Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Front Cell Dev Biol ; 10: 878142, 2022.
Article in English | MEDLINE | ID: mdl-35517494

ABSTRACT

Axon-dendrite formation is a crucial milestone in the life history of neurons. During this process, historically referred as "the establishment of polarity," newborn neurons undergo biochemical, morphological and functional transformations to generate the axonal and dendritic domains, which are the basis of neuronal wiring and connectivity. Since the implementation of primary cultures of rat hippocampal neurons by Gary Banker and Max Cowan in 1977, the community of neurobiologists has made significant achievements in decoding signals that trigger axo-dendritic specification. External and internal cues able to switch on/off signaling pathways controlling gene expression, protein stability, the assembly of the polarity complex (i.e., PAR3-PAR6-aPKC), cytoskeleton remodeling and vesicle trafficking contribute to shape the morphology of neurons. Currently, the culture of hippocampal neurons coexists with alternative model systems to study neuronal polarization in several species, from single-cell to whole-organisms. For instance, in vivo approaches using C. elegans and D. melanogaster, as well as in situ imaging in rodents, have refined our knowledge by incorporating new variables in the polarity equation, such as the influence of the tissue, glia-neuron interactions and three-dimensional development. Nowadays, we have the unique opportunity of studying neurons differentiated from human induced pluripotent stem cells (hiPSCs), and test hypotheses previously originated in small animals and propose new ones perhaps specific for humans. Thus, this article will attempt to review critical mechanisms controlling polarization compiled over decades, highlighting points to be considered in new experimental systems, such as hiPSC neurons and human brain organoids.

2.
Biochim Biophys Acta Mol Basis Dis ; 1868(4): 166324, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34954343

ABSTRACT

BACKGROUND: Myelin-associated glycoprotein (MAG) is a key molecule involved in the nurturing effect of myelin on ensheathed axons. MAG also inhibits axon outgrowth after injury. In preclinical stroke models, administration of a function-blocking anti-MAG monoclonal antibody (mAb) aimed to improve axon regeneration demonstrated reduced lesion volumes and a rapid clinical improvement, suggesting a mechanism of immediate neuroprotection rather than enhanced axon regeneration. In addition, it has been reported that antibody-mediated crosslinking of MAG can protect oligodendrocytes (OLs) against glutamate (Glu) overload by unknown mechanisms. PURPOSE: To unravel the molecular mechanisms underlying the protective effect of anti-MAG therapy with a focus on neuroprotection against Glu toxicity. RESULTS: MAG activation (via antibody crosslinking) triggered the clearance of extracellular Glu by its uptake into OLs via high affinity excitatory amino acid transporters. This resulted not only in protection of OLs but also nearby neurons. MAG activation led to a PKC-dependent activation of factor Nrf2 (nuclear-erythroid related factor-2) leading to antioxidant responses including increased mRNA expression of metabolic enzymes from the glutathione biosynthetic pathway and the regulatory chain of cystine/Glu antiporter system xc- increasing reduced glutathione (GSH), the main antioxidant in cells. The efficacy of early anti-MAG mAb administration was demonstrated in a preclinical model of excitotoxicity induced by intrastriatal Glu administration and extended to a model of Experimental Autoimmune Encephalitis showing axonal damage secondary to demyelination. CONCLUSIONS: MAG activation triggers Glu uptake into OLs under conditions of Glu overload and induces a robust protective antioxidant response.


Subject(s)
Antibodies, Monoclonal/immunology , Glutamic Acid/metabolism , Myelin-Associated Glycoprotein/metabolism , Amino Acid Transport Systems, Acidic/genetics , Amino Acid Transport Systems, Acidic/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Axons/metabolism , Cells, Cultured , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Glutamic Acid/administration & dosage , Glutamic Acid/pharmacology , Glutathione/metabolism , Mice , Mice, Inbred C57BL , Myelin-Associated Glycoprotein/immunology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neurons/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Oxidative Stress/drug effects , Protein Kinase C/metabolism , Rats , Receptors, Glutamate/metabolism , Signal Transduction/drug effects
3.
Mol Ther ; 26(3): 730-743, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29433936

ABSTRACT

Analysis of microRNA (miR) expression in the central nervous system white matter of SJL mice infected with the BeAn strain of Theiler's murine encephalomyelitis virus (TMEV) revealed a significant reduction of miR-219, a critical regulator of myelin assembly and repair. Restoration of miR-219 expression by intranasal administration of a synthetic miR-219 mimic before disease onset ameliorates clinical disease, reduces neurogliosis, and partially recovers motor and sensorimotor function by negatively regulating proinflammatory cytokines and virus RNA replication. Moreover, RNA sequencing of host lesions showed that miR-219 significantly downregulated two genes essential for the biosynthetic cholesterol pathway, Cyp51 (lanosterol 14-α-demethylase) and Srebf1 (sterol regulatory element-binding protein-1), and reduced cholesterol biosynthesis in infected mice and rat CG-4 glial precursor cells in culture. The change in cholesterol biosynthesis had both anti-inflammatory and anti-viral effects. Because RNA viruses hijack endoplasmic reticulum double-layered membranes to provide a platform for RNA virus replication and are dependent on endogenous pools of cholesterol, miR-219 interference with cholesterol biosynthesis interfered virus RNA replication. These findings demonstrate that miR-219 inhibits TMEV-induced demyelinating disease through its anti-inflammatory and anti-viral properties.


Subject(s)
Cardiovirus Infections/complications , Cardiovirus Infections/virology , Demyelinating Diseases/etiology , Demyelinating Diseases/pathology , MicroRNAs/genetics , Theilovirus , Viral Load , Animals , Biomarkers , Cell Line , Cholesterol/metabolism , Cytokines/metabolism , Demyelinating Diseases/metabolism , Disease Models, Animal , Female , Fibrinogen/metabolism , Gene Expression Regulation , Inflammation Mediators/metabolism , Lipid Metabolism/genetics , Mice , Microglia/metabolism , RNA Interference , Rats
4.
PLoS One ; 12(5): e0178103, 2017.
Article in English | MEDLINE | ID: mdl-28531236

ABSTRACT

In prior studies, our laboratory showed that psychosine accumulates and disrupts lipid rafts in brain membranes of Krabbe's disease. A model of lipid raft disruption helped explaining psychosine's effects on several signaling pathways important for oligodendrocyte survival and differentiation but provided more limited insight in how this sphingolipid caused demyelination. Here, we have studied how this cationic inverted coned lipid affects the fluidity, stability and structure of myelin and plasma membranes. Using a combination of cutting-edge imaging techniques in non-myelinating (red blood cell), and myelinating (oligodendrocyte) cell models, we show that psychosine is sufficient to disrupt sphingomyelin-enriched domains, increases the rigidity of localized areas in the plasma membrane, and promotes the shedding of membranous microvesicles. The same physicochemical and structural changes were measured in myelin membranes purified from the mutant mouse Twitcher, a model for Krabbe's disease. Areas of higher rigidity were measured in Twitcher myelin and correlated with higher levels of psychosine and of myelin microvesiculation. These results expand our previous analyses and support, for the first time a pathogenic mechanism where psychosine's toxicity in Krabbe disease involves deregulation of cell signaling not only by disruption of membrane rafts, but also by direct local destabilization and fragmentation of the membrane through microvesiculation. This model of membrane disruption may be fundamental to introduce focal weak points in the myelin sheath, and consequent diffuse demyelination in this leukodystrophy, with possible commonality to other demyelinating disorders.


Subject(s)
Cell-Derived Microparticles/metabolism , Leukodystrophy, Globoid Cell/metabolism , Myelin Sheath/metabolism , Oligodendroglia/cytology , Psychosine/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Female , Humans , Male , Membrane Microdomains , Mice , Myelin Sheath/chemistry , Oligodendroglia/metabolism
5.
Dev Cell ; 40(6): 566-582.e5, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28350989

ABSTRACT

A lack of sufficient oligodendrocyte myelination contributes to remyelination failure in demyelinating disorders. miRNAs have been implicated in oligodendrogenesis; however, their functions in myelin regeneration remained elusive. Through developmentally regulated targeted mutagenesis, we demonstrate that miR-219 alleles are critical for CNS myelination and remyelination after injury. Further deletion of miR-338 exacerbates the miR-219 mutant hypomyelination phenotype. Conversely, miR-219 overexpression promotes precocious oligodendrocyte maturation and regeneration processes in transgenic mice. Integrated transcriptome profiling and biotin-affinity miRNA pull-down approaches reveal stage-specific miR-219 targets in oligodendrocytes and further uncover a novel network for miR-219 targeting of differentiation inhibitors including Lingo1 and Etv5. Inhibition of Lingo1 and Etv5 partially rescues differentiation defects of miR-219-deficient oligodendrocyte precursors. Furthermore, miR-219 mimics enhance myelin restoration following lysolecithin-induced demyelination as well as experimental autoimmune encephalomyelitis, principal animal models of multiple sclerosis. Together, our findings identify context-specific miRNA-regulated checkpoints that control myelinogenesis and a therapeutic role for miR-219 in CNS myelin repair.


Subject(s)
Central Nervous System/metabolism , Central Nervous System/pathology , MicroRNAs/metabolism , Myelin Sheath/metabolism , Myelin Sheath/pathology , Nerve Regeneration , Wound Healing , Animals , Cell Differentiation/drug effects , Cell Lineage/drug effects , Central Nervous System/drug effects , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Deletion , Lecithins/pharmacology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Multiple Sclerosis/therapy , Myelin Sheath/drug effects , Nerve Regeneration/drug effects , Nerve Regeneration/genetics , Nerve Tissue Proteins/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Optic Nerve/pathology , Optic Nerve/ultrastructure , Phenotype , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , Wound Healing/drug effects , Wound Healing/genetics
6.
J Neurosci Res ; 94(11): 1333-40, 2016 11.
Article in English | MEDLINE | ID: mdl-27638615

ABSTRACT

The discovery that most cells produce extracellular vesicles (EVs) and release them in the extracellular milieu has spurred the idea that these membranous cargoes spread pathogenic mechanisms. In the brain, EVs may have multifold and important physiological functions, from deregulating synaptic activity to promoting demyelination to changes in microglial activity. The finding that small EVs (exosomes) contain α-synuclein and ß-amyloid, among other pathogenic proteins, is an example of this notion, underscoring their potential role in the brains of patients with Parkinson's and Alzheimer's diseases. Given that they are membranous vesicles, we speculate that EVs also have an intrinsic capacity to incorporate sphingolipids. In conditions under which these lipids are elevated to toxic levels, such as in Krabbe's disease and metachromatic leukodystrophy, EVs may contribute to spread disease from sick to healthy cells. In this essay, we discuss a working hypothesis that brain cells in sphingolipidoses clear some of the accumulated lipid material to attempt restoring cell homeostasis via EV secretion. We hypothesize that secreted sphingolipid-loaded EVs shuttle pathogenic lipids to cells that are not intrinsically affected, contributing to establishing non-cell-autonomous defects. © 2016 Wiley Periodicals, Inc.


Subject(s)
Biological Transport/physiology , Brain/cytology , Cell Communication/physiology , Extracellular Vesicles/metabolism , Sphingolipids/metabolism , Animals , Humans , Models, Biological , Sphingolipidoses/pathology , Sphingolipids/toxicity
7.
J Neurosci Res ; 94(12): 1579-1587, 2016 12.
Article in English | MEDLINE | ID: mdl-27557608

ABSTRACT

Extracellular vesicles (EVs) are membrane nanovesicles of diverse sizes secreted by different cell types and are involved in intercellular communication. EVs shuttle proteins, nucleic acids, and lipids that reflect their cellular origin and could mediate their biological function in recipient cells. EVs circulate in biological fluids and are considered as potential biomarkers that could be used to analyze and characterize disease development, course and response to treatment. EVs exhibit specific distribution of glycolipids and membrane organization, but little is known about the biological significance of this distribution or how it could contribute to pathological conditions such as multiple sclerosis (MS). We provide the first description of sulfatide composition in plasma-derived EVs by ultra-high-performance liquid chromatography tandem mass spectrometry. We found that EVs of different sizes showed C16:0 sulfatide but no detectable levels of C18:0, C24:0, or C24:1 sulfatide species. Small EVs isolated at 100,000 × g-enriched in exosomes-from plasma of patients with MS showed a significant increase of C16:0 sulfatide compared with healthy controls. Nanoparticle tracking analysis showed that the particle size distribution in MS plasma was significantly different compared with healthy controls. Characterization of small EVs isolated from MS plasma showed similar protein content and similar levels of exosomal markers (Alix, Rab-5B) and vesicular marker MHC class I (major histocompatibility complex class I) compared with healthy controls. Our findings indicate that C16:0 sulfatide associated with small EVs is a candidate biomarker for MS that could potentially reflect pathological changes associated with this disease and/or the effects of its treatment. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cytoplasmic Vesicles/metabolism , Extracellular Vesicles/metabolism , Multiple Sclerosis/metabolism , Sulfoglycosphingolipids/metabolism , Adult , Biomarkers , Chromatography, High Pressure Liquid , Cytoplasmic Vesicles/chemistry , Extracellular Vesicles/chemistry , Female , Genes, MHC Class I , Humans , Male , Middle Aged , Multiple Sclerosis/blood , Nanoparticles/chemistry , Nanoparticles/metabolism , Particle Size , Sulfoglycosphingolipids/analysis , Sulfoglycosphingolipids/blood , Tandem Mass Spectrometry , Young Adult
8.
Anal Biochem ; 467: 31-9, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25205652

ABSTRACT

Sulfated galactosylceramides (sulfatides) are glycosphingolipids associated with cholesterol- and sphingolipid-enriched membrane microdomains (lipid rafts) and are highly expressed in brain tissue. Although it is known that sulfatide species show heterogeneity in their fatty acid acyl group composition throughout brain development, their lipid raft distribution and biological relevance is poorly understood. We validated a fast and sensitive ultra-high-pressure liquid chromatography tandem mass spectrometry (UHPLC-MS/MS) method to measure developmentally regulated sulfatide species (C16:0, C18:0, C24:1, and C24:0) in central nervous system (CNS) lipid rafts isolated without using detergent. Our UHPLC-MS/MS assay showed good accuracy and precision with a linear range of 5 to 1,000 nM for C18:0 and C24:1 sulfatides and 10 to 1,000 nM for C16:0 and C24:0 sulfatides. We applied this quantitative analysis to detergent-free lipid rafts isolated from wild-type mice and arylsulfatase A-deficient (ASA knockout) mice that accumulate sulfatides. All four sulfatide species were more abundant in raft membranes than in non-raft membranes, with a significant increase in lipid rafts isolated from ASA knockout mice. This is the first description of an analytical method to study these sulfatide species in raft and non-raft membranes and has the potential to be applied to preparations from other tissues.


Subject(s)
Brain/metabolism , Cerebroside-Sulfatase/physiology , Membrane Microdomains/metabolism , Sulfoglycosphingolipids/analysis , Animals , Animals, Newborn , Blotting, Western , Cholesterol/analysis , Chromatography, High Pressure Liquid , Mice , Mice, Knockout , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...