Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Can J Infect Dis Med Microbiol ; 2020: 4783062, 2020.
Article in English | MEDLINE | ID: mdl-33062082

ABSTRACT

OBJECTIVE: To explore the clinical characteristics of patients with different severity in the early outbreak of COVID-19, hoping to provide reference for clinical diagnosis and treatment. METHODS: We retrospectively analyzed the clinical data of 95 COVID-19 patients in Wuhan Red Cross Hospital of China from January 17 to February 13, 2020. All patients were investigated with epidemiological questionnaires. Outcomes were followed up until April 1, 2020. RESULTS: There were 53 males and 42 females, aged 22-84 years (mean 57.3 years). Clinical classification included 54 cases of common type, 27 cases of severe type, and 14 cases of critical type. Six patients had been exposed to the local Huanan seafood market. There were 38 clusters of COVID-19, including 27 family clusters and 11 work unit clusters. Common symptoms included fever (86 (90.5%) of 95), cough (73 (76.8%)), and fatigue (50 (52.6%)). Laboratory findings showed that the most common abnormalities were lymphopenia (75 (78.9%)), elevated D-dimer (60 (63.2%)), and elevated C-reactive protein (56 (58.9%)) on admission. All patients had abnormal chest computed tomography, showing patchy shadows or ground-glass opacities. Severe and critical cases were older, more likely to have shortness of breath, more likely to have underlying comorbidities, and more likely to have abnormal laboratory findings than common cases. The prognosis of patients with different degrees of severity was significantly different. All common and severe patients (100%) were cured and discharged from the hospital, while 10 (71.4%) of 14 critical patients died. CONCLUSIONS: COVID-19 has fast transmission speed and high pathogenicity. We must assess the severity of the disease and take corresponding treatment measures as early as possible.

2.
Aging Dis ; 8(4): 458-470, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28840060

ABSTRACT

Oxidative stress and iron accumulation are tightly associated with neurodegenerative diseases. Mitochondrial ferritin (FtMt) is identified as an iron-storage protein located in the mitochondria, and its role in regulation of iron hemeostasis in neurodegenerative diseases has been reported. However, the role of FtMt in hydrogen peroxide (H2O2)-induced oxidative stress and iron accumulation in neuronal cells has not been studied. Here, we overexpressed FtMt in neuroblastoma SH-SY5Y cells and induced oxidative stress by treating with extracellular H2O2. We found that overexpression of FtMt significantly prevented cell death induced by H2O2, particularly the apoptosis-dependent cell death. The protective effects involved inhibiting the generation of cellular reactive oxygen species, sustaining mitochondrial membrane potential, maintaining the level of anti-apoptotic protein Bcl-2, and inhibiting the activation of pro-apoptotic protein caspase 3. We further explored the mechanism of these protective effects and found that FtMt expression markedly altered iron homeostasis of the H2O2 treated cells as compared to that of controls. The FtMt overexpression significantly reduced cellular labile iron pool (LIP) and protected H2O2-induced elevation on LIP. While in H2O2 treated SH-SY5Y cells, the increased iron uptake and reduced iron release, in correlation with levels of DMT1(-IRE) and ferroportin 1, resulted in heavy iron accumulation, the FtMt overexpressing cells didn't show any significant changes in levels of iron transport proteins and in the level of LIP. These results implicate a neuroprotective role of FtMt on H2O2-induced oxidative stress, which may provide insights into the treatment of iron accumulation associated neurodegenerative diseases.

3.
Brain Res ; 1642: 33-42, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27017962

ABSTRACT

Our previous work showed that mitochondrial ferritin (MtFt) played an important role in preventing neuronal damage in 6-OHDA-induced Parkinson's disease (PD). However, the role of MtFt in a PD model induced by MPTP is not clear. Here, we found that methyl-4-phenyl-1, 2, 3, 6-tetra-pyridine (MPTP) significantly upregulated MtFt in the mouse hippocampus, substantia nigra (SN) and striatum. To explore the effect of MtFt upregulation on the MPTP-mediated injury to neural cells, MtFt-/- mice and MtFt-overexpressing cells were used to construct models of PD induced by MPTP. Our results showed that MPTP dramatically downregulated expression of transferrin receptor 1 (TfR1) and tyrosine hydroxylase and upregulated L-ferritin expression in the mouse striatum and SN. Interestingly, MPTP induced high levels of MtFt in these tissues, indicating that MtFt was involved in iron metabolism and influenced dopamine synthesis induced by MPTP. Meanwhile, the Bcl2/Bax ratio was decreased significantly by MPTP in the striatum and SN of MtFt knockout (MtFt-/-) mice compared with controls. Overexpression of MtFt increased TfR1 and decreased ferroportin 1 induced by 1-methyl-4-phenylpyridinium ions (MPP+). MtFt strongly inhibited mitochondrial damage through maintaining the mitochondrial membrane potential and protecting the integrity of the mitochondrial membrane. It also suppressed the increase of the labile iron pool, decreased production of reactive oxygen species and dramatically rescued the apoptosis induced by MPP+. In conclusion, this study demonstrates that MtFt plays an important role in preventing neuronal damage in the MPTP-induced parkinsonian phenotype by inhibiting cellular iron accumulation and subsequent oxidative stress.


Subject(s)
Brain/metabolism , Ferritins/metabolism , Iron/metabolism , MPTP Poisoning/metabolism , Mitochondria/metabolism , Oxidative Stress , Parkinson Disease/metabolism , 1-Methyl-4-phenylpyridinium/administration & dosage , Animals , Apoferritins/metabolism , Apoptosis/drug effects , Brain/drug effects , Cation Transport Proteins/metabolism , Cell Survival/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Ferritins/genetics , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Mice, Knockout , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Receptors, Transferrin/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/metabolism
4.
Cell Mol Life Sci ; 72(5): 983-97, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25213357

ABSTRACT

Mitochondrial ferritin (FtMt) has a significant effect on the regulation of cytosolic and mitochondrial iron levels. However, because of the deficiency of iron regulatory elements (IRE) in FtMt's gene sequence, the exact function of FtMt remains unclear. In the present study, we found that FtMt dramatically inhibited SH-SY5Y cell proliferation and tumor growth in nude mice. Interestingly, excess FtMt did not adversely affect the development of drosophila. Additionally, we found that the expression of FtMt in human normal brain tissue was significantly higher than that of neuroblastoma, but not higher than that of neurospongioma. However, the expression of transferrin receptor 1 is completely opposite. We therefore hypothesized that increased expression of FtMt may negatively affect the vitality of neuronal tumor cells. Therefore, we further investigated the underlying mechanisms of FtMt's inhibitory effects on neuronal tumor cell proliferation. As expected, FtMt overexpression disturbed the iron homeostasis of tumor cells and significantly downregulated the expression of proliferating cell nuclear antigen. Moreover, FtMt affected cell cycle, causing G1/S arrest by modifying the expression of cyclinD1, cyclinE, Cdk2, Cdk4 and p21. Remarkably, FtMt strongly upregulated the expression of the tumor suppressors, p53 and N-myc downstream-regulated gene-1 (NDRG1), but dramatically decreased C-myc, N-myc and p-Rb levels. This study demonstrates for the first time a new role and mechanism for FtMt in the regulation of cell cycle. We thus propose FtMt as a new candidate target for inhibiting neuronal tumor cell proliferation. Appropriate regulation of FtMt expression may prevent tumor cell growth. Our study may provide a new strategy for neuronal cancer therapy.


Subject(s)
Ferritins/metabolism , Mitochondria/metabolism , Animals , Apoptosis , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Cyclin D1/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Ferritins/genetics , G1 Phase Cell Cycle Checkpoints , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neuroblastoma/metabolism , Neuroblastoma/pathology , Proto-Oncogene Proteins c-myc/metabolism , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism
5.
J Gastroenterol Hepatol ; 30(3): 513-20, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25318588

ABSTRACT

BACKGROUND AND AIM: The body's requirement for iron is different at different developmental stages. However, the molecular mechanisms of age-dependent iron metabolism are poorly understood. In the present study, we investigated the expression of iron transport proteins in the duodenum of Sprague-Dawley rats at five different age stages. METHODS: Male Sprague-Dawley rats at postnatal week (PNW) 1, 3, 12, 44, and 88 were employed in the study. Serum iron status and tissue non-heme iron concentrations in the spleen, liver, bone marrow, heart, kidney, duodenal epithelium, and gastrocnemius were examined at each age stage. The expression of duodenal cytochrome b (DcytB), divalent metal transporter 1 (DMT1), ferroportin 1 (FPN1), hephaestin, and hepcidin were measured by real-time polymerase chain reaction or Western blot. RESULTS: The levels of serum iron and transferrin saturation were higher in the rats at PNW1 and 3 than in those at PNW12, 44, and 88. Non-heme iron contents decreased from PNW1 to PNW3 and then increased thereafter. Duodenal DcytB, DMT1, and FPN1 increased to the highest level at PNW3 and then decreased from PNW12 to 88. The hepatic hepcidin mRNA level decreased to the lowest level at PNW3 and then increased with age. CONCLUSION: Our findings showed that age had a significant effect on body iron status. The increased duodenal DcytB, DMT1, and FPN1 expression can enhance intestinal iron absorption to meet the high iron requirements in infants. Hepcidin or enterocyte iron levels may be involved in the regulation of age-dependent FPN1, DMT1, and DcytB expression in the duodenum.


Subject(s)
Aging/genetics , Aging/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cytochromes b/genetics , Cytochromes b/metabolism , Duodenum/metabolism , Gene Expression Regulation, Developmental/genetics , Gene Expression , Iron/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Animals , Blotting, Western , Enterocytes/metabolism , Hepcidins/metabolism , Intestinal Absorption/genetics , Male , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Tissue Distribution , Transferrin/metabolism
6.
Biomed Res Int ; 2015: 105325, 2015.
Article in English | MEDLINE | ID: mdl-26788496

ABSTRACT

In female, inadequate iron supply is a highly prevalent problem that often leads to iron-deficiency anemia. This study aimed to understand the effects of pregnancy and lactation on iron metabolism. Rats with different days of gestation and lactation were used to determine the variations in iron stores and serum iron level and the changes in expression of iron metabolism-related proteins, including ferritin, ferroportin 1 (FPN1), ceruloplasmin (Cp), divalent metal transporter 1 (DMT1), transferrin receptor 1 (TfR1), and the major iron-regulatory molecule-hepcidin. We found that iron stores decline dramatically at late-pregnancy period, and the low iron store status persists throughout the lactation period. The significantly increased FPN1 level in small intestine facilitates digestive iron absorption, which maintains the serum iron concentration at a near-normal level to meet the increase of iron requirements. Moreover, a significant decrease of hepcidin expression is observed during late-pregnancy and early-lactation stages, suggesting the important regulatory role that hepcidin plays in iron metabolism during pregnancy and lactation. These results are fundamental to the understanding of iron homeostasis during pregnancy and lactation and may provide experimental bases for future studies to identify key molecules expressed during these special periods that regulate the expression of hepcidin, to eventually improve the iron-deficiency status.


Subject(s)
Anemia, Iron-Deficiency/genetics , Cation Transport Proteins/blood , Hepcidins/blood , Iron/blood , Lactation/metabolism , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/pathology , Animals , Antigens, CD/blood , Ceruloplasmin/biosynthesis , Ceruloplasmin/metabolism , Female , Ferritins/blood , Gene Expression , Humans , Pregnancy , Rats , Receptors, Transferrin/blood
7.
Biomaterials ; 35(9): 3110-20, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24411335

ABSTRACT

Our previous work showed that a charge-reversal layer-by-layer nanosystem, PEI/PAH-Cit/AuNP-CS, effectively facilitates cellular uptake of siRNA and enhances the silencing efficacy of MDR1 siRNA. Here, the plasmid loading capacity of this vehicle was examined using EGFP-N1, and the plasmid release profile was determined in response to pH changes. The cytotoxicity of the EGFP-N1/PEI/PAH-Cit/AuNP-CS complex against HeLa and 293T cells was almost negligible. PEI/PAH-Cit/AuNP-CS efficaciously delivered the plasmids EGFP-N1 (encoding green fluorescent protein) and pGL3.0 (encoding luciferase) into 293T and HeLa cells, thus verifying the universality of PEI/PAH-Cit/AuNP-CS as a gene carrier. The results of an inverted fluorescence microscopy, flow cytometry (FCM) and western blotting methods demonstrated that PC-3 prostate cancer cells treated with EGFP-p53/PEI/PAH-Cit/AuNP-CS expressed higher levels of GFP than cells treated with EGFP-p53/PEI. Furthermore, PC-3 cells treated with EGFP-p53/PEI/PAH-Cit/AuNP-CS showed reduced cellular viability and increased nuclear fragmentation, consistent with elevated p53 expression. Propidium iodide (PI) flow cytometric assays were conducted to demonstrate that EGFP-p53/PEI/PAH-Cit/AuNP-CS elevated the level of apoptosis in PC-3 cells. Western blotting and caspase activation studies revealed that EGFP-p53/PEI/PAH-Cit/AuNP-CS complexes may induce PC-3 apoptosis via the mitochondria-mediated signaling pathway by up-regulation of Bax, down-regulation of Bcl-2, and activation of caspase-3.


Subject(s)
Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Transfection , Tumor Suppressor Protein p53/metabolism , Cell Proliferation , Cell Survival , Chitosan/chemistry , Citraconic Anhydrides/chemistry , Gold/chemistry , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Male , Metal Nanoparticles/chemistry , Metal Nanoparticles/ultrastructure , Microscopy, Fluorescence , Plasmids/metabolism , Polyamines/chemistry , Polyethyleneimine/chemistry , Prostatic Neoplasms/pathology , bcl-2-Associated X Protein/metabolism
8.
Yi Chuan ; 35(3): 255-61, 2013 Mar.
Article in Chinese | MEDLINE | ID: mdl-23575531

ABSTRACT

The mammalian SWI/SNF complex is one of ATP-dependent chromatin-remodeling complexes, which plays important roles in cell proliferation, differentiation, development and tumor suppression. ARID1A (AT-rich interactive domain-containing protein 1A) is a large subunit of SWI/SNF complex, and also an ARID family member with non- sequence-specific DNA binding activity. ARID1A is a tumor suppressor gene which is frequently mutated in many cancers, such as ovarian, bladder and gastric cancers. ARID1A can suppress cell proliferation through the up-regulation of p21 and the down-regulation of E2F-responsive genes. These findings on ARID1A and its role of tumor suppression contribute to understanding the mechanism of cancer development and developing new therapy for cancer.It is introduced in the review that ARID1A basic characteristic, related to cancer development, and biological role for full understanding of ARID1A.


Subject(s)
Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins , Humans , Mutation , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
9.
Antioxid Redox Signal ; 18(2): 158-69, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-22746342

ABSTRACT

AIMS: Mitochondrial ferritin (MtFt), which was recently discovered, plays an important role in preventing neuronal damage in 6-hydroxydopamine-induced Parkinsonism by maintaining mitochondrial iron homeostasis. Disruption of iron regulation also plays a key role in the etiology of Alzheimer's disease (AD). To explore the potential neuroprotective roles of MtFt, rats and cells were treated with Aß(25-35) to establish an AD model. RESULTS: We report that knockdown of MtFt expression significantly enhanced Aß(25-35)-induced neurotoxicity as shown by dysregulation of iron homeostasis, enhanced oxidative stress, and increased cell apoptosis. Opposite results were obtained when MtFt was overexpressed in SH-SY5Y cells prior to treatment with Aß(25-35). Further, MtFt inhibited Aß(25-35)-induced P38 mitogen-activated protein kinase and activated extracellular signal-regulated kinase (Erk) signaling. INNOVATION: MtFt attenuated Aß(25-35)-induced neurotoxicity and reduced oxidative damage through Erk/P38 kinase signaling. CONCLUSION: Our results show a protective role of MtFt in AD and suggest that regulation of MtFt expression in neuronal cells may provide a new neuroprotective strategy for AD.


Subject(s)
Amyloid beta-Peptides/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Ferritins/physiology , Mitochondria/metabolism , Oxidative Stress , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis , Caspase 3/metabolism , Cytochromes c/metabolism , Enzyme Activation , Ferritins/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Malondialdehyde/metabolism , RNA, Small Interfering , Rats , Reactive Oxygen Species/metabolism
10.
PLoS One ; 6(9): e25324, 2011.
Article in English | MEDLINE | ID: mdl-21957487

ABSTRACT

Oxidative stress plays an important role in neuronal injuries caused by cerebral ischemia. It is well established that free iron increases significantly during ischemia and is responsible for oxidative damage in the brain. However, the mechanism of this ischemia-induced increase in iron is not completely understood. In this report, the middle cerebral artery occlusion (MCAO) rat model was performed and the mechanism of iron accumulation in cerebral ischemia-reperfusion was studied. The expression of L-ferritin was significantly increased in the cerebral cortex, hippocampus, and striatum on the ischemic side, whereas H-ferritin was reduced in the striatum and increased in the cerebral cortex and hippocampus. The expression level of the iron-export protein ferroportin1 (FPN1) significantly decreased, while the expression of transferrin receptor 1 (TfR1) was increased. In order to elucidate the mechanisms of FPN1 regulation, we studied the expression of the key regulator of FPN1, hepcidin. We observed that the hepcidin level was significantly elevated in the ischemic side of the brain. Knockdown hepcidin repressed the increasing of L-ferritin and decreasing of FPN1 invoked by ischemia-reperfusion. The results indicate that hepcidin is an important contributor to iron overload in cerebral ischemia. Furthermore, our results demonstrated that the levels of hypoxia-inducible factor-1α (HIF-1α) were significantly higher in the cerebral cortex, hippocampus and striatum on the ischemic side; therefore, the HIF-1α-mediated TfR1 expression may be another contributor to the iron overload in the ischemia-reperfusion brain.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Infarction, Middle Cerebral Artery/metabolism , Iron/metabolism , Animals , Antimicrobial Cationic Peptides/deficiency , Antimicrobial Cationic Peptides/genetics , Brain/metabolism , Cation Transport Proteins/metabolism , Ferritins/metabolism , Gene Knockdown Techniques , Hepcidins , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/genetics , Interleukin-6/genetics , Iron Overload/complications , Iron Overload/genetics , Iron Overload/metabolism , Mice , Rats , Receptors, Transferrin/metabolism , Reperfusion Injury/complications , Up-Regulation
11.
Int J Sport Nutr Exerc Metab ; 21(1): 19-26, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21411831

ABSTRACT

Some athletes are diagnosed as suffering from sports anemia because of iron deficiency, but the regulatory mechanism remains poorly understood. It is reported that hepcidin may provide a way to illuminate the regulatory mechanism of exercise-associated anemia. Here the authors investigate the hepcidin-involved iron absorption in exercise-associated anemia. Twelve male Wistar rats (300 ± 10 g) were randomly divided into 2 groups, 6 in a control group (CG) and 6 in an exercise group (EG, 5 wk treadmill exercise of different intensities with progressive loading). Serum samples were analyzed for circulating levels of IL-6 by means of enzyme-linked immunosorbent assay (ELISA). The expression of hepatic hepcidin mRNA was examined by real-time polymerase chain reaction analysis. The protein levels of divalent metal transporter 1 (DMT1), ferroportin1 (FPN1), and heme-carrier protein 1 (HCP1) of duodenum epithelium were examined by Western blot. The results showed that the amount of iron and ferritin in serum were lower in EG than in CG (p < .05). The levels of IL-6 and white blood cells were greater in EG than in CG (p < .01). The expression of DMT1, HCP1, and FPN1 was significantly lower in EG than in CG (p < .01). The mRNA expressions of hepatic hepcidin and hemojuvelin in skeletal muscle were remarkably higher in EG than in CG. The data indicated that inflammation was induced by strenuous exercise, and as a result, the transcriptional level of the hepatic hepcidin gene was increased, which further inhibited the expression of iron-absorption proteins and led to exercise-associated anemia.


Subject(s)
Anemia, Iron-Deficiency/etiology , Antimicrobial Cationic Peptides/metabolism , Physical Exertion/physiology , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/metabolism , Animals , Antimicrobial Cationic Peptides/genetics , Case-Control Studies , Cation Transport Proteins/metabolism , Duodenum/metabolism , Enzyme-Linked Immunosorbent Assay , GPI-Linked Proteins , Hemochromatosis Protein , Hepcidins , Interleukin-6/metabolism , Iron/blood , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Muscle, Skeletal/metabolism , Proton-Coupled Folate Transporter/metabolism , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
13.
Antioxid Redox Signal ; 13(6): 783-96, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20121342

ABSTRACT

Neuronal iron homeostasis disruption and oxidative stress are closely related to the pathogenesis of Parkinson's disease (PD). Adult iron-regulatory protein 2 knockout (Ireb2(-/-)) mice develop iron accumulation in white matter tracts and nuclei in different brain area and display severe neurodegeneration in Purkinje cells of the cerebrum. Mitochondrial ferritin (MtFt), a newly discovered ferritin, specifically expresses in high energy-consuming cells, including neurons of brain and spinal cord. Interestingly, the decreased expression of MtFt in cerebrum, but not in striatum, matches the differential neurodegeneration pattern in these Ireb2(-/-) mice. To explore its effect on neurodegeneration, the effects of MtFt expression on 6-hydrodopamine (6-OHDA)-induced neuronal damage was examined. The overexpression of MtFt led to a cytosolic iron deficiency in the neuronal cells and significantly prevented the alteration of iron redistribution induced by 6-OHDA. Importantly, MtFt strongly inhibited mitochondrial damage, decreased production of the reactive oxygen species and lipid peroxidation, and dramatically rescued apoptosis by regulating Bcl-2, Bax and caspase-3 pathways. In conclusion, this study demonstrates that MtFt plays an important role in preventing neuronal damage in an 6-OHDA-induced parkinsonian phenotype by maintaining iron homeostasis. Regulation of MtFt expression in neuronal cells may provide a new neuroprotective strategy for PD.


Subject(s)
Apoptosis , Cytoprotection , Ferritins/metabolism , Hydroxydopamines/metabolism , Mitochondria/metabolism , Parkinson Disease/metabolism , Animals , Caspase 3/metabolism , Cell Line , Ferritins/genetics , Iron Regulatory Protein 2/deficiency , Iron Regulatory Protein 2/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/metabolism
15.
J Trace Elem Med Biol ; 23(4): 290-9, 2009.
Article in English | MEDLINE | ID: mdl-19747625

ABSTRACT

Aceruloplasminemia is an autosomal recessive disorder caused by mutations in the ceruloplasmin (CP) gene. It is characterized by iron accumulation in the brain and in visceral organs. However, little is known about the mechanism of iron transport in these regions. Adult CP null (CP(-/-)) mice show increased iron deposition in several regions of brain, such as the cerebellum and brainstem. In this study, we investigated the expression of the ceruloplasmin homolog hephaestin (Heph) in the brain of CP(-/-) mice as a function of age. In the cerebral cortex and caudate putamen of 80-week-old CP(-/-) mice, the expression of Heph increased significantly whilst iron levels remain normal [Patel BN, Dunn RJ, Jeong SY, Zhu Q, Julien JP, David S. Ceruloplasmin regulates iron levels in the CNS and prevents free radical injury. J Neurosci 2002;22(15):6578-6], indicating that Heph might compensate for the loss of CP. In contrast, the substantia nigra and cerebellum of 80-week-old CP(-/-) mice accumulate iron but do not express high levels or significant decrease of Heph, suggesting that Heph does not replace CP in these regions. These data suggest that Heph may compensate for the loss of CP in a region-specific manner.


Subject(s)
Aging/metabolism , Brain/metabolism , Ceruloplasmin/deficiency , Membrane Proteins/metabolism , Aging/genetics , Animals , Brain/pathology , Ceruloplasmin/genetics , Ceruloplasmin/metabolism , Ependyma/metabolism , Ependyma/pathology , Gene Deletion , Genotype , Immunohistochemistry , Iron Overload/metabolism , Mice , Mice, Inbred BALB C , Phenotype
16.
Guang Pu Xue Yu Guang Pu Fen Xi ; 29(4): 1127-30, 2009 Apr.
Article in Chinese | MEDLINE | ID: mdl-19626918

ABSTRACT

In the present study, the authors performed the solid sampling and detected the iron levels in cortex, hippocampus and striatum of rat brain by GFAAS. The authors' results showed that there are no remarkable difference between the data obtained by solid sampling graphite furnace atomic absorption and liquid sampling graphite furnace atomic absorption. Compared to liquid sampling graphite furnace atomic absorption, the sample pre-treatment stage was obviously simplified, the cost was reduced significantly, and the time was shortened significantly in the solid sampling GFAAS. This study will be beneficial to the mensuration of iron content in micro-tissue of animal by solid sampling GFASS.


Subject(s)
Brain Chemistry , Brain/growth & development , Iron/analysis , Spectrophotometry, Atomic/methods , Age Factors , Animals , Brain/metabolism , Female , Iron/metabolism , Male , Rats , Rats, Sprague-Dawley , Spectrophotometry, Atomic/instrumentation
17.
Peptides ; 30(2): 262-6, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18948155

ABSTRACT

Hepcidin, a principle regulator of iron metabolism, is synthesized by the liver. Contradictory results have been reported on the regulation of hepcidin expression in response to serum transferrin saturation and liver iron content. In the present study, we explore the expression of murine hepcidin mRNA and further analyze the relationship between liver hepcidin mRNA expression, liver iron stores, and serum iron level utilizing ceruloplasmin gene knockout mice. We find that hepcidin expression correlates significantly with serum transferrin saturation, whereas there is a negative correlation of hepcidin expression with liver tissue iron level.


Subject(s)
Anti-Infective Agents/metabolism , Antimicrobial Cationic Peptides/genetics , Ceruloplasmin/genetics , Down-Regulation/genetics , Animals , Antimicrobial Cationic Peptides/metabolism , Ceruloplasmin/metabolism , Genotype , Hepcidins , Iron/blood , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred BALB C , Phenotype , RNA, Messenger/metabolism , Transferrin/metabolism
18.
Anat Rec (Hoboken) ; 292(2): 225-33, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19051252

ABSTRACT

The cellular localization of DMT1 and its functional characterization suggest that DMT1 may play an important role in the physiological brain iron transport. But the regulation of DMT1 expression by iron in the brain is still not clearly understood. In this study, both the contents of ferric and ferrous iron as well as DMT1 expression were evaluated in CPu and SN after ICV of 500 microg iron dextran/rat/day for 3 or 7 days. It was found that the iron levels in CPu and SN were not altered obviously until ICV for 7 days. Immunohistochemistry results indicated that the expression of DMT1 (-IRE) in CPu and SN was not altered significantly after 3 days of ICV. Whereas the expression of DMT1 (-IRE) decreased significantly after 7 days of ICV when ferrous iron was increased significantly. Contrary to that of DMT1 (-IRE) in the same regions, there were no significant alterations in DMT1 (+IRE) expression in CPu and SN in spite of the existence of the altered iron levels, compared with that of control groups. The results demonstrate that DMT1 (-IRE) expression was correlated probably with brain iron levels; especially, its regulation was correlated with ferrous iron (not ferric iron) in CPu and SN in adult rats, compared with those of saline-injected control rats. The effect of ferrous iron on the expression of DMT1 (-IRE) in the brain also suggests that it might play a major physiological role in brain iron uptake and transport, but further studies are needed to clarify these issues.


Subject(s)
Cation Transport Proteins/metabolism , Caudate Nucleus/drug effects , Caudate Nucleus/metabolism , Iron-Dextran Complex/administration & dosage , Iron/metabolism , Putamen/drug effects , Putamen/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Animals , Cation Transport Proteins/genetics , Immunohistochemistry , Injections, Intraventricular , Male , Rats , Rats, Sprague-Dawley , Time Factors
19.
J Gastroenterol ; 43(2): 136-43, 2008.
Article in English | MEDLINE | ID: mdl-18306987

ABSTRACT

BACKGROUND: Erythropoietin (Epo) is the central regulator of red blood cell production and can stimulate proliferation and differentiation of erythroid progenitor cells. Now, recombinant human erythropoietin (rHuEpo) is widely used in patients with renal disease, chronic anemia, and iron deficiency of early childhood. It has been reported that the enhanced erythropoiesis associated with erythropoietin therapy increases intestinal iron absorption, but the molecular mechanisms underlying are unknown. Therefore, we have investigated the effect of rHuEpo on duodenal iron transport protein synthesis in rats. METHODS: Male Sprague-Dawley rats weighing 250 g were randomly divided into two groups: (1) rHuEpo injection group (rHuEpo, 500 IU/day, s.c.), and (2) control group (injection of the same volume of saline). After 3 days injection, blood parameters, serum iron status, and non-heme iron concentrations in the liver and duodenum were examined at the fifth day. The mRNA levels and protein synthesis of duodenal divalent metal transporter 1 (DMT1), ferroportin 1 (FPN1), and hephaestin (Hp) were measured by reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blot analysis. Hepatic hepcidin mRNA expression was analyzed by RT-PCR. RESULTS: rHuEpo injection significantly stimulated erythropoiesis and decreased serum iron status, non-heme iron concentrations in the liver and duodenum. DMT1 (+IRE) and Hp expression in duodenum were increased significantly. However, DMT1 (-IRE) and FPN1 expression had no apparent change. Hepatic hepcidin mRNA expression was decreased dramatically, reaching an almost undetectable level in rHuEpo-treated rats. CONCLUSIONS: rHuEpo administration improved the duodenal iron absorption by increasing the expression of DMT1 (+IRE) and Hp.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Cation Transport Proteins/metabolism , Erythropoietin/pharmacology , Hematinics/pharmacology , Intestinal Absorption/drug effects , Iron-Regulatory Proteins/metabolism , Membrane Proteins/metabolism , Animals , Antimicrobial Cationic Peptides/genetics , Blotting, Western , Cation Transport Proteins/genetics , Duodenum/drug effects , Duodenum/metabolism , Gene Expression Regulation/drug effects , Hepcidins , Immunohistochemistry , Iron-Regulatory Proteins/genetics , Male , Models, Animal , Rats , Rats, Sprague-Dawley , Recombinant Proteins , Reverse Transcriptase Polymerase Chain Reaction
20.
J Cell Biochem ; 104(2): 629-41, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18189270

ABSTRACT

Recycled iron from reticuloendothelial macrophages to erythroid precursors is important to maintain the iron homeostasis. However, the molecular mechanisms underlying iron homeostasis in macrophages are poorly understood. In this study, male Sprague-Dawley rats were treated with recombinant human erythropoietin (rHuEpo, 500 IU/day, s.c.) for 3 days. At the fifth day, peritoneal exudate macrophages were harvested, and then (55)Fe uptake and release were measured by liquid scintillation counting method. The expression of divalent metal transporter 1 (DMT1) and ferroportin 1 (FPN1) in peritoneal exudate macrophages was detected by RT-PCR and Western blot. In order to exclude the direct effect of rHuEpo on macrophages, the parallel experiments were performed with incubation normal peritoneal exudate macrophages with rHuEpo (2 IU/ml). Our results showed rHuEpo injection reduced the peritoneal exudate macrophages iron retention. The uptake of Fe(II) was decreased via the suppression of DMT1 (+IRE) expression and the release of Fe(II) was increased with increasing the expression of FPN1 in macrophages. Moreover, the expression of HAMP mRNA was four times lower in rHuEpo-treated liver of rats than control group (CG). HAMP mRNA expression was increased; the synthesis of DMT1 had no significant change, whereas the FPN1 was decreased in normal peritoneal exudate macrophages after treatment with rHuEpo in vitro. We conclude that hepcidin may play a major, causative role in the change of FPN1 synthesis and that decreased the iron retention in macrophages of rHuEpo-treated rats.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Cation Transport Proteins/biosynthesis , Erythropoietin/pharmacology , Iron/metabolism , Macrophages, Peritoneal/metabolism , Animals , Hepcidins , Homeostasis , Humans , Male , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...