Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Exp Cell Res ; 429(1): 113656, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37245583

ABSTRACT

In this study, we investigated the effect of doxorubicin and tunicamycin treatment alone or in combination on MDM-, Cul9-and prion protein (PrP)-mediated subcellular regulation of p53 in the context of apoptosis and autophagy. MTT analysis was performed to determine the cytotoxic effect of the agents. Apoptosis was monitorized by ELISA, flow cytometry and JC-1 assay. Monodansylcadaverine assay was performed for autophagy. Western blotting and immunofluorescence were performed to determine p53, MDM2, CUL9 and PrP levels. Doxorubicin increased p53, MDM2 and CUL9 levels in a dose-dependent manner. Expression of p53 and MDM2 was higher at the 0.25 µM concentration of tunicamycin compared to the control, but it decreased at 0.5 µM and 1 µM concentrations. CUL9 expression was significantly decreased only after treatment of tunicamycin at 0.25 µM. According to its glycosylation status, the upper band of PrP increased only in combination treatment. In combination treatment, p53 expression was higher than control, whereas MDM2 and CUL9 expressions were decreased. Combination treatments may make MCF-7 cells more susceptible to apoptosis rather than autophagy. In conclusion, PrP may be important in determining the fate of cell death through crosstalk between proteins such as p53 and MDM2 under endoplasmic reticulum (ER) stress conditions. Further studies are needed to obtain in-depth information on these potential molecular networks.


Subject(s)
Breast Neoplasms , Prions , Humans , Female , Prion Proteins/genetics , Prion Proteins/metabolism , Prion Proteins/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tunicamycin/pharmacology , MCF-7 Cells , Breast Neoplasms/drug therapy , Doxorubicin/pharmacology , Endoplasmic Reticulum Stress , Prions/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Apoptosis , Cell Line, Tumor
2.
Curr Issues Mol Biol ; 37: 21-32, 2020.
Article in English | MEDLINE | ID: mdl-31814573

ABSTRACT

Worldwide spread of influenza A virus (IAV) strains, which are resistant to currently available anti- influenza agents such as viral neuraminidase inhibitors, has encouraged identification of new target molecules for anti-influenza agents. Reactive oxygen species (ROS) causing oxidative stress play a pivotal role in the pathogenesis of lung injuries induced by infection with IAVs, therefore suggesting that anti-oxidative therapeutics targeting cellular molecules could be beneficial against IAV infection without inducing drug-resistant IAV strains. We recently found that the normal cellular prion protein, PrPC, whose conformational conversion into the amyloidogenic isoform, PrPSc, in the brain is a key pathogenic event in prion diseases, is expressed by lung epithelial cells and exerts a protective role against IAV infection in mice by reducing ROS in infected lungs. The Cu content and activity of anti- oxidative enzyme Cu/Zn-superoxide dismutase, or SOD1, were lower in the lungs of PrPC-knockout mice, suggesting that the anti-oxidative activity of PrPC is probably attributable to its function of activating SOD1 through regulating Cu content in lungs. Here, we introduce PrPC as a novel modulator of influenza and its potential implication for anti-oxidative therapies for IAV infection. We also introduce other candidate targets reported for anti- oxidative anti-influenza therapies.


Subject(s)
Antiviral Agents/pharmacology , Influenza A virus/drug effects , Influenza A virus/physiology , Influenza, Human/virology , Prion Proteins/pharmacology , Animals , Antioxidants/pharmacology , Antiviral Agents/therapeutic use , Host-Pathogen Interactions , Humans , Influenza, Human/drug therapy , Influenza, Human/genetics , Influenza, Human/metabolism , Oxidation-Reduction/drug effects , Protective Agents/pharmacology , Reactive Oxygen Species/metabolism , Virus Replication/drug effects
3.
PLoS Pathog ; 14(5): e1007049, 2018 05.
Article in English | MEDLINE | ID: mdl-29723291

ABSTRACT

The cellular prion protein, designated PrPC, is a membrane glycoprotein expressed abundantly in brains and to a lesser extent in other tissues. Conformational conversion of PrPC into the amyloidogenic isoform is a key pathogenic event in prion diseases. However, the physiological functions of PrPC remain largely unknown, particularly in non-neuronal tissues. Here, we show that PrPC is expressed in lung epithelial cells, including alveolar type 1 and 2 cells and bronchiolar Clara cells. Compared with wild-type (WT) mice, PrPC-null mice (Prnp0/0) were highly susceptible to influenza A viruses (IAVs), with higher mortality. Infected Prnp0/0 lungs were severely injured, with higher inflammation and higher apoptosis of epithelial cells, and contained higher reactive oxygen species (ROS) than control WT lungs. Treatment with a ROS scavenger or an inhibitor of xanthine oxidase (XO), a major ROS-generating enzyme in IAV-infected lungs, rescued Prnp0/0 mice from the lethal infection with IAV. Moreover, Prnp0/0 mice transgenic for PrP with a deletion of the Cu-binding octapeptide repeat (OR) region, Tg(PrPΔOR)/Prnp0/0 mice, were also highly susceptible to IAV infection. These results indicate that PrPC has a protective role against lethal infection with IAVs through the Cu-binding OR region by reducing ROS in infected lungs. Cu content and the activity of anti-oxidant enzyme Cu/Zn-dependent superoxide dismutase, SOD1, were lower in Prnp0/0 and Tg(PrPΔOR)/Prnp0/0 lungs than in WT lungs. It is thus conceivable that PrPC functions to maintain Cu content and regulate SOD1 through the OR region in lungs, thereby reducing ROS in IAV-infected lungs and eventually protecting them from lethal infection with IAVs. Our current results highlight the role of PrPC in protection against IAV infection, and suggest that PrPC might be a novel target molecule for anti-influenza therapeutics.


Subject(s)
PrPC Proteins/metabolism , Prion Proteins/metabolism , Animals , Brain/pathology , Copper/metabolism , Disease Susceptibility/metabolism , Influenza A virus/metabolism , Influenza A virus/pathogenicity , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , PrPC Proteins/physiology , Prion Diseases/metabolism , Prion Proteins/pharmacology , Prions/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
4.
J Immunol ; 199(1): 224-232, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28533442

ABSTRACT

HIV-1 enters the CNS soon after peripheral infection and causes chronic neuroinflammation and neuronal damage that leads to cognitive impairment in 40-70% of HIV-infected people. The nonpathogenic cellular isoform of the human prion protein (PrPc) is an adhesion molecule constitutively expressed in the CNS. Previously, our laboratory showed that shed PrPc (sPrPc) is increased in the cerebrospinal fluid of HIV-infected people with cognitive deficits as compared with infected people with no impairment. In this article, we demonstrate that CCL2 and TNF-α, inflammatory mediators that are elevated in the CNS of HIV-infected people, increase shedding of PrPc from human astrocytes by increasing the active form of the metalloprotease ADAM10. We show that the consequence of this shedding can be the production of inflammatory mediators, because treatment of astrocytes with rPrPc increased secretion of CCL2, CXCL-12, and IL-8. Supernatants from rPrPc-treated astrocytes containing factors produced in response to this treatment, but not rPrPc by itself, cause increased chemotaxis of both uninfected and HIV-infected human monocytes, suggesting a role for sPrPc in monocyte recruitment into the brain. Furthermore, we examined whether PrPc participates in glutamate uptake and found that rPrPc decreased uptake of this metabolite in astrocytes, which could lead to neurotoxicity and neuronal loss. Collectively, our data characterize mediators involved in PrPc shedding and the effect of this sPrPc on monocyte chemotaxis and glutamate uptake from astrocytes. We propose that shedding of PrPc could be a potential target for therapeutics to limit the cognitive impairment characteristic of neuroAIDS.


Subject(s)
Astrocytes/metabolism , Central Nervous System/physiopathology , HIV Infections/physiopathology , HIV/physiology , Monocytes/immunology , Monocytes/virology , Prion Proteins/metabolism , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Astrocytes/drug effects , Astrocytes/immunology , Cells, Cultured , Central Nervous System/virology , Chemokine CCL2/metabolism , Chemokine CCL2/pharmacology , Chemokine CXCL1/immunology , Chemokine CXCL1/metabolism , Chemotaxis, Leukocyte , Dipeptides/pharmacology , HIV/immunology , HIV Infections/virology , Humans , Hydroxamic Acids/pharmacology , Interleukin-8/immunology , Interleukin-8/metabolism , Membrane Proteins/metabolism , Prion Proteins/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
5.
Neurobiol Dis ; 91: 124-131, 2016 07.
Article in English | MEDLINE | ID: mdl-26949218

ABSTRACT

The pathogenic process in Alzheimer's disease (AD) appears to be closely linked to the neurotoxic action of amyloid-ß (Aß) oligomers. Recent studies have shown that these oligomers bind with high affinity to the membrane-anchored cellular prion protein (PrP(C)). It has also been proposed that this binding might mediate some of the toxic effects of the oligomers. Here, we show that the soluble (membrane anchor-free) recombinant human prion protein (rPrP) and its N-terminal fragment N1 block Aß oligomers-induced inhibition of long-term potentiation (LTP) in hippocampal slices, an important surrogate marker of cognitive deficit associated with AD. rPrP and N1 are also strikingly potent inhibitors of Aß cytotoxicity in primary hippocampal neurons. Furthermore, experiments using hippocampal slices and neurons from wild-type and PrP(C) null mice (as well as rat neurons in which PrP(C) expression was greatly reduced by gene silencing) indicate that, in contrast to the impairment of synaptic plasticity by Aß oligomers, the cytotoxic effects of these oligomers, and the inhibition of these effects by rPrP and N1, are independent of the presence of endogenous PrP(C). This suggests fundamentally different mechanisms by which soluble rPrP and its fragments inhibit these two toxic responses to Aß. Overall, these findings provide strong support to recent suggestions that PrP-based compounds may offer new avenues for pharmacological intervention in AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Long-Term Potentiation/drug effects , Neuronal Plasticity/physiology , Prion Proteins/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Animals , Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Long-Term Potentiation/physiology , Neurons/drug effects , Neurons/metabolism , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Prion Proteins/metabolism , Rats, Sprague-Dawley
6.
Pharmacol Res ; 113(Pt A): 500-514, 2016 11.
Article in English | MEDLINE | ID: mdl-27667770

ABSTRACT

We tested the efficacy of novel cyclooxygenase 2 (COX-2) inhibitors in counteracting glia-driven neuroinflammation induced by the amyloidogenic prion protein fragment PrP90-231 or lipopolysaccharide (LPS). In search for molecules with higher efficacy than celecoxib, we focused our study on its 2,3-diaryl-1,3-thiazolidin-4-one analogues. As experimental models, we used the immortalized microglial cell line N9, rat purified microglial primary cultures, and mixed cultures of astrocytes and microglia. Microglia activation in response to PrP90-231 or LPS was characterized by growth arrest, morphology changes and the production of reactive oxygen species (ROS). Moreover, PrP90-231 treatment caused the overexpression of the inducible nitric oxide synthase (iNOS) and COX-2, with the consequent nitric oxide (NO), and prostaglandin E2 (PGE2) accumulation. These effects were challenged by different celecoxib analogues, among which Q22 (3-[4-(sulfamoyl)phenyl]-2-(4-tolyl)thiazolidin-4-one) inhibited microglia activation more efficiently than celecoxib, lowering both iNOS and COX-2 activity and reducing ROS release. During neurodegenerative diseases, neuroinflammation induced by amyloidogenic peptides causes the activation of both astrocytes and microglia with these cell populations mutually regulating each other. Thus the effects of PrP90-231 and LPS were also studied on mixed glial cultures containing astrocytes and microglia. PrP90-231 treatment elicited different responses in the co-cultures induced astrocyte proliferation and microglia growth arrest, resulting in a differential ability to release proinflammatory molecules with the production of NO and ROS mainly attributable on microglia, while COX-2 expression was induced also in astrocytes. Q22 effects on both NO and PGE2 secretion were more significant in the mixed glial cultures than in purified microglia, demonstrating Q22 ability to revert the functional interaction between astrocytes and microglia. These results demonstrate that Q22 is a powerful drug able to revert glial neuroinflammatory responses and might represent a lead to explore the chemical space around celecoxib frameworks to design even more effective agents, paving the way to novel approaches to contrast the neuroinflammation-dependent toxicity.


Subject(s)
Celecoxib/pharmacology , Dinoprostone/metabolism , Inflammation/drug therapy , Lipopolysaccharides/pharmacology , Neuroglia/drug effects , Nitric Oxide/metabolism , Prion Proteins/pharmacology , Reactive Oxygen Species/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cell Line , Cell Proliferation/drug effects , Cyclooxygenase 2/metabolism , Inflammation/metabolism , Mice , Microglia/drug effects , Microglia/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neuroglia/metabolism , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Sprague-Dawley
7.
Virus Res ; 338: 199249, 2023 12.
Article in English | MEDLINE | ID: mdl-37858731

ABSTRACT

Flaviviruses are a major cause of viral diseases worldwide, for which effective treatments have yet to be discovered. The prion protein (PrPc) is abundantly expressed in brain cells and has been shown to play a variety of roles, including neuroprotection, cell homeostasis, and regulation of cellular signaling. However, it is still unclear whether PrPc can protect against flaviviruses. In this study, we investigated the role of PrPc in regulating autophagy flux and its potential antiviral activity during Japanese encephalitis virus (JEV) infection. Our in vivo experiment showed that JEV was more lethal to the PrPc knocked out mice which was further supported by histological analysis, western blot and rtPCR results from infected mice brain samples. Role of PrPc against viral propagation in vitro was verified through cell survival study, protein expression and RNA replication analysis, and adenoviral vector assay by overexpressing PrPc. Further analysis indicated that after virus entry, PrPc inhibited autophagic flux that prevented JEV replication inside the host cell. Our results from in vivo and in vitro investigations demonstrate that prion protein effectively inhibited JEV propagation by regulating autophagy flux which is used by JEV to release its genetic material and replication after entering the host cell, suggesting that prion protein may be a promising therapeutic target for flavivirus infection.


Subject(s)
Encephalitis Virus, Japanese , Encephalitis, Japanese , Animals , Mice , Prion Proteins/genetics , Prion Proteins/pharmacology , Cell Line , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Virus Replication
8.
JCI Insight ; 7(6)2022 03 22.
Article in English | MEDLINE | ID: mdl-35133987

ABSTRACT

Prion protein (PrP) concentration controls the kinetics of prion replication and is a genetically and pharmacologically validated therapeutic target for prion disease. In order to evaluate PrP concentration as a pharmacodynamic biomarker and assess its contribution to known prion disease risk factors, we developed and validated a plate-based immunoassay reactive for PrP across 6 species of interest and applicable to brain and cerebrospinal fluid (CSF). PrP concentration varied dramatically across different brain regions in mice, cynomolgus macaques, and humans. PrP expression did not appear to contribute to the known risk factors of age, sex, or common PRNP genetic variants. CSF PrP was lowered in the presence of rare pathogenic PRNP variants, with heterozygous carriers of P102L displaying 55%, and D178N just 31%, of the CSF PrP concentration of mutation-negative controls. In rodents, pharmacologic reduction of brain Prnp RNA was reflected in brain parenchyma PrP and, in turn in CSF PrP, validating CSF as a sampling compartment for the effect of PrP-lowering therapy. Our findings support the use of CSF PrP as a pharmacodynamic biomarker for PrP-lowering drugs and suggest that relative reduction from individual baseline CSF PrP concentration may be an appropriate marker for target engagement.


Subject(s)
Prion Diseases , Prion Proteins , Prions , Animals , Biomarkers/cerebrospinal fluid , Genotype , Humans , Mice , Prion Diseases/diagnosis , Prion Diseases/drug therapy , Prion Proteins/cerebrospinal fluid , Prion Proteins/genetics , Prion Proteins/pharmacology , Prions/genetics , Prions/metabolism
9.
Behav Brain Res ; 419: 113680, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34822947

ABSTRACT

Conversion of the cellular prion protein (PrPC) into the scrapie form (PrPSc) is the leading step to the development of transmissible spongiform encephalopathies (TSEs), still incurable neurodegenerative disorders. Interaction of PrPC with cellular and synthetic ligands that induce formation of scrapie-like conformations has been deeply investigated in vitro. Different nucleic acid (NA) sequences bind PrP and convert it to ß-sheet-rich or unfolded species; among such NAs, a 21-mer double-stranded DNA, D67, was shown to induce formation of PrP aggregates that were cytotoxic. However, in vivo effects of these PrP-DNA complexes were not explored. Herein, aggregates of recombinant full-length PrP (rPrP23-231) induced by interaction with the D67 aptamer were inoculated into the lateral ventricle of Swiss mice and acute effects were investigated. The aggregates had no influence on emotional, locomotor and motor behavior of mice. In contrast, mice developed cognitive impairment and hippocampal synapse loss, which was accompanied by intense activation of glial cells in this brain region. Our results suggest that the i.c.v. injection of rPrP:D67 aggregates is an interesting model to study the neurotoxicity of aggregated PrP in vivo, and that glial cell activation may be an important step for behavioral and cognitive dysfunction in prion diseases.


Subject(s)
Aptamers, Nucleotide/pharmacology , Behavior, Animal/drug effects , Cognitive Dysfunction/chemically induced , Hippocampus/drug effects , Prion Proteins/pharmacology , Synapses/drug effects , Animals , Disease Models, Animal , Lateral Ventricles/drug effects , Male , Mice
11.
Sci Rep ; 10(1): 6654, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32313035

ABSTRACT

Microglia act as the protective immune cell of the brain. By surveying the tissue to identify and rectify problems, they function to maintain the health of brain cells. The prion protein N-terminal cleavage fragment, N1, has demonstrated neuroprotective activities in vitro and in vivo. This study aimed to elucidate whether N1 could modulate microglial function and, if so, determine the consequences for the surrounding tissue. Using a mixed neuronal lineage and microglia co-culture system, we showed that N1 stimulation changed overall morphology and metabolism, suggesting enhanced cellular viability. Furthermore, N1 induced an increase in Cxcl10 secretion in the co-cultures. Recombinant Cxcl10, administered exogenously, mediated the changes in the mixed neuronal lineage culture morphology and metabolism in the absence of microglia, but no effect of Cxcl10 was observed on microglia cultured on their own. Direct cell-to-cell contact was required for N1 to influence microglia in the co-cultures, and this was linked with restructuring of microglial membrane composition to include a higher GM1 content at interaction sites with surrounding cells. Our findings show that N1 can play a regulatory role in microglial function in the context of an inter-connected network of cells by changing both cellular interaction sites and cytokine secretion.


Subject(s)
Microglia/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , Prion Proteins/pharmacology , Animals , Brain/cytology , Brain/metabolism , Cell Communication/drug effects , Cell Differentiation , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Coculture Techniques , G(M1) Ganglioside/metabolism , Gene Expression/drug effects , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Knockout , Microglia/cytology , Microglia/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Primary Cell Culture , Prion Proteins/chemistry , Prion Proteins/metabolism , Prions/chemistry , Prions/metabolism , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , Single-Cell Analysis , Red Fluorescent Protein
12.
Brain Res ; 1710: 125-135, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30593771

ABSTRACT

BACKGROUND: The prion protein (PrP) is known to bind certain soluble aggregates of the amyloid ß-protein (Aß), and two regions of PrP, one centered around residues 19-33, and the other around 87-112, are thought to be particularly important for this interaction. When either of these sequences are grafted into a human IgG the resulting antibodies react with disease-associated PrP conformers, whereas the parental b12 IgG does not. METHODS: Human antibodies containing grafts of PrP 19-33 or 87-112 were prepared as before (Solforosi et al., 2007) and tested for their ability to recognize synthetic and Alzheimer's disease (AD) brain-derived Aß. Since aqueous extracts of AD brain contain a complex mixture of active and inactive Aß species, we also assessed whether PrP-grafted antibodies could protect against neuritotoxicity mediated by AD brain-derived Aß. For these experiments, human iPSC-derived neurons were grown in 96-well plates at 5000 cells per well and on post-induction day 21, AD brain extracts were added +/- test antibodies. Neurons were imaged for 3 days using an IncuCyte live-cell imaging system, and neurite number and density quantified. RESULTS: Grafted antibodies bound a significant portion of aggregated Aß in aqueous AD extracts, but when these antibodies were co-incubated with neurons treated with brain extracts they did not reduce toxicity. By contrast, the PrP fragment N1 did protect against Aß. CONCLUSIONS: These results further demonstrate that not all Aß oligomers are toxic and suggest that PrP derivatives may allow development of agents that differentially recognize toxic and innocuous Aß aggregates.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/pharmacology , Antibodies/pharmacology , Brain/metabolism , Prion Proteins/pharmacology , Protein Aggregation, Pathological/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Antibodies/genetics , Antibodies/metabolism , Brain/drug effects , Brain/pathology , Female , Humans , Mice, Inbred C57BL , Middle Aged , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Prion Proteins/genetics , Prion Proteins/metabolism , Protein Binding
13.
J Trace Elem Med Biol ; 39: 50-53, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27908423

ABSTRACT

In Creutzfeldt Jakob, Alzheimer and Parkinson diseases, copper metalloproteins such as prion, amyloid protein precursor and α-synuclein are able to protect against free radicals by reduction from cupric Cu+2 to cupreous Cu+. In these pathologies, a regional copper (Cu) brain decrease correlated with an iron, zinc or manganese (Mn) increase has previously been observed, leading to local neuronal death and abnormal deposition of these metalloproteins in ß-sheet structures. In this study we demonstrate the protective effect of Cu metalloproteins against deleterious free-radical effects. With neuroblastoma SH-SY5Y cell cultures, we show that bovine brain prion protein in Cu but not Mn form prevents free radical-induced neuronal death. The survival ratio of SH-SY5Y cells has been measured after UV irradiation (free radical production), when the incubating medium is supplemented with bovine brain homogenate in native, Cu or Mn forms. This ratio, about 28% without any addition or with bovine brain protein added in Mn form, increases by as much as 54.73% with addition to the culture medium of native bovine brain protein and by as much as 95.95% if the addition is carried out in cupric form. This protective effect of brain copper protein against free radical-induced neuronal death has been confirmed with Inductively Coupled Plasma Mass Spectrometry Mn and Cu measurement in bovine brain homogenates: respectively lower than detection limit and 9.01µg/g dry weight for native form; lower than detection limit and 825.85µg/g dry weight for Cu-supplemented form and 1.75 and 68.1µg/g dry weight in Mn-supplemented brain homogenate.


Subject(s)
Copper/metabolism , Neuroblastoma/pathology , Neurons/drug effects , Neurons/pathology , Prion Proteins/chemistry , Prion Proteins/metabolism , Animals , Cattle , Cell Death/drug effects , Cell Survival/drug effects , Copper/pharmacology , Free Radicals/antagonists & inhibitors , Free Radicals/chemistry , Free Radicals/pharmacology , Humans , Manganese/metabolism , Manganese/pharmacology , Prion Proteins/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL