Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Article En | MEDLINE | ID: mdl-38658189

Iron is a fundamental element for biological life, starting from bacteria till humans. Iron is essential for cell function and survival, energy production and metabolism, whereas increased levels cause oxidative stress. It is also a constituent of haemoglobin and thus it is necessary for oxygen transportation through the body. Given these multiple functions, the regulation of iron metabolism is complex and tight coupled with oxygen homeostasis at tissue and cellular levels, thanks to the interaction with the hypoxia inducible factor (HIF) system. In patients with chronic kidney disease (CKD), iron deficiency significantly contributes to anaemia development. This frequently overlaps with chronic inflammation, causing iron- restricted erythropoiesis. To add further complexity, metabolic hyperferritinemia may, on one side, increase the risk for CKD and, on the other, overlaps with functional iron deficiency. Excessive intracellular iron in certain cell types during CKD can also mediate cellular death (called ferroptosis), and contribute to the pathogenesis of kidney damage, atherosclerosis and vascular calcifications. This review is aimed at broadening the perspective of iron metabolism in the setting of CKD not just as a contributor to anaemia in CKD patients, but also as an important player with an impact on cell metabolism, renal fibrosis, and the cardiovascular system.

2.
Mol Neurobiol ; 61(4): 2006-2020, 2024 Apr.
Article En | MEDLINE | ID: mdl-37833459

Both neuroinflammation and iron accumulation play roles in the pathogenesis of Parkinson's disease (PD). However, whether inflammation induces iron dyshomeostasis in dopaminergic neurons at an early stage of PD, at which no quantifiable dopaminergic neuron loss can be observed, is still unknown. As for the inflammation mediators, although several cytokines have been reported to increase in PD, the functions of these cytokines in the SN are double-edged and controversial. In this study, whether inflammation could induce iron dyshomeostasis in dopaminergic neurons through high mobility group protein B1 (HMGB1) in the early stage of PD is explored. Lipopolysaccharide (LPS), a toxin that primarily activates glia cells, and 6-hydroxydopamine (6-OHDA), the neurotoxin that firstly impacts dopaminergic neurons, were utilized to mimic PD in rats. We found a common and exceedingly early over-production of HMGB1, followed by an increase of divalent metal transporter 1 with iron responsive element (DMT1+) in the dopaminergic neurons before quantifiable neuronal loss. HMGB1 neutralizing antibody suppressed inflammation in the SN, DMT1+ elevation in dopaminergic neurons, and dopaminergic neuronal loss in both LPS and 6-OHDA administration- induced PD models. On the contrary, interleukin-1ß inhibitor diacerein failed to suppress these outcomes induced by 6-OHDA. Our findings not only demonstrate that inflammation could be one of the causes of DMT1+ increase in dopaminergic neurons, but also highlight HMGB1 as a pivotal early mediator of inflammation-induced iron increase and subsequent neurodegeneration, thereby HMGB1 could serve as a potential target for early-stage PD treatment.


HMGB1 Protein , Parkinson Disease , Parkinsonian Disorders , Animals , Rats , Cytokines/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , HMGB1 Protein/metabolism , Inflammation/pathology , Iron/metabolism , Lipopolysaccharides , Oxidopamine , Parkinson Disease/pathology , Parkinsonian Disorders/metabolism
3.
Trends Endocrinol Metab ; 34(7): 404-413, 2023 07.
Article En | MEDLINE | ID: mdl-37210298

Despite several decades of study, whether iron is involved in the development of atherosclerosis remains a controversial and unresolved issue. Here, we focus on the up-to-date advances in studies on role of iron in atherosclerosis and discuss possible reasons why patients with hereditary hemochromatosis (HH) do not show any increased incidence of atherosclerosis. In addition, we analyze conflicting results concerning the role of iron in atherogenesis from several epidemiological and animal studies. We argue that atherosclerosis is not observed in HH because iron homeostasis in the arterial wall, the actual location of atherosclerosis, is not significantly affected, and support a causal link between iron in the arterial wall and atherosclerosis.


Atherosclerosis , Hemochromatosis , Animals , Hemochromatosis/genetics , Hemochromatosis/metabolism , Iron/metabolism , Homeostasis
4.
Autophagy ; 17(11): 3607-3621, 2021 11.
Article En | MEDLINE | ID: mdl-33563064

RASAL2 (RAS protein activator like 2), a RASGTPase activating protein, can catalyze the hydrolysis of RAS-GTP into RAS-GDP to inactivate the RAS pathway in various types of cancer cells. However, the cellular function of RASAL2 remains elusive. Here we showed that RASAL2 can attenuate PRKAA/AMPKα phosphorylation by recruiting phosphatase PPM1B/pp2cß, thus inhibiting the initiation of basal autophagy under normal conditions. In addition, we found that glucose starvation could induce dissociation of PPM1B from RASAL2 and then RASAL2 at S351 be phosphorylated by PRKAA, followed by the binding of phosphorylated-RASAL2 with to PIK3C3/VPS34-ATG14-BECN1/Beclin1 complex to increase PIK3C3 activity and autophagy. Furthermore, RASAL2 S351 phosphorylation facilitated breast tumor growth and correlated to poor clinical outcomes in breast cancer patients. Our study demonstrated that the phosphorylation status of RASAL2 S351 can function as a molecular switch to either suppress or promote AMPK-mediated autophagy. Inhibition of RASAL2 S351 phosphorylation might be a potential therapeutic strategy to overcome the resistance of AMPK-activation agents.Abbreviations: AICAR: aminoimidazole carboxamide ribonucleotide; AMPK: adenosine 5'-monophosphate (AMP)-activated protein kinase; ATG14: autophagy related 14; C.C: compound C; CQ: chloroquine; DKO: double-knockout; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PIK3R4/VPS15: phosphoinositide-3-kinase regulatory subunit 4; PPM1B/pp2cß: protein phosphatase, Mg2+/Mn2+ dependent 1B; PRKAA/AMPKα: protein kinase AMP-activated catalytic subunit alpha; PtdIns: phosphatidylinositol; PtdIns3P: phosphatidylinositol-3-phosphate; RASAL2: RAS protein activator like 2; RasGAPs: RasGTPase activating proteins; SQSTM1/p62: sequestosome 1; TNBC: triple-negative breast cancer.


AMP-Activated Protein Kinases/metabolism , Autophagy , GTPase-Activating Proteins/metabolism , Binding Sites , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , GTPase-Activating Proteins/physiology , Glucose/deficiency , Humans , Phosphorylation , Protein Phosphatase 2C/metabolism
5.
J Cell Physiol ; 236(6): 4515-4527, 2021 06.
Article En | MEDLINE | ID: mdl-33442879

Ischemic preconditioned (IP) neurons protect astrocytes against ischemia/reperfusion (I/R)-induced injury by inhibiting oxidative stress. However, the relevant mechanisms are unknown. Based on the role of nuclear factor-κB (NF-κB) in cell survival and adaption to oxidative stress, we hypothesized that NF-κB might be associated with astroprotection induced by IP neurons via upregulation of antioxidant enzymes. Here, we investigated the effects of IP neurons on NF-κB activation, cell viability, reactive oxygen species (ROS), expression of antioxidant enzymes, erythropoietin (EPO), and tumor necrosis factor α (TNF-α), in the presence or absence of BAY11-7082 (an NF-κB inhibitor), anti-EPO, and anti-TNF-α antibodies, in astrocytes treated with or without I/R. We found that IP neurons could keep NF-κB activation at a relatively higher but beneficial level, and in turn, upregulated the activity of antioxidant enzymes and hence enhanced cell viability and reduced ROS in I/R treated astrocytes. The results collectively indicated that IP neurons are able to significantly inhibit the I/R-induced NF-κB overactivation, probably via EPO and TNF-α, being essential for IP neuron-induced astroprotection under the conditions of I/R. We concluded that NF-κB-mediated antioxidative stress is one of the mechanisms by which IP neurons protect astrocytes against I/R injury.


Astrocytes/metabolism , Cerebral Cortex/metabolism , NF-kappa B/metabolism , Neurons/metabolism , Paracrine Communication , Reperfusion Injury/prevention & control , Animals , Antioxidants/metabolism , Astrocytes/enzymology , Astrocytes/pathology , Cell Hypoxia , Cells, Cultured , Cerebral Cortex/pathology , Culture Media, Conditioned/metabolism , Erythropoietin/metabolism , Glucose/deficiency , Neurons/pathology , Oxidative Stress , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
6.
Mol Cell Neurosci ; 111: 103589, 2021 03.
Article En | MEDLINE | ID: mdl-33422672

Iron has a key role in the activation of the autophagic pathway in rats with intracerebral hemorrhage (ICH), and hepcidin has the ability to reduce brain iron in ICH-rats. We therefore hypothesized that hepcidin might be able to inhibit autophagy by reducing iron in an ICH brain. Here, we investigated the effects of Ad-hepcidin and/or hepcidin peptide on autophagic activities in ICH models in vitro and in vivo. We demonstrated that ad-hepcidin and hepcidin peptide both inhibited hemin-induced increase in LC3-II/LC3-I conversion ratio and reversed the reduction in p62 content in cortical neurons in vitro. We also showed that ad-hepcidin inhibited ICH-induced increase in LC3-II/LC3-I conversion ratio and reversed ICH-induced reduction in p62 content in the brain cortex of rats in vivo. Based on these findings plus previous data on the effects of ad-hepcidin and/or hepcidin peptide on iron contents in ICH models, we suggested that hepcidin-induced inhibition of autophagy might be mediated via reducing iron in hemin-treated neurons in vitro and ICH-rat brain in vivo.


Autophagy , Cerebral Hemorrhage/metabolism , Hepcidins/metabolism , Adenoviridae/genetics , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Genetic Vectors/genetics , Hepcidins/genetics , Male , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequestosome-1 Protein/metabolism
7.
Redox Biol ; 40: 101865, 2021 04.
Article En | MEDLINE | ID: mdl-33493903

Association of both iron/hepcidin and apolipoprotein E (ApoE) with development of Alzheimer disease (AD) and atherosclerosis led us to hypothesize that ApoE might be required for body iron homeostasis. Here, we demonstrated that ApoE knock-out (KO) induced a progressive accumulation of iron with age in the liver and spleen of mice. Subsequent investigations showed that the increased iron in the liver and spleen was due to phosphorylated extracellular regulated protein kinases (pERK) mediated up-regulation of transferrin receptor 1 (TfR1), and nuclear factor erythroid 2-related factor-2 (Nrf2)-dependent down-regulation of ferroportin 1. Furthermore, replenishment of ApoE could partially reverse the iron-related phenotype in ApoE KO mice. The findings imply that ApoE may be essential for body iron homeostasis and also suggest that clinical late-onset diseases with unexplained iron abnormality may partly be related to deficiency or reduced expression of ApoE.


Cation Transport Proteins , Iron Overload , Animals , Apolipoproteins E/genetics , Cation Transport Proteins/genetics , Hepcidins , Iron/metabolism , Mice , Mice, Knockout , Receptors, Transferrin/genetics
8.
Transl Res ; 229: 53-68, 2021 03.
Article En | MEDLINE | ID: mdl-32932001

Iron plays a key role in secondary neuronal injury after intracerebral hemorrhage (ICH), and hepcidin is able to reduce brain iron in iron-overloaded rats by down-regulating iron transport proteins including ferroportin 1 and transferrin receptor 1. These led us to hypothesize that hepcidin might reduce iron-mediated neurotoxicity by inhibiting iron accumulation in ICH brain. Here, we examined effects of Ad-hepcidin (hepcidin expression adenovirus) on the nonheme iron contents, expression of hepcidin, ferritin and iron transport proteins, neuronal cell survival, water contents in the brain and/or cerebrospinal fluid (CSF), and ICH-induced apoptosis, neurological deficit by RT-PCR, Western blot analysis, NeuN Immunofluorescence, TUNEL, Fluoro-Jade B staining, behavioral performance and Morris water-maze tests in 510 rats. We demonstrated that hepcidin could significantly suppress the ICH-induced increase in iron and ferritin in brain tissues and CSF by inhibiting expression of iron transport proteins, increase neuronal survival by attenuating ICH-induced apoptosis, reactive oxygen species, neurodegeneration and brain edema, as well as effectively improve ICH-induced behavioral and cognitive deficit in rats. The findings collectively showed that hepcidin could effectively attenuate iron-mediated secondary neuronal injury after ICH in rats. This naturally existing protein can potentially be developed into a therapeutic drug for the treatment of ICH patients.


Cerebral Hemorrhage/pathology , Hepcidins/genetics , Iron/metabolism , Neurons/pathology , Adenoviridae/genetics , Animals , Apoptosis/genetics , Behavior, Animal , Brain Edema/metabolism , Brain Edema/pathology , Cation Transport Proteins/metabolism , Cell Survival/genetics , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/metabolism , Corpus Striatum/pathology , Corpus Striatum/physiology , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Gene Expression , Hepcidins/metabolism , Iron/cerebrospinal fluid , Male , Rats, Sprague-Dawley , Receptors, Transferrin/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
10.
EBioMedicine ; 50: 144-155, 2019 Dec.
Article En | MEDLINE | ID: mdl-31759919

BACKGROUND: Rasal2 has diametric effects on progression of oestrogen receptor-positive (ER+) and -negative (ER-) breast cancers. The relevant causes are unknown. It is also unknown whether the effects of Rasal2 are mediated by an exosome-transport process. METHODS: Exosomes were purified from breast cancer cells and identified by transmission electron microscopy and flow cytometry analysis. In vivo and in vitro experiments were conducted to investigate the role of Rasal2 in exosome-mediated breast cancer progression. Western blot analysis was performed to detect Rasal2 and p-Rasal2 (phosphorylated Rasal2) expression in ER+/ER- breast cancer cells and in exosomes, cancer tissues and blood of patients with ER+ or ER- breast cancer. FINDINGS: Phosphorylation of Rasal2 at Serine 237 promoted tumour growth in both ER+ and ER- tumour cells and tissues. The functions of both p-Rasal2 and non-p-Rasal2 (non-phosphorylated-Rasal2) in the modulation of breast cancer progression are exosome-mediated. p-Rasal2 expression in ER+ breast cancer cells and exosomes, cancer tissues and blood was significantly lower than in ER- tumour cells and patients. INTERPRETATION: p-Rasal2 facilitates tumour progression in both ER+ and ER- breast cancers. The ratio of p-Rasal2/non-p-Rasal2 in ER+ and ER- breast cancers is one of the factors deciding the role of Rasal2 (or total Rasal2) as a suppressor in ER+ breast cancers or as a promoter in ER- breast cancers. Targeting the phosphorylation of Rasal2 machinery may therefore be useful as a therapy to restrain breast cancer progression by reducing p-Rasal2/non-p-Rasal2 ratio, especially in ER- breast cancers. FUND: NSFC and Hong Kong Research Grants Council.


Biomarkers, Tumor , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , GTPase-Activating Proteins/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Exosomes/metabolism , Extracellular Vesicles/metabolism , Female , GTPase-Activating Proteins/genetics , Gene Expression , Gene Knockdown Techniques , Gene Targeting , Humans , Immunohistochemistry , Mice , Models, Biological , Phosphorylation , Xenograft Model Antitumor Assays
11.
Cell Death Dis ; 10(10): 708, 2019 09 24.
Article En | MEDLINE | ID: mdl-31551410

The reduced iron usage induced by the suppression of erythropoiesis is a major cause of the systemic iron overload in CBS knockout (CBS-/-) mice. However, the relevant mechanisms are unknown. Here, we examined changes in granulocyte/erythroid cell ratios, iron content, and expression of iron-metabolism proteins, including; two key enzymes involved in the heme biosynthetic pathway, ALAS2 (delta-aminolevulinate synthase 2) and FECH (ferrochelatase), a heme exporter from the cytosol and mitochondria, FLVCR (feline leukemia virus subgroup C cellular receptor) as well as EPO (erythropoietin), EPOR (erythropoietin receptor) and HIF-2α (hypoxia inducible factor-2 subunit α), in the blood, bone marrow or liver of CBS-/- (homozygous), CBS+/- (heterozygous) and CBS+/+ (Wild Type) mice. Our findings demonstrate that CBS deficiency can induce a significant reduction in the expression of ALAS2, FECH, FLVCR, HIF-2α, EPO, and EPOR as well as an increase in interleukin-6 (IL-6), hepcidin and iron content in the blood, bone marrow or liver of mice. We conclude that the suppression of erythropoiesis is mainly due to the CBS deficiency-induced disruption in the expression of heme biosynthetic enzymes and heme-transporter.


Cystathionine beta-Synthase/deficiency , Heme/metabolism , Animals , Cystathionine beta-Synthase/metabolism , Erythropoiesis , Humans , Mice
12.
J Cell Physiol ; 234(4): 3158-3169, 2019 04.
Article En | MEDLINE | ID: mdl-30370692

Hydrogen sulfide (H2 S) has a significant effect on the regulation of interleukin-6 (IL-6) and signal transducer and activator of transcription 3 (STAT3) activities, while IL-6 directly regulates hepcidin expression via STAT3. We therefore hypothesized that H 2 S has a role in body iron homeostasis by regulating the expression of iron transport proteins via the IL-6/STAT3/Hepcidin pathway. Here, we investigated the effects of two H 2 S donors sodium hydrosulfide and GYY4137 on the expression of ferroportin-1 (Fpn1), transferrin receptor-1 (TfR1), hepcidin, IL-6 and pSTAT3 in the spleen of mice in vivo and peritoneal macrophage in vitro. We also examined the effects of H 2 S on serum iron, transferrin saturation, and ferritin light chain contents in the spleen, and on nitrite content, nuclear factor erythroid 2-related factor-2 (Nrf2) and iron regulatory protein 1 (IRP1) in the macrophages. We demonstrated that H 2 S regulates the expression of TfR1 and Fpn1 in the spleen in vivo and in peritoneal macrophages in vitro predominantly via the IL-6/pSTAT3/hepcidin pathway, under the conditions of inflammation induced by lipopolysaccharides. We also provide evidence that under uninflamed conditions, the regulation of Fpn1 and TfR1 expression by H 2 S, both in vivo and in vitro, are mediated by the nitric oxide (NO)/Nrf2 and iron regulatory protein/iron responsive element pathways, respectively, which are independent of IL-6/pSTAT3/hepcidin signals. These findings show that H 2 S is a key player in iron homeostasis under not only the inflamed conditions but also uninflamed conditions.


Cation Transport Proteins/metabolism , Hydrogen Sulfide/pharmacology , Iron/metabolism , Macrophages, Peritoneal/drug effects , Morpholines/pharmacology , Organothiophosphorus Compounds/pharmacology , Receptors, Transferrin/metabolism , Spleen/drug effects , Sulfides/pharmacology , Animals , Cells, Cultured , Hepcidins/genetics , Hepcidins/metabolism , Inflammation/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Iron-Regulatory Proteins/metabolism , Macrophages, Peritoneal/metabolism , Male , Mice, Inbred C57BL , Morpholines/metabolism , NF-E2-Related Factor 2/metabolism , Nitric Oxide/metabolism , Organothiophosphorus Compounds/metabolism , Phosphorylation , STAT3 Transcription Factor/metabolism , Spleen/metabolism , Sulfides/metabolism
13.
Hepatology ; 67(1): 21-35, 2018 01.
Article En | MEDLINE | ID: mdl-28859237

Cystathionine ß-synthase (CBS) catalyzes the transsulfuration pathway and contributes, among other functions, to the generation of hydrogen sulfide. In view of the exceptionally high expression of CBS in the liver and the common interleukin-6 pathway used in the regulatory systems of hydrogen sulfide and hepcidin, we speculate that CBS is involved in body iron homeostasis. We found that CBS knockout (CBS-/- ) mice exhibited anemia and a significant increase in iron content in the serum, liver, spleen, and heart, along with severe damage to the liver, displaying a hemochromatosis-like phenotype. A high level of hepatic and serum hepcidin was also found. A major cause of the systemic iron overload is the reduced iron usage due to suppressed erythropoiesis, which is consistent with an increase in interleukin-6 and reduced expression of erythropoietin. Importantly, in the liver, absence of CBS caused both a reduction in the transcriptional factor nuclear factor erythroid 2-related factor-2 and an up-regulation of hepcidin that led to a decrease in the iron export protein ferroportin 1. The resulting suppression of iron export exacerbates iron retention, causing damage to hepatocytes. Finally, administration of CBS-overexpressing adenovirus into CBS mutant mice could partially reverse the iron-related phenotype. CONCLUSION: Our findings point to a critical role of CBS in iron homeostasis of the body, and the liver in particular; it is likely that a hemochromatosis-like phenotype in patients can be induced by aberration not only in the expression of key molecules in the hepcidin pathway but also of those related to CBS. (Hepatology 2018;67:21-35).


Anemia, Iron-Deficiency/enzymology , Anemia, Iron-Deficiency/pathology , Cystathionine beta-Synthase/metabolism , Hepatocytes/enzymology , Iron/metabolism , Liver/enzymology , Anemia, Iron-Deficiency/metabolism , Animals , Biopsy, Needle , Blotting, Western , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Hepatocytes/metabolism , Hepcidins/metabolism , Homeostasis , Humans , Immunohistochemistry , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multivariate Analysis , Random Allocation , Real-Time Polymerase Chain Reaction/methods , Reference Values
14.
Redox Biol ; 13: 20-31, 2017 10.
Article En | MEDLINE | ID: mdl-28551085

The extensive existing knowledge on bi-directional communication between astrocytes and neurons led us to hypothesize that not only ischemia-preconditioned (IP) astrocytes can protect neurons but also IP neurons protect astrocytes from lethal ischemic injury. Here, we demonstrated for the first time that neurons have a significant role in protecting astrocytes from ischemic injury. The cultured medium from IP neurons (IPcNCM) induced a remarkable reduction in LDH and an increase in cell viability in ischemic astrocytes in vitro. Selective neuronal loss by kainic acid injection induced a significant increase in apoptotic astrocyte numbers in the brain of ischemic rats in vivo. Furthermore, TUNEL analysis, DNA ladder assay, and the measurements of ROS, GSH, pro- and anti-apoptotic factors, anti-oxidant enzymes and signal molecules in vitro and/or in vivo demonstrated that IP neurons protect astrocytes by an EPO-mediated inhibition of pro-apoptotic signals, activation of anti-apoptotic proteins via the P13K/ERK/STAT5 pathways and activation of anti-oxidant proteins via up-regulation of anti-oxidant enzymes. We demonstrated the existence of astro-protection by IP neurons under ischemia and proposed that the bi-directionally protective communications between cells might be a common activity in the brain or peripheral organs under most if not all pathological conditions.


Astrocytes/metabolism , Cell Communication , Neurons/metabolism , Oxygen/metabolism , Animals , Astrocytes/physiology , Cell Hypoxia , Cells, Cultured , DNA Fragmentation , Glutathione/metabolism , L-Lactate Dehydrogenase/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/physiology , Phosphatidylinositol 3-Kinases/metabolism , Rats , Reactive Oxygen Species/metabolism , STAT5 Transcription Factor/metabolism
15.
Pharmacol Rep ; 69(1): 1-5, 2017 Feb.
Article En | MEDLINE | ID: mdl-27755990

BACKGROUND: The antioxidant properties of alpha-lipoic acid (ALA) are associated with its ability to reduce iron in cells and tissues, which is partly due to its inhibiting effect on iron uptake from transferrin and its promoting effect on iron deposition into ferritin. However, the relevant mechanisms are unknown. METHODS: We therefore investigated the effects of ALA on the expression of transferrin receptor 1 (TfR1), divalent metal transporter 1 (DMT1), ferroportin 1 (Fpn1) and ferritin in BV-2 microglia cells. RESULTS: We demonstrated that ALA significantly inhibited DMT1 expression, lowered ferritin-light-chain (Ft-L) and ferritin-heavy-chain (Ft-H) content, and had no effect on TfR1 and Fpn1 in BV-2 microglia cells. This indicated that the inhibiting effect of ALA on DMT1 might be one of the causes of the ALA-induced reduction in cellular transferrin-bound-iron uptake. We also demonstrated that ALA enhanced DMT1 and TfR1 expression in ferric ammonium citrate (FAC)-treated cells. FAC treatment led to a significant increase in Ft-L, Ft-H and Fpn1, and pre-treatment with ALA resulted in a further increase in the contents of Ft-L and Ft-H but not Fpn1 in cells. CONCLUSIONS: ALA could up-regulate TfR1, DMT1 and ferritin expression when iron is increased outside of the cell, promoting iron deposition into ferritin by increasing cell iron uptake, and then reducing free iron both inside and outside of the cell.


Cation Transport Proteins/metabolism , Ferritins/metabolism , Iron/metabolism , Microglia/metabolism , Receptors, Transferrin/metabolism , Thioctic Acid/pharmacology , Animals , Antioxidants/pharmacology , Biological Transport/drug effects , Biological Transport/physiology , Cell Line , Gene Expression Regulation , Mice , Microglia/drug effects
16.
Sci Rep ; 6: 21970, 2016 Feb 22.
Article En | MEDLINE | ID: mdl-26898550

Association of a high-serum ferritin with poor outcome showed that iron might play a detrimental role in the brain after intracerebral hemorrhage (ICH). Here, we investigated changes in serum iron, ferritin, transferrin (Tf) and ceruloplasmin (CP) in patients with ICH (n = 100) at day 1 (admission), 3, 7, 14 and 21 and those in control subjects (n = 75). The hematoma and edema volumes were also determined in ICH-patients on admission and at day 3. The Modified Rankin Scale (mRS) of 59 patients was ≥3 (poor outcome) and 41 < 3 (good outcome) at day 90. Serum ferritin was significantly higher and serum iron and Tf markedly lower in patients with poor-outcome than the corresponding values in patients with good-outcome at day 1 to 7 and those in the controls. There was a significant positive correlation between serum ferritin and relative edema volume or ratio at day 1 and 3 and hematoma volume at day 1 (n = 28), and a negative correlation between serum iron or Tf and hematoma volume at day 1 (n = 100). We concluded that not only increased serum ferritin but also reduced serum iron and Tf are associated with outcome as well as hematoma volume.


Cerebral Hemorrhage/blood , Ferritins/blood , Iron/blood , Transferrin/metabolism , Adult , Aged , Aged, 80 and over , Cerebral Hemorrhage/diagnostic imaging , Cerebral Hemorrhage/physiopathology , Ceruloplasmin/metabolism , Female , Hematoma/blood , Humans , Male , Middle Aged , Prognosis , Time Factors , Tomography, X-Ray Computed
17.
Glia ; 59(6): 936-45, 2011 Jun.
Article En | MEDLINE | ID: mdl-21438013

Hepcidin, an iron-regulatory hormone, plays a central role in iron homeostasis in peripheral tissues. The widespread distribution of hepcidin in the brain implies that the hormone may be essential for brain iron homeostasis. Here, we investigated the effects of hepcidin on the expression of iron uptake proteins, including transferrin receptor 1 (TfR1) and divalent metal transporter1 (DMT1) and the release protein ferroportin1 (Fpn1) in the cultured astrocytes. The effects of hepcidin on iron uptake, including transferrin-bound iron (Tf-Fe) and non-transferrin-bound iron (NTBI), and iron release were also studied. Our results demonstrated that astrocytes, when treated with hepcidin peptide or infected with hepcidin expression adenovirus (ad-hepcidin), showed a significant ability in reducing iron uptake (both Tf-Fe and NTBI), and iron release, which were accompanied by decreased expressions of TfR1, DMT1, and Fpn1. Moreover, we found that the effect of hepcidin in reducing TfR1 expression, which is dependent on the cyclic AMP-protein kinase A pathway, was the primary and dominant event. In conclusion, our results demonstrated that hepcidin controlled iron uptake and release by regulating expression of iron transport proteins. The findings also implied the existence of a novel hepcidin-receptor on the membrane of astrocytes.


Antimicrobial Cationic Peptides/physiology , Astrocytes/enzymology , Cyclic AMP-Dependent Protein Kinases/physiology , Iron/metabolism , Receptors, Transferrin/antagonists & inhibitors , Signal Transduction/physiology , Animals , Animals, Newborn , Antimicrobial Cationic Peptides/metabolism , Astrocytes/metabolism , Cell Membrane/enzymology , Cell Membrane/genetics , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/genetics , Hepcidins , Homeostasis/genetics , Rats , Rats, Sprague-Dawley , Receptors, Transferrin/biosynthesis , Receptors, Transferrin/genetics , Signal Transduction/genetics
18.
J Cell Biochem ; 109(1): 30-7, 2010 Jan 01.
Article En | MEDLINE | ID: mdl-19899109

Primary astrocyte cultures are the most commonly used in vitro model for neurobiological studies. We speculated that different protocols might induce differences not only in the percentage of astrocytes but also in their biological characteristics. In this study, we investigated the effects of four major protocols on the purity of astrocytes, cell viability, expression of glial fibrillary acidic protein (GFAP) and bystin of cultured astrocytes using MTT assay, immunocytochemical staining, and Western blot analysis. We demonstrated that the purity of astrocytes (98.9%) generated by the subculture (SC) procedure is significantly higher than those generated by primary culture (PC), shaken once culture (SK-1) or shaken twice culture (SK-2). We also showed that expressions of GFAP and bystin in astrocytes that are purified by the SK-2 or SK-1 procedures are significantly higher than those in astrocytes prepared by PC or SC. In addition, astrocytes cultured by SK-2 or SK-1 have a higher level of cell viabilities at most time points after ischemia compared with astrocytes cultured by PC or SC. These suggested that physical stimulation induced by "shaken" or culture operation might be able to activate astrocytes and implied that different procedures induce differences not only in the purity but also in the biological characteristics of astrocytes, such as the percentage of activated astrocytes, GFAP, and bystin expressions and responses to ischemia. A more detailed analysis about the effect of "culture protocol factor" on the biological characteristics of astrocytes is absolutely needed.


Astrocytes/cytology , Astrocytes/metabolism , Cell Adhesion Molecules/biosynthesis , Cell Culture Techniques/methods , Glial Fibrillary Acidic Protein/biosynthesis , Animals , Animals, Newborn , Blotting, Western , Cell Survival , Cells, Cultured/cytology , Cells, Cultured/metabolism , Immunohistochemistry , Mice , Mice, Inbred ICR
...