Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 146: 112551, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34923336

ABSTRACT

This study tested the hypothesis that valsartan (Val) and melatonin (Mel)-assisted adipose-derived mesenchymal stem cells (ADMSCs) preserved the residual renal function in chronic kidney disease (CKD) rat through promoting cellular-prior-protein (PrPC) to upregulate PI3K/Akt/mTOR signaling and cell proliferation. In vitro study demonstrated that as compared with CKD-derived-ADMSCs, Val/Mel/overexpression of PrPC-treated CKD derived-ADMSCs significantly upregulated cell proliferation and protein expressions of PrPC and phosphorylated (p)-PI3K/p-Akt/p-mTOR, and downregulated oxidative stress (all p < 0.001). Rats (n = 42) were categorized into group 1 (sham-operated-control), group 2 (CKD), group 3 (CKD + ADMSCs/1.2 ×106 cells) + Mel/20 mg/kg/day), group 4 (CKD + siRNA-PrPC-ADMSCs/1.2 ×106 cells), group 5 (CKD + ADMSCs/1.2 ×106 cells + Val/20 mg/kg/day) and group 6 (CKD + Val + Mel). By day 35, the kidney specimens were harvested and the result showed that the protein expression of PrPC was highest in group 1, lowest in groups 2/4 and significantly lower in group 6 than in groups 3/5, but it was similar in groups 3/5 (all p < 0.0001). The protein expressions of cell-stress-signaling (p-PI3K/p-Akt/p-mTOR) and cell-cycle activity (cyclin-D1/clyclin-E2/Cdk2/Cdk4) exhibited an identical pattern, whereas the protein expressions of oxidative-stress (NOX-1/NOX-2)/mitochondrial fission (PINK1/DRP1)/apoptosis (cleaved-capsase3/cleaved-PARP) and fibrosis (TFG-ß/Smad3) as well as creatinine/BUN levels, ratio of urine-protein to urine-creatine and kidney-injured score exhibited an opposite pattern of PrPC among the groups (all p < 0.0001). In conclusion, Mel/Val facilitated-ADMSCs preserved renal architecture and function in CKD rat through promoting PrPC to regulate the cell proliferation/oxidative-stress/cell-stress signalings.


Subject(s)
Melatonin/pharmacology , Mesenchymal Stem Cells/metabolism , Renal Insufficiency, Chronic/pathology , Valsartan/pharmacology , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Kidney/drug effects , Male , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Prion Proteins/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/drug effects , Up-Regulation
2.
J Biol Chem ; 297(3): 101073, 2021 09.
Article in English | MEDLINE | ID: mdl-34390689

ABSTRACT

The study of prions and the discovery of candidate therapeutics for prion disease have been facilitated by the ability of prions to replicate in cultured cells. Paradigms in which prion proteins from different species are expressed in cells with low or no expression of endogenous prion protein (PrP) have expanded the range of prion strains that can be propagated. In these systems, cells stably expressing a PrP of interest are typically generated via coexpression of a selectable marker and treatment with an antibiotic. Here, we report the unexpected discovery that the aminoglycoside G418 (Geneticin) interferes with the ability of stably transfected cultured cells to become infected with prions. In G418-resistant lines of N2a or CAD5 cells, the presence of G418 reduced levels of protease-resistant PrP following challenge with the RML or 22L strains of mouse prions. G418 also interfered with the infection of cells expressing hamster PrP with the 263K strain of hamster prions. Interestingly, G418 had minimal to no effect on protease-resistant PrP levels in cells with established prion infection, arguing that G418 selectively interferes with de novo prion infection. As G418 treatment had no discernible effect on cellular PrP levels or its localization, this suggests that G418 may specifically target prion assemblies or processes involved in the earliest stages of prion infection.


Subject(s)
Gentamicins/pharmacology , Prion Proteins/drug effects , Prions/antagonists & inhibitors , Aminoglycosides/metabolism , Aminoglycosides/pharmacology , Animals , Cell Line , Cell Line, Tumor , Gentamicins/metabolism , Mice , PrPC Proteins/drug effects , PrPC Proteins/metabolism , PrPSc Proteins/drug effects , PrPSc Proteins/metabolism , Prion Diseases/prevention & control , Prion Proteins/metabolism , Prions/metabolism , Protein Synthesis Inhibitors
3.
J Mol Med (Berl) ; 99(3): 383-402, 2021 03.
Article in English | MEDLINE | ID: mdl-33409554

ABSTRACT

Endoplasmic reticulum (ER) stress is a common threat to photoreceptors during the pathogenesis of chronic retinopathies and often results in irreversible visual impairment. 2,3,5,6-Tetramethylpyrazine (TMP), which possesses many beneficial pharmacological activities, is a potential drug that could be used to protect photoreceptors. In the present study, we found that the cellular growth rate of 661 W cells cultured under low glucose conditions was lower than that of control cells, while the G2/M phase of the cell cycle was longer. We further found that the mitochondrial membrane potential (ΔΨm) was lower and that ER stress factor expression was increased in 661 W cells cultured under low glucose conditions. TMP reversed these trends. Visual function and cell counts in the outer nuclear layer (ONL) were low and the TUNEL-positive rate in the ONL was high in a C3H mouse model of spontaneous retinal degeneration. Similarly, visual function was decreased, and the TUNEL-positive rate in the ONL was increased in fasted C57/BL6j mice compared with control mice. On the other hand, ER stress factor expression was found to be increased in the retinas of both mouse models, as shown by reverse transcription real-time PCR (RT-qPCR) and western blotting. TMP reversed the physiological and molecular biological variations observed in both mouse models, and ATF4 expression was enhanced again. Further investigation by using western blotting illustrated that the proportion of insoluble prion protein (PRP) versus soluble PRP was reduced both in vitro and in vivo. Taken together, these results suggest that TMP increased the functions of photoreceptors by alleviating ER stress in vitro and in vivo, and the intrinsic mechanism was the ATF4-mediated inhibition of PRP aggregation. TMP may potentially be used clinically as a therapeutic agent to attenuate the functional loss of photoreceptors during the pathogenesis of chronic retinopathies. KEY MESSAGES: • Already known: TMP is a beneficial drug mainly used in clinic to enhance organ functions, and the intrinsic mechanism is still worthy of exploring. • New in the study: We discovered that TMP ameliorated retinal photoreceptors function via ER stress alleviation, which was promoted by ATF4-mediated inhibition of PRP aggregation. • Application prospect: In prospective clinical practices, TMP may potentially be used in the clinic as a therapeutic agent to attenuate the photoreceptors functional reduction in chronic retinopathies.


Subject(s)
Activating Transcription Factor 4/physiology , Endoplasmic Reticulum Stress/drug effects , Prion Proteins/drug effects , Protein Aggregates/drug effects , Protein Aggregation, Pathological/prevention & control , Pyrazines/pharmacology , Retinal Cone Photoreceptor Cells/drug effects , Retinal Degeneration/prevention & control , Animals , Cell Line, Tumor , Disease Models, Animal , Drug Evaluation, Preclinical , Electroretinography , Eye Proteins/biosynthesis , Eye Proteins/genetics , Fasting , Female , Glucose/pharmacology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Prion Proteins/chemistry , Protein Aggregation, Pathological/metabolism , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/physiopathology , Single-Blind Method , Solubility , Specific Pathogen-Free Organisms , Transcription, Genetic/drug effects
4.
Int J Mol Sci ; 21(23)2020 Nov 24.
Article in English | MEDLINE | ID: mdl-33255444

ABSTRACT

The formation of amyloid fibrils is linked to multiple neurodegenerative disorders, including Alzheimer's and Parkinson's disease. Despite years of research and countless studies on the topic of such aggregate formation, as well as their resulting structure, the current knowledge is still fairly limited. One of the main aspects prohibiting effective aggregation tracking is the environment's effect on amyloid-specific dyes, namely thioflavin-T (ThT). Currently, there are only a few studies hinting at ionic strength being one of the factors that modulate the dye's binding affinity and fluorescence intensity. In this work we explore this effect under a range of ionic strength conditions, using insulin, lysozyme, mouse prion protein, and α-synuclein fibrils. We show that ionic strength is an extremely important factor affecting both the binding affinity, as well as the fluorescence intensity of ThT.


Subject(s)
Amyloid/drug effects , Benzothiazoles/pharmacology , Protein Aggregates/drug effects , Protein Aggregation, Pathological/prevention & control , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Benzothiazoles/chemistry , Binding Sites/drug effects , Fluorescence , Humans , Insulin/chemistry , Kinetics , Mice , Osmolar Concentration , Parkinson Disease/metabolism , Parkinson Disease/prevention & control , Prion Proteins/chemistry , Prion Proteins/drug effects , Protein Aggregation, Pathological/metabolism , Protein Binding/drug effects , alpha-Synuclein/chemistry , alpha-Synuclein/drug effects
5.
Neurotherapeutics ; 17(4): 1836-1849, 2020 10.
Article in English | MEDLINE | ID: mdl-32767031

ABSTRACT

The accumulation of abnormal prion protein (PrPSc) produced by the structure conversion of PrP (PrPC) in the brain induces prion disease. Although the conversion process of the protein is still not fully elucidated, it has been known that the intramolecular chemical bridging in the most fragile pocket of PrP, known as the "hot spot," stabilizes the structure of PrPC and inhibits the conversion process. Using our original structure-based drug discovery algorithm, we identified the low molecular weight compounds that predicted binding to the hot spot. NPR-130 and NPR-162 strongly bound to recombinant PrP in vitro, and fragment molecular orbital (FMO) analysis indicated that the high affinity of those candidates to the PrP is largely dependent on nonpolar interactions, such as van der Waals interactions. Those NPRs showed not only significant reduction of the PrPSc levels but also remarkable decrease of the number of aggresomes in persistently prion-infected cells. Intriguingly, treatment with those candidate compounds significantly prolonged the survival period of prion-infected mice and suppressed prion disease-specific pathological damage, such as vacuole degeneration, PrPSc accumulation, microgliosis, and astrogliosis in the brain, suggesting their possible clinical use. Our results indicate that in silico drug discovery using NUDE/DEGIMA may be widely useful to identify candidate compounds that effectively stabilize the protein.


Subject(s)
Computer Simulation , Disease Progression , Drug Discovery/methods , Prion Diseases/diagnosis , Prion Diseases/drug therapy , Prion Proteins/drug effects , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Humans , Male , Mice , Mice, Transgenic , Prion Diseases/genetics , Prion Proteins/genetics , Prion Proteins/metabolism , Protein Binding/physiology , Structure-Activity Relationship
7.
PLoS Pathog ; 15(12): e1008139, 2019 12.
Article in English | MEDLINE | ID: mdl-31815959

ABSTRACT

Prion or PrPSc is the proteinaceous infectious agent causing prion diseases in various mammalian species. Despite decades of research, the structural basis for PrPSc formation and prion infectivity remains elusive. To understand the role of the hydrophobic region in forming infectious prion at the molecular level, we report X-ray crystal structures of mouse (Mo) prion protein (PrP) (residues 89-230) in complex with a nanobody (Nb484). Using the recombinant prion propagation system, we show that the binding of Nb484 to the hydrophobic region of MoPrP efficiently inhibits the propagation of proteinase K resistant PrPSc and prion infectivity. In addition, when added to cultured mouse brain slices in high concentrations, Nb484 exhibits no neurotoxicity, which is drastically different from other neurotoxic anti-PrP antibodies, suggesting that the Nb484 can be a potential therapeutic agent against prion disease. In summary, our data provides the first structure-function evidence supporting a crucial role of the hydrophobic region of PrP in forming an infectious prion.


Subject(s)
PrPSc Proteins/chemistry , PrPSc Proteins/drug effects , Prion Proteins/chemistry , Prion Proteins/drug effects , Single-Domain Antibodies/pharmacology , Animals , Mice , Protein Conformation , Protein Domains/drug effects , Structure-Activity Relationship
8.
J Biol Inorg Chem ; 24(8): 1231-1244, 2019 12.
Article in English | MEDLINE | ID: mdl-31401689

ABSTRACT

Misfolded prion protein (PrPSc) is known for its role in fatal neurodegenerative conditions, such as Creutzfeldt-Jakob disease. PrP fragments and their mutants represent important tools in the investigation of the neurotoxic mechanisms and in the evaluation of new compounds that can interfere with the processes involved in neuronal death. Metal-catalyzed oxidation of PrP has been implicated as a trigger for the conformational changes in protein structure, which, in turn, lead to misfolding. Targeting redox-active biometals copper and iron is relevant in the context of protection against the oxidation of biomolecules and the generation of oxidative stress, observed in several conditions and considered an event that might promote sporadic prion diseases as well as other neurodegenerative disorders. In this context, ortho-pyridine aroylhydrazones are of interest, as they can act as moderate tridentate ligands towards divalent metal ions such as copper(II). In the present work, we explore the potentiality of this chemical class as peptide protecting agents against the deleterious metal-catalyzed oxidation in the M112A mutant fragment of human PrP, which mimics relevant structural features that may play an important role in the neurotoxicity observed in prion pathologies. The compounds inhere studied, especially HPCFur, showed an improved stability in aqueous solution compared to our patented lead hydrazone INHHQ, displaying a very interesting protective effect toward the oxidation of methionine and histidine, processes that are related to both physiological and pathological aging.


Subject(s)
Chelating Agents/chemistry , Copper/chemistry , Hydrazones/chemistry , Prion Proteins/drug effects , Pyridines/chemistry , Chelating Agents/chemical synthesis , Humans , Hydrazones/chemical synthesis , Ligands , Methionine/chemistry , Mutation , Oxidation-Reduction , Prion Proteins/chemistry , Prion Proteins/genetics , Pyridines/chemical synthesis
9.
J Integr Neurosci ; 18(4): 439-444, 2019 Dec 30.
Article in English | MEDLINE | ID: mdl-31912703

ABSTRACT

The effects of cellular prion protein on rapid eye movement sleep deprivation-induced spatial memory impairment were investigated, and the related mechanisms explored. Male C57BL/6 mice were randomly divided into four groups: environment control, sleep deprivation control, sleep-deprived-plasmid adeno-associated virus-green fluorescent protein group, and sleep-deprived-plasmid adeno-associated virus-cellular prion protein-green fluorescent protein group. Overexpression of cellular prion protein was induced by stereotaxic injection of adeno-associated viral plasmids-CAG-enhanced green fluorescent protein-cellular prion protein-Flag (a small label, which can be detected with corresponding tagged antibodies) into the hippocampus. Sleep-deprived mice were allowed no rapid eye movement sleep for 72 hours. Morris water maze was used to assess the effects of cellular prion protein on spatial learning and memory. The expression of amyloid-ß was also investigated in all groups. The sleep-deprived- plasmid adeno-associated virus- cellular prion protein-green fluorescent protein group spent significantly more time in a goal quadrant compared with the sleep-deprived- plasmid adeno-associated virus-green fluorescent protein group. Sleep deprivation resulted in increased amyloid-ß in the hippocampus, which was reversed by the overexpression of hippocampus cellular prion protein. Overexpression of cellular prion protein in the hippocampus rescues rapid eye movement sleep deprivation-induced spatial memory impairment in mice. It is shown that amyloid-ß in the hippocampus might be one of the mechanisms.


Subject(s)
Amyloid beta-Peptides/metabolism , Behavior, Animal/physiology , Hippocampus/metabolism , Memory Disorders/metabolism , Memory Disorders/physiopathology , Prion Proteins/metabolism , Sleep Deprivation/metabolism , Sleep Deprivation/physiopathology , Spatial Memory/physiology , Animals , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Prion Proteins/drug effects , Random Allocation , Sleep, REM
10.
Vet Microbiol ; 224: 1-7, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30269782

ABSTRACT

Prion diseases are characterized by the conformational conversion of the cellular prion protein (PrPC) to the pathogenic isoform (PrPSc). Lipids have been found to interact with PrPC and contribute to the efficient formation of PrPSc. Non-mammalian PrPs are not readily to undergo the conversion process into an infectious isoform, yet the effect of lipid on the conformational conversion of non-mammalian PrPC remains to be explored. Herein, the effects of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoglycerol (POPG) on full-length recombinant chicken PrP (ChPrP) 24-249 and murine PrP (MoPrP) 23-230 were investigated. Firstly, it was found that in the presence of chemical denaturant, POPG remarkably inhibited MoPrP amyloid fibril growth, while had slight effect on that of ChPrP as estimated by amyloid fibril growth and transmissible electronic microscope assays. Secondly, under physiological condition, POPG induced conformation changes in both MoPrP and ChPrP using Thioflavin T (ThT) fluorescence, circular dichroism, proteinase K digestion and transmission electron microscopy assays. With a POPG:PrP molar ratio of 30:1, the ThT fluorescence of MoPrP was found to be lower than that of ChPrP, however, the POPG-induced MoPrP had higher ß-sheet content and was more proteinase K-resistant than POPG-induced ChPrP. In summary, the present results suggested that the effects of POPG on conformational conversion of MoPrP and ChPrP were different under both denaturation and physiological conditions.


Subject(s)
Phosphatidylglycerols/pharmacology , Prion Proteins/chemistry , Prion Proteins/drug effects , Amyloid/drug effects , Amyloid/physiology , Animals , Chickens , Mice , Microscopy, Electron, Transmission , Prion Proteins/genetics , Protein Conformation , Recombinant Proteins/drug effects
11.
ACS Chem Neurosci ; 9(12): 2898-2903, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30074759

ABSTRACT

Several neurodegenerative diseases have a common pathophysiology where selective damage to neurons results from the accumulation of amyloid oligomer proteins formed via fibrilization. Considering that the formation of amyloid oligomers leads to cytotoxicity, the development of chemical compounds that are able to effectively cross the blood-brain barrier (BBB) and inhibit this conversion to oligomers and/or fibrils is essential for neurodegenerative disease therapy. We previously reported that pyrroloquinoline quinone (PQQ) prevented aggregation and fibrillation of α-synuclein, amyloid ß1-42 (Aß1-42), and mouse prion protein. To develop a novel drug against neurodegenerative diseases based on PQQ, it is necessary to improve the insufficient BBB permeability of PQQ. Here, we show that an esterified compound of PQQ, PQQ-trimethylester (PQQ-TME), has twice the BBB permeability than PQQ in vitro. Moreover, PQQ-TME exhibited greater inhibitory activity against fibrillation of α-synuclein, Aß1-42, and prion protein. These results indicated that esterification of PQQ could be a useful approach in developing a novel PQQ-based amyloid inhibitor.


Subject(s)
Amyloid beta-Peptides/drug effects , Amyloid/drug effects , Amyloidogenic Proteins/drug effects , Blood-Brain Barrier/metabolism , Neurons/drug effects , PQQ Cofactor/analogs & derivatives , Peptide Fragments/drug effects , Prion Proteins/drug effects , Protein Aggregation, Pathological/metabolism , alpha-Synuclein/drug effects , Amyloid beta-Peptides/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , Esterification , Esters/chemical synthesis , Esters/pharmacology , Humans , Mice , Neurons/metabolism , PQQ Cofactor/chemical synthesis , PQQ Cofactor/pharmacology , Peptide Fragments/metabolism , Permeability , Prion Proteins/metabolism , alpha-Synuclein/metabolism
12.
Bioorg Med Chem ; 25(20): 5875-5888, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28951092

ABSTRACT

Prion diseases are fatal neurodegenerative disorders of the central nervous system characterized by the accumulation of a protease resistant form (PrPSc) of the cellular prion protein (PrPC) in the brain. Two types of cellular prion (PrPC) compounds have been identified that appear to affect prion conversion are known as Effective Binders (EBs) and Accelerators (ACCs). Effective binders shift the balance in favour of PrPC, whereas Accelerators favour the formation of PrPSc. Molecular docking indicates EBs and ACCs both bind to pocket-D of the SHaPrPC molecule. However, EBs and ACCs may have opposing effects on the stability of the salt bridge between Arg156 and Glu196/Glu200. Computational docking data indicate that the hydrophobic benzamide group of the EB, GFP23 and the 1-(3,3-dimethylcyclohexylidene)piperidinium group of the ACC, GFP22 play an important role in inhibition and conversion from SHaPrPC to SHaPrPSc, respectively. Experimentally, NMR confirmed the amide chemical shift perturbations observed upon the binding of GFP23 to pocket-D of SHaPrPC. Consistent with its role as an ACC, titration of GFP22 resulted in widespread chemical shift changes and signal intensity loss due to protein unfolding. Virtual screening of a ligand database using the molecular scaffold developed from the set of EBs identified six of our compounds (previously studied using fluorescence quenching) as being among the top 100 best binders. Among them, compounds 5 and 6 were found to be particularly potent in decreasing the accumulation SHaPrPSc in ScN2a cells with an IC50 of ∼35µM and 20µM.


Subject(s)
Benzamides/pharmacology , Molecular Docking Simulation , PrPC Proteins/metabolism , PrPSc Proteins/metabolism , Prion Proteins/drug effects , Prion Proteins/metabolism , Benzamides/chemistry , Biological Assay , Cell Line , Chromones/chemistry , Chromones/pharmacology , Humans , Inhibitory Concentration 50 , Ligands , Models, Molecular , PrPC Proteins/chemistry , PrPSc Proteins/chemistry , Prion Proteins/classification , Protein Binding , Protein Folding , Thiadiazoles/chemistry , Thiadiazoles/pharmacology
13.
Arch Ital Biol ; 155(1-2): 81-97, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28715601

ABSTRACT

The cellular prion protein (PrPc) is physiologically expressed within selective brain areas of mammals. Alterations in the secondary structure of this protein lead to scrapie-like prion protein (PrPsc), which precipitates in the cell. PrPsc has been detected in infectious, inherited or sporadic neurodegenerative disorders. Prion protein metabolism is dependent on autophagy and ubiquitin proteasome. Despite not being fully elucidated, the physiological role of prion protein relates to chaperones which rescue cells under stressful conditions.Methamphetamine (METH) is a widely abused drug which produces oxidative stress in various brain areas causing mitochondrial alterations and protein misfolding. These effects produce a compensatory increase of chaperones while clogging cell clearing pathways. In the present study, we explored whether METH administration modifies the amount of PrPc. Since high levels of PrPc when the clearing systems are clogged may lead to its misfolding into PrPsc, we further tested whether METH exposure triggers the appearance of PrPsc. We analysed the effects of METH and dopamine administration in PC12 and striatal cells by using SDS-PAGE Coomassie blue, immune- histochemistry and immune-gold electron microscopy. To analyze whether METH administration produces PrPsc aggregates we used antibodies directed against PrP following exposure to proteinase K or sarkosyl which digest folded PrPc but misfolded PrPsc. We fond that METH triggers PrPsc aggregates in DA-containing cells while METH is not effective in primary striatal neurons which do not produce DA. In the latter cells exogenous DA is needed to trigger PrPsc accumulation similarly to what happens in DA containing cells under the effects of METH. The present findings, while fostering novel molecular mechanisms involving prion proteins, indicate that, cell pathology similar to prion disorders can be mimicked via a DA-dependent mechanism by a drug of abuse.


Subject(s)
Dopamine Agents/pharmacology , Methamphetamine/pharmacology , Neurons/drug effects , Oxidative Stress/drug effects , PrPSc Proteins/drug effects , Prion Proteins/drug effects , Adrenal Gland Neoplasms , Animals , Cell Line, Tumor , Dopamine/metabolism , Electrophoresis, Polyacrylamide Gel , Endopeptidase K/pharmacology , Mice , Microglia/drug effects , Neostriatum/cytology , Neurons/metabolism , Pheochromocytoma , PrPSc Proteins/metabolism , Prion Proteins/metabolism , Rats , Sarcosine/analogs & derivatives , Sarcosine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...