Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Brain Behav ; 10(1): e01498, 2020 01.
Article in English | MEDLINE | ID: mdl-31837111

ABSTRACT

INTRODUCTION: Several experimental studies have suggested the potential remyelinating effects of vitamin D (VitD) supplements regardless of the presence of VitD deficiency. This study aims to analyze neurogenesis in a model of toxic demyelination in order to evaluate the effects of VitD on demyelination and remyelination. MATERIAL AND METHODS: We used 24 male Wistar rats that had received surgical lesions to the corpus callosum and were injected with lysolecithin. Rats were divided into three groups: Group 1 included eight rats with lesions to the corpus callosum but not lysolecithin injections (sham group), group 2 included eight rats with lesions to the corpus callosum that were injected with lysolecithin (lysolecithin group), and group 3 included eight rats with lesions that were injected with lysolecithin and received VitD (VitD group). We analyzed neurogenesis both in the subventricular zone and at the lesion site. RESULTS: Administration of VitD promotes the proliferation and differentiation of neural stem cells in the subventricular zone and the migration of these cells to the lesion site in the corpus callosum; these cells subsequently differentiate into oligodendrocyte lineage cells and produce myelin basic protein. This phenomenon was not caused by microglial activation, which was less marked in rats receiving VitD. Megalin expression did not increase at the lesion site, which suggests that VitD is internalized by other mechanisms. CONCLUSION: Our results support the hypothesis that regardless of the presence of VitD deficiency, treatment with VitD may contribute to remyelination by promoting the proliferation of oligodendrocyte precursor cells.


Subject(s)
Oligodendroglia/physiology , Remyelination , Vitamin D , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Lineage/drug effects , Male , Multiple Sclerosis/therapy , Neural Stem Cells/physiology , Rats , Rats, Wistar , Remyelination/drug effects , Remyelination/physiology , Treatment Outcome , Vitamin D/metabolism , Vitamin D/pharmacology , Vitamins/metabolism , Vitamins/pharmacology
2.
Cells ; 8(10)2019 10 22.
Article in English | MEDLINE | ID: mdl-31652490

ABSTRACT

Microglia originate from yolk sac-primitive macrophages and auto-proliferate into adulthood without replacement by bone marrow-derived circulating cells. In inflammation, stroke, aging, or infection, microglia have been shown to contribute to brain pathology in both deleterious and beneficial ways, which have been studied extensively. However, less is known about their role in the healthy adult brain. Astrocytes and oligodendrocytes are widely accepted to strongly contribute to the maintenance of brain homeostasis and to modulate neuronal function. On the other hand, contribution of microglia to cognition and behavior is only beginning to be understood. The ability to probe their function has become possible using microglial depletion assays and conditional mutants. Studies have shown that the absence of microglia results in cognitive and learning deficits in rodents during development, but this effect is less pronounced in adults. However, evidence suggests that microglia play a role in cognition and learning in adulthood and, at a cellular level, may modulate adult neurogenesis. This review presents the case for repositioning microglia as key contributors to the maintenance of homeostasis and cognitive processes in the healthy adult brain, in addition to their classical role as sentinels coordinating the neuroinflammatory response to tissue damage and disease.


Subject(s)
Brain/physiology , Cognition/physiology , Learning/physiology , Microglia/physiology , Adult , Animals , Astrocytes/cytology , Astrocytes/physiology , Brain/cytology , Humans , Microglia/cytology , Oligodendroglia/cytology , Oligodendroglia/physiology
3.
PLoS Comput Biol ; 14(9): e1006378, 2018 09.
Article in English | MEDLINE | ID: mdl-30180157

ABSTRACT

Clustering of genes and/or samples is a common task in gene expression analysis. The goals in clustering can vary, but an important scenario is that of finding biologically meaningful subtypes within the samples. This is an application that is particularly appropriate when there are large numbers of samples, as in many human disease studies. With the increasing popularity of single-cell transcriptome sequencing (RNA-Seq), many more controlled experiments on model organisms are similarly creating large gene expression datasets with the goal of detecting previously unknown heterogeneity within cells. It is common in the detection of novel subtypes to run many clustering algorithms, as well as rely on subsampling and ensemble methods to improve robustness. We introduce a Bioconductor R package, clusterExperiment, that implements a general and flexible strategy we entitle Resampling-based Sequential Ensemble Clustering (RSEC). RSEC enables the user to easily create multiple, competing clusterings of the data based on different techniques and associated tuning parameters, including easy integration of resampling and sequential clustering, and then provides methods for consolidating the multiple clusterings into a final consensus clustering. The package is modular and allows the user to separately apply the individual components of the RSEC procedure, i.e., apply multiple clustering algorithms, create a consensus clustering or choose tuning parameters, and merge clusters. Additionally, clusterExperiment provides a variety of visualization tools for the clustering process, as well as methods for the identification of possible cluster signatures or biomarkers. The R package clusterExperiment is publicly available through the Bioconductor Project, with a detailed manual (vignette) as well as well documented help pages for each function.


Subject(s)
Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation , Hypothalamus/physiology , Olfactory Mucosa/physiology , Algorithms , Animals , Astrocytes/physiology , Biomarkers , Cluster Analysis , Databases, Factual , Humans , Microglia/physiology , Multigene Family , Neurons/physiology , Oligodendroglia/physiology , Programming Languages , Sequence Analysis, RNA , Software
4.
Glia ; 66(7): 1302-1316, 2018 07.
Article in English | MEDLINE | ID: mdl-29423921

ABSTRACT

Oligodendrocyte-formed myelin sheaths play important roles in the neuronal functions in the central nervous system. In demyelinating diseases, such as Multiple Sclerosis, the myelin sheaths are damaged and the remyelinating process is somehow hindered. Restoration of the myelin sheaths requires the differentiation of the oligodendrocyte precursor cells (OPCs) into mature oligodendrocytes (OLs). To discover small molecule compounds that might promote the OPC to OL differentiation, a high-throughput screening system is established and L-ascorbyl-2-phosphate (As-2P), a stable form of Vitamin C (Vc), is found to greatly enhance the OPC to OL differentiation. As-2P promotes gradual expression of OL lineage markers, including O4, CNPase and MBP, in a dose- and time-dependent manner. It also facilitates the formation of myelin sheaths in OPC-neuron co-culture. As-2P also promotes the repair of the myelin sheaths in vivo and provides significant therapeutic effect in a cuprizone-mediated demyelination animal model. Interestingly, As-2P's function in promoting OPC differentiation is not related to its antioxidant activity. And an intracellular rather than an extracellular mechanism might be involved. Considering the safe use of Vc as a dietary supplement for many years, it might also be used as an alternative medicine for CNS demyelinating diseases.


Subject(s)
Ascorbic Acid/analogs & derivatives , Cell Differentiation/drug effects , Demyelinating Diseases/drug therapy , Neuroprotective Agents/pharmacology , Oligodendroglia/drug effects , Remyelination/drug effects , Animals , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Brain/cytology , Brain/drug effects , Brain/pathology , Brain/physiology , Cell Differentiation/physiology , Coculture Techniques , Cuprizone , Demyelinating Diseases/pathology , Demyelinating Diseases/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Oligodendroglia/cytology , Oligodendroglia/physiology , Remyelination/physiology , Time Factors
5.
Exp Neurol ; 300: 222-231, 2018 02.
Article in English | MEDLINE | ID: mdl-29199131

ABSTRACT

We investigated whether electroacupuncture (EA) and treadmill (TM) exercise improve behaviors related to motor and memory dysfunction in a cerebral palsy-like rat model via activation of oligodendrogenesis. A neonatal hypoxia-ischemia model was created using Sprague-Dawley rats (P7), and these underwent EA stimulation and treadmill training from 3 to 5weeks after hypoxia-ischemia induction. EA treatment was delivered via electrical stimulation (2Hz, 1mA) at two acupoints, Baihui (GV20) and Zusanli (ST36). Behavioral tests showed that EA alleviated motor dysfunction caused by hypoxia-ischemia on a rotarod test, and TM exercise alleviated motor and memory dysfunction seen on cylinder and passive avoidance tests. Combined therapy with EA and TM exercise showed synergistic effects on the cylinder, rotarod, and catwalk tests. TM exercise significantly restored corpus callosum thickness, and combined therapy with EA and TM restored myelin basic protein (MBP) levels in this region. While EA stimulation only increased activation of cAMP-response element binging protein (CREB) in oligodendrocytes of the corpus callosum, TM exercise increased newly generated oligodendrocyte progenitor cells or oligodendrocytes via activation of CREB. Synergistic effects on oligodendrogenesis were also observed by the combined therapy. Furthermore, the combined therapy induced mature brain-derived neurotrophic factor (BDNF) expression in the cerebral cortex. These results demonstrate that combined therapy with EA and TM exercise may restore myelin components following neonatal hypoxia-ischemia via upregulation of oligodendrogenesis involving CREB/BDNF signaling, which subsequently improves motor and memory function. Therefore, combined therapy with EA and TM exercise offers another treatment option for functional recovery from injuries caused by neonatal hypoxia-ischemia, such as cerebral palsy.


Subject(s)
Combined Modality Therapy/methods , Demyelinating Diseases/therapy , Electroacupuncture/methods , Exercise Test/methods , Hypoxia-Ischemia, Brain/therapy , Oligodendroglia/physiology , Animals , Animals, Newborn , Cell Proliferation/physiology , Corpus Callosum/cytology , Corpus Callosum/metabolism , Corpus Callosum/pathology , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Disease Models, Animal , Female , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Male , Pregnancy , Rats , Rats, Sprague-Dawley
6.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(9): 1013-1023, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28647405

ABSTRACT

Docosahexaenoic acid (DHA) is an essential omega-3 fatty acid known to be neuroprotective in several models of human diseases, including multiple sclerosis. The protective effects of DHA are largely attributed to its ability to interfere with the activity of transcription factors controlling immune and inflammatory responses, including the agonist-dependent transcription factor peroxisome proliferator-activated receptor-γ (PPAR-γ). In this study, we used primary oligodendrocyte progenitor (OP) cultures from neonatal rat brain to investigate whether DHA could influence OP maturation and directly promote myelination, as previously reported for selective PPAR-γ agonists. We show that, similarly to the selective PPAR-γ agonist pioglitazone (PGZ), DHA promotes OP maturation and counteracts the maturational arrest induced by TNF-α, used to mimic inflammatory conditions. The PPAR-γ antagonist GW9662 prevented both DHA-induced OP maturation and PPAR-γ nuclear translocation, supporting the hypothesis that DHA acts through the activation of PPAR-γ. In addition, both PGZ and DHA induced the phosphorylation of extracellular signal-regulated-kinase 1-2 (ERK1/2), in a PPAR-γ-dependent manner. ERK1/2 activity is known to regulate the transition from OPs to immature oligodendrocytes and the presence of specific inhibitors of ERK1/2 phosphorylation (U0126 or PD98059) prevented the differentiating effects of both DHA and PGZ. These results indicate that DHA might influence the process of OP maturation through its PPAR-γ agonistic activity and provide novel molecular mechanisms for the action of this dietary fatty acid, further supporting the nutritional intervention in demyelinating diseases such as multiple sclerosis.


Subject(s)
Cell Differentiation/drug effects , Docosahexaenoic Acids/pharmacology , Oligodendroglia/drug effects , PPAR gamma/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Demyelinating Diseases/metabolism , Fatty Acids, Omega-3/pharmacology , Inflammation/metabolism , MAP Kinase Signaling System/drug effects , Neurogenesis/drug effects , Oligodendroglia/metabolism , Oligodendroglia/physiology , Phosphorylation/drug effects , Pioglitazone , Rats , Rats, Wistar , Thiazolidinediones/pharmacology , Transcription Factors/metabolism
7.
Nat Commun ; 8: 14241, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28117328

ABSTRACT

Multiple Sclerosis (MS) is an inflammatory demyelinating disorder in which remyelination failure contributes to persistent disability. Cholesterol is rate-limiting for myelin biogenesis in the developing CNS; however, whether cholesterol insufficiency contributes to remyelination failure in MS, is unclear. Here, we show the relationship between cholesterol, myelination and neurological parameters in mouse models of demyelination and remyelination. In the cuprizone model, acute disease reduces serum cholesterol levels that can be restored by dietary cholesterol. Concomitant with blood-brain barrier impairment, supplemented cholesterol directly supports oligodendrocyte precursor proliferation and differentiation, and restores the balance of growth factors, creating a permissive environment for repair. This leads to attenuated axon damage, enhanced remyelination and improved motor learning. Remarkably, in experimental autoimmune encephalomyelitis, cholesterol supplementation does not exacerbate disease expression. These findings emphasize the safety of dietary cholesterol in inflammatory diseases and point to a previously unrecognized role of cholesterol in promoting repair after demyelinating episodes.


Subject(s)
Cholesterol, Dietary/administration & dosage , Cholesterol/blood , Multiple Sclerosis/therapy , Myelin Proteins/biosynthesis , Animals , Axons/pathology , Biomarkers/blood , Brain/cytology , Brain/pathology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cholesterol/metabolism , Cholesterol, Dietary/adverse effects , Cuprizone/toxicity , Dietary Supplements , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/blood , Multiple Sclerosis/chemically induced , Oligodendroglia/cytology , Oligodendroglia/pathology , Oligodendroglia/physiology , Primary Cell Culture , Stem Cells/physiology
8.
Sci Rep ; 6: 28646, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27350403

ABSTRACT

We modeled prolonged cerebral hypoperfusion in mice using bilateral common carotid artery stenosis (BCAS) and electroacupuncture (EA) stimulation was applied at two acupoints, Baihui (GV20) and Dazhui (GV14). In behavioral tests of memory, BCAS produced impairments in spatial and short-term memory in mice that were attenuated by therapeutic EA stimulation. Therapeutic use of EA in BCAS also enhanced oligodendrocyte (OL) differentiation from oligodendrocyte precursor cells (OPCs), in association with white matter improvements in the corpus callosum (CC). In PCR analyses of growth factor gene expression, significant positive changes in 3 genes were observed following EA stimulation in BCAS, and here we highlight alterations in neurotrophin-4/5 (NT4/5). We confirmed EA-mediated positive changes in the expression of NT4/5 and its receptor, tyrosine receptor kinase B (TrkB). Treatment of naïve and BCAS + EA animals with a selective TrkB antagonist, ANA-12, produced losses of myelin and cognitive function that were ameliorated by EA therapy. Moreover, following BCAS we observed an EA-dependent increase in phospho-activated CREB (a downstream mediator of NT4/5-TrkB signaling) in OPCs and OLs of the CC. Our results suggest that EA stimulation promotes the recovery of memory function following white matter injury via a mechanism that promotes oligodendrocyte regeneration and involves NT4/5-TrkB signaling.


Subject(s)
Brain Ischemia/therapy , Brain/pathology , Electroacupuncture , Memory Disorders/therapy , Nerve Regeneration , Oligodendroglia/physiology , Acupuncture Points , Animals , Behavior, Animal , Cell Differentiation , Corpus Callosum/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Male , Memory, Short-Term , Mice , Mice, Inbred C57BL , Nerve Growth Factors/metabolism , Oligodendroglia/cytology , Perfusion , Phosphorylation , Polymerase Chain Reaction , Signal Transduction
9.
BMC Neurosci ; 17: 16, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-27103572

ABSTRACT

BACKGROUND: Regeneration of new myelin is impaired in persistent multiple sclerosis (MS) lesions, leaving neurons unable to function properly and subject to further degeneration. Current MS therapies attempt to ameliorate autoimmune-mediated demyelination, but none directly promote the regeneration of lost and damaged myelin of the central nervous system (CNS). Development of new drugs that stimulate remyelination has been hampered by the inability to evaluate axonal myelination in a rapid CNS culture system. RESULTS: We established a high throughput cell-based assay to identify compounds that promote myelination. Culture methods were developed for initiating myelination in vitro using primary embryonic rat cortical cells. We developed an immunofluorescent phenotypic image analysis method to quantify the morphological alignment of myelin characteristic of the initiation of myelination. Using γ-secretase inhibitors as promoters of myelination, the optimal growth, time course and compound treatment conditions were established in a 96 well plate format. We have characterized the cortical myelination assay by evaluating the cellular composition of the cultures and expression of markers of differentiation over the time course of the assay. We have validated the assay scalability and consistency by screening the NIH clinical collection library of 727 compounds and identified ten compounds that promote myelination. Half maximal effective concentration (EC50) values for these compounds were determined to rank them according to potency. CONCLUSIONS: We have designed the first high capacity in vitro assay that assesses myelination of live axons. This assay will be ideal for screening large compound libraries to identify new drugs that stimulate myelination. Identification of agents capable of promoting the myelination of axons will likely lead to the development of new therapeutics for MS patients.


Subject(s)
Axons/drug effects , Cerebral Cortex/drug effects , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Multiple Sclerosis/drug therapy , Myelin Sheath/drug effects , Nerve Regeneration/drug effects , Amyloid Precursor Protein Secretases/pharmacology , Animals , Axons/physiology , Cell Culture Techniques , Cell Differentiation/drug effects , Cerebral Cortex/physiology , Culture Media, Conditioned/pharmacology , Fluorescent Antibody Technique/methods , Multiple Sclerosis/physiopathology , Myelin Sheath/physiology , Oligodendroglia/drug effects , Oligodendroglia/physiology , Rats
10.
Mol Med Rep ; 13(3): 2892-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26821234

ABSTRACT

Hyperoxia is a high risk factor for neurodevelopmental disorders and can cause nerve cell death. 17ß­Estradiol (E2) has been demonstrated as a neuroprotective agent. In the present study, the effect of hyperoxia on rat oligodendrocyte precursor cells (OPCs) in vivo and the neuroprotective effects of E2 on these cells were investigated. OPCs were treated with various concentrations of E2 and were harvested for reverse transcription­quantitiative polymerase chain reaction (RT­qPCR) analysis at various time­points. RT­qPCR analysis demonstrated that paired immunoglobin­like receptor B (PriB) PriB mRNA expression levels were markedly decreased following treatment with 10(­6), 10(­7) and 10(­8) M E2. Cells treated with 10(­7) M E2 for 24 h were selected for subsequent experiments. PriB was silenced with small interfering (si)RNA and the effects of E2 treatment and silencing of PriB on the viability and apoptosis of OPCs under hyperoxic stimulation was detected using 3­(4,5­dimethyl­2­thiazolyl)­2,5­diphenyl­2­H­tetrazoliu­bromide (MTT) assay and flow cytometry analysis. The results revealed that hyperoxia induced apoptosis in OPCs and decreased their viability. Hyperoxia also induced the expression of caspases­3 and ­8, and Fas cell surface death receptor (Fas). E2 treatment markedly downregulated the expression of PirB. E2 treatment or PirB silencing markedly decreased hyperoxia­induced apoptosis, increased cell viability and decreased the expression of caspases­3 and ­8, and Fas in OPCs, indicating that E2 protects OPCs from hyperoxia­induced apoptosis, predominantly through the downregulation of PirB The results of the present study provide a theoretical basis for the reasonable use of oxygen in Neonatal Intensive Care Units.


Subject(s)
Apoptosis , Estradiol/pharmacology , Neuroprotective Agents/pharmacology , Oligodendroglia/physiology , Receptors, Immunologic/metabolism , Animals , Caspase 3/metabolism , Caspase 8/metabolism , Cell Hypoxia , Drug Evaluation, Preclinical , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley
11.
Cell Death Dis ; 6: e1922, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26469970

ABSTRACT

We demonstrate that EphB3 receptors mediate oligodendrocyte (OL) cell death in the injured spinal cord through dependence receptor mechanism. OLs in the adult spinal cord express EphB3 as well as other members of the Eph receptor family. Spinal cord injury (SCI) is associated with tissue damage, cellular loss and disturbances in EphB3-ephrinB3 protein balance acutely (days) after the initial impact creating an environment for a dependence receptor-mediated cell death to occur. Genetic ablation of EphB3 promotes OL survival associated with increased expression of myelin basic protein and improved locomotor function in mice after SCI. Moreover, administration of its ephrinB3 ligand to the spinal cord after injury also promotes OL survival. Our in vivo findings are supported by in vitro studies showing that ephrinB3 administration promotes the survival of both oligodendroglial progenitor cells and mature OLs cultured under pro-apoptotic conditions. In conclusion, the present study demonstrates a novel dependence receptor role of EphB3 in OL cell death after SCI, and supports further development of ephrinB3-based therapies to promote recovery.


Subject(s)
Apoptosis , Oligodendroglia/physiology , Receptor, EphB3/physiology , Spinal Cord Injuries/metabolism , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Ephrin-B3/pharmacology , Ephrin-B3/therapeutic use , Female , Mice, Knockout , Recovery of Function , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology
12.
Nat Med ; 20(8): 954-960, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24997607

ABSTRACT

Functional screening for compounds that promote remyelination represents a major hurdle in the development of rational therapeutics for multiple sclerosis. Screening for remyelination is problematic, as myelination requires the presence of axons. Standard methods do not resolve cell-autonomous effects and are not suited for high-throughput formats. Here we describe a binary indicant for myelination using micropillar arrays (BIMA). Engineered with conical dimensions, micropillars permit resolution of the extent and length of membrane wrapping from a single two-dimensional image. Confocal imaging acquired from the base to the tip of the pillars allows for detection of concentric wrapping observed as 'rings' of myelin. The platform is formatted in 96-well plates, amenable to semiautomated random acquisition and automated detection and quantification. Upon screening 1,000 bioactive molecules, we identified a cluster of antimuscarinic compounds that enhance oligodendrocyte differentiation and remyelination. Our findings demonstrate a new high-throughput screening platform for potential regenerative therapeutics in multiple sclerosis.


Subject(s)
High-Throughput Screening Assays/methods , Multiple Sclerosis/drug therapy , Muscarinic Antagonists/isolation & purification , Nerve Fibers, Myelinated/drug effects , Animals , Cell Differentiation/drug effects , Cells, Cultured , Clemastine/pharmacology , Drug Evaluation, Preclinical/methods , Female , Histamine H1 Antagonists/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscarinic Antagonists/pharmacology , Nanostructures , Oligodendroglia/cytology , Oligodendroglia/drug effects , Oligodendroglia/physiology , Rats , Rats, Sprague-Dawley , Regeneration/drug effects
13.
Article in English | MEDLINE | ID: mdl-24360206

ABSTRACT

Recently, we investigated the effects of eicosapentaenoic acid (EPA), a fatty acid which modulates immune response and stimulates myelin gene expression, in an established model of multiple sclerosis (MS): the experimental autoimmune encephalomyelitis (EAE) induced in Dark Agouti rats. As scientific evidences and our previous studies have suggested that EPA could directly affect oligodendrocytes, we have now evaluated the effects of EPA in the non-immune mediate MS model characterized by selective oligodendrocytes damage induced by cuprizone (CPZ). We found that feeding weanling rats diets containing 0.6% CPZ for 2 weeks induced variation of whole brain and myelin biochemical composition representative of a severe myelin damage. We thus administered daily and by gavage EPA or PBS to 2-day old rats up to 21 days. Afterwards, rats were fed CPZ diet for 9 days. The results show that compared to PBS/CPZ fed rats, the whole brain cerebroside content in EPA pre-treated rats was statistically increased as well as there was an overall trend of increase of all other biochemical components.


Subject(s)
Brain/metabolism , Eicosapentaenoic Acid/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Myelin Sheath/metabolism , Animals , Brain/drug effects , Cells, Cultured , Cerebrosides/metabolism , Cuprizone , Drug Evaluation, Preclinical , Eicosapentaenoic Acid/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/metabolism , Erythrocytes/metabolism , Fatty Acids/blood , Inflammation Mediators/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oligodendroglia/drug effects , Oligodendroglia/physiology , Rats , Rats, Wistar , Weaning
14.
Neurodegener Dis ; 13(1): 45-52, 2014.
Article in English | MEDLINE | ID: mdl-23949302

ABSTRACT

BACKGROUND: inefficient remyelination of demyelinated plaques in multiple sclerosis (ms) leads to secondary axon degeneration and progressive disability. therapies that potentiate remyelination would be of immense help for managing MS. OBJECTIVE: Here, we report the effects of valproic acid (VPA) on focal experimental autoimmune encephalomyelitis (fEAE). METHODS: fEAE was induced in Wistar rats by immunizing the animals with guinea pig spinal cord homogenate emulsified in complete Freund's adjuvant and with pertussis toxin (PT) injection into the spinal cord at the level of T8 vertebra on day 18 after immunization. VPA 300 mg/kg was applied for 4 days after or 8 days before PT administration. Behavioral evaluation, histological assessment and immunohistofluorescence assays were used to evaluate the outcomes. RESULTS: VPA administration had no effect on the development of symptoms, but after discontinuing VPA, animals showed faster recovery. Eight days of pretreatment with VPA accelerated the recovery phase of EAE and increased the number of remyelinated axons in the lesion area. VPA pretreatment also increased the recruitment of neural stem cells and oligodendrocyte precursors within the lesion. CONCLUSIONS: Results suggest VPA as a potential therapy for remyelinating the lesions in MS and for faster recovery from disease relapses. The effect of VPA seems to be mediated by endogenous progenitors recruitment.


Subject(s)
Central Nervous System Agents/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Nerve Fibers, Myelinated/drug effects , Neural Stem Cells/drug effects , Stem Cells/drug effects , Valproic Acid/therapeutic use , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Fluorescent Antibody Technique , Guinea Pigs , Nerve Fibers, Myelinated/pathology , Neural Stem Cells/physiology , Oligodendroglia/drug effects , Oligodendroglia/physiology , Rats , Rats, Wistar , Severity of Illness Index , Spinal Cord/drug effects , Spinal Cord/pathology , Stem Cells/pathology
15.
Neuroscience ; 258: 131-46, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24215977

ABSTRACT

Voluntary exercise (VEx) has profound effects on neural and behavioral plasticity, including recovery of CNS trauma and disease. However, the unique regional cortical adaption to VEx has not been elucidated. In a series of experiments, we first examined whether VEx would restore and retain neurotrophin levels in several cortical regions (frontal cortex [FC], retrosplenial cortex [RSC], occipital cortex [OC]) in an animal model (pyrithiamine-induced thiamine deficiency [PTD]) of the amnestic disorder Wernicke-Korsakoff syndrome. In addition, we assessed the time-dependent effect of VEx to rescue performance on a spontaneous alternation task. Following 2-weeks of VEx or stationary housing conditions (Stat), rats were behaviorally tested and brains were harvested either the day after VEx (24-h) or after an additional 2-week period (2-wk). In both control pair-fed (PF) rats and PTD rats, all neurotrophin levels (brain-derived neurotrophic factor [BDNF], nerve growth factor [NGF], and vascular endothelial growth factor) increased at the 24-h period after VEx in the FC and RSC, but not OC. Two-weeks following VEx, BDNF remained elevated in both FC and RSC, whereas NGF remained elevated in only the FC. Interestingly, VEx only recovered cognitive performance in amnestic rats when there was an additional 2-wk adaptation period after VEx. Given this unique temporal profile, Experiment 2 examined the cortical cytogenetic responses in all three cortical regions following a 2-wk adaptation period after VEx. In healthy (PF) rats, VEx increased the survival of progenitor cells in both the FC and RSC, but only increased oligodendrocyte precursor cells (OLPs) in the FC. Furthermore, VEx had a selective effect of only recovering OLPs in the FC in PTD rats. These data reveal the therapeutic potential of exercise to restore cortical plasticity in the amnestic brain, and that the FC is one of the most responsive cortical regions to VEx.


Subject(s)
Amnesia/physiopathology , Cerebral Cortex/physiopathology , Motor Activity/physiology , Nerve Growth Factors/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cell Survival , Cytogenetic Analysis , Frontal Lobe/physiopathology , Housing, Animal , Male , Nerve Growth Factor/metabolism , Occipital Lobe/physiopathology , Oligodendroglia/physiology , Pyrithiamine , Rats , Rats, Sprague-Dawley , Stem Cells/physiology , Thiamine Deficiency/physiopathology , Time Factors , Vascular Endothelial Growth Factor A/metabolism
16.
Sci Rep ; 3: 1806, 2013.
Article in English | MEDLINE | ID: mdl-23657670

ABSTRACT

The migration of oligodendrocyte progenitor cells (OPCs) to the white matter is an indispensable requirement for an intact brain function. The mechanism of cell migration in general is not yet completely understood. Nevertheless, evidence is accumulating that besides the coordinated rearrangement of the cytoskeleton, a finetuned interplay of ion and water fluxes across the cell membrane is essential for cell migration. One part of a general hypothesis is that a local volume increase towards the direction of movement triggers a mechano-activated calcium influx that regulates various procedures at the rear end of a migrating cell. Here, we investigated cell volume changes of migrating OPCs using scanning ion conductance microscopy. We found that during accelerated migration OPCs undergo an increase in the frontal cell body volume. These findings are supplemented with time lapse calcium imaging data that hint an increase in calcium content the frontal part of the cell soma.


Subject(s)
Brain/metabolism , Calcium/metabolism , Cell Movement/physiology , Cell Size , Cytoskeleton/metabolism , Oligodendroglia/physiology , Stem Cells/physiology , Animals , Brain/cytology , Cell Differentiation , Cell Nucleus/metabolism , Cells, Cultured , Mechanotransduction, Cellular , Microscopy, Scanning Probe/instrumentation , Microscopy, Scanning Probe/methods , Molecular Imaging/methods , Oligodendroglia/cytology , Rats , Rats, Sprague-Dawley , Stem Cells/cytology
17.
PLoS One ; 8(5): e63415, 2013.
Article in English | MEDLINE | ID: mdl-23650566

ABSTRACT

Repair in multiple sclerosis involves remyelination, a process in which axons are provided with a new myelin sheath by new oligodendrocytes. Bone morphogenic proteins (BMPs) are a family of growth factors that have been shown to influence the response of oligodendrocyte progenitor cells (OPCs) in vivo during demyelination and remyelination in the adult brain. We have previously shown that BMP4 infusion increases numbers of OPCs during cuprizone-induced demyelination, while infusion of Noggin, an endogenous antagonist of BMP4 increases numbers of mature oligodendrocytes and remyelinated axons following recovery. Additional studies have shown that insulin-like growth factor-1 (IGF-1) promotes the survival of OPCs during cuprizone-induced demyelination. Based on these data, we investigated whether myelin repair could be further enhanced by sequential infusion of these agents firstly, BMP4 to increase OPC numbers, followed by either Noggin or IGF-1 to increase the differentiation and survival of the newly generated OPCs. We identified that sequential delivery of BMP4 and IGF-1 during cuprizone challenge increased the number of mature oligodendrocytes and decreased astrocyte numbers following recovery compared with vehicle infused mice, but did not alter remyelination. However, sequential delivery of BMP4 and Noggin during cuprizone challenge did not alter numbers of oligodendrocytes or astrocytes in the corpus callosum compared with vehicle infused mice. Furthermore, electron microscopy analysis revealed no change in average myelin thickness in the corpus callosum between vehicle infused and BMP4-Noggin infused mice. Our results suggest that while single delivery of Noggin or IGF-1 increased the production of mature oligodendrocytes in vivo in the context of demyelination, only Noggin infusion promoted remyelination. Thus, sequential delivery of BMP4 and Noggin or IGF-1 does not further enhance myelin repair above what occurs with delivery of Noggin alone.


Subject(s)
Bone Morphogenetic Protein 4/administration & dosage , Demyelinating Diseases/drug therapy , Myelin Sheath/physiology , Animals , Astrocytes/drug effects , Bone Morphogenetic Protein 4/pharmacology , Carrier Proteins/administration & dosage , Cell Differentiation , Corpus Callosum/drug effects , Corpus Callosum/pathology , Cuprizone , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Drug Evaluation, Preclinical , Drug Therapy, Combination , Insulin-Like Growth Factor I/administration & dosage , Mice , Mice, Inbred C57BL , Neural Stem Cells/physiology , Oligodendroglia/drug effects , Oligodendroglia/physiology
18.
Nat Neurosci ; 15(9): 1192-4, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22885848

ABSTRACT

We found that, during the formation of the mouse barrel cortex, NG2 cells received glutamatergic synapses from thalamocortical fibers and preferentially accumulated along septa separating the barrels. Sensory deprivation reduced thalamocortical inputs on NG2 cells and increased their proliferation, leading to a more uniform distribution in the deprived barrels. Thus, early sensory experience regulates thalamocortical innervation on NG2 cells, as well as their proliferation and distribution during development.


Subject(s)
Neural Stem Cells/physiology , Somatosensory Cortex/physiology , Animals , Cell Count , Cell Proliferation , DNA-Binding Proteins , Darkness , Excitatory Postsynaptic Potentials/physiology , Glutamates/physiology , Immunohistochemistry , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Fluorescence , Nerve Fibers/physiology , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Oligodendroglia/physiology , Patch-Clamp Techniques , Thalamus/physiology , Vesicular Glutamate Transport Protein 2/physiology , Vibrissae/innervation , Vibrissae/physiology
19.
Stem Cells ; 29(12): 2062-76, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22038821

ABSTRACT

Adult spinal cord has little regenerative potential, thus limiting patient recovery following injury. In this study, we describe a new population of cells resident in the adult rat spinal cord meninges that express the neural stem/precursor markers nestin and doublecortin. Furthermore, from dissociated meningeal tissue a neural stem cell population was cultured in vitro and subsequently shown to differentiate into functional neurons or mature oligodendrocytes. Proliferation rate and number of nestin- and doublecortin-positive cells increased in vivo in meninges following spinal cord injury. By using a lentivirus-labeling approach, we show that meningeal cells, including nestin- and doublecortin-positive cells, migrate in the spinal cord parenchyma and contribute to the glial scar formation. Our data emphasize the multiple roles of meninges in the reaction of the parenchyma to trauma and indicate for the first time that spinal cord meninges are potential niches harboring stem/precursor cells that can be activated by injury. Meninges may be considered as a new source of adult stem/precursor cells to be further tested for use in regenerative medicine applied to neurological disorders, including repair from spinal cord injury.


Subject(s)
Intermediate Filament Proteins/metabolism , Meninges/metabolism , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuropeptides/metabolism , Spinal Cord Injuries/therapy , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Adult Stem Cells/physiology , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Doublecortin Domain Proteins , Doublecortin Protein , Electrophysiologic Techniques, Cardiac , Gene Expression Profiling , Intermediate Filament Proteins/genetics , Laminectomy , Lentivirus/genetics , Lentivirus/metabolism , Meninges/cytology , Meninges/physiology , Microtubule-Associated Proteins/genetics , Nerve Tissue Proteins/genetics , Nestin , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neurogenesis , Neuropeptides/genetics , Oligodendroglia/cytology , Oligodendroglia/metabolism , Oligodendroglia/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Regenerative Medicine , Stem Cell Niche
20.
Front Biosci (Landmark Ed) ; 16(7): 2695-733, 2011 06 01.
Article in English | MEDLINE | ID: mdl-21622204

ABSTRACT

Psychotropic treatments such as second generation or atypical antipsychotics are efficacious in a wide spectrum of psychiatric disorders ranging from schizophrenia to depression, bipolar disorder, and autism. These treatments are associated with peripheral metabolic derangements that are often also present in drug-naive patients. Furthermore, altering lipid composition/levels (with omega 3 fatty acids) and ameliorating oxidative toxicities may treat/prevent disease. The above observations are reexamined from the perspective of a myelin-centered model of the human brain. The model proposes that the human brain's extensive myelination required higher metabolic resources that caused evolutionary adaptations resulting in our quadratic (inverted U) myelination trajectory that peaks in the sixth decade of life. It further proposes that optimal brain function depends on exquisite action potential synchronization that myelin makes possible and that myelin's exceptional vulnerability to subtle metabolic/oxidative abnormalities may promote both developmental and degenerative diseases. Available data are integrated herein to suggest that widely used psychotropic treatments have under-appreciated CNS metabolic and neurotransmitter effects on myelination, its plasticity, and repair that may substantially contribute to their mechanisms of action.


Subject(s)
Bipolar Disorder/physiopathology , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/physiopathology , Mental Disorders/drug therapy , Mental Disorders/physiopathology , Psychotropic Drugs/therapeutic use , Schizophrenia/physiopathology , Animals , Epigenesis, Genetic , Humans , Lipid Metabolism , Models, Neurological , Myelin Sheath/genetics , Myelin Sheath/physiology , Nerve Net/physiology , Neuropharmacology , Neurotransmitter Agents/physiology , Oligodendroglia/physiology
SELECTION OF CITATIONS
SEARCH DETAIL