Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Oxid Med Cell Longev ; 2021: 5521503, 2021.
Article in English | MEDLINE | ID: mdl-33815654

ABSTRACT

BACKGROUND: Bu Shen Yi Sui capsule (BSYS) is a traditional Chinese medicine prescription that has shown antineuroinflammatory and neuroprotective effects in treating multiple sclerosis (MS) and its animal model of experimental autoimmune encephalomyelitis (EAE). Microglia play an important role in neuroinflammation. The M1 phenotype of microglia is involved in the proinflammatory process of the disease, while the M2 phenotype plays an anti-inflammatory role. Promoting the polarization of microglia to M2 in MS/EAE is a promising therapeutic strategy. This study is aimed at exploring the effects of BSYS on microglial polarization in mice with EAE. METHODS: The EAE model was established by the intraperitoneal injection of pertussis toxin and subcutaneous injection of myelin oligodendrocyte glycoprotein (MOG)35-55 in C57BL/6J mice. The mice were treated with BSYS (3.02 g/kg), FTY720 (0.3 mg/kg), or distilled water by intragastric administration. H&E and LFB staining, transmission electron microscopy, qRT-PCR, immunofluorescence, ELISA, fluorescence in situ hybridization, and western blotting were used to detect the histological changes in myelin, microglial M1/M2 polarization markers, and the expression of key genes involved in EAE. Results and Conclusions. BSYS treatment of EAE mice increased the body weight, decreased the clinical score, and reduced demyelination induced by inflammatory infiltration. BSYS also inhibited the mRNA expression of M1 microglial markers while increasing the mRNA level of M2 markers. Additionally, BSYS led to a marked decrease in the ratio of M1 microglia (iNOS+/Iba1+) and an obvious increase in the number of M2 microglia (Arg1+/Iba1+). In the EAE mouse model, miR-124 expression was decreased, and miR-155 expression was increased, while BSYS treatment significantly reversed this effect and modulated the levels of C/EBP α, PU.1, and SOCS1 (target genes of miR-124 and miR-155). Therefore, the neuroprotective effect of BSYS against MS/EAE was related to promoting microglia toward M2 polarization, which may be correlated with changes in miR-124 and miR-155 in vivo.


Subject(s)
Brain/pathology , Demyelinating Diseases/genetics , Drugs, Chinese Herbal/pharmacology , Encephalomyelitis, Autoimmune, Experimental/genetics , Inflammation/pathology , MicroRNAs/metabolism , Microglia/pathology , Animals , Body Weight/drug effects , CCAAT-Enhancer-Binding Proteins/metabolism , Capsules , Cell Differentiation/drug effects , Cell Polarity/drug effects , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/pathology , Exosomes/metabolism , Female , Inflammation/genetics , Mice, Inbred C57BL , MicroRNAs/blood , MicroRNAs/genetics , Oligodendroglia/drug effects , Oligodendroglia/pathology , Phenotype , Proto-Oncogene Proteins/metabolism , Spinal Cord/pathology , Trans-Activators/metabolism , Up-Regulation/genetics
2.
Biochim Biophys Acta Mol Basis Dis ; 1866(7): 165779, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32224154

ABSTRACT

Oligodendrocytes not only produce myelin to facilitate nerve impulse conduction, but are also essential metabolic partners of the axon. Oligodendrocyte loss and myelin destruction, as occurs in multiple sclerosis (MS), leaves axons vulnerable to degeneration and permanent neurological deficits ensue. Many studies now propose that lifestyle factors such as diet may impact demyelinating conditions, including MS. Most prior reviews have focused on the regulatory role of diet in the inflammatory events that drive MS pathogenesis, however the potential for dietary factors to modulate oligodendrocyte biology, myelin injury and myelin regeneration remain poorly understood. Here we review the current evidence from clinical and animal model studies regarding the impact of diet or dietary factors on myelin integrity and other pathogenic features of MS. Some limited evidence exists that certain foods may decrease risk or influence the progression of MS, such as increased intake of fish or polyunsaturated fatty acids, caloric restriction and fasting-mimicking diets. In addition, evidence suggests adolescent obesity or insufficient vitamin D levels increase the risk for developing MS. However, no clear or consistent evidence exists that dietary components exacerbate disease progression. Cumulatively, current evidence highlights the need for more extensive clinical trials to validate dietary effects on MS and to identify diets or supplements that may be beneficial as food-based strategies in the management of MS alone or in combination with conventional disease modifying therapies.


Subject(s)
Central Nervous System/metabolism , Demyelinating Diseases/metabolism , Multiple Sclerosis/etiology , Myelin Sheath/metabolism , Animals , Axons/metabolism , Axons/pathology , Central Nervous System/injuries , Central Nervous System/pathology , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Dietary Supplements , Disease Models, Animal , Humans , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Myelin Sheath/drug effects , Nerve Regeneration/genetics , Oligodendroglia/metabolism , Oligodendroglia/pathology
3.
Med Sci Monit ; 25: 9679-9689, 2019 Dec 18.
Article in English | MEDLINE | ID: mdl-31848329

ABSTRACT

BACKGROUND The aim of this study was to determine the association between white matter lesions (WML) and diabetes-associated cognitive decline (DACD) in rat models of type 2 diabetes (T2DM). MATERIAL AND METHODS Sixty Sprague-Dawley male rats were divided into 4 groups: control, control+metformin, T2DM, and T2DM+metformin groups. The T2DM groups were fed a diet high in fat and glucose to induce impaired glucose tolerance (IGT) and then were injected with streptozotocin to induce T2DM. The Morris water maze test was used to evaluate cognitive function. Brain diffusion tensor imaging scans were performed for WML. The expression of myelin basic protein (MBP), oligodendrocyte transcription factor 1 (OLIG1), and OLIG2 (markers of brain damage and repair) was determined using immunofluorescence. After IGT, the fractional anisotropy (FA) values of the right thalamus area were significantly lower in both T2DM groups compared with controls. RESULTS Eight weeks after streptozotocin injection, the FA values of the thalamus were lower in the T2DM (bilateral thalamus) group and T2DM+metformin (left thalamus) group than in controls, while the FA values in the left thalamus area were lower in the T2DM+metformin group than in the control and control+metformin groups. The maze escape latency was longer and the number of rats passing through the platform was smaller in the T2DM and T2DM+metformin groups than in the control group. MBP levels were lower and OLIG1 and OLIG2 levels were higher in both T2DM groups than in controls. CONCLUSIONS WML is associated with DACD and appears before the onset of T2DM and signs of DACD and plays a role in diabetes-associated cognitive decline. Metformin reduces WMLs but does not rescue cognitive dysfunction.


Subject(s)
Cognitive Dysfunction/complications , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Prediabetic State/complications , White Matter/pathology , Animals , Anisotropy , Cognitive Dysfunction/physiopathology , Demyelinating Diseases/complications , Demyelinating Diseases/pathology , Demyelinating Diseases/physiopathology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Diffusion Tensor Imaging , Disease Models, Animal , Male , Maze Learning , Nerve Tissue Proteins/metabolism , Oligodendroglia/pathology , Prediabetic State/physiopathology , Rats, Sprague-Dawley , Swimming , Thalamus/pathology , Thalamus/physiopathology , White Matter/physiopathology
4.
Biomed Res Int ; 2019: 5249675, 2019.
Article in English | MEDLINE | ID: mdl-31687391

ABSTRACT

The broad spectrum of disabilities caused by white matter injury (WMI) cannot be explained simply by hypomyelination. Synaptic injury in the thalamus may be related to disabilities in WMI survivors. Neuronal injury in the thalamus has been found most commonly in autopsy cases of preterm WMI. We hypothesized that hypoxia/ischemia (HI) in neonatal rats results in synaptic abnormalities in the thalamus that contribute to disabilities in WMI survivors. We examined changes in synapses in a neonatal rat model of HI-induced WMI. Right common carotid artery ligation and hypoxia (8% oxygen for 2.5 hours (h)) were performed in three-day-old Sprague-Dawley rats. We found HI rats performed worse in the Morris water maze test than sham rats, suggesting long-term cognition impairment after HI injury. A loss of synapses in the thalamus accompanied by hypomyelination and oligodendrocytes (OLs) reduction was observed. At the ultrastructural level, reductions in active zone (AZ) length and postsynaptic density (PSD) thickness were detected at 2 weeks after HI exposure. Furthermore, increased expression of synaptophysin and PSD-95 in both groups was observed from 3 days (d) to 21 d after hypoxic/ischemic (HI) injury. PSD-95 expression was significantly lower in HI rats than in sham rats from 14 d to 21 d after HI injury, and synaptophysin expression was significantly lower in HI rats from 7 d to 14 d after HI injury. However, no significant difference in synaptophysin expression was observed between HI rats and sham rats at 21 d after HI injury. The results demonstrated synaptic abnormalities in the thalamus accompanied by hypomyelination in WMI in response to HI exposure, which may contribute to the diverse neurological defects observed in WMI patients. Although synaptic reorganization occurred as a compensatory response to HI injury, the impairments in synaptic transmission were not reversed.


Subject(s)
Brain Injuries/pathology , Hypoxia-Ischemia, Brain/pathology , Hypoxia/pathology , Synapses/pathology , Thalamus/pathology , White Matter/pathology , Animals , Animals, Newborn , Disease Models, Animal , Maze Learning/physiology , Oligodendroglia/pathology , Post-Synaptic Density/pathology , Rats , Rats, Sprague-Dawley , Synaptic Transmission/physiology
5.
BMC Neurosci ; 20(1): 33, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31291887

ABSTRACT

BACKGROUND: Hypothalamic inflammation including astrogliosis and microglia activation occurs after intake of high fat diet (HFD) in rodent models or in obese individuals. However, the effect of chronic HFD feeding on oligodendrocytes (OLGs), a myelin-producing glial population in the central nervous system (CNS), remains unclear. In this study, we used 8-week old male C57BL/6 mice fed by HFD for 3-6 months to induce chronic obesity. RESULTS: The transmission electron microscopy imaging analysis showed that the integrity of hypothalamic myelin was disrupted after HFD feeding for 4 and 6 months. Moreover, the accumulation of Iba1+-microglia with an amoeboid hypertrophic form was continually observed in arcuate nucleus of HFD-fed mice during the entire feeding time period. Interleukin-33 (IL-33), a tissue alarmin upon injury to the CNS, was detected with an increased level in hypothalamus after HFD feeding for 3 and 4 months. Furthermore, the in vitro study indicated that exposure of mature OLGs to IL-33 impaired OLG cell structure along with a decline in the expression of myelin basic protein. CONCLUSIONS: Altogether, our findings demonstrate that chronic HFD feeding triggers hypothalamic myelin disruption in accompany with IL-33 upregulation and prolonged microglial activation in hypothalamus. Given that the addition of exogenous IL-33 was harmful for the maturation of OLGs, an increase in IL-33 by chronic HFD feeding might contribute to the induction of hypothalamic myelin disruption.


Subject(s)
Diet, High-Fat/adverse effects , Hypothalamus/metabolism , Interleukin-33/metabolism , Myelin Sheath/pathology , Up-Regulation , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/pathology , Hypothalamus/pathology , Male , Mice , Myelin Basic Protein/biosynthesis , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Primary Cell Culture , Rats , Time Factors
6.
Neurotox Res ; 36(1): 144-162, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31049881

ABSTRACT

A murine model used to investigate the osmotic demyelination syndrome (ODS) demonstrated ultrastructural damages in thalamus nuclei. Following chronic hyponatremia, significant myelinolysis was merely detected 48 h after the rapid reinstatement of normonatremia (ODS 48 h). In ODS samples, oligodendrocytes and astrocytes revealed injurious changes associated with a few cell deaths while both cell types seemed to endure a sort of survival strategy: (a) ODS 12 h oligodendrocytes displayed nucleoplasm with huge heterochromatic compaction, mitochondria hypertrophy, and most reclaimed an active NN cell aspect at ODS 48 h. (b) Astrocytes responded to the osmotic stress by overall cell shrinkage with clasmatodendrosis, these changes accompanied nucleus wrinkling, compacted and segregated nucleolus, destabilization of astrocyte-oligodendrocyte junctions, loss of typical GFAP filaments, and detection of round to oblong woolly, proteinaceous aggregates. ODS 48 h astrocytes regained an active nucleus aspect, without restituting GFAP filaments and still contained cytoplasmic proteinaceous deposits. (c) Sustaining minor shrinking defects at ODS 12 h, neurons showed slight axonal injury. At ODS 48 h, neuron cell bodies emerged again with deeply indented nucleus and, owing nucleolus translational activation, huge amounts of polysomes along with secretory-like activities. (d) In ODS, activated microglial cells got stuffed with huge lysosome bodies out of captures cell damages, leaving voids in interfascicular and sub-vascular neuropil. Following chronic hyponatremia, the murine thalamus restoration showed macroglial cells acutely turned off transcriptional and translational activities during ODS and progressively recovered activities, unless severely damaged cells underwent cell death, leading to neuropil disruption and demyelination.


Subject(s)
Demyelinating Diseases/pathology , Osmotic Pressure , Thalamus/pathology , Thalamus/ultrastructure , Animals , Astrocytes/pathology , Astrocytes/ultrastructure , Axons/pathology , Axons/ultrastructure , Demyelinating Diseases/etiology , Disease Models, Animal , Hyponatremia/complications , Hyponatremia/pathology , Male , Mice, Inbred C57BL , Neurons/pathology , Neurons/ultrastructure , Oligodendroglia/pathology , Oligodendroglia/ultrastructure
7.
Exp Physiol ; 104(6): 876-886, 2019 06.
Article in English | MEDLINE | ID: mdl-30811744

ABSTRACT

NEW FINDINGS: What is the central question of this study? Oligomeric proanthocyanidin has the capacity to alleviate abnormalities in neurological functioning. However, whether oligomeric proanthocyanidin can reduce the progression of demyelination or promote remyelination in demyelinating diseases remains unknown. What is the main finding and its importance? Oligomeric proanthocyanidin can improve cuprizone-induced demyelination by inhibiting immune cell infiltration, reversing overactivated microglia, decreasing the inflammatory cytokines secreted by inflammatory cells and decreasing the production of myelin oligodendrocyte glycoprotein35-55 -specific antibody in the brain. ABSTRACT: Demyelinating diseases of the CNS, including multiple sclerosis, neuromyelitis optica and acute disseminated encephalomylitis, are characterized by recurrent primary demyelination-remyelination and progressive neurodegeneration. In the present study, we investigated the therapeutic effect of oligomeric proanthocyanidin (OPC), the most effective component of grape seed extract, in cuprizone-fed C57BL/6 mice, a classic demyelination-remyelination model. Our results showed that OPC attenuated abnormal behaviour, reduced demyelination and increased expression of myelin basic protein and expression of O4+ oligodendrocytes in the corpus callosum. Oligomeric proanthocyanidin also reduced the numbers of B and T cells, activated microglia in the corpus callosum and inhibited secretion of inflammatory factors. Furthermore, concentrations of myelin oligodendrocyte glycoprotein-specific antibodies were significantly reduced in serum and brain homogenates after OPC treatment. Together, these results demonstrate a potent therapeutic effect for OPC in cuprizone-mediated demyelination and clearly highlight multiple effects of this natural product in attenuating myelin-specific autoantibodies and the inflammatory microenvironment in the brain.


Subject(s)
Corpus Callosum/drug effects , Demyelinating Diseases/drug therapy , Oligodendroglia/drug effects , Proanthocyanidins/therapeutic use , Animals , Corpus Callosum/pathology , Cuprizone , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Disease Models, Animal , Mice , Oligodendroglia/pathology , Proanthocyanidins/pharmacology , Treatment Outcome
8.
Folia Morphol (Warsz) ; 78(3): 564-574, 2019.
Article in English | MEDLINE | ID: mdl-30402879

ABSTRACT

BACKGROUND: Humans are widely exposed to acrylamide (ACR) and its neurotoxicity is a significant public health issue attracting wide attention. The aim of the study was to investigate ACR-induced adverse cerebellar changes in rats and study the possible oligodendrogenic effect of omega 3 and green tea. MATERIALS AND METHODS: Twenty-four adult albino rats weighing 150-200 g were randomly divided into four equal groups (6 rats each): control group (Group I), the rats that received ACR 45 mg/kg/day (Group II), the rats that received ACR concomitant with omega 3 at a dosage of 200 mg/kg/day (Group III), the rats that received ACR concomitant with green tea dissolved in drinking water at a dosage of 5 g/L (Group IV). The rats were euthanized after 8 weeks of the experiment. Malondialdehyde (MDA) and glutathione (GSH) were measured in cerebellar homogenates. Sections of 5 µm thickness from specimens from the cerebellum were stained with haematoxylin and eosin, silver stain and immunohistochemical stains: platelet-derived growth factor alpha (PDGFα; for oligodendrocytes), glial fibrillary acidic protein (GFAP; for astrocytes) and BCL2 (antiapoptotic). RESULTS: Omega 3 and green tea had improved MDA and GSH as compared to the ACR group. Histologically, the ACR group showed variable degrees of cellular degeneration. Omega 3 had induced oligodendrogenesis in Group III. The optical density of silver stain was significantly (p < 0.05) increased in Groups III and IV as compared to the ACR group. Area per cent of positive PDGFα was significantly increased in the ACR + omega 3 group as compared to the ACR group. Area per cent of positive GFAP was significantly decreased in Groups III and IV as compared to the ACR group. Area per cent of positive BCL2 was significantly increased in the omega 3-trated group as compared to the ACR group. CONCLUSIONS: Concomitant administration of omega 3 or green tea with ACR might mitigate the adverse cerebellar changes caused by ACR thanks to an oligodendrogenic effect of omega 3.


Subject(s)
Acrylamide/toxicity , Cerebellum/pathology , Fatty Acids, Omega-3/pharmacology , Oligodendroglia/pathology , Tea/chemistry , Animals , Antioxidants/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Biomarkers/metabolism , Body Weight/drug effects , Cerebellum/drug effects , Male , Oligodendroglia/drug effects , PC12 Cells , Proto-Oncogene Proteins c-bcl-2/metabolism , Purkinje Cells/drug effects , Purkinje Cells/pathology , Rats
9.
Glia ; 66(12): 2589-2603, 2018 12.
Article in English | MEDLINE | ID: mdl-30325069

ABSTRACT

Gap junctions (GJs) coupling oligodendrocytes to astrocytes and to other oligodendrocytes are formed mainly by connexin47 (Cx47) and a smaller portion by connexin32 (Cx32). Mutations in both connexins cause inherited demyelinating disorders, but their expression is also disrupted in multiple sclerosis (MS). To clarify whether the loss of either Cx47 or Cx32 could modify the outcome of inflammation and myelin loss, we induced experimental autoimmune encephalomyelitis (EAE) in fully backcrossed Cx32 knockout (KO) and Cx47KO mice and compared their outcome with wild type (WT, C57BI/6 N) mice. Cx47KO EAE mice developed the most severe phenotype assessed by clinical scores and behavioral testing, followed by Cx32KO and WT mice. Cx47KO more than Cx32KO EAE mice developed more microglial activation, myelin, and axonal loss than did WT mice. Oligodendrocyte apoptosis and precursor proliferation was also higher in Cx47KO than in Cx32KO or WT EAE mice. Similarly, blood-spinal cord barrier (BSCB) disruption and inflammatory infiltrates of macrophages, T- and B-cells were more severe in Cx47KO than either Cx32KO or WT EAE groups. Finally, expression profiling revealed that several proinflammatory cytokines were higher at the peak of inflammation in the Cx47KO mice and persisted at later stages of EAE in contrast to reduction of their levels in WT EAE mice. Thus, loss of oligodendrocyte GJs aggravates BSCB disruption and inflammatory myelin loss, likely due to dysregulation of proinflammatory cytokines. This mechanism may play an important role in MS brain with reduced connexin expression, as well as in patients with inherited mutations in oligodendrocyte connexins and secondary inflammation.


Subject(s)
Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Gap Junctions/metabolism , Gene Expression Regulation/physiology , Hand Strength/physiology , Oligodendroglia/metabolism , Animals , Apoptosis/genetics , Astrocytes/metabolism , Astrocytes/pathology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Calcium-Binding Proteins/metabolism , Cell Proliferation/genetics , Connexins/genetics , Connexins/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Freund's Adjuvant/toxicity , Gap Junctions/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Macrophages/pathology , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Motor Activity/drug effects , Motor Activity/genetics , Myelin-Oligodendrocyte Glycoprotein/toxicity , Oligodendroglia/pathology , Peptide Fragments/toxicity , Gap Junction beta-1 Protein
10.
J Neuroinflammation ; 15(1): 121, 2018 Apr 24.
Article in English | MEDLINE | ID: mdl-29690885

ABSTRACT

BACKGROUND: Tumor necrosis factor (TNF) is associated with several neurodegenerative disorders including multiple sclerosis (MS). Although TNF-targeted therapies have been largely unsuccessful in MS, recent preclinical data suggests selective soluble TNF inhibition can promote remyelination. This has renewed interest in regulation of TNF signaling in demyelinating disease, especially given the limited treatment options for progressive MS. Using a mouse model of progressive MS, this study evaluates the effects of sustained TNF on oligodendrocyte (OLG) apoptosis and OLG precursor cell (OPC) differentiation. METHODS: Induction of experimental autoimmune encephalomyelitis (EAE) in transgenic mice expressing a dominant-negative interferon-γ receptor under the human glial fibrillary acidic protein promoter (GFAPγR1Δ) causes severe non-remitting disease associated with sustained TNF. Therapeutic effects in GFAPγR1Δ mice treated with anti-TNF compared to control antibody during acute EAE were evaluated by assessing demyelinating lesion size, remyelination, OLG apoptosis, and OPC differentiation. RESULTS: More severe and enlarged demyelinating lesions in GFAPγR1Δ compared to wild-type (WT) mice were associated with increased OLG apoptosis and reduced differentiated CC1+Olig2+ OLG within lesions, as well as impaired upregulation of TNF receptor-2, suggesting impaired OPC differentiation. TNF blockade during acute EAE in GFAPγR1Δ both limited OLG apoptosis and enhanced OPC differentiation consistent with reduced lesion size and clinical recovery. TNF neutralization further limited increasing endothelin-1 (ET-1) expression in astrocytes and myeloid cells noted in lesions during disease progression in GFAPγR1Δ mice, supporting inhibitory effects of ET-1 on OPC maturation. CONCLUSION: Our data implicate that IFNγ signaling to astrocytes is essential to limit a detrimental positive feedback loop of TNF and ET-1 production, which increases OLG apoptosis and impairs OPC differentiation. Interference of this cycle by TNF blockade promotes repair independent of TNFR2 and supports selective TNF targeting to mitigate progressive forms of MS.


Subject(s)
Antibodies/therapeutic use , Apoptosis/genetics , Encephalomyelitis, Autoimmune, Experimental , Oligodendroglia/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Calcium-Binding Proteins/metabolism , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Freund's Adjuvant/toxicity , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Mutation/genetics , Nerve Tissue Proteins/metabolism , Tumor Necrosis Factor-alpha/immunology , beta-Galactosidase/metabolism
11.
Glia ; 66(3): 538-561, 2018 03.
Article in English | MEDLINE | ID: mdl-29148104

ABSTRACT

Oligodendroglial cell death and demyelination are hallmarks of neurotrauma and multiple sclerosis that cause axonal damage and functional impairments. Remyelination remains a challenge as the ability of endogenous precursor cells for oligodendrocyte replacement is hindered in the unfavorable milieu of demyelinating conditions. Here, in a rat model of lysolecithin lysophosphatidyl-choline (LPC)-induced focal demyelination, we report that Neuregulin-1 (Nrg-1), an important factor for oligodendrocytes and myelination, is dysregulated in demyelinating lesions and its bio-availability can promote oligodendrogenesis and remyelination. We delivered recombinant human Nrg-1ß1 (rhNrg-1ß1) intraspinally in the vicinity of LPC demyelinating lesion in a sustained manner using poly lactic-co-glycolic acid microcarriers. Availability of Nrg-1 promoted generation and maturation of new oligodendrocytes, and accelerated endogenous remyelination by both oligodendrocyte and Schwann cell populations in demyelinating foci. Importantly, Nrg-1 enhanced myelin thickness in newly remyelinated spinal cord axons. Our complementary in vitro studies also provided direct evidence that Nrg-1 significantly promotes maturation of new oligodendrocytes and facilitates their transition to a myelinating phenotype. Nrg-1 therapy remarkably attenuated the upregulated expression chondroitin sulfate proteoglycans (CSPGs) specific glycosaminoglycans in the extracellular matrix of demyelinating foci and promoted interleukin-10 (IL-10) production by immune cells. CSPGs and IL-10 are known to negatively and positively regulate remyelination, respectively. We found that Nrg-1 effects are mediated through ErbB2 and ErbB4 receptor activation. Our work provides novel evidence that dysregulated levels of Nrg-1 in demyelinating lesions of the spinal cord pose a challenge to endogenous remyelination, and appear to be an underlying cause of myelin thinning in newly remyelinated axons.


Subject(s)
Demyelinating Diseases/therapy , Immunomodulation , Neuregulin-1/administration & dosage , Neuroprotective Agents/administration & dosage , Remyelination/physiology , Spinal Cord/immunology , Animals , Cells, Cultured , Chondroitin Sulfate Proteoglycans/metabolism , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Disease Models, Animal , Drug Carriers , Extracellular Matrix/immunology , Extracellular Matrix/pathology , Female , Ganglia, Spinal/immunology , Ganglia, Spinal/pathology , Humans , Lactic Acid , Male , Neural Stem Cells/immunology , Neural Stem Cells/pathology , Oligodendroglia/immunology , Oligodendroglia/pathology , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Spinal Cord/pathology , Spinal Cord Diseases/immunology , Spinal Cord Diseases/pathology , Spinal Cord Diseases/therapy
12.
Dev Cell ; 42(1): 52-67.e4, 2017 07 10.
Article in English | MEDLINE | ID: mdl-28697333

ABSTRACT

The childhood-onset motor disorder DYT6 dystonia is caused by loss-of-function mutations in the transcription factor THAP1, but the neurodevelopmental processes in which THAP1 participates are unknown. We find that THAP1 is essential for the timing of myelination initiation during CNS maturation. Conditional deletion of THAP1 in the CNS retards maturation of the oligodendrocyte (OL) lineage, delaying myelination and causing persistent motor deficits. The CNS myelination defect results from a cell-autonomous requirement for THAP1 in the OL lineage and is recapitulated in developmental assays performed on OL progenitor cells purified from Thap1 null mice. Loss of THAP1 function disrupts a core set of OL maturation genes and reduces the DNA occupancy of YY1, a transcription factor required for OL maturation. These studies establish a role for THAP1 transcriptional regulation at the inception of myelination and implicate abnormal timing of myelination in the pathogenesis of childhood-onset dystonia.


Subject(s)
Cell Lineage , DNA-Binding Proteins/metabolism , Dystonia/metabolism , Dystonia/pathology , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Animals , Cell Differentiation , Central Nervous System/pathology , Chromatin Immunoprecipitation , DNA-Binding Proteins/deficiency , Dystonia/genetics , Dystonia/physiopathology , Gene Deletion , Gene Expression Regulation , Mice, Knockout , Motor Activity , Stem Cells/metabolism , YY1 Transcription Factor/metabolism
13.
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
14.
Brain Res ; 1657: 130-139, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27956122

ABSTRACT

Iron deficiency has a critical impact on maturational mechanisms of the brain and the damage related to neuroanatomical parameters is not satisfactorily reversed after iron replacement. However, emerging evidence suggest that enriched early experience may offer great therapeutic efficacy in cases of nutritional disorders postnatally, since the brain is remarkably responsive to its interaction with the environment. Given the fact that tactile stimulation (TS) treatment has been previously shown to be an effective therapeutic approach and with potential application to humans, here we ask whether exposure to TS treatment, from postnatal day (P) 1 to P32 for 3min/day, could also be employed to prevent neuroanatomical changes in the optic nerve of rats maintained on an iron-deficient diet during brain development. We found that iron deficiency changed astrocyte, oligodendrocyte, damaged fiber, and myelinated fiber density, however, TS reversed the iron-deficiency-induced alteration in oligodendrocyte, damaged fiber and myelinated fiber density, but failed to reverse astrocyte density. Our results suggest that early iron deficiency may act by disrupting the timing of key steps in visual system development thereby modifying the normal progression of optic nerve maturation. However, optic nerve development is sensitive to enriching experiences, and in the current study we show that this sensitivity can be used to prevent damage from postnatal iron deficiency during the critical period.


Subject(s)
Iron Deficiencies , Musculoskeletal Manipulations , Optic Nerve/growth & development , Visual Pathways/growth & development , Animals , Animals, Newborn , Astrocytes/metabolism , Astrocytes/pathology , Body Weight , Diet , Disease Models, Animal , Handling, Psychological , Male , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Neuroprotection , Oligodendroglia/metabolism , Oligodendroglia/pathology , Optic Nerve/blood supply , Optic Nerve/metabolism , Optic Nerve/pathology , Physical Stimulation , Random Allocation , Rats, Wistar , Visual Pathways/blood supply , Visual Pathways/metabolism , Visual Pathways/pathology
15.
Sci Rep ; 6: 33736, 2016 09 21.
Article in English | MEDLINE | ID: mdl-27650712

ABSTRACT

The aim of the present study was to examine involvement of MBD3 (methyl-CpG-binding domain protein 3), a protein involved in reading DNA methylation patterns, in epileptogenesis and epilepsy. We used a well-characterized rat model of temporal lobe epilepsy that is triggered by status epilepticus, evoked by electrical stimulation of the amygdala. Stimulated and sham-operated animals were sacrificed 14 days after stimulation. We found that MBD3 transcript was present in neurons, oligodendrocytes, and astrocytes in both control and epileptic animals. We detected the nuclear localization of MBD3 protein in neurons, mature oligodendrocytes, and a subpopulation of astrocytes but not in microglia. Amygdala stimulation significantly increased the level of MBD3 immunofluorescence. Immunoprecipitation followed by mass spectrometry and Western blot revealed that MBD3 in the adult brain assembles the NuRD complex, which also contains MTA2, HDAC2, and GATAD2B. Using chromatin immunoprecipitation combined with deep sequencing, we observed differences in the occupancy of DNA regions by MBD3 protein between control and stimulated animals. This was not followed by subsequent changes in the mRNA expression levels of selected MBD3 targets. Our data demonstrate for the first time alterations in the MBD3 expression and DNA occupancy in the experimental model of epilepsy.


Subject(s)
Amygdala/metabolism , DNA-Binding Proteins/biosynthesis , DNA/metabolism , Epilepsy, Temporal Lobe/metabolism , Gene Expression Regulation , Neurons/metabolism , Oligodendroglia/metabolism , Amygdala/pathology , Animals , Disease Models, Animal , Electric Stimulation Therapy , Epilepsy, Temporal Lobe/pathology , Epilepsy, Temporal Lobe/therapy , Humans , Male , Neurons/pathology , Oligodendroglia/pathology , Protein Binding , Rats , Rats, Sprague-Dawley
16.
Brain Res ; 1639: 28-37, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26944297

ABSTRACT

We investigated the effects of auraptene on mouse oligodendroglial cell lineage in an animal model of demyelination induced by cuprizone. Auraptene, a citrus coumarin, was intraperitoneally administered to mice fed the demyelinating agent cuprizone. Immunohistochemical analysis of the corpus callosum and/or Western blotting analysis of brain extracts revealed that cuprizone reduced immunoreactivity for myelin-basic protein, a marker of myelin, whereas it increased immunoreactivity to platelet derived-growth factor receptor-α, a marker of oligodendrocyte precursor cells. Administration of auraptene enhanced the immunoreactivity to oligodendrocyte transcription factor 2, a marker of oligodendrocyte precursor cells and oligodendrocyte lineage precursor cells, but had no effect on immunoreactivity to myelin-basic protein or platelet-derived growth factor receptor-α. These findings suggest that auraptene promotes the production of oligodendrocyte lineage precursor cells in an animal model of demyelination and may be useful for individuals with demyelinating diseases.


Subject(s)
Coumarins/pharmacology , Demyelinating Diseases/drug therapy , Neuroprotective Agents/pharmacology , Oligodendroglia/drug effects , Stem Cells/drug effects , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/drug effects , Brain/metabolism , Brain/pathology , Cuprizone , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Gene Expression/drug effects , Injections, Intraperitoneal , Male , Mice, 129 Strain , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Myelin Basic Protein/metabolism , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/metabolism , Oligodendroglia/pathology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Stem Cells/physiology
17.
Exp Mol Pathol ; 100(2): 337-43, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26681653

ABSTRACT

Inflammation, demyelination, oligodendrocyte (OLG) death, and axonal degeneration are primary characteristics of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). OLGs generate myelin sheaths that surround axons, while damage to OLGs leads to demyelination and neurological functional deficit. Matrine (MAT), a quinolizidine alkaloid derived from the herb Radix Sophorae Flave, has been recently found to effectively ameliorate clinical signs in EAE. Its therapeutic mechanism has, however, not been completely elucidated. In the present study, we found that MAT retarded the disease process, attenuated the clinical severity of EAE rats, ameliorated inflammation and demyelination, and suppressed the apoptosis of OLGs in the central nervous system (CNS) of EAE rats. In addition, MAT markedly blocked increased expression of the proNGF-p75(NTR) death signaling complex, which is known to mediate OLG death in EAE animals. At the same time, MAT also prevented a decrease in the levels of NGF and its receptor TrkA, which together mediate the cell survival pathway and differentiation of OLGs. ProNGF, NGF, and the downstream effector proteins play an important role in the growth, differentiation, and apoptosis of OLGs as well as the reparative response to neuronal damage. These findings thus indicate that MAT improves clinical severity of EAE in part by reducing OLG apoptosis via restoring the ratios of proNGF:NGF and the respective receptors p75(NTR):TrkA in vivo. Taken together, these results suggest that MAT may be a promising agent for MS treatment based on its protective effect on OLGs.


Subject(s)
Alkaloids/pharmacology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Nerve Growth Factors/metabolism , Protein Precursors/metabolism , Quinolizines/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Axons/drug effects , Cell Survival/drug effects , Central Nervous System/drug effects , Central Nervous System/metabolism , Central Nervous System/pathology , Demyelinating Diseases/metabolism , Demyelinating Diseases/prevention & control , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Fluorescent Antibody Technique , Immunohistochemistry , Neuroprotective Agents/pharmacology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/pathology , Phytotherapy/methods , Rats, Wistar , Sophora/chemistry , Matrines
18.
Neurobiol Dis ; 82: 504-515, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26388399

ABSTRACT

Spinal cord injury leads to major neurological impairment for which there is currently no effective treatment. Recent clinical trials have demonstrated the efficacy of Fortasyn® Connect in Alzheimer's disease. Fortasyn® Connect is a specific multi-nutrient combination containing DHA, EPA, choline, uridine monophosphate, phospholipids, and various vitamins. We examined the effect of Fortasyn® Connect in a rat compression model of spinal cord injury. For 4 or 9 weeks following the injury, rats were fed either a control diet or a diet enriched with low, medium, or high doses of Fortasyn® Connect. The medium-dose Fortasyn® Connect-enriched diet showed significant efficacy in locomotor recovery after 9 weeks of supplementation, along with protection of spinal cord tissue (increased neuronal and oligodendrocyte survival, decreased microglial activation, and preserved axonal integrity). Rats fed the high-dose Fortasyn® Connect-enriched diet for 4 weeks showed a much greater enhancement of locomotor recovery, with a faster onset, than rats fed the medium dose. Bladder function recovered quicker in these rats than in rats fed the control diet. Their spinal cord tissues showed a smaller lesion, reduced neuronal and oligodendrocyte loss, decreased neuroinflammatory response, reduced astrocytosis and levels of inhibitory chondroitin sulphate proteoglycans, and better preservation of serotonergic axons than those of rats fed the control diet. These results suggest that this multi-nutrient preparation has a marked therapeutic potential in spinal cord injury, and raise the possibility that this original approach could be used to support spinal cord injured patients.


Subject(s)
Dietary Supplements , Docosahexaenoic Acids , Eicosapentaenoic Acid , Phospholipids , Spinal Cord Injuries/diet therapy , Animals , Astrocytes/immunology , Astrocytes/pathology , Cell Death , Cell Survival , Cicatrix/diet therapy , Cicatrix/pathology , Cicatrix/physiopathology , Disease Models, Animal , Female , Gliosis/diet therapy , Gliosis/pathology , Gliosis/physiopathology , Motor Activity , Neurons/immunology , Neurons/pathology , Oligodendroglia/immunology , Oligodendroglia/pathology , Rats, Sprague-Dawley , Rats, Wistar , Recovery of Function , Spinal Cord/immunology , Spinal Cord/pathology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Thoracic Vertebrae , Treatment Outcome , Urinary Bladder/physiopathology
19.
Schizophr Res ; 166(1-3): 235-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26004690

ABSTRACT

BACKGROUND: Our previous lipidomics studies demonstrated elevated sulfatides, plasmalogens, and N-acylphosphatidylserines in the frontal cortex of schizophrenia subjects. These data suggest that there may be an abnormal function of glycosynapses in schizophrenia. We further examined the disease and anatomical specificity of these observations. METHODS: We undertook a targeted lipidomics analysis of plasmalogens, sulfatides, and N-acyl-phosphatidylserines in the frontal cortex obtained from schizophrenia, bipolar, and ALS subjects and the cerebellum of schizophrenia subjects. RESULTS: We demonstrate that sulfatides, plasmalogens, and N-acyl-phosphatidylserines are significantly elevated in the frontal cortex of patients suffering from schizophrenia and bipolar depression but not in ALS patients. These lipids were unchanged in the cerebellum of subjects with schizophrenia. CONCLUSIONS: Our data suggest that dysfunction of oligodendrocyte glycosynapses may be specific to limbic circuits in schizophrenia and that this dysfunction is also detected in bipolar depression, suggesting that these disorders possess several common pathophysiological features.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Bipolar Disorder/metabolism , Cerebellum/metabolism , Frontal Lobe/metabolism , Schizophrenia/metabolism , Adult , Aged , Aged, 80 and over , Amyotrophic Lateral Sclerosis/pathology , Bipolar Disorder/pathology , Cerebellum/pathology , Female , Frontal Lobe/pathology , Humans , Male , Middle Aged , Oligodendroglia/metabolism , Oligodendroglia/pathology , Phosphatidylserines/metabolism , Plasmalogens/metabolism , Schizophrenia/pathology , Sulfoglycosphingolipids/metabolism
20.
J Cereb Blood Flow Metab ; 33(9): 1474-84, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23801244

ABSTRACT

Dietary supplementation with omega-3 (ω-3) fatty acids is a safe, economical mean of preventive medicine that has shown protection against several neurologic disorders. The present study tested the hypothesis that this method is protective against controlled cortical impact (CCI). Indeed, mice fed with ω-3 polyunsaturated fatty acid (PUFA)-enriched diet for 2 months exhibited attenuated short and long-term behavioral deficits due to CCI. Although ω-3 PUFAs did not decrease cortical lesion volume, these fatty acids did protect against hippocampal neuronal loss after CCI and reduced pro-inflammatory response. Interestingly, ω-3 PUFAs prevented the loss of myelin basic protein (MPB), preserved the integrity of the myelin sheath, and maintained the nerve fiber conductivity in the CCI model. ω-3 PUFAs also directly protected oligodendrocyte cultures from excitotoxicity and blunted the microglial activation-induced death of oligodendrocytes in microglia/oligodendrocyte cocultures. In sum, ω-3 PUFAs elicit multifaceted protection against behavioral dysfunction, hippocampal neuronal loss, inflammation, and loss of myelination and impulse conductivity. The present report is the first demonstration that ω-3 PUFAs protect against white matter injury in vivo and in vitro. The protective impact of ω-3 PUFAs supports the clinical use of this dietary supplement as a prophylaxis against traumatic brain injury and other nervous system disorders.


Subject(s)
Behavior, Animal/drug effects , Brain Injuries , Cerebral Cortex , Dietary Supplements , Fatty Acids, Omega-3/pharmacology , Hippocampus , Animals , Brain Injuries/drug therapy , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/physiopathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Male , Mice , Myelin Basic Protein/metabolism , Neurons/metabolism , Neurons/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology
SELECTION OF CITATIONS
SEARCH DETAIL