Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Toxicol Appl Pharmacol ; 336: 94-100, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29054681

ABSTRACT

Manganese neurotoxicity is characterized by Parkinson-like symptoms with degeneration of dopaminergic neurons in the basal ganglia as the principal pathological feature. Manganese neurotoxicity studies may contribute to a good understanding of the mechanism of Parkinson's disease (PD). In this study, we first confirmed that MnCl2 can promote the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) protein in the nucleus or cytoplasm while increasing the binding activity of Nrf2 and antioxidant response elements, further promoting the expression of downstream target gene heme oxygenase 1 (HO-1) and leading to increase levels of reactive oxygen species (ROS) and reduce the levels of reduced glutathione (GSH). Second, we investigated the role of histone acetylation in the activation of Nrf2/HO-1 pathway by manganese chloride in rat adrenal pheochromocytoma (PC12) cells. Histone acetyltransferase inhibitor (anacardic acid) and histone deacetylase inhibitor (trichostatin A, TSA) were used as pretreatment reagents to adjust the level of histone acetylation. Here, we show that downregulation of histone acetylation can inhibit Mn-induced Nrf2 nuclear translocation and further inhibits the Mn-activated Nrf2/HO-1 pathway. This downregulation also promotes manganese-induced increase of ROS and decrease of GSH in neurons. These results suggest that the downregulation of histone acetylation may play an important role in the neurotoxicity caused by manganese and that TSA may provide new ideas and targets in treating manganese-induced Parkinson's syndrome and PD.


Subject(s)
Chlorides/toxicity , Heme Oxygenase (Decyclizing)/metabolism , Histones/metabolism , Manganese Poisoning/etiology , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Acetylation , Active Transport, Cell Nucleus , Anacardic Acids/pharmacology , Animals , Glutathione/metabolism , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Manganese Compounds , Manganese Poisoning/drug therapy , Manganese Poisoning/enzymology , Manganese Poisoning/genetics , NF-E2-Related Factor 2/genetics , Neurons/enzymology , PC12 Cells , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
2.
Glia ; 59(11): 1732-43, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21812036

ABSTRACT

Manganese (Mn) is a trace element essential for normal human development and is required for the proper functioning of a variety of physiological processes. Chronic exposure to Mn can cause manganism, a neurodegenerative disorder resembling idiopathic Parkinson's disease (PD). Mn(II) neurotoxicity is characterized by astrocytic impairment both in the expression and activity of glutamine (Gln) transporters. Because protein kinase C (PKC) activation leads to the downregulation of a number of neurotransmitter transporters and Mn(II) increases PKC activity, we hypothesized that the PKC signaling pathway contributes to the Mn(II)-mediated disruption of Gln turnover. Our results have shown that Mn exposure increases the phosphorylation of both the PKCα and PKCδ isoforms. PKC activity was also shown to be increased in response to Mn(II) treatment. Corroborating our earlier observations, Mn(II) also caused a decrease in Gln uptake. This effect was blocked by PKC inhibitors. Notably, PKC activation caused a decrease in Gln uptake mediated by systems ASC and N, but had no effect on the activities of systems A and L. Exposure to α-phorbol 12-myristate 13-acetate significantly decreased SNAT3 (system N) and ASCT2 (system ASC) protein levels. Additionally, a co-immunoprecipitation study demonstrated the association of SNAT3 and ASCT2 with the PKCδ isoform, and Western blotting revealed the Mn(II)-mediated activation of PKCδ by proteolytic cleavage. PKC activation was also found to increase SNAT3 and ubiquitin ligase Nedd4-2 binding and to induce hyperubiquitination. Taken together, these findings demonstrate that the Mn(II)-induced dysregulation of Gln homeostasis in astrocytes involves PKCδ signaling accompanied by an increase in ubiquitin-mediated proteolysis.


Subject(s)
Astrocytes/metabolism , Glutamine/metabolism , Manganese Poisoning/metabolism , Protein Kinase C/physiology , Amino Acid Transport System ASC/biosynthesis , Amino Acid Transport System ASC/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Biotinylation , Blotting, Western , Cells, Cultured , Chlorides/toxicity , Endosomal Sorting Complexes Required for Transport/metabolism , Immunoprecipitation , Manganese Compounds , Manganese Poisoning/enzymology , Membrane Proteins/biosynthesis , Minor Histocompatibility Antigens , Nedd4 Ubiquitin Protein Ligases , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Ubiquitin/physiology , Ubiquitin-Protein Ligases/metabolism
3.
Toxicol Lett ; 295: 357-368, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-30040983

ABSTRACT

Heme Oxygenase-1 (HO-1), a stress- responsive enzyme which catalyzes heme degradation into iron, carbon monoxide, and biliverdin, exerts a neuroprotective role involving many different signaling pathways. In Parkinson disease patients, elevated HO-1 expression levels in astrocytes are involved in antioxidant defense. In the present work, employing an in vitro model of Mn2+-induced Parkinsonism in astroglial C6 cells, we investigated the role of HO-1 in both apoptosis and mitochondrial quality control (MQC). HO-1 exerted a protective effect against Mn2+ injury. In fact, HO-1 decreased both intracellular and mitochondrial reactive oxygen species as well as the appearance of apoptotic features. Considering that Mn2+ induces mitochondrial damage and a defective MQC has been implicated in neurodegenerative diseases, we hypothesized that HO-1 could mediate cytoprotection by regulating the MQC processes. Results obtained provide the first evidence that the beneficial effects of HO-1 in astroglial cells are mediated by the maintenance of both mitochondrial fusion/fission and biogenesis/mitophagy balances. Altogether, our data demonstrate a pro-survival function for HO-1 in Mn2+-induced apoptosis that involves the preservation of a proper MQC. These findings point to HO-1 as a new therapeutic target linked to mitochondrial pathophysiology in Manganism and probably Parkinson´s disease.


Subject(s)
Astrocytes/drug effects , Chlorides/toxicity , Heme Oxygenase-1/metabolism , Manganese Poisoning/etiology , Mitochondria/drug effects , Oxidative Stress/drug effects , Parkinsonian Disorders/chemically induced , Animals , Apoptosis/drug effects , Astrocytes/enzymology , Astrocytes/pathology , Cell Line, Tumor , Dose-Response Relationship, Drug , Manganese Compounds , Manganese Poisoning/enzymology , Manganese Poisoning/pathology , Mitochondria/enzymology , Mitochondria/pathology , Mitochondrial Dynamics/drug effects , Mitophagy/drug effects , Parkinsonian Disorders/enzymology , Parkinsonian Disorders/pathology , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Time Factors
4.
Neurotoxicology ; 28(3): 548-54, 2007 May.
Article in English | MEDLINE | ID: mdl-17320182

ABSTRACT

Recent studies have suggested that the globus pallidus may be a particularly sensitive target of manganese (Mn), however, in vitro studies of the effects of Mn on GABAergic neurons have been restricted by the lack of a cell model expressing GABAergic properties. Here, we investigated the effects of low-level Mn treatment on cellular GABA and glutamate metabolism using the newly characterized AF5 rat neural-derived cell line, which displays GABAergic properties during culture in vitro. Intracellular GABA and glutamate levels were measured along with measurement of the release of GABA and glutamate into the culture medium, glutamine uptake from the culture medium, and the specific effects of Mn on the enzymes directly responsible for the synthesis and degradation of GABA, glutamate decarboxylase (GAD) and GABA transaminase (GABA-T). Our results demonstrate that Mn had no effect on the activities of GAD or GABA-T. Similarly, low-level Mn treatment of AF5 cultures had only a small effect on intracellular GABA levels (114% of control) and no effect on the release of GABA. In contrast, intracellular and extracellular glutamate levels were enhanced to 170 and 198% of control during Mn treatment, respectively, while extracellular glutamine decreased to 73% of controls. Together, these results suggest that glutamate homeostasis may be preferentially affected over GABA in AF5 cells during low-level Mn treatment, suggesting a novel mechanism by which Mn-induced excitotoxicity might arise.


Subject(s)
Glutamic Acid/metabolism , Manganese Poisoning/metabolism , gamma-Aminobutyric Acid/metabolism , 4-Aminobutyrate Transaminase/metabolism , Amino Acids/metabolism , Animals , Cell Line , Culture Media , Extracellular Space/drug effects , Extracellular Space/metabolism , Glutamate Decarboxylase/metabolism , L-Lactate Dehydrogenase/metabolism , Manganese Poisoning/enzymology , Neurons/drug effects , Neurons/enzymology , Neurons/metabolism , Rats
5.
Biol Trace Elem Res ; 180(2): 297-305, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28361387

ABSTRACT

There were many studies about the effect of excess manganese (Mn) on nervous system apoptosis; however, Mn-induced apoptosis in chicken cerebrums and embryonic neurocytes was unclear. The purpose of this study was to investigate the effect of excess Mn on chicken cerebrum and embryonic neurocyte apoptosis. Seven-day-old Hyline male chickens were fed either a commercial diet or three levels of manganese chloride (MnCl2)-added commercial diets containing 600-, 900-, and 1800-mg/kg-Mn diet, respectively. On the 30th, 60th, and 90th days, cerebrums were collected. Fertilized Hyline chicken eggs were hatched for 6-8 days and were selected. Embryonic neurocytes with 0, 0.5, 1, 1.5, 2, 2.5, and 3 mM Mn were collected and were cultured for 12, 24, 36, and 48 h, respectively. The following research contents were performed: superoxide dismutase (SOD) and total antioxidant capacity (T-AOC) activities; tumor protein p53 (p53), B cell lymphoma-2 (Bcl-2), B cell lymphoma extra large (Bcl-x), Bcl-2-associated X protein (Bax), Bcl-2 homologous antagonist/killer (Bak), fas, and caspase-3 messenger RNA (mRNA) expression; and morphologic observation. The results indicated that excess Mn inhibited SOD and T-AOC activities; induced p53, Bax, Bak, fas, and caspase-3 mRNA expression; and inhibited Bcl-2 and Bcl-x mRNA expression in chicken cerebrums and embryonic neurocytes. There were dose-dependent manners on all the above factors at all the time points and time-dependent manners on SOD activity of 1800-mg/kg-Mn group, T-AOC activity, and apoptosis-related gene mRNA expression in all the treatment groups in chicken cerebrums. Excess Mn induced chicken cerebrum and embryonic neurocyte apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/agonists , Apoptosis/drug effects , Cerebrum/drug effects , Gene Expression Regulation, Developmental/drug effects , Manganese/adverse effects , Neurons/drug effects , Oxidative Stress/drug effects , Administration, Oral , Animals , Animals, Inbred Strains , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Avian Proteins/agonists , Avian Proteins/antagonists & inhibitors , Avian Proteins/genetics , Avian Proteins/metabolism , Biomarkers/metabolism , Cells, Cultured , Cerebrum/metabolism , Cerebrum/pathology , Cerebrum/ultrastructure , Chick Embryo , Chickens , China , Chlorides/administration & dosage , Dose-Response Relationship, Drug , Male , Manganese/administration & dosage , Manganese Compounds/administration & dosage , Manganese Poisoning/enzymology , Manganese Poisoning/metabolism , Manganese Poisoning/pathology , Microscopy, Electron, Transmission , Neurons/metabolism , Neurons/pathology , Neurons/ultrastructure , Random Allocation
6.
Hum Exp Toxicol ; 34(3): 240-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24972623

ABSTRACT

This study was conducted to investigate the protective effects of sodium p-aminosalicylic acid (PAS-Na) on learning and memory via increasing the number of basal forebrain choline acetyltransferase (ChAT) neurons in manganese (Mn)-exposed rats. Male Sprague Dawley rats were divided into following groups: the normal control I, II, and III groups, the model I, II, and III groups, low- and high-dose PAS-Na treatment (L- and H-PAS) group, PAS-Na prevention (PAS-P) group, and PAS-Na treatment (PAS-T) group. The model I, II, and III groups, L- and H-PAS, and PAS-T groups received intraperitoneal (i.p.) injection of 15 mg/kg manganese chloride tetrahydrate (MnCl2·4H2O) for 3 or 12 weeks, while the normal control I, II, and III groups received i.p. injection of an equal volume of saline; L- and H-PAS and PAS-T groups received back subcutaneous (s.c.) injection of PAS-Na (100 and 200 mg/kg) for the next 5 or 6 weeks, whereas model I and II group received back s.c. injection of an equal volume of saline. However, PAS-P group received back s.c. injection of 200 mg/kg PAS-Na + i.p. injection of 15 mg/kg MnCl2·4H2O for 12 weeks. Mn exposure significantly reduced the ability of spatial learning and memory capability, while PAS-Na prevention recovered it. Mn decreased the number of ChAT-positive neurons in vertical limb nucleus of the basal forebrain diagonal band/horizontal limb nucleus of the basal forebrain diagonal band and ChAT protein activity and treatment or prevention with PAS-Na restored those comparable with control. In brief, our results showed that PAS-Na may have protective effects on learning and memory against Mn via increasing the number of ChAT-positive neurons and activity of ChAT protein.


Subject(s)
Aminosalicylic Acid/pharmacology , Choline O-Acetyltransferase/metabolism , Cognition Disorders/enzymology , Manganese Poisoning/enzymology , Neuroprotective Agents/pharmacology , Aminosalicylic Acid/therapeutic use , Animals , Basal Forebrain/drug effects , Basal Forebrain/enzymology , Cognition Disorders/drug therapy , Learning/drug effects , Male , Manganese Poisoning/drug therapy , Memory/drug effects , Neurons/drug effects , Neurons/enzymology , Neuroprotective Agents/therapeutic use , Rats, Sprague-Dawley
7.
Toxicol Lett ; 235(1): 17-27, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25791630

ABSTRACT

Overexposure to manganese (Mn) has been known to induce neuronal death and neurodegenerative symptoms. However, the precise mechanisms underlying Mn neurotoxicity remain incompletely understood. In the present study, we established a Mn-exposed rat model and found that downregulation of wild type p53-induced phosphatase 1 (Wip1) might contribute to p53 activation and resultant neuronal apoptosis following Mn exposure. Western blot and immunohistochemical analyses revealed that the expression of Wip1 was markedly decreased following Mn exposure. In addition, immunofluorescence assay demonstrated that Mn exposure led to significant reduction in the number of Wip1-positive neurons. Accordingly, the expression of Mdm2 was progressively decreased, which was accompanied with markedly increased expression of p53, as well as the ratio of Bax/Bcl-xl. Furthermore, we showed that Mn exposure decreased the viability and induced apparent apoptosis in NFG-differentiated neuron-like PC12 cells. Importantly, the expression of Wip1 decreased progressively, whereas the level of cellular p53 and the ratio of Bax/Bcl-xl were elevated, which resembled the expression of the proteins in animal model studies. Depletion of p53 significantly ameliorated Mn-mediated cytotoxic effect in PC12 cells. In addition, ectopic expression of Wip1 attenuated Mn-induced p53 signaling as well as apoptosis in PC12 cells. Finally, we observed that depletion of Wip1 augmented Mn-induced apoptosis in PC12 cells. Collectively, these findings suggest that downregulated Wip1 expression plays an important role in Mn-induced neuronal death in the brain striatum via the modulation of p53 signaling.


Subject(s)
Apoptosis , Basal Ganglia/enzymology , Manganese Poisoning/enzymology , Neurons/enzymology , Phosphoprotein Phosphatases/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Basal Ganglia/pathology , Chlorides , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Manganese Compounds , Manganese Poisoning/etiology , Manganese Poisoning/genetics , Manganese Poisoning/pathology , Nerve Degeneration , Neurons/drug effects , Neurons/pathology , PC12 Cells , Phosphoprotein Phosphatases/genetics , Protein Phosphatase 2C , Proto-Oncogene Proteins c-mdm2/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transfection , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
8.
Neurotoxicology ; 38: 51-60, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23721876

ABSTRACT

Exposure to high levels of manganese (Mn) results in a neurological condition termed manganism, which is characterized by oxidative stress, abnormal dopamine (DA) signaling, and cell death. Epidemiological evidence suggests correlations with occupational exposure to Mn and the development of the movement disorder Parkinson's disease (PD), yet the molecular determinants common between the diseases are ill-defined. Glutathione S-transferases (GSTs) of the class pi (GSTπ) are phase II detoxification enzymes that conjugate both endogenous and exogenous compounds to glutathione to reduce cellular oxidative stress, and their decreased expression has recently been implicated in PD progression. In this study we demonstrate that a Caenorhabditis elegans GSTπ homologue, GST-1, inhibits Mn-induced DA neuron degeneration. We show that GST-1 is expressed in DA neurons, Mn induces GST-1 gene and protein expression, and GST-1-mediated neuroprotection is dependent on the PD-associated transcription factor Nrf2/SKN-1, as a reduction in SKN-1 gene expression results in a decrease in GST-1 protein expression and an increase in DA neuronal death. Furthermore, decreases in gene expression of the SKN-1 inhibitor WDR-23 or the GSTπ-binding cell death activator JNK/JNK-1 result in an increase in resistance to the metal. Finally, we show that the Mn-induced DA neuron degeneration is independent of the dopamine transporter DAT, but is largely dependent on the caspases CED-3 and the novel caspase CSP-1. This study identifies a C. elegans Nrf2/SKN-1-dependent GSTπ homologue, cell death effectors of GSTπ-associated xenobiotic-induced pathology, and provides the first in vivo evidence that a phase II detoxification enzyme may modulate DA neuron vulnerability in manganism.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , DNA-Binding Proteins/metabolism , Dopaminergic Neurons/enzymology , Dopaminergic Neurons/pathology , Glutathione S-Transferase pi/metabolism , Manganese Poisoning/enzymology , Manganese Poisoning/pathology , Nerve Degeneration/prevention & control , Transcription Factors/metabolism , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/enzymology , Caenorhabditis elegans Proteins/genetics , Caspases/genetics , Caspases/metabolism , Disease Models, Animal , Dopamine Plasma Membrane Transport Proteins/genetics , Gene Expression , Glutathione S-Transferase pi/biosynthesis , Glutathione S-Transferase pi/genetics , Mitogen-Activated Protein Kinases/genetics , Repressor Proteins/genetics
9.
Neurotoxicology ; 33(4): 697-702, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22285144

ABSTRACT

INTRODUCTION: A higher prevalence of individuals affected by Parkinsonism was found in Valcamonica, Italy. This may be related to ferro-alloy smelters in the area, releasing manganese (Mn) in the air, soil and water for about a century. There exists individual susceptibility for Mn neurotoxicity. AIM: To analyse how polymorphism in genes regulating Mn metabolism and toxicity can modify neurophysiological effects of Mn exposure. MATERIALS AND METHODS: Elderly (N=255) and adolescents (N=311) from Northern Italy were examined for neuromotor and olfactory functions. Exposure to Mn was assessed in blood and urine by atomic absorption spectroscopy and in soil by a portable instrument based on X-Ray fluorescence technology. Polymorphisms in the Parkinson-related gene ATPase type 13A2 (ATP13A2, also called PARK9: rs3738815, rs2076602, rs4920608, rs2871776 and rs2076600), and in the secretory pathway Ca(2+)/Mn(2+) ATPase isoform 1 gene (SPCA1: rs218498, rs3773814 and rs2669858) were analysed by TaqMan probes. RESULTS: For both adolescents and elderly, negative correlations between Mn in soil and motor coordination (R(s)=-0.20, p<0.001; R(s)=-0.13, p=0.05, respectively) were demonstrated. Also among adolescents, negative correlations were seen between Mn in soil with odor identification (R(s)=-0.17, p<0.01). No associations were seen for Mn in blood or urine. ATP13A2 polymorphisms rs4920608 and rs2871776 significantly modified the effects of Mn exposure on impaired motor coordination in elderly (p for interaction=0.029, p=0.041, respectively), also after adjustments for age and gender. The rs2871776 altered a binding site for transcription factor insulinoma-associated 1. CONCLUSIONS: ATP13A2 variation may be a risk marker for neurotoxic effects of Mn in humans.


Subject(s)
Environmental Exposure/adverse effects , Iron/adverse effects , Manganese Poisoning/genetics , Manganese/adverse effects , Parkinson Disease, Secondary/genetics , Proton-Translocating ATPases/genetics , Soil Pollutants/adverse effects , Adolescent , Age Factors , Aged , Aged, 80 and over , Chi-Square Distribution , Child , Cross-Sectional Studies , Environmental Monitoring , Female , Gene-Environment Interaction , Genetic Predisposition to Disease , Humans , Iron/blood , Iron/urine , Italy , Linear Models , Male , Manganese/blood , Manganese/urine , Manganese Poisoning/blood , Manganese Poisoning/diagnosis , Manganese Poisoning/enzymology , Manganese Poisoning/physiopathology , Manganese Poisoning/urine , Middle Aged , Motor Activity/drug effects , Motor Activity/genetics , Multivariate Analysis , Neuropsychological Tests , Olfaction Disorders/chemically induced , Olfaction Disorders/physiopathology , Parkinson Disease, Secondary/blood , Parkinson Disease, Secondary/diagnosis , Parkinson Disease, Secondary/enzymology , Parkinson Disease, Secondary/physiopathology , Parkinson Disease, Secondary/urine , Phenotype , Polymerase Chain Reaction , Proton-Translocating ATPases/metabolism , Residence Characteristics , Risk Assessment , Risk Factors , Smell/drug effects , Smell/genetics , Soil/chemistry , Soil Pollutants/blood , Soil Pollutants/urine , Spectrometry, X-Ray Emission , Spectrophotometry, Atomic
10.
Basic Clin Pharmacol Toxicol ; 103(2): 171-5, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18816301

ABSTRACT

Manganese (Mn) is an essential metalloenzyme component that in high doses can exert serious oxidative and neurotoxic effects. The aim of this study was to investigate the potential effect of the antioxidant L-cysteine (Cys, 7 mg/kg) on the adult rat brain total antioxidant status (TAS) and the activities of acetylcholinesterase (AChE), Na+,K+-ATPase and Mg2+-ATPase induced by short-term Mn administration (as Mn chloride, 50 mg/kg). Twenty-eight male Wistar rats were divided into four groups: A (saline-treated control), B (Mn), C (Cys) and D (Mn and Cys). All rats were treated once daily, for 1 week with intraperitoneal injections of the tested compounds. Rats were killed by decapitation and mentioned parameters were measured spectrophotometrically. Rats treated with Mn exhibited a significant reduction in brain TAS (-39%, P < 0.001, B versus A) that was partially reversed by Cys co-administration (-13%, P < 0.01, D versus A), while Cys (group C) had no effect on TAS. The rat brain AChE activity was found significantly increased by both Mn (+21%, P < 0.001, B versus A) and Cys (+61%, P < 0.001, C versus A), while it was adjusted into the control levels by the co-administration of Mn and Cys. The activity of rat brain Na+,K+-ATPase was not affected by Mn administration, while Mg2+-ATPase exhibited a slight but statistically significant reduction in its activity (-9%, P < 0.01, B versus A) due to Mn, which was further reduced by Cys co-administration. The above findings suggest that short-term Mn in vivo administration causes a statistically significant decrease in the rat brain TAS and an increase in AChE activity. Both effects can be, partially or totally, reversed into the control levels by Cys co-administration (which could thus be considered for future applications as a neuroprotective agent against chronic exposure to Mn and the treatment of manganism). The activity of Na+,K+-ATPase is not affected by Mn, while Mg2+-ATPase activity is slightly (but significantly) inhibited by Mn, possibly due to Mg replacement.


Subject(s)
Acetylcholinesterase/metabolism , Antioxidants/metabolism , Brain/drug effects , Ca(2+) Mg(2+)-ATPase/metabolism , Chlorides/toxicity , Cysteine/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Brain/enzymology , Male , Manganese Compounds , Manganese Poisoning/enzymology , Manganese Poisoning/etiology , Manganese Poisoning/prevention & control , Rats , Rats, Wistar , Time Factors
11.
Neuroimage ; 39(2): 669-79, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-17949999

ABSTRACT

Noncystic periventricular leukomalacia (PVL) in premature infants is becoming a predominant lesion form. However, detection of the gray matter (GM) lesions involved in this disease, which closely relate to the later cognitive and behavioral deficits, is challenging because of their subtle and transient nature observed by conventional MRI. This study evaluated manganese-enhanced MRI (MEMRI) for detecting such GM lesions in 7-day-old rats with mild hypoxic-ischemic (H-I) insult, a characteristic model of noncystic PVL. Group 1 (n=6) and Group 2 (n=8) were administered intraperitoneally with MnCl(2) at hour 3 and day 7 after H-I insult, respectively. Control Group (n=6) received no MnCl(2). T1-, T2- and diffusion-weighted imaging (T1WI, T2WI and DWI, respectively) was performed. Animals were sacrificed for H&E staining, and immunohistochemical staining for glutamine synthetase (GS) and Mn-superoxide dismutase (Mn-SOD), which are two Mn-binding enzymes against glutamate toxicity and oxidative stress respectively in neurodegeneration. In Control Group, MRI appearance of H-I lesions normalized by day 7 after H-I insult. In Group 1, MEMRI provided the enhanced and prolonged GM lesion detection from day 3 up to day 21. In Group 2, similar Mn enhancement was observed, enabling day 8 detection of GM lesions that were invisible before Mn injection at day 7. These in vivo Mn-induced GM lesion enhancements were found to correlate with increased immunoactivities of GS and Mn-SOD. These findings suggest the potential utility of MEMRI in detecting the GM lesions that are otherwise undetectable using the conventional MRI techniques in late phase of mild H-I injury.


Subject(s)
Cerebral Cortex/pathology , Hypoxia-Ischemia, Brain/pathology , Magnetic Resonance Imaging/methods , Manganese Poisoning/pathology , Animals , Animals, Newborn , Brain Mapping , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Data Interpretation, Statistical , Female , Glutamate-Ammonia Ligase/metabolism , Glutamic Acid/toxicity , Hypoxia-Ischemia, Brain/enzymology , Hypoxia-Ischemia, Brain/metabolism , Image Processing, Computer-Assisted , Manganese Poisoning/enzymology , Manganese Poisoning/metabolism , Oxidative Stress/physiology , Pregnancy , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL