Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167187, 2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38653354

ABSTRACT

Clinical relevance of miRNAs as biomarkers is growing due to their stability and detection in biofluids. In this, diagnosis at asymptomatic stages of Alzheimer's disease (AD) remains a challenge since it can only be made at autopsy according to Braak NFT staging. Achieving the objective of detecting AD at early stages would allow possible therapies to be addressed before the onset of cognitive impairment. Many studies have determined that the expression pattern of some miRNAs is dysregulated in AD patients, but to date, none has been correlated with downregulated expression of cellular prion protein (PrPC) during disease progression. That is why, by means of cross studies of miRNAs up-regulated in AD with in silico identification of potential miRNAs-binding to 3'UTR of human PRNP gene, we selected miR-519a-3p for our study. Then, in vitro experiments were carried out in two ways. First, we validated miR-519a-3p target on 3'UTR-PRNP, and second, we analyzed the levels of PrPC expression after using of mimic technology on cell culture. In addition, RT-qPCR was performed to analyzed miR-519a-3p expression in human cerebral samples of AD at different stages of disease evolution. Additionally, samples of other neurodegenerative diseases such as other non-AD tauopathies and several synucleinopathies were included in the study. Our results showed that miR-519a-3p overlaps with PRNP 3'UTR in vitro and promotes downregulation of PrPC. Moreover, miR-519a-3p was found to be up-regulated exclusively in AD samples from stage I to VI, suggesting its potential use as a novel label of preclinical stages of the disease.

2.
Eur J Med Chem ; 250: 115169, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36753881

ABSTRACT

A set of twenty-five thioxanthene-9-one and xanthene-9-one derivatives, that were previously shown to inhibit cholinesterases (ChEs) and amyloid ß (Aß40) aggregation, were evaluated for the inhibition of tau protein aggregation. All compounds exhibited a good activity, and eight of them (5-8, 10, 14, 15 and 20) shared comparable low micromolar inhibitory potency versus Aß40 aggregation and human acetylcholinesterase (AChE), while inhibiting human butyrylcholinesterase (BChE) even at submicromolar concentration. Compound 20 showed outstanding biological data, inhibiting tau protein and Aß40 aggregation with IC50 = 1.8 and 1.3 µM, respectively. Moreover, at 0.1-10 µM it also exhibited neuroprotective activity against tau toxicity induced by okadoic acid in human neuroblastoma SH-SY5Y cells, that was comparable to that of estradiol and PD38. In preliminary toxicity studies, these interesting results for compound 20 are somewhat conflicting with a narrow safety window. However, compound 10, although endowed with a little lower potency for tau and Aß aggregation inhibition additionally demonstrated good inhibition of ChEs and rather low cytotoxicity. Compound 4 is also worth of note for its high potency as hBChE inhibitor (IC50 = 7 nM) and for the three order of magnitude selectivity versus hAChE. Molecular modelling studies were performed to explain the different behavior of compounds 4 and 20 towards hBChE. The observed balance of the inhibitory potencies versus the relevant targets indicates the thioxanthene-9-one derivatives as potential MTDLs for AD therapy, provided that the safety window will be improved by further structural variations, currently under investigation.


Subject(s)
Alzheimer Disease , Neuroblastoma , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Butyrylcholinesterase/metabolism , Amyloid beta-Peptides/metabolism , Acetylcholinesterase/metabolism , Cholinesterase Inhibitors/chemistry , Molecular Structure , Structure-Activity Relationship , Neuroblastoma/drug therapy , Drug Design , Molecular Docking Simulation
3.
Int J Mol Sci ; 22(10)2021 May 20.
Article in English | MEDLINE | ID: mdl-34065232

ABSTRACT

Tau protein is largely responsible for tauopathies, including Alzheimer's disease (AD), where it accumulates in the brain as insoluble aggregates. Tau mRNA is regulated by alternative splicing, and inclusion or exclusion of exon 10 gives rise to the 3R and 4R isoforms respectively, whose balance is physiologically regulated. In this sense, one of the several factors that regulate alternative splicing of tau is GSK3ß, whose activity is inhibited by the cellular prion protein (PrPC), which has different physiological functions in neuroprotection and neuronal differentiation. Moreover, a relationship between PrPC and tau expression levels has been reported during AD evolution. For this reason, in this study we aimed to analyze the role of PrPC and the implication of GSK3ß in the regulation of tau exon 10 alternative splicing. We used AD human samples and mouse models of PrPC ablation and tau overexpression. In addition, we used primary neuronal cultures to develop functional studies. Our results revealed a paralleled association between PrPC expression and tau 4R isoforms in all models analyzed. In this sense, reduction or ablation of PrPC levels induces an increase in tau 3R/4R balance. More relevantly, our data points to GSK3ß activity downstream from PrPC in this phenomenon. Our results indicate that PrPC plays a role in tau exon 10 inclusion through the inhibitory capacity of GSK3ß.


Subject(s)
Down-Regulation/genetics , Exons/genetics , Glycogen Synthase Kinase 3 beta/genetics , Prions/genetics , tau Proteins/genetics , Adult , Aged , Aged, 80 and over , Alternative Splicing/genetics , Alzheimer Disease/genetics , Animals , Brain/pathology , Disease Models, Animal , Female , Humans , Inclusion Bodies/genetics , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neurons/pathology , Protein Isoforms/genetics , RNA, Messenger/genetics , Tauopathies/genetics
4.
Mol Neurobiol ; 57(10): 4170-4186, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32683652

ABSTRACT

Cellular prion protein (PrPC) is largely responsible for transmissible spongiform encephalopathies (TSEs) when it becomes the abnormally processed and protease resistant form PrPSC. Physiological functions of PrPC include protective roles against oxidative stress and excitotoxicity. Relevantly, PrPC downregulates tau levels, whose accumulation and modification are a hallmark in the advance of Alzheimer's disease (AD). In addition to the accumulation of misfolded proteins, in the initial stages of AD-affected brains display both increased reactive oxygen species (ROS) markers and levels of PrPC. However, the factors responsible for the upregulation of PrPC are unknown. Thus, the aim of this study was to uncover the different molecular actors promoting PrPC overexpression. In order to mimic early stages of AD, we used ß-amyloid-derived diffusible ligands (ADDLs) and tau cellular treatments, as well as ROS generation, to elucidate their particular roles in human PRNP promoter activity. In addition, we used specific chemical inhibitors and site-specific mutations of the PRNP promoter sequence to analyze the contribution of the main transcription factors involved in PRNP transcription under the analyzed conditions. Our results revealed that tau is a new modulator of PrPC expression independently of ADDL treatment and ROS levels. Lastly, we discovered that the JNK/c-jun-AP-1 pathway is involved in increased PRNP transcription activity by tau but not in the promoter response to ROS.


Subject(s)
Prion Proteins/metabolism , Transcription, Genetic , tau Proteins/metabolism , Amyloid beta-Peptides/metabolism , Animals , Binding Sites , HEK293 Cells , Humans , Ligands , Mice, Inbred C57BL , Mice, Transgenic , Prion Proteins/genetics , Prion Proteins/ultrastructure , Promoter Regions, Genetic/genetics , Protein Kinase Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Solubility , tau Proteins/ultrastructure
5.
Cells ; 9(5)2020 05 19.
Article in English | MEDLINE | ID: mdl-32438605

ABSTRACT

Reelin is an extracellular glycoprotein that modulates neuronal function and synaptic plasticity in the adult brain. Decreased levels of Reelin activity have been postulated as a key factor during neurodegeneration in Alzheimer´s disease (AD) and in aging. Thus, changes in levels of full-length Reelin and Reelin fragments have been revealed in cerebrospinal fluid (CSF) and in post-mortem brains samples of AD patients with respect to non-AD patients. However, conflicting studies have reported decreased or unchanged levels of full-length Reelin in AD patients compared to control (nND) cases in post-mortem brains and CSF samples. In addition, a compelling analysis of Reelin levels in neurodegenerative diseases other than AD is missing. In this study, we analyzed brain levels of RELN mRNA and Reelin protein in post-mortem frontal cortex samples from different sporadic AD stages, Parkinson's disease with dementia (PDD), and Creutzfeldt-Jakob disease (sCJD), obtained from five different Biobanks. In addition, we measured Reelin protein levels in CSF samples of patients with mild cognitive impairment (MCI), dementia, or sCJD diagnosis and a group of neurologically healthy cases. The results indicate an increase in RELN mRNA in the frontal cortex of advanced stages of AD and in sCJD(I) compared to controls. This was not observed in PDD and early AD stages. However, Reelin protein levels in frontal cortex samples were unchanged between nND and advanced AD stages and PDD. Nevertheless, they decreased in the CSF of patients with dementia in comparison to those not suffering with dementia and patients with MCI. With respect to sCJD, there was a tendency to increase in brain samples in comparison to nND and to decrease in the CSF with respect to nND. In conclusion, Reelin levels in CSF cannot be considered as a diagnostic biomarker for AD or PDD. However, we feel that the CSF Reelin changes observed between MCI, patients with dementia, and sCJD might be helpful in generating a biomarker signature in prodromal studies of unidentified dementia and sCJD.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/genetics , Nerve Tissue Proteins/genetics , Neurodegenerative Diseases/genetics , Serine Endopeptidases/genetics , Brain/metabolism , Brain/pathology , Cell Adhesion Molecules, Neuronal/cerebrospinal fluid , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/cerebrospinal fluid , Extracellular Matrix Proteins/metabolism , Humans , Nerve Tissue Proteins/cerebrospinal fluid , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/cerebrospinal fluid , Neurodegenerative Diseases/pathology , Postmortem Changes , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reelin Protein , Serine Endopeptidases/cerebrospinal fluid , Serine Endopeptidases/metabolism
6.
Cells ; 9(3)2020 03 02.
Article in English | MEDLINE | ID: mdl-32131451

ABSTRACT

Cellular (also termed 'natural') prion protein has been extensively studied for many years for its pathogenic role in prionopathies after misfolding. However, neuroprotective properties of the protein have been demonstrated under various scenarios. In this line, the involvement of the cellular prion protein in neurodegenerative diseases other than prionopathies continues to be widely debated by the scientific community. In fact, studies on knock-out mice show a vast range of physiological functions for the protein that can be supported by its ability as a cell surface scaffold protein. In this review, we first summarize the most commonly described roles of cellular prion protein in neuroprotection, including antioxidant and antiapoptotic activities and modulation of glutamate receptors. Second, in light of recently described interaction between cellular prion protein and some amyloid misfolded proteins, we will also discuss the molecular mechanisms potentially involved in protection against neurodegeneration in pathologies such as Alzheimer's, Parkinson's, and Huntington's diseases.


Subject(s)
Brain/pathology , Nervous System Diseases/physiopathology , Prion Proteins/metabolism , Aged , Animals , Humans , Mice , Mice, Knockout
7.
Brain Pathol ; 30(2): 298-318, 2020 03.
Article in English | MEDLINE | ID: mdl-31397930

ABSTRACT

Human tau seeding and spreading occur following intracerebral inoculation of brain homogenates obtained from tauopathies in transgenic mice expressing natural or mutant tau, and in wild-type (WT) mice. The present study was geared to learning about the patterns of tau seeding, the cells involved and the characteristics of tau following intracerebral inoculation of homogenates from primary age-related tauopathy (PART: neuronal 4Rtau and 3Rtau), aging-related tau astrogliopathy (ARTAG: astrocytic 4Rtau) and globular glial tauopathy (GGT: 4Rtau with neuronal deposits and specific tau inclusions in astrocytes and oligodendrocytes). For this purpose, young and adult WT mice were inoculated unilaterally in the hippocampus or in the lateral corpus callosum with sarkosyl-insoluble fractions from PART, ARTAG and GGT cases, and were killed at variable periods of three to seven months. Brains were processed for immunohistochemistry in paraffin sections. Tau seeding occurred in the ipsilateral hippocampus and corpus callosum and spread to the septal nuclei, periventricular hypothalamus and contralateral corpus callosum, respectively. Tau deposits were mainly found in neurons, oligodendrocytes and threads; the deposits were diffuse or granular, composed of phosphorylated tau, tau with abnormal conformation and 3Rtau and 4Rtau independently of the type of tauopathy. Truncated tau at the aspartic acid 421 and ubiquitination were absent. Tau deposits had the characteristics of pre-tangles. A percentage of intracellular tau deposits co-localized with active (phosphorylated) tau kinases p38 and ERK 1/2. Present study shows that seeding and spreading of human tau into the brain of WT mice involves neurons and glial cells, mainly oligodendrocytes, thereby supporting the idea of a primary role of oligodendrogliopathy, together with neuronopathy, in the progression of tauopathies. In addition, it suggests that human tau inoculation modifies murine tau metabolism with the production and deposition of 3Rtau and 4Rtau, and by activation of specific tau kinases in affected cells.


Subject(s)
Brain/pathology , Tauopathies/pathology , tau Proteins/metabolism , Animals , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic
8.
Prog Neurobiol ; 165-167: 87-102, 2018.
Article in English | MEDLINE | ID: mdl-29530723

ABSTRACT

Several studies have indicated that certain misfolded amyloids composed of tau, ß-amyloid or α-synuclein can be transferred from cell to cell, suggesting the contribution of mechanisms reminiscent of those by which infective prions spread through the brain. This process of a 'prion-like' spreading between cells is also relevant as a novel putative therapeutic target that could block the spreading of proteinaceous aggregates throughout the brain which may underlie the progressive nature of neurodegenerative diseases. The relevance of ß-amyloid oligomers and cellular prion protein (PrPC) binding has been a focus of interest in Alzheimer's disease (AD). At the molecular level, ß-amyloid/PrPC interaction takes place in two differently charged clusters of PrPC. In addition to ß-amyloid, participation of PrPC in α-synuclein binding and brain spreading also appears to be relevant in α-synucleopathies. This review summarizes current knowledge about PrPC as a putative receptor for amyloid proteins and the physiological consequences of these interactions.


Subject(s)
Amyloid beta-Peptides/metabolism , Interneurons/metabolism , Neurodegenerative Diseases/metabolism , Prion Proteins/metabolism , alpha-Synuclein/metabolism , tau Proteins/metabolism , Animals , Humans
9.
Mol Neurobiol ; 55(4): 3033-3048, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28466265

ABSTRACT

Gerstmann-Sträussler-Scheinker (GSS) syndrome is a fatal autosomal dominant neurodegenerative prionopathy clinically characterized by ataxia, spastic paraparesis, extrapyramidal signs and dementia. In some GSS familiar cases carrying point mutations in the PRNP gene, patients also showed comorbid tauopathy leading to mixed pathologies. In this study we developed an induced pluripotent stem (iPS) cell model derived from fibroblasts of a GSS patient harboring the Y218N PRNP mutation, as well as an age-matched healthy control. This particular PRNP mutation is unique with very few described cases. One of the cases presented neurofibrillary degeneration with relevant Tau hyperphosphorylation. Y218N iPS-derived cultures showed relevant astrogliosis, increased phospho-Tau, altered microtubule-associated transport and cell death. However, they failed to generate proteinase K-resistant prion. In this study we set out to test, for the first time, whether iPS cell-derived neurons could be used to investigate the appearance of disease-related phenotypes (i.e, tauopathy) identified in the GSS patient.


Subject(s)
Gerstmann-Straussler-Scheinker Disease/genetics , Gerstmann-Straussler-Scheinker Disease/pathology , Induced Pluripotent Stem Cells/pathology , Mutation/genetics , Prion Proteins/genetics , tau Proteins/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Base Sequence , Brain/pathology , Cell Differentiation , Cells, Cultured , Female , Gliosis/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Middle Aged , Mitochondria/metabolism , Neurons/metabolism , Neurons/pathology , Phosphorylation
11.
Mol Neurobiol ; 55(3): 1847-1860, 2018 03.
Article in English | MEDLINE | ID: mdl-28229331

ABSTRACT

The cellular prion protein, encoded by the gene Prnp, has been reported to be a receptor of ß-amyloid. Their interaction is mandatory for neurotoxic effects of ß-amyloid oligomers. In this study, we aimed to explore whether the cellular prion protein participates in the spreading of α-synuclein. Results demonstrate that Prnp expression is not mandatory for α-synuclein spreading. However, although the pathological spreading of α-synuclein can take place in the absence of Prnp, α-synuclein expanded faster in PrPC-overexpressing mice. In addition, α-synuclein binds strongly on PrPC-expressing cells, suggesting a role in modulating the effect of α-synuclein fibrils.


Subject(s)
Neurons/metabolism , PrPC Proteins/genetics , PrPC Proteins/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Prion Proteins/genetics , Prion Proteins/metabolism , Protein Transport/physiology
12.
Prion ; 11(4): 226-233, 2017 07 04.
Article in English | MEDLINE | ID: mdl-28759332

ABSTRACT

The term 'prion-like' is used to define some misfolded protein species that propagate intercellularly, triggering protein aggregation in recipient cells. For cell binding, both direct plasma membrane interaction and membrane receptors have been described for particular amyloids. In this respect, emerging evidence demonstrates that several ß-sheet enriched proteins can bind to the cellular prion protein (PrPC). Among other interactions, the physiological relevance of the binding between ß-amyloid and PrPC has been a relevant focus of numerous studies. At the molecular level, published data point to the second charged cluster domain of the PrPC molecule as the relevant binding domain of the ß-amyloid/PrPC interaction. In addition to ß-amyloid, participation of PrPC in binding α-synuclein, responsible for neurodegenerative synucleopathies, has been reported. Although results indicate relevant participation of PrPC in the spreading of α-synuclein in living mice, the physiological relevance of the interaction remains elusive. In this comment, we focus our attention on summarizing current knowledge of PrPC as a receptor for amyloid proteins and its physiological significance, with particular focus on α-synuclein.


Subject(s)
Amyloid beta-Peptides/metabolism , Neurodegenerative Diseases/metabolism , PrPC Proteins/metabolism , Sensory Receptor Cells/metabolism , alpha-Synuclein/metabolism , Animals , Mice
13.
Mol Neurobiol ; 54(8): 6412-6425, 2017 Oct.
Article in English | MEDLINE | ID: mdl-27726110

ABSTRACT

Reelin is an extracellular glycoprotein involved in key cellular processes in developing and adult nervous system, including regulation of neuronal migration, synapse formation, and plasticity. Most of these roles are mediated by the intracellular phosphorylation of disabled-1 (Dab1), an intracellular adaptor molecule, in turn mediated by binding Reelin to its receptors. Altered expression and glycosylation patterns of Reelin in cerebrospinal and cortical extracts have been reported in Alzheimer's disease. However, putative changes in Reelin are not described in natural prionopathies or experimental models of prion infection or toxicity. With this is mind, in the present study, we determined that Reelin protein and mRNA levels increased in CJD human samples and in mouse models of human prion disease in contrast to murine models of prion infection. However, changes in Reelin expression appeared only at late terminal stages of the disease, which prevent their use as an efficient diagnostic biomarker. In addition, increased Reelin in CJD and in in vitro models does not correlate with Dab1 phosphorylation, indicating failure in its intracellular signaling. Overall, these findings widen our understanding of the putative changes of Reelin in neurodegeneration.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Creutzfeldt-Jakob Syndrome/metabolism , Extracellular Matrix Proteins/metabolism , Nerve Tissue Proteins/metabolism , Prion Diseases/metabolism , Serine Endopeptidases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Aged, 80 and over , Animals , Brain/metabolism , Creutzfeldt-Jakob Syndrome/genetics , Disease Models, Animal , Female , Humans , Male , Mice , Middle Aged , Nerve Tissue Proteins/genetics , Neurons/metabolism , Phosphorylation , Prion Diseases/genetics , Reelin Protein
14.
Prion ; 10(1): 25-40, 2016.
Article in English | MEDLINE | ID: mdl-26890218

ABSTRACT

Since its discovery the cellular prion protein (encoded by the Prnp gene) has been associated with a large number of functions. The proposed functions rank from basic cellular processes such as cell cycle and survival to neural functions such as behavior and neuroprotection, following a pattern similar to that of Moore's law for electronics. In addition, particular interest is increasing in the participation of Prnp in neurodegeneration. However, in recent years a redefinition of these functions has begun, since examples of previously attributed functions were increasingly re-associated with other proteins. Most of these functions are linked to so-called "Prnp-flanking genes" that are close to the genomic locus of Prnp and which are present in the genome of some Prnp mouse models. In addition, their role in neuroprotection against convulsive insults has been confirmed in recent studies. Lastly, in recent years a large number of models indicating the participation of different domains of the protein in apoptosis have been uncovered. However, after more than 10 years of molecular dissection our view is that the simplest mechanistic model in PrP(C)-mediated cell death should be considered, as Ockham's razor theory suggested.


Subject(s)
Prion Proteins , Animals , Mice , Mice, Knockout , Neuronal Plasticity , Prion Diseases , Prion Proteins/genetics , Prion Proteins/metabolism , Prion Proteins/physiology
15.
Mol Neurobiol ; 53(7): 4438-48, 2016 09.
Article in English | MEDLINE | ID: mdl-26250617

ABSTRACT

The biological functions of the cellular prion protein remain poorly understood. In fact, numerous studies have aimed to determine specific functions for the different protein domains. Studies of cellular prion protein (PrP(C)) domains through in vivo expression of molecules carrying internal deletions in a mouse Prnp null background have provided helpful data on the implication of the protein in signalling cascades in affected neurons. Nevertheless, understanding of the mechanisms underlying the neurotoxicity induced by these PrP(C) deleted forms is far from complete. To better define the neurotoxic or neuroprotective potential of PrP(C) N-terminal domains, and to overcome the heterogeneity of results due to the lack of a standardized model, we used neuroblastoma cells to analyse the effects of overexpressing PrP(C) deleted forms. Results indicate that PrP(C) N-terminal deleted forms were properly processed through the secretory pathway. However, PrPΔF35 and PrPΔCD mutants led to death by different mechanisms sharing loss of alpha-cleavage and activation of caspase-3. Our data suggest that both gain-of-function and loss-of-function pathogenic mechanisms may be associated with N-terminal domains and may therefore contribute to neurotoxicity in prion disease. Dissecting the molecular response induced by PrPΔF35 may be the key to unravelling the physiological and pathological functions of the prion protein.


Subject(s)
Death Domain Receptor Signaling Adaptor Proteins , Neuroblastoma/metabolism , Prion Proteins/metabolism , Animals , Cell Death , Cell Line, Tumor , Membrane Microdomains/metabolism , Mice , Models, Biological , Neuroblastoma/pathology , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Transfection
16.
Sci Rep ; 5: 11971, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-26155834

ABSTRACT

The cellular prion protein (PrP(C)) has been associated with a plethora of cellular functions ranging from cell cycle to neuroprotection. Mice lacking PrP(C) show an increased susceptibility to epileptic seizures; the protein, then, is neuroprotective. However, lack of experimental reproducibility has led to considering the possibility that other factors besides PrP(C) deletion, such as the genetic background of mice or the presence of so-called "Prnp flanking genes", might contribute to the reported susceptibility. Here, we performed a comparative analysis of seizure-susceptibility using characterized Prnp(+/+) and Prnp(0/0) mice of B6129, B6.129, 129/Ola or FVB/N genetic backgrounds. Our study indicates that PrP(C) plays a role in neuroprotection in KA-treated cells and mice. For this function, PrP(C) should contain the aa32-93 region and needs to be linked to the membrane. In addition, some unidentified "Prnp-flanking genes" play a role parallel to PrP(C) in the KA-mediated responses in B6129 and B6.129 Prnp(0/0) mice.


Subject(s)
Kainic Acid/toxicity , PrPC Proteins/genetics , Animals , Biomarkers , Cell Death , Cell Line , Cytokines/metabolism , Disease Models, Animal , Gene Expression , Genetic Predisposition to Disease , Gliosis/etiology , Gliosis/metabolism , Gliosis/pathology , Hippocampus/metabolism , Hippocampus/pathology , Inflammation Mediators/metabolism , Mice , Mice, Knockout , Seizures/etiology , Seizures/metabolism , Seizures/pathology , Transfection
17.
Cell Mol Life Sci ; 72(14): 2719-37, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25708702

ABSTRACT

Olfactory ensheathing cell (OEC) transplantation emerged some years ago as a promising therapeutic strategy to repair injured spinal cord. However, inhibitory molecules are present for long periods of time in lesioned spinal cord, inhibiting both OEC migration and axonal regrowth. Two families of these molecules, chondroitin sulphate proteoglycans (CSPG) and myelin-derived inhibitors (MAIs), are able to trigger inhibitory responses in lesioned axons. Mounting evidence suggests that OEC migration is inhibited by myelin. Here we demonstrate that OEC migration is largely inhibited by CSPGs and that inhibition can be overcome by the bacterial enzyme Chondroitinase ABC. In parallel, we have generated a stable OEC cell line overexpressing the Nogo receptor (NgR) ectodomain to reduce MAI-associated inhibition in vitro and in vivo. Results indicate that engineered cells migrate longer distances than unmodified OECs over myelin or oligodendrocyte-myelin glycoprotein (OMgp)-coated substrates. In addition, they also show improved migration in lesioned spinal cord. Our results provide new insights toward the improvement of the mechanisms of action and optimization of OEC-based cell therapy for spinal cord lesion.


Subject(s)
Myelin Proteins/metabolism , Myelin Sheath/metabolism , Nerve Regeneration/physiology , Neuroglia/physiology , Animals , Axons/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Chondroitin Sulfate Proteoglycans/pharmacology , Cloning, Molecular , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Microfluidic Analytical Techniques , Myelin Proteins/genetics , Neuroglia/metabolism , Nogo Receptor 1 , Olfactory Bulb/cytology , Oligodendrocyte-Myelin Glycoprotein/pharmacology , Protein Structure, Tertiary , Rats , Receptors, Cell Surface/genetics , Spinal Cord Injuries/therapy , Time-Lapse Imaging
18.
PLoS One ; 8(8): e70881, 2013.
Article in English | MEDLINE | ID: mdl-23940658

ABSTRACT

The physiological functions of PrP(C) remain enigmatic, but the central domain, comprising highly conserved regions of the protein may play an important role. Indeed, a large number of studies indicate that synthetic peptides containing residues 106-126 (CR) located in the central domain (CD, 95-133) of PrP(C) are neurotoxic. The central domain comprises two chemically distinct subdomains, the charge cluster (CC, 95-110) and a hydrophobic region (HR, 112-133). The aim of the present study was to establish the individual cytotoxicity of CC, HR and CD. Our results show that only the CD peptide is neurotoxic. Biochemical, Transmission Electron Microscopy and Atomic Force Microscopy experiments demonstrated that the CD peptide is able to activate caspase-3 and disrupt the cell membrane, leading to cell death.


Subject(s)
Neurons/physiology , Peptide Fragments/physiology , PrPC Proteins/physiology , Amino Acid Sequence , Animals , Apoptosis , Benzothiazoles , Caspase 3/metabolism , Cell Membrane Permeability/drug effects , Cells, Cultured , Dimyristoylphosphatidylcholine/chemistry , Enzyme Activation , Fluorescent Dyes/chemistry , Kinetics , Lipid Bilayers/chemistry , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Molecular Mimicry , Molecular Sequence Data , Neurons/drug effects , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , PrPC Proteins/chemistry , PrPC Proteins/pharmacology , Primary Cell Culture , Protein Multimerization , Protein Structure, Tertiary , Thiazoles/chemistry
19.
Neurobiol Aging ; 34(12): 2793-804, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23831375

ABSTRACT

Alzheimer's disease and prion diseases are neuropathological disorders that are caused by abnormal processing and aggregation of amyloid and prion proteins. Interactions between amyloid precursor protein (APP) and PrP(c) proteins have been described at the neuron level. Accordingly to this putative interaction, we investigated whether ß-amyloid accumulation may affect prion infectivity and, conversely, whether different amounts of PrP may affect ß-amyloid accumulation. For this purpose, we used the APPswe/PS1dE9 mouse line, a common model of Alzheimer's disease, crossed with mice that either overexpress (Tga20) or that lack prion protein (knock-out) to generate mice that express varying amounts of prion protein and deposit ß-amyloid. On these mouse lines, we investigated the influence of each protein on the evolution of both diseases. Our results indicated that although the presence of APP/PS1 and ß-amyloid accumulation had no effect on prion infectivity, the accumulation of ß-amyloid deposits was dependent on PrP(c), whereby increasing levels of prion protein were accompanied by a significant increase in ß-amyloid aggregation associated with aging.


Subject(s)
Aging/genetics , Aging/metabolism , Amyloid beta-Protein Precursor/physiology , PrPC Proteins/physiology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Mice , Mice, Transgenic , Presenilin-1/physiology , Prions/metabolism , Prions/pathogenicity
20.
PLoS One ; 7(4): e33872, 2012.
Article in English | MEDLINE | ID: mdl-22529900

ABSTRACT

There are numerous studies describing the signaling mechanisms that mediate oligodendrocyte precursor cell (OPC) proliferation and differentiation, although the contribution of the cellular prion protein (PrP(c)) to this process remains unclear. PrP(c) is a glycosyl-phosphatidylinositol (GPI)-anchored glycoprotein involved in diverse cellular processes during the development and maturation of the mammalian central nervous system (CNS). Here we describe how PrP(c) influences oligodendrocyte proliferation in the developing and adult CNS. OPCs that lack PrP(c) proliferate more vigorously at the expense of a delay in differentiation, which correlates with changes in the expression of oligodendrocyte lineage markers. In addition, numerous NG2-positive cells were observed in cortical regions of adult PrP(c) knockout mice, although no significant changes in myelination can be seen, probably due to the death of surplus cells.


Subject(s)
Cell Differentiation/physiology , Central Nervous System/embryology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , PrPC Proteins/metabolism , Animals , Cell Proliferation , Central Nervous System/cytology , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Female , Gene Expression , Hippocampus/cytology , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Neurogenesis , PrPC Proteins/genetics , Telencephalon/embryology , Telencephalon/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...