitochondrial dysfunction. Here we have evaluated the role of Iron Regulatory Protein 1 (IRP1) in the death of SH-SY5Y dopaminergic neuroblastoma cells subjected to mitochondria complex I inhibition. We found that complex I inhibition was associated with increased levels of transferrin receptor 1 (TfR1) and iron uptake transporter divalent metal transporter 1 (DMT1), and decreased levels of iron efflux transporter Ferroportin 1 (FPN1), together with increased 55Fe uptake activity and an increased cytoplasmic labile iron pool. Complex I inhibition also resulted in increased oxidative modifications and increased cysteine oxidation that were inhibited by the iron chelators desferoxamine, M30 and Q1. Silencing of IRP1 abolished the rotenone-induced increase in 55Fe uptake activity and it protected cells from death induced by complex I inhibition. IRP1 knockdown cells presented higher ferritin levels, a lower iron labile pool, increased resistance to cysteine oxidation and decreased oxidative modifications. These results support the concept that IRP1 is an oxidative stress biosensor that mediates iron accumulation and cell death when deregulated by mitochondrial dysfunction. IRP1 activation, secondary to mitochondrial dysfunction, may underlie the events leading to iron dyshomeostasis and neuronal death observed in neurodegenerative disorders with an iron accumulation component.


Subject(s)
Electron Transport Complex I/antagonists & inhibitors , Iron Regulatory Protein 1/metabolism , Mitochondria/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Death , Cell Line, Tumor , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Humans , Iron Regulatory Protein 1/genetics , Mitochondria/genetics , Mitochondria/pathology , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism
5.
ACS Chem Neurosci ; 8(1): 178-185, 2017 01 18.
Article in English | MEDLINE | ID: mdl-27806193

ABSTRACT

Disturbed iron homeostasis, often coupled to mitochondrial dysfunction, plays an important role in the progression of common neurodegenerative diseases such as Parkinson's disease (PD). Recent studies have underlined the relevance of iron chelation therapy for the treatment of these diseases. Here we describe the synthesis, chemical, and biological characterization of the multifunctional chelator 7,8-dihydroxy-4-((methylamino)methyl)-2H-chromen-2-one (DHC12). Metal selectivity of DHC12 was Cu2+ ∼ Fe2+ > Zn2+ > Fe3+. No binding capacity was detected for Hg2+, Co2+, Ca2+, Mn2+, Mg2+, Ni2+, Pb2+, or Cd2+. DHC12 accessed cells colocalizing with Mitotracker Orange, an indication of mitochondrial targeting. In addition, DHC12 chelated mitochondrial and cytoplasmic labile iron. Upon mitochondrial complex I inhibition, DHC12 protected plasma membrane and mitochondria against lipid peroxidation, as detected by the reduced formation of 4-hydroxynonenal adducts and oxidation of C11-BODIPY581/591. DHC12 also blocked the decrease in mitochondrial membrane potential, detected by tetramethylrhodamine distribution. DHC12 inhibited MAO-A and MAO-B activity. Oral administration of DHC12 to mice (0.25 mg/kg body weight) protected substantia nigra pars compacta (SNpc) neurons against MPTP-induced death. Taken together, our results support the concept that DHC12 is a mitochondrial-targeted neuroprotective iron-copper chelator and MAO-B inhibitor with potent antioxidant and mitochondria protective activities. Oral administration of low doses of DHC12 is a promising therapeutic strategy for the treatment of diseases with a mitochondrial iron accumulation component, such as PD.


Subject(s)
Coumarins/chemical synthesis , Coumarins/therapeutic use , MPTP Poisoning/pathology , MPTP Poisoning/prevention & control , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/therapeutic use , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Copper/metabolism , Coumarins/chemistry , Cytosol/drug effects , Cytosol/metabolism , Disease Models, Animal , Humans , Iron/metabolism , Iron Chelating Agents/chemical synthesis , Iron Chelating Agents/chemistry , Iron Chelating Agents/therapeutic use , MPTP Poisoning/chemically induced , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Monoamine Oxidase/metabolism , Neuroblastoma/pathology , Neuroprotective Agents/chemistry , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism
6.
Parkinsons Dis ; 2016: 7049108, 2016.
Article in English | MEDLINE | ID: mdl-27293957

ABSTRACT

Mitochondrial dysfunction, iron accumulation, and oxidative damage are conditions often found in damaged brain areas of Parkinson's disease. We propose that a causal link exists between these three events. Mitochondrial dysfunction results not only in increased reactive oxygen species production but also in decreased iron-sulfur cluster synthesis and unorthodox activation of Iron Regulatory Protein 1 (IRP1), a key regulator of cell iron homeostasis. In turn, IRP1 activation results in iron accumulation and hydroxyl radical-mediated damage. These three occurrences-mitochondrial dysfunction, iron accumulation, and oxidative damage-generate a positive feedback loop of increased iron accumulation and oxidative stress. Here, we review the evidence that points to a link between mitochondrial dysfunction and iron accumulation as early events in the development of sporadic and genetic cases of Parkinson's disease. Finally, an attempt is done to contextualize the possible relationship between mitochondria dysfunction and iron dyshomeostasis. Based on published evidence, we propose that iron chelation-by decreasing iron-associated oxidative damage and by inducing cell survival and cell-rescue pathways-is a viable therapy for retarding this cycle.

7.
PLoS One ; 10(12): e0144848, 2015.
Article in English | MEDLINE | ID: mdl-26658949

ABSTRACT

Neuronal death in Parkinson's disease (PD) is often preceded by axodendritic tree retraction and loss of neuronal functionality. The presence of non-functional but live neurons opens therapeutic possibilities to recover functionality before clinical symptoms develop. Considering that iron accumulation and oxidative damage are conditions commonly found in PD, we tested the possible neuritogenic effects of iron chelators and antioxidant agents. We used three commercial chelators: DFO, deferiprone and 2.2'-dypyridyl, and three 8-hydroxyquinoline-based iron chelators: M30, 7MH and 7DH, and we evaluated their effects in vitro using a mesencephalic cell culture treated with the Parkinsonian toxin MPP+ and in vivo using the MPTP mouse model. All chelators tested promoted the emergence of new tyrosine hydroxylase (TH)-positive processes, increased axodendritic tree length and protected cells against lipoperoxidation. Chelator treatment resulted in the generation of processes containing the presynaptic marker synaptophysin. The antioxidants N-acetylcysteine and dymetylthiourea also enhanced axodendritic tree recovery in vitro, an indication that reducing oxidative tone fosters neuritogenesis in MPP+-damaged neurons. Oral administration to mice of the M30 chelator for 14 days after MPTP treatment resulted in increased TH- and GIRK2-positive nigra cells and nigrostriatal fibers. Our results support a role for oral iron chelators as good candidates for the early treatment of PD, at stages of the disease where there is axodendritic tree retraction without neuronal death.


Subject(s)
Antioxidants/pharmacology , Iron Chelating Agents/pharmacology , MPTP Poisoning/drug therapy , Nerve Fibers/drug effects , Neurites/drug effects , Neuroprotective Agents/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , 2,2'-Dipyridyl/pharmacology , Animals , Deferiprone , Deferoxamine/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/agonists , G Protein-Coupled Inwardly-Rectifying Potassium Channels/biosynthesis , Hydroxyquinolines/pharmacology , Lipid Peroxidation/drug effects , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Male , Mesencephalon/drug effects , Mesencephalon/metabolism , Mesencephalon/pathology , Mice , Mice, Inbred C57BL , Nerve Fibers/metabolism , Nerve Fibers/pathology , Neurites/metabolism , Neurites/pathology , Primary Cell Culture , Pyridones/pharmacology , Rats , Rats, Sprague-Dawley , Synaptophysin/agonists , Synaptophysin/biosynthesis , Tyrosine 3-Monooxygenase/biosynthesis
8.
J Neurochem ; 126(4): 541-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23506423

ABSTRACT

Inflammation and iron accumulation are present in a variety of neurodegenerative diseases that include Alzheimer's disease and Parkinson's disease. The study of the putative association between inflammation and iron accumulation in central nervous system cells is relevant to understand the contribution of these processes to the progression of neuronal death. In this study, we analyzed the effects of the inflammatory cytokines tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) and of lipopolysaccharide on total cell iron content and on the expression and abundance of the iron transporters divalent metal transporter 1 (DMT1) and Ferroportin 1 (FPN1) in neurons, astrocytes and microglia obtained from rat brain. Considering previous reports indicating that inflammatory stimuli induce the systemic synthesis of the master iron regulator hepcidin, we identified brain cells that produce hepcidin in response to inflammatory stimuli, as well as hepcidin-target cells. We found that inflammatory stimuli increased the expression of DMT1 in neurons, astrocytes, and microglia. Inflammatory stimuli also induced the expression of hepcidin in astrocytes and microglia, but not in neurons. Incubation with hepcidin decreased the expression of FPN1 in the three cell types. The net result of these changes was increased iron accumulation in neurons and microglia but not in astrocytes. The data presented here establish for the first time a causal association between inflammation and iron accumulation in brain cells, probably promoted by changes in DMT1 and FPN1 expression and mediated in part by hepcidin. This connection may potentially contribute to the progression of neurodegenerative diseases by enhancing iron-induced oxidative damage.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Cation Transport Proteins/genetics , Encephalitis/immunology , Encephalitis/metabolism , Iron/metabolism , Animals , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/metabolism , Astrocytes/cytology , Astrocytes/immunology , Astrocytes/metabolism , Brain/cytology , Brain/immunology , Cation Transport Proteins/immunology , Cation Transport Proteins/metabolism , Encephalitis/genetics , Female , Fetus/cytology , Hepcidins , Interleukin-6/immunology , Interleukin-6/pharmacology , Lipopolysaccharides/pharmacology , Male , Microglia/cytology , Microglia/immunology , Microglia/metabolism , Nerve Degeneration/genetics , Nerve Degeneration/immunology , Nerve Degeneration/metabolism , Neurons/cytology , Neurons/immunology , Neurons/metabolism , Primary Cell Culture , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology
9.
Biometals ; 25(4): 795-803, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22302610

ABSTRACT

Hallmarks of idiopathic and some forms of familial Parkinson's disease are mitochondrial dysfunction, iron accumulation and oxidative stress in dopaminergic neurons of the substantia nigra. There seems to be a causal link between these three conditions, since mitochondrial dysfunction can give rise to increased electron leak and reactive oxygen species production. In turn, recent evidence indicates that diminished activity of mitochondrial complex I results in decreased Fe­S cluster synthesis and anomalous activation of Iron Regulatory Protein 1. Thus, mitochondrial dysfunction could be a founding event in the process that leads to neuronal death. Here, we present evidence showing that at low micromolar concentrations, the dopamine metabolite aminochrome inhibits complex I and ATP production in SH-SY5Y neuroblastoma cells differentiated into a dopaminergic phenotype. This effect is apparently direct, since it is replicated in isolated mitochondria. Additionally, overnight treatment with aminochrome increased the expression of the iron import transporter divalent metal transporter 1 and decreased the expression of the iron export transporter ferroportin 1. In accordance with these findings, cells treated with aminochrome presented increased iron uptake. These results suggest that aminochrome is an endogenous toxin that inhibits by oxidative modifications mitochondrial complex I and modifies the levels of iron transporters in a way that leads to iron accumulation.


Subject(s)
Cation Transport Proteins/metabolism , Dopamine/metabolism , Electron Transport Complex I/metabolism , Indolequinones/metabolism , Indolequinones/pharmacology , Mitochondria/metabolism , Cation Transport Proteins/genetics , Cell Line, Tumor , Cell Survival/drug effects , Electron Transport Complex I/drug effects , Humans , Mitochondria/drug effects , Reverse Transcriptase Polymerase Chain Reaction
10.
Biometals ; 25(4): 761-76, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22318507

ABSTRACT

Iron is an essential element for life on earth, participating in a plethora of cellular processes where one-electron transfer reactions are required. Its essentiality, coupled to its scarcity in aqueous oxidative environments, has compelled living organisms to develop mechanisms that ensure an adequate iron supply, at times with disregard to long-term deleterious effects derived from iron accumulation. However, iron is an intrinsic producer of reactive oxygen species, and increased levels of iron promote neurotoxicity because of hydroxyl radical formation, which results in glutathione consumption, protein aggregation, lipid peroxidation and nucleic acid modification. Neurons from brain areas sensitive to degeneration accumulate iron with age and thus are subjected to an ever increasing oxidative stress with the accompanying cellular damage. The ability of these neurons to survive depends on the adaptive mechanisms developed to cope with the increasing oxidative load. Here, we describe the chemical and thermodynamic peculiarities of iron chemistry in living matter, review the components of iron homeostasis in neurons and elaborate on the mechanisms by which iron homeostasis is lost in Parkinson's disease, Alzheimer's disease and other diseases in which iron accumulation has been demonstrated.


Subject(s)
Iron/metabolism , Neurodegenerative Diseases/metabolism , Animals , Glutathione/metabolism , Homeostasis , Humans , Models, Biological , Oxidative Stress/physiology , Thermodynamics
11.
J Neural Transm (Vienna) ; 118(3): 421-31, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20890618

ABSTRACT

Studies in post-mortem tissues of patients with Parkinson's disease (PD) and in mice treated with 6-hydroxydopamine have shown a decrease in the length of axon and dendrites of striatal neurons. However, the etiology of the morphological changes and their relationship to inhibition of mitochondrial complex I and the cellular levels of iron and glutathione (GSH) have not been described. In this study, we characterized the effect of MPP+, an inhibitor of mitochondria complex I, on the integrity of the neuritic tree of midbrain dopaminergic neurons, and determined the influence of iron and cellular levels of GSH on this degeneration. Sub-maximal concentrations of MPP+ induced a drastic dose-dependent reduction of neurites, without modification of the soma or apparent cell death. Concurrent treatment with MPP+ and non-toxic concentrations of iron accelerated the process of degeneration, whereas neurons grown on a medium low in iron showed enhanced resistance to MPP+ treatment. MPP+-induced neurite shortening depended on the redox state of neurons. Pre-treatment with the general antioxidant N-acetyl cysteine protected neurons from degeneration. Treatment with sub-maximal concentrations of the inhibitor of GSH synthesis buthionine sulfoximine (BSO), in conjunction with iron and MPP+, produced massive cell death, whereas treatment with BSO plus MPP+ under low iron conditions did not damage neurons. These results suggest that under conditions of inhibition of mitochondrial complex I caused by MPP+, the accumulation of iron and the concurrent decrease in GSH results in the loss of the dendritic tree prior to cell death, of dopaminergic neurons in PD.


Subject(s)
1-Methyl-4-phenylpyridinium/pharmacology , Iron/metabolism , Mesencephalon/drug effects , Neurites/drug effects , Neurons/drug effects , Analysis of Variance , Animals , Cell Death/drug effects , Cells, Cultured , Dopamine/metabolism , Immunohistochemistry , Iron/pharmacology , Mesencephalon/metabolism , Mesencephalon/pathology , Neurites/metabolism , Neurites/pathology , Neurons/metabolism , Neurons/pathology , Rats , Rats, Sprague-Dawley
12.
Am J Physiol Cell Physiol ; 298(3): C477-85, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20007457

ABSTRACT

Intestinal iron absorption comprises the coordinated activity of the influx transporter divalent metal transporter 1 (DMT1) and the efflux transporter ferroportin (FPN). In this work, we studied the movement of DMT1 and FPN between cellular compartments as a function of iron supply. In rat duodenum, iron gavage resulted in the relocation of DMT1 to basal domains and the internalization of basolateral FPN. Considerable FPN was also found in apical domains. In Caco-2 cells, the apical-to-basal movement of cyan fluorescent protein-tagged DMT1 was complete 90 min after the addition of iron. Steady-state membrane localization studies in Caco-2 cells revealed that iron status determined the apical/basolateral membrane distribution of DMT1 and FPN. In agreement with the membrane distribution of the transporters, (55)Fe flux experiments revealed inward and outward iron fluxes at both membrane domains. Antisense oligonucleotides targeted to DMT1 or FPN inhibited basolateral iron uptake and apical iron efflux, respectively, indicating the participation of DMT1 and FPN in these fluxes. The fluxes were regulated by the iron supply; increased iron reduced apical uptake and basal efflux and increased basal uptake and apical efflux. These findings suggest a novel mechanism of regulation of intestinal iron absorption based on inward and outward fluxes at both membrane domains, and repositioning of DMT1 and FPN between membrane and intracellular compartments as a function of iron supply. This mechanism should be complementary to those based in the transcriptional or translational regulation of iron transport proteins.


Subject(s)
Cation Transport Proteins/metabolism , Cell Membrane/metabolism , Duodenum/metabolism , Intestinal Absorption , Intestinal Mucosa/metabolism , Iron/metabolism , Animals , Caco-2 Cells , Cation Transport Proteins/genetics , Cell Polarity , Chlorides , Ferric Compounds/metabolism , Ferrous Compounds/administration & dosage , Ferrous Compounds/metabolism , Humans , Kinetics , Microscopy, Confocal , Microscopy, Fluorescence , Protein Transport , RNA Interference , Rats , Rats, Sprague-Dawley
13.
Am J Physiol Cell Physiol ; 292(6): C2197-203, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17344309

ABSTRACT

Reactive iron is an important prooxidant factor, whereas GSH is a crucial component of a long-term adaptive system that allows cells to function during extended periods of high oxidative stress. In this work, the adaptive response of the GSH system to prolonged iron loads was characterized in human dopaminergic SH-SY5Y neuroblastoma cells. After the initial death of a substantial portion of the cell population, the surviving cells increased their GSH content by up to fivefold. This increase was traced to increased expression of the catalytic and modulatory subunits of gamma-glutamate-cysteine ligase. Under conditions of high iron load, cells maintained a low GSSG content through two mechanisms: 1) GSSG reductase-mediated recycling of GSSG to GSH and 2) multidrug resistant protein 1-mediated extrusion of GSSG. Increased GSH synthesis and low GSSG levels contributed to recover the cell reduction potential from -290 mV at the time of cell death to about -320 mV. These results highlight the fundamental role of GSH homeostasis in the antioxidant response to cellular iron accumulation and provide novel insights into the adaptive mechanisms of neurons subjected to increased iron loads, such as those observed in Parkinson's disease.


Subject(s)
Adaptation, Physiological , Gene Expression Regulation, Enzymologic , Glutamate-Cysteine Ligase/metabolism , Iron/metabolism , Neurons/metabolism , Up-Regulation , Cell Line, Tumor , Glutamate-Cysteine Ligase/genetics , Glutathione/metabolism , Glutathione Disulfide/metabolism , Humans , Oxidative Stress , Protein Subunits , Time Factors
14.
Biometals ; 19(4): 379-88, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16841247

ABSTRACT

Mutations in the HFE gene result in hereditary hemochromatosis, a disorder of iron metabolism characterized by increased intestinal iron absorption. Based on the observation that ectopic expression of HFE strongly inhibits apical iron uptake (Arredondo et al., 2001, FASEB J 15, 1276-1278), a negative regulation of HFE on the apical membrane transporter DMT1 was proposed as a mechanism by which HFE regulates iron absorption. To test this hypothesis, we investigated: (i) the effect of HFE antisense oligonucleotides on apical iron uptake by polarized Caco-2 cells; (ii) the apical/basolateral membrane distribution of HFE, beta-2 microglobulin and DMT1; (iii) the putative molecular association between HFE and DMT1. We found that HFE antisense treatment reduced HFE expression and increased apical iron uptake, whereas transfection with wild-type HFE inhibited iron uptake. Thus, an inverse relationship was established between HFE levels and apical iron uptake activity. Selective apical or basolateral biotinylation indicated preferential localization of DMT1 to the apical membrane and of HFE and beta-2 microglobulin (beta2m) to the basolateral membrane. Ectopic expression of HFE resulted in increased distribution of HFE-beta2m to the apical membrane. The amount of HFE-beta2m in the apical membrane inversely correlated with apical iron uptake rates. Immunoprecipitations of HFE or beta2m with specific antibodies resulted in the co-precipitation of DMT1. These results sustain a model by which direct interaction between DMT1 and HFE-beta2m in the apical membrane of Caco-2 cells result in down-regulation of apical iron uptake activity.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Intestinal Mucosa/metabolism , Iron/pharmacokinetics , Membrane Proteins/metabolism , beta 2-Microglobulin/metabolism , Caco-2 Cells , Cation Transport Proteins/metabolism , DNA, Antisense/genetics , Hemochromatosis/metabolism , Hemochromatosis Protein , Histocompatibility Antigens Class I/genetics , Humans , Immunoprecipitation/methods , Intestinal Mucosa/cytology , Iron/metabolism , Iron-Binding Proteins/metabolism , Membrane Proteins/genetics , Models, Biological , Transfection/methods , beta 2-Microglobulin/genetics
15.
J Neurochem ; 98(1): 11-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16805792

ABSTRACT

The brain uses massive amounts of oxygen, generating large quantities of reactive oxygen species (ROS). Because of its lipid composition, rich in unsaturated fatty acids, the brain is especially vulnerable to ROS. Furthermore, oxidative damage in the brain is often associated with iron, which has pro-oxidative properties. Iron-mediated oxidative damage in the brain is compounded by the fact that brain iron distribution is non-uniform, being particularly high in areas sensitive to neurodegeneration. This work was aimed to further our understanding of the cellular mechanisms by which SHSY5Y neuroblastoma cells adapt to, and survive increasing iron loads. Using an iron accumulation protocol that kills about 50% of the cell population, we found by cell sorting analysis that the SHSY5Y sub-population that survived the iron loading arrested in the G(0) phase of the cell cycle. These cells expressed neuronal markers, while their electrical properties remained largely unaltered. These results suggest that upon iron challenge, neuroblastoma cells respond by entering the G(0) phase, somehow rendering them resistant to oxidative stress. A similar physiological condition might be involved in neuronal survival in tissues known to accumulate iron with age, such as the hippocampus and the substantia nigra pars compacta.


Subject(s)
Iron/pharmacology , Neuroblastoma , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Resting Phase, Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Fluorescent Antibody Technique/methods , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Microtubule-Associated Proteins/metabolism , Oxidative Stress/physiology , Patch-Clamp Techniques/methods
16.
Biol Res ; 39(1): 103-4, 2006.
Article in English | MEDLINE | ID: mdl-16629170

ABSTRACT

Brain cells have a highly active oxidative metabolism, yet they contain only low to moderate superoxide dismutase and catalase activities. Thus, their antioxidant defenses rely mainly on cellular reduced glutathione levels. In this work, in cortical neurons we characterized viability and changes in reduced and oxidized glutathione levels in response to a protocol of iron accumulation. We found that massive death occurred after 2 days in culture with 10 microM Fe. Surviving cells developed an adaptative response that included increased synthesis of GSH and the maintenance of a glutathione-based reduction potential. These results highlight the fundamental role of glutathione homeostasis in the antioxidant response and provide novel insights into the adaptative mechanisms of neurons subjected to progressive iron loads.


Subject(s)
Cerebral Cortex/cytology , Glutathione/metabolism , Iron/metabolism , Neurons/metabolism , Oxidative Stress , Animals , Cell Death/drug effects , Cerebral Cortex/metabolism , Glutathione Disulfide/metabolism , Homeostasis , Iron/pharmacology , Neurons/chemistry , Oxidation-Reduction , Rats , Time Factors
17.
Biol Res ; 39(1): 157-65, 2006.
Article in English | MEDLINE | ID: mdl-16629175

ABSTRACT

Neurons, as non-dividing cells, encounter a myriad of stressful conditions throughout their lifespan. In particular, there is increasing evidence that iron progressively accumulates in the brain with age and that iron induced oxidative stress is the cause of several forms of neurodegeneration. Here, we review recent evidence that gives support to the following notions: 1) neuronal iron accumulation leads to oxidative stress and cell death; 2) neuronal survival to iron accumulation associates with decreased expression of the iron import transporter DMT1 and increased expression of the efflux transporter IREG1; and 3) the adaptive process of neurons towards iron-induced oxidative stress includes a marked increase in both the expression of the catalytic subunit of gamma glutamate-cysteine ligase and glutathione. These findings may help to understand aging-related neurodegeneration hallmarks: oxidative damage, functional impairment and cell death.


Subject(s)
Glutathione/metabolism , Iron/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Oxidative Stress , Adult , Aged , Cation Transport Proteins/metabolism , Cell Death , Glutamate-Cysteine Ligase/metabolism , Humans , Middle Aged , Neurons/pathology , Oxidation-Reduction
18.
Biol Res ; 39(1): 189-90, 2006.
Article in English | MEDLINE | ID: mdl-16629179

ABSTRACT

Recent evidence suggests that reactive oxygen species function as second messenger molecules in normal physiological processes. For example, activation of N-Methyl-D-Aspartate receptor results in the production of ROS, which appears to be critical for synaptic plasticity, one of the cellular mechanisms that underlie learning and memory. In this work, we studied the effect of iron in the activation of MAPK/ERK pathway and on Ca2+ signaling in neuronal PC12 cells. We found that iron-dependent generation of hydroxyl radicals is likely to modulate Ca2+ signaling through RyR calcium channel activation, which, in turn, activates the MAPK/ERK pathway. These findings underline the relevance of iron in normal neuronal function.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/drug effects , Iron/pharmacology , Neuroblastoma/enzymology , Reactive Oxygen Species/metabolism , Ryanodine Receptor Calcium Release Channel/drug effects , Animals , Blotting, Western , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , PC12 Cells/metabolism , Phosphorylation/drug effects , Rats , Ryanodine Receptor Calcium Release Channel/metabolism , Signal Transduction/drug effects
19.
Biol. Res ; 39(1): 103-104, 2006. ilus
Article in English | LILACS | ID: lil-430703

ABSTRACT

Brain cells have a highly active oxidative metabolism, yet they contain only low to moderate superoxide dismutase and catalase activities. Thus, their antioxidant defenses rely mainly on cellular reduced glutathione levels. In this work, in cortical neurons we characterized viability and changes in reduced and oxidized glutathione levels in response to a protocol of iron accumulation. We found that massive death occurred after 2 days in culture with 10 mM Fe. Surviving cells developed an adaptative response that included increased synthesis of GSH and the maintenance of a glutathione-based reduction potential. These results highlight the fundamental role of glutathione homeostasis in the antioxidant response and provide novel insights into the adaptative mechanisms of neurons subjected to progressive iron loads.


Subject(s)
Animals , Rats , Cerebral Cortex/cytology , Glutathione/metabolism , Iron/metabolism , Neurons/metabolism , Oxidative Stress , Cell Death/drug effects , Cerebral Cortex/metabolism , Glutathione Disulfide/metabolism , Homeostasis , Iron/pharmacology , Neurons/chemistry , Oxidation-Reduction , Time Factors
20.
Biol. Res ; 39(1): 157-165, 2006. ilus
Article in English | LILACS | ID: lil-430708

ABSTRACT

Neurons, as non-dividing cells, encounter a myriad of stressful conditions throughout their lifespan. In particular, there is increasing evidence that iron progressively accumulates in the brain with age and that iron-induced oxidative stress is the cause of several forms of neurodegeneration. Here, we review recent evidence that gives support to the following notions: 1) neuronal iron accumulation leads to oxidative stress and cell death; 2) neuronal survival to iron accumulation associates with decreased expression of the iron import transporter DMT1 and increased expression of the efflux transporter IREG1; and 3) the adaptive process of neurons towards iron-induced oxidative stress includes a marked increase in both the expression of the catalytic subunit of gamma glutamate-cysteine ligase and glutathione. These findings may help to understand aging-related neurodegeneration hallmarks: oxidative damage, functional impairment and cell death.


Subject(s)
Adult , Aged , Humans , Middle Aged , Glutathione/metabolism , Iron/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Oxidative Stress , Cell Death , Cation Transport Proteins/metabolism , Glutamate-Cysteine Ligase/metabolism , Neurons/pathology , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL
...