Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 897
Filter
1.
Nat Commun ; 15(1): 6744, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39112447

ABSTRACT

Demyelination is a common pathological feature in a wide range of diseases, characterized by the loss of myelin sheath and myelin-supporting oligodendrocytes. These losses lead to impaired axonal function, increased vulnerability of axons to damage, and result in significant brain atrophy and neuro-axonal degeneration. Multiple pathomolecular processes contribute to neuroinflammation, oligodendrocyte cell death, and progressive neuronal dysfunction. In this study, we use the cuprizone mouse model of demyelination to investigate long-term non-invasive gamma entrainment using sensory stimulation as a potential therapeutic intervention for promoting myelination and reducing neuroinflammation in male mice. Here, we show that multisensory gamma stimulation mitigates demyelination, promotes oligodendrogenesis, preserves functional integrity and synaptic plasticity, attenuates oligodendrocyte ferroptosis-induced cell death, and reduces brain inflammation. Thus, the protective effects of multisensory gamma stimulation on myelin and anti-neuroinflammatory properties support its potential as a therapeutic approach for demyelinating disorders.


Subject(s)
Cuprizone , Demyelinating Diseases , Disease Models, Animal , Myelin Sheath , Oligodendroglia , Animals , Cuprizone/toxicity , Male , Demyelinating Diseases/chemically induced , Demyelinating Diseases/therapy , Demyelinating Diseases/pathology , Mice , Oligodendroglia/metabolism , Oligodendroglia/pathology , Myelin Sheath/metabolism , Mice, Inbred C57BL , Ferroptosis , Neuronal Plasticity , Brain/pathology , Brain/metabolism , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/pathology
2.
Cell Mol Life Sci ; 81(1): 346, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39134808

ABSTRACT

In people with multiple sclerosis (MS), newborn and surviving oligodendrocytes (OLs) can contribute to remyelination, however, current therapies are unable to enhance or sustain endogenous repair. Low intensity repetitive transcranial magnetic stimulation (LI-rTMS), delivered as an intermittent theta burst stimulation (iTBS), increases the survival and maturation of newborn OLs in the healthy adult mouse cortex, but it is unclear whether LI-rTMS can promote remyelination. To examine this possibility, we fluorescently labelled oligodendrocyte progenitor cells (OPCs; Pdgfrα-CreER transgenic mice) or mature OLs (Plp-CreER transgenic mice) in the adult mouse brain and traced the fate of each cell population over time. Daily sessions of iTBS (600 pulses; 120 mT), delivered during cuprizone (CPZ) feeding, did not alter new or pre-existing OL survival but increased the number of myelin internodes elaborated by new OLs in the primary motor cortex (M1). This resulted in each new M1 OL producing ~ 471 µm more myelin. When LI-rTMS was delivered after CPZ withdrawal (during remyelination), it significantly increased the length of the internodes elaborated by new M1 and callosal OLs, increased the number of surviving OLs that supported internodes in the corpus callosum (CC), and increased the proportion of axons that were myelinated. The ability of LI-rTMS to modify cortical neuronal activity and the behaviour of new and surviving OLs, suggests that it may be a suitable adjunct intervention to enhance remyelination in people with MS.


Subject(s)
Cuprizone , Demyelinating Diseases , Myelin Sheath , Oligodendroglia , Remyelination , Transcranial Magnetic Stimulation , Animals , Transcranial Magnetic Stimulation/methods , Oligodendroglia/metabolism , Demyelinating Diseases/therapy , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Mice , Myelin Sheath/metabolism , Disease Models, Animal , Mice, Transgenic , Motor Cortex/pathology , Motor Cortex/metabolism , Cell Survival , Mice, Inbred C57BL , Multiple Sclerosis/therapy , Multiple Sclerosis/pathology
3.
Commun Biol ; 7(1): 813, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965401

ABSTRACT

Strategies for treating progressive multiple sclerosis (MS) remain limited. Here, we found that miR-145-5p is overabundant uniquely in chronic lesion tissues from secondary progressive MS patients. We induced both acute and chronic demyelination in miR-145 knockout mice to determine its contributions to remyelination failure. Following acute demyelination, no advantage to miR-145 loss could be detected. However, after chronic demyelination, animals with miR-145 loss demonstrated increased remyelination and functional recovery, coincident with altered presence of astrocytes and microglia within the corpus callosum relative to wild-type animals. This improved response in miR-145 knockout animals coincided with a pathological upregulation of miR-145-5p in wild-type animals with chronic cuprizone exposure, paralleling human chronic lesions. Furthermore, miR-145 overexpression specifically in oligodendrocytes (OLs) severely stunted differentiation and negatively impacted survival. RNAseq analysis showed altered transcriptome in these cells with downregulated major pathways involved in myelination. Our data suggest that pathological accumulation of miR-145-5p is a distinctive feature of chronic demyelination and is strongly implicated in the failure of remyelination, possibly due to the inhibition of OL differentiation together with alterations in other glial cells. This is mirrored in chronic MS lesions, and thus miR-145-5p serves as a potential relevant therapeutic target in progressive forms of MS.


Subject(s)
Demyelinating Diseases , Disease Models, Animal , Mice, Knockout , MicroRNAs , Remyelination , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Remyelination/genetics , Mice , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Humans , Oligodendroglia/metabolism , Oligodendroglia/pathology , Recovery of Function , Male , Mice, Inbred C57BL , Cuprizone/toxicity , Female , Chronic Disease , Myelin Sheath/metabolism
4.
Neurosci Lett ; 836: 137869, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-38852766

ABSTRACT

Dietary administration of a copper chelator, cuprizone (CPZ), has long been reported to induce intense and reproducible demyelination of several brain structures such as the corpus callosum. Despite the widespread use of CPZ as an animal model for demyelinating diseases such as multiple sclerosis (MS), the mechanism by which it induces demyelination and then allows robust remyelination is still unclear. An intensive mapping of the cell dynamics of oligodendrocyte (OL) lineage during the de- and remyelination course would be particularly important for a deeper understanding of this model. Here, using a panel of OL lineage cell markers as in situ hybridization (ISH) probes, including Pdgfra, Plp, Mbp, Mog, Enpp6, combined with immunofluorescence staining of CC1, SOX10, we provide a detailed dynamic profile of OL lineage cells during the entire course of the model from 1, 2, 3.5 days, 1, 2, 3, 4,5 weeks of CPZ treatment, as well as after 1, 2, 3, 4 weeks of recovery from CPZ treatment. The result showed an unexpected early death of mature OLs and response of OL progenitor cells (OPCs) in vivo upon CPZ challenge, and a prolonged upregulation of myelin-forming OLs compared to the intact control even 4 weeks after CPZ withdrawal. These data may serve as a basic reference system for future studies of the effects of any intervention on de- and remyelination using the CPZ model, and imply the need to optimize the timing windows for the introduction of pro-remyelination therapies in demyelinating diseases such as MS.


Subject(s)
Cell Lineage , Cuprizone , Demyelinating Diseases , Oligodendroglia , Cuprizone/toxicity , Animals , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Oligodendroglia/drug effects , Oligodendroglia/pathology , Oligodendroglia/metabolism , Disease Models, Animal , In Situ Hybridization/methods , Mice, Inbred C57BL , Mice , Remyelination/drug effects , Remyelination/physiology , Male , Chelating Agents/toxicity , Chelating Agents/pharmacology , Myelin Sheath/pathology , Myelin Sheath/drug effects , Myelin Sheath/metabolism
5.
Glia ; 72(10): 1801-1820, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38899723

ABSTRACT

The kappa opioid receptor has been identified as a promising therapeutic target for promoting remyelination. In the current study, we evaluated the ability of nalfurafine to promote oligodendrocyte progenitor cell (OPC) differentiation and myelination in vitro, and its efficacy in an extended, cuprizone-induced demyelination model. Primary mouse (C57BL/6J) OPC-containing cultures were treated with nalfurafine (0.6-200 nM), clemastine (0.01-100 µM), T3 (30 ng/mL), or vehicle for 5 days. Using immunocytochemistry and confocal microscopy, we found that nalfurafine treatment increased OPC differentiation, oligodendrocyte (OL) morphological complexity, and myelination of nanofibers in vitro. Adult male mice (C57BL/6J) were given a diet containing 0.2% cuprizone and administered rapamycin (10 mg/kg) once daily for 12 weeks followed by 6 weeks of treatment with nalfurafine (0.01 or 0.1 mg/kg), clemastine (10 mg/kg), or vehicle. We quantified the number of OLs using immunofluorescence, gross myelination using black gold staining, and myelin thickness using electron microscopy. Cuprizone + rapamycin treatment produced extensive demyelination and was accompanied by a loss of mature OLs, which was partially reversed by therapeutic administration of nalfurafine. We also assessed these mice for functional behavioral changes in open-field, horizontal bar, and mouse motor skill sequence tests (complex wheel running). Cuprizone + rapamycin treatment resulted in hyperlocomotion, poorer horizontal bar scores, and less distance traveled on the running wheels. Partial recovery was observed on both the horizontal bar and complex running wheel tests over time, which was facilitated by nalfurafine treatment. Taken together, these data highlight the potential of nalfurafine as a remyelination-promoting therapeutic.


Subject(s)
Cuprizone , Demyelinating Diseases , Mice, Inbred C57BL , Morphinans , Myelin Sheath , Sirolimus , Spiro Compounds , Animals , Morphinans/pharmacology , Male , Spiro Compounds/pharmacology , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Demyelinating Diseases/drug therapy , Mice , Myelin Sheath/drug effects , Myelin Sheath/pathology , Myelin Sheath/metabolism , Sirolimus/pharmacology , Cuprizone/toxicity , Cells, Cultured , Disease Models, Animal , Oligodendrocyte Precursor Cells/drug effects , Oligodendrocyte Precursor Cells/metabolism , Cell Differentiation/drug effects
6.
Sci Rep ; 14(1): 13988, 2024 06 18.
Article in English | MEDLINE | ID: mdl-38886527

ABSTRACT

Demyelination is generated in several nervous system illnesses. Developing strategies for effective clinical treatments requires the discovery of promyelinating drugs. Increased GABAergic signaling through γ-aminobutyric acid type A receptor (GABAAR) activation in oligodendrocytes has been proposed as a promyelinating condition. GABAAR expressed in oligodendroglia is strongly potentiated by n-butyl-ß-carboline-3-carboxylate (ß-CCB) compared to that in neurons. Here, mice were subjected to 0.3% cuprizone (CPZ) added in the food to induce central nervous system demyelination, a well-known model for multiple sclerosis. Then ß-CCB (1 mg/Kg) was systemically administered to analyze the remyelination status in white and gray matter areas. Myelin content was evaluated using Black-Gold II (BGII) staining, immunofluorescence (IF), and magnetic resonance imaging (MRI). Evidence indicates that ß-CCB treatment of CPZ-demyelinated animals promoted remyelination in several white matter structures, such as the fimbria, corpus callosum, internal capsule, and cerebellar peduncles. Moreover, using IF, it was observed that CPZ intake induced an increase in NG2+ and a decrease in CC1+ cell populations, alterations that were importantly retrieved by ß-CCB treatment. Thus, the promyelinating character of ß-CCB was confirmed in a generalized demyelination model, strengthening the idea that it has clinical potential as a therapeutic drug.


Subject(s)
Carbolines , Cuprizone , Demyelinating Diseases , Disease Models, Animal , Remyelination , Animals , Cuprizone/toxicity , Remyelination/drug effects , Mice , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Demyelinating Diseases/metabolism , Carbolines/pharmacology , Carbolines/administration & dosage , Myelin Sheath/metabolism , Myelin Sheath/drug effects , Male , Mice, Inbred C57BL , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Multiple Sclerosis/chemically induced , Multiple Sclerosis/pathology , White Matter/drug effects , White Matter/metabolism , White Matter/pathology , Magnetic Resonance Imaging
7.
Neurochem Res ; 49(8): 2087-2104, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38819698

ABSTRACT

Addressing inflammation, demyelination, and associated neurodegeneration in inflammatory demyelinating diseases like multiple sclerosis (MS) remains challenging. ACT-1004-1239, a first-in-class and potent ACKR3 antagonist, currently undergoing clinical development, showed promise in preclinical MS models, reducing neuroinflammation and demyelination. However, its effectiveness in treating established disease and impact on remyelination after the occurrence of demyelinated lesions remain unexplored. This study assessed the therapeutic effect of ACT-1004-1239 in two demyelinating disease models. In the proteolipid protein (PLP)-induced experimental autoimmune encephalomyelitis (EAE) model, ACT-1004-1239 administered upon the detection of the first signs of paralysis, resulted in a dose-dependent reduction in EAE disease severity, concomitant with diminished immune cell infiltrates in the CNS and reduced demyelination. Notably, efficacy correlated with elevated plasma concentrations of CXCL11 and CXCL12, two pharmacodynamic biomarkers of ACKR3 antagonism. Combining ACT-1004-1239 with siponimod, an approved immunomodulatory treatment for MS, synergistically reduced EAE severity. In the cuprizone-induced demyelination model, ACT-1004-1239 administered after 5 weeks of cuprizone exposure, significantly accelerated remyelination, already quantifiable one week after cuprizone withdrawal. Additionally, ACT-1004-1239 penetrated the CNS, elevating brain CXCL12 concentrations. These results demonstrate that ACKR3 antagonism significantly reduces the severity of experimental demyelinating diseases, even when treatment is initiated therapeutically, after the occurrence of lesions. It confirms the dual mode of action of ACT-1004-1239, exhibiting both immunomodulatory effects by reducing neuroinflammation and promyelinating effects by accelerating myelin repair. The results further strengthen the rationale for evaluating ACT-1004-1239 in clinical trials for patients with demyelinating diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Mice, Inbred C57BL , Remyelination , Animals , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Remyelination/drug effects , Mice , Female , Demyelinating Diseases/drug therapy , Demyelinating Diseases/chemically induced , Cuprizone , Azetidines/pharmacology , Azetidines/therapeutic use , Immunomodulating Agents/pharmacology , Immunomodulating Agents/therapeutic use , Benzyl Compounds/therapeutic use , Benzyl Compounds/pharmacology , Myelin Sheath/drug effects , Myelin Sheath/metabolism
8.
J Neurosci ; 44(28)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38749703

ABSTRACT

Dysregulation of oligodendrocyte progenitor cell (OPC) recruitment and oligodendrocyte differentiation contribute to failure of remyelination in human demyelinating diseases such as multiple sclerosis (MS). Deletion of muscarinic receptor enhances OPC differentiation and remyelination. However, the role of ligand-dependent signaling versus constitutive receptor activation is unknown. We hypothesized that dysregulated acetylcholine (ACh) release upon demyelination contributes to ligand-mediated activation hindering myelin repair. Following chronic cuprizone (CPZ)-induced demyelination (male and female mice), we observed a 2.5-fold increase in ACh concentration. This increase in ACh concentration could be attributed to increased ACh synthesis or decreased acetylcholinesterase-/butyrylcholinesterase (BChE)-mediated degradation. Using choline acetyltransferase (ChAT) reporter mice, we identified increased ChAT-GFP expression following both lysolecithin and CPZ demyelination. ChAT-GFP expression was upregulated in a subset of injured and uninjured axons following intraspinal lysolecithin-induced demyelination. In CPZ-demyelinated corpus callosum, ChAT-GFP was observed in Gfap+ astrocytes and axons indicating the potential for neuronal and astrocytic ACh release. BChE expression was significantly decreased in the corpus callosum following CPZ demyelination. This decrease was due to the loss of myelinating oligodendrocytes which were the primary source of BChE. To determine the role of ligand-mediated muscarinic signaling following lysolecithin injection, we administered neostigmine, a cholinesterase inhibitor, to artificially raise ACh. We identified a dose-dependent decrease in mature oligodendrocyte density with no effect on OPC recruitment. Together, these results support a functional role of ligand-mediated activation of muscarinic receptors following demyelination and suggest that dysregulation of ACh homeostasis directly contributes to failure of remyelination in MS.


Subject(s)
Demyelinating Diseases , Oligodendroglia , Signal Transduction , Animals , Demyelinating Diseases/metabolism , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Mice , Oligodendroglia/metabolism , Oligodendroglia/drug effects , Female , Male , Signal Transduction/drug effects , Signal Transduction/physiology , Mice, Inbred C57BL , Acetylcholine/metabolism , Cuprizone/toxicity , Lysophosphatidylcholines/toxicity , Cell Differentiation/drug effects , Cell Differentiation/physiology , Choline O-Acetyltransferase/metabolism , Remyelination/physiology , Remyelination/drug effects , Myelin Sheath/metabolism , Myelin Sheath/drug effects , Mice, Transgenic
9.
Br J Pharmacol ; 181(17): 3263-3281, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38742374

ABSTRACT

BACKGROUND AND PURPOSE: ApTOLL is an aptamer selected to antagonize toll-like receptor 4 (TLR4), a relevant actor for innate immunity involved in inflammatory responses in multiple sclerosis (MS) and other diseases. The currently available therapeutic arsenal to treat MS is composed of immunomodulators but, to date, there are no (re)myelinating drugs available in clinics. In our present study, we studied the effect of ApTOLL on different animal models of MS. EXPERIMENTAL APPROACH: The experimental autoimmune encephalomyelitis (EAE) model was used to evaluate the effect of ApTOLL on reducing the inflammatory component. A more direct effect on oligodendroglia was studied with the cuprizone model and purified primary cultures of murine and human oligodendrocyte precursor cells (OPCs) isolated through magnetic-activated cell sorting (MACS) from samples of brain cortex. Also, we tested these effects in an ex vivo model of organotypic cultures demyelinated with lysolecithin (LPC). KEY RESULTS: ApTOLL treatment positively impacted the clinical symptomatology of mice in the EAE and cuprizone models, which was associated with better preservation plus restoration of myelin and oligodendrocytes in the demyelinated lesions of animals. Restoration was corroborated on purified cultures of rodent and human OPCs. CONCLUSION AND IMPLICATIONS: Our findings reveal a new therapeutic approach for the treatment of inflammatory and demyelinating diseases such as MS. The molecular nature of the aptamer exerts not only an anti-inflammatory effect but also neuroprotective and remyelinating effects. The excellent safety profile demonstrated by ApTOLL in animals and humans opens the door to future clinical trials in MS patients.


Subject(s)
Aptamers, Nucleotide , Encephalomyelitis, Autoimmune, Experimental , Mice, Inbred C57BL , Multiple Sclerosis , Animals , Humans , Multiple Sclerosis/drug therapy , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Mice , Aptamers, Nucleotide/pharmacology , Female , Cuprizone , Oligodendroglia/drug effects , Cells, Cultured , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/antagonists & inhibitors , Myelin Sheath/drug effects , Myelin Sheath/metabolism
10.
Mol Biol Rep ; 51(1): 674, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38787497

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system (CNS) with inflammation and immune dysfunction. OBJECTIVES: We compared the remyelination and immunomodulation properties of mesenchymal stem cells (MSCs) with their conditioned medium (CM) in the cuprizone model. METHODS: Twenty-four C57BL/ 6 mice were divided into four groups. After cuprizone demyelination, MSCs and their CM were injected into the right lateral ventricle of mice. The expression level of IL-1ß, TNF-α, and BDNF genes was evaluated using the qRT-PCR. APC antibody was used to assess the oligodendrocyte population using the immunofluorescent method. The remyelination and axonal repair were studied by specific staining of the LFB and electron microscopy techniques. RESULTS: Transplantation of MSCs and CM increased the expression of the BDNF gene and decreased the expression of IL-1ß and TNF-α genes compared to the cuprizone group, and these effects in the cell group were more than CM. Furthermore, cell transplantation resulted in a significant improvement in myelination and axonal repair, which was measured by luxol fast blue and transmission electron microscope images. The cell group had a higher number of oligodendrocytes than other groups. CONCLUSIONS: According to the findings, injecting MSCs intraventricularly versus cell-conditioned medium can be a more effective approach to improving chronic demyelination in degenerative diseases like MS.


Subject(s)
Cuprizone , Demyelinating Diseases , Disease Models, Animal , Inflammation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Mice, Inbred C57BL , Animals , Mesenchymal Stem Cell Transplantation/methods , Mice , Mesenchymal Stem Cells/metabolism , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Culture Media, Conditioned/pharmacology , Inflammation/pathology , Inflammation/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Oligodendroglia/metabolism , Remyelination , Multiple Sclerosis/pathology , Multiple Sclerosis/therapy , Multiple Sclerosis/metabolism , Multiple Sclerosis/chemically induced , Tumor Necrosis Factor-alpha/metabolism , Male , Myelin Sheath/metabolism
11.
Glia ; 72(8): 1392-1401, 2024 08.
Article in English | MEDLINE | ID: mdl-38572807

ABSTRACT

Multiple sclerosis is an autoimmune disease of the central nervous system (CNS) characterized by demyelination, axonal damage and, for the majority of people, a decline in neurological function in the long-term. Remyelination could assist in the protection of axons and their functional recovery, but such therapies are not, as yet, available. The TAM (Tyro3, Axl, and MERTK) receptor ligand GAS6 potentiates myelination in vitro and promotes recovery in pre-clinical models of MS. However, it has remained unclear which TAM receptor is responsible for transducing this effect and whether post-translational modification of GAS6 is required. In this study, we show that the promotion of myelination requires post-translational modification of the GLA domain of GAS6 via vitamin K-dependent γ-carboxylation. We also confirmed that the intracerebroventricular provision of GAS6 for 2 weeks to demyelinated wild-type (WT) mice challenged with cuprizone increased the density of myelinated axons in the corpus callosum by over 2-fold compared with vehicle control. Conversely, the provision of GAS6 to Tyro3 KO mice did not significantly improve the density of myelinated axons. The improvement in remyelination following the provision of GAS6 to WT mice was also accompanied by an increased density of CC1+ve mature oligodendrocytes compared with vehicle control, whereas this improvement was not observed in the absence of Tyro3. This effect occurs independent of any influence on microglial activation. This work therefore establishes that the remyelinative activity of GAS6 is dependent on Tyro3 and includes potentiation of oligodendrocyte numbers.


Subject(s)
Cuprizone , Demyelinating Diseases , Intercellular Signaling Peptides and Proteins , Mice, Inbred C57BL , Mice, Knockout , Receptor Protein-Tyrosine Kinases , Remyelination , Animals , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Remyelination/physiology , Remyelination/drug effects , Receptor Protein-Tyrosine Kinases/metabolism , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Cuprizone/toxicity , Mice , Disease Models, Animal , Myelin Sheath/metabolism , Myelin Sheath/drug effects , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Male , Female
12.
Brain Behav ; 14(4): e3487, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38648385

ABSTRACT

INTRODUCTION: Demyelination is a key factor in axonal degeneration and neural loss, leading to disability in multiple sclerosis (MS) patients. Transforming growth factor beta activated kinase 1 (TAK1) is a critical molecule involved in immune and inflammatory signaling pathways. Knockout of microglia TAK1 can inhibit autoimmune inflammation of the brain and spinal cord and improve the outcome of MS. However, it is unclear whether inhibiting TAK1 can alleviate demyelination. METHODS: Eight-week-old male c57bl/6j mice were randomly divided into five groups: (a) the control group, (b) the group treated with cuprizone (CPZ) only, (c) the group treated with 5Z-7-Oxozaenol (OZ) only, and (d) the group treated with both cuprizone and 15 µg/30 µg OZ. Demyelination in the mice of this study was induced by administration of CPZ (ig) at a daily dose of 400 mg/kg for consecutive 5 weeks. OZ was intraperitoneally administered at mentioned doses twice a week, starting from week 3 after beginning cuprizone treatment. Histology, rotarod test, grasping test, pole test, Western blot, RT-PCR, and ELISA were used to evaluate corpus callosum demyelination, behavioral impairment, oligodendrocyte differentiation, TAK1 signaling pathway expression, microglia, and related cytokines. RESULTS: Our results demonstrated that OZ protected against myelin loss and behavior impairment caused by CPZ. Additionally, OZ rescued the loss of oligodendrocytes in CPZ-induced mice. OZ inhibited the activation of JNK, p65, and p38 pathways, transformed M1 polarized microglia into M2 phenotype, and increased brain-derived neurotrophic factor (BDNF) expression to attenuate demyelination in CPZ-treated mice. Furthermore, OZ reduced the expression of proinflammatory cytokines and increases anti-inflammatory cytokines in CPZ-treated mice. CONCLUSION: These findings suggest that inhibiting TAK1 may be an effective approach for treating demyelinating diseases.


Subject(s)
Cuprizone , Demyelinating Diseases , Lactones , Mice, Inbred C57BL , Microglia , Resorcinols , Zearalenone/administration & dosage , Animals , Cuprizone/pharmacology , Microglia/drug effects , Microglia/metabolism , Demyelinating Diseases/drug therapy , Demyelinating Diseases/chemically induced , Mice , Male , MAP Kinase Kinase Kinases/metabolism , Zearalenone/pharmacology , Zearalenone/analogs & derivatives , Cell Polarity/drug effects , Corpus Callosum/drug effects , Corpus Callosum/pathology , Corpus Callosum/metabolism , Disease Models, Animal
13.
Inflammopharmacology ; 32(2): 1295-1315, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38512652

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory neurodegenerative disease of the central nervous system that injures the myelin sheath, provoking progressive axonal degeneration and functional impairments. No efficient therapy is available at present to combat such insults, and hence, novel safe and effective alternatives for MS therapy are extremely required. Rutin (RUT) is a flavonoid that exhibits antioxidant, anti-inflammatory, and neuroprotective effects in several brain injuries. The present study evaluated the potential beneficial effects of two doses of RUT in a model of pattern-III lesion of MS, in comparison to the conventional standard drug; dimethyl fumarate (DMF). Demyelination was induced in in male adult C57BL/6 mice by dietary 0.2% (w/w) cuprizone (CPZ) feeding for 6 consecutive weeks. Treated groups received either oral RUT (50 or 100 mg/kg) or DMF (15 mg/kg), along with CPZ feeding, for 6 consecutive weeks. Mice were then tested for behavioral changes, followed by biochemical analyses and histological examinations of the corpus callosum (CC). Results revealed that CPZ caused motor dysfunction, demyelination, and glial activation in demyelinated lesions, as well as significant oxidative stress, and proinflammatory cytokine elevation. Six weeks of RUT treatment significantly improved locomotor activity and motor coordination. Moreover, RUT considerably improved remyelination in the CC of CPZ + RUT-treated mice, as revealed by luxol fast blue staining and transmission electron microscopy. Rutin also significantly attenuated CPZ-induced oxidative stress and inflammation in the CC of tested animals. The effect of RUT100 was obviously more marked than either that of DMF, regarding most of the tested parameters, or even its smaller tested dose. In silico docking revealed that RUT binds tightly within NF-κB at the binding site of the protein-DNA complex, with a good negative score of -6.79 kcal/mol. Also, RUT-Kelch-like ECH-associated protein 1 (Keap1) model clarifies the possible inhibition of Keap1-Nrf2 protein-protein interaction. Findings of the current study provide evidence for the protective effect of RUT in CPZ-induced demyelination and behavioral dysfunction in mice, possibly by modulating NF-κB and Nrf2 signaling pathways. The present study may be one of the first to indicate a pro-remyelinating effect for RUT, which might represent a potential additive benefit in treating MS.


Subject(s)
Demyelinating Diseases , Multiple Sclerosis , Neurodegenerative Diseases , Neuroprotective Agents , Male , Animals , Mice , Multiple Sclerosis/chemically induced , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Cuprizone/adverse effects , Kelch-Like ECH-Associated Protein 1/metabolism , Demyelinating Diseases/chemically induced , Demyelinating Diseases/drug therapy , Demyelinating Diseases/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , NF-kappa B/metabolism , Rutin/pharmacology , NF-E2-Related Factor 2/metabolism , Neurodegenerative Diseases/drug therapy , Mice, Inbred C57BL , Disease Models, Animal
14.
EBioMedicine ; 100: 104982, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38306899

ABSTRACT

BACKGROUND: Inflammatory demyelinating diseases of the central nervous system, such as multiple sclerosis, are significant sources of morbidity in young adults despite therapeutic advances. Current murine models of remyelination have limited applicability due to the low white matter content of their brains, which restricts the spatial resolution of diagnostic imaging. Large animal models might be more suitable but pose significant technological, ethical and logistical challenges. METHODS: We induced targeted cerebral demyelinating lesions by serially repeated injections of lysophosphatidylcholine in the minipig brain. Lesions were amenable to follow-up using the same clinical imaging modalities (3T magnetic resonance imaging, 11C-PIB positron emission tomography) and standard histopathology protocols as for human diagnostics (myelin, glia and neuronal cell markers), as well as electron microscopy (EM), to compare against biopsy data from two patients. FINDINGS: We demonstrate controlled, clinically unapparent, reversible and multimodally trackable brain white matter demyelination in a large animal model. De-/remyelination dynamics were slower than reported for rodent models and paralleled by a degree of secondary axonal pathology. Regression modelling of ultrastructural parameters (g-ratio, axon thickness) predicted EM features of cerebral de- and remyelination in human data. INTERPRETATION: We validated our minipig model of demyelinating brain diseases by employing human diagnostic tools and comparing it with biopsy data from patients with cerebral demyelination. FUNDING: This work was supported by the DFG under Germany's Excellence Strategy within the framework of the Munich Cluster for Systems Neurology (EXC 2145 SyNergy, ID 390857198) and TRR 274/1 2020, 408885537 (projects B03 and Z01).


Subject(s)
Demyelinating Diseases , Multiple Sclerosis , White Matter , Swine , Humans , Animals , Mice , Demyelinating Diseases/diagnostic imaging , Demyelinating Diseases/pathology , Cuprizone , Swine, Miniature , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/pathology , Myelin Sheath/pathology , White Matter/pathology , Microscopy, Electron , Disease Models, Animal
15.
Int J Mol Sci ; 25(3)2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38338724

ABSTRACT

Multiple sclerosis (MS) is an autoimmune and inflammatory disorder affecting the central nervous system whose cause is still largely unknown. Oligodendrocyte degeneration results in demyelination of axons, which can eventually be repaired by a mechanism called remyelination. Prevention of demyelination and the pharmacological support of remyelination are two promising strategies to ameliorate disease progression in MS patients. The cuprizone model is commonly employed to investigate oligodendrocyte degeneration mechanisms or to explore remyelination pathways. During the last decades, several different protocols have been applied, and all have their pros and cons. This article intends to offer guidance for conducting pre-clinical trials using the cuprizone model in mice, focusing on discovering new treatment approaches to prevent oligodendrocyte degeneration or enhance remyelination.


Subject(s)
Demyelinating Diseases , Multiple Sclerosis , Remyelination , Humans , Mice , Animals , Cuprizone , Myelin Sheath/metabolism , Demyelinating Diseases/metabolism , Oligodendroglia/metabolism , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Mice, Inbred C57BL , Disease Models, Animal
16.
J Neurosci ; 44(13)2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38395617

ABSTRACT

Myelinating oligodendrocytes die in human disease and early in aging. Despite this, the mechanisms that underly oligodendrocyte death are not resolved and it is also not clear whether these mechanisms change as oligodendrocyte lineage cells are undergoing differentiation and maturation. Here, we used a combination of intravital imaging, single-cell ablation, and cuprizone-mediated demyelination, in both female and male mice, to discover that oligodendrocyte maturation dictates the dynamics and mechanisms of cell death. After single-cell phototoxic damage, oligodendrocyte precursor cells underwent programmed cell death within hours, differentiating oligodendrocytes died over several days, while mature oligodendrocytes took weeks to die. Importantly cells at each maturation stage all eventually died but did so with drastically different temporal dynamics and morphological features. Consistent with this, cuprizone treatment initiated a caspase-3-dependent form of rapid cell death in differentiating oligodendrocytes, while mature oligodendrocytes never activated this executioner caspase. Instead, mature oligodendrocytes exhibited delayed cell death which was marked by DNA damage and disruption in poly-ADP-ribose subcellular localization. Thus, oligodendrocyte maturation plays a key role in determining the mechanism of death a cell undergoes in response to the same insult. This means that oligodendrocyte maturation is important to consider when designing strategies for preventing cell death and preserving myelin while also enhancing the survival of new oligodendrocytes in demyelinating conditions.


Subject(s)
Cuprizone , Demyelinating Diseases , Humans , Mice , Male , Female , Animals , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Demyelinating Diseases/metabolism , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Apoptosis/physiology , Cell Differentiation , Mice, Inbred C57BL
17.
Exp Neurol ; 375: 114730, 2024 May.
Article in English | MEDLINE | ID: mdl-38401853

ABSTRACT

Demyelination is a proper syndrome in plenty of central nervous system (CNS) diseases, which is the main obstacle to recovery and still lacks an effective treatment. To overcome the limitations of the brain-blood barrier on drug permeability, we modified an exosome secreted by neural stem cells (NSCs), which had transfected with lentivirus armed with platelet-derived growth factors A (PDGFA)-ligand. Through the in vivo and in vitro exosomes targeting test, the migration ability to the lesion areas and OPCs significantly improved after ligand modification. Furthermore, the targeted exosomes loaded with 3,5, 30-L-triiodothyronine (T3) have a critical myelination ability in CNS development, administrated to the cuprizone animal model treatment. The data shows that the novel drug vector loaded with T3 significantly promotes remyelination compared with T3 alone. At the same time, it improved the CNS microenvironment by reducing astrogliosis, inhibiting pro-inflammatory microglia, and alleviating axon damage. This investigation provides a straightforward strategy to produce a targeting exosome and indicates a possible therapeutic manner for demyelinating disease.


Subject(s)
Demyelinating Diseases , Exosomes , Animals , Mice , Demyelinating Diseases/therapy , Demyelinating Diseases/drug therapy , Oligodendroglia , Ligands , Exosomes/metabolism , Triiodothyronine/metabolism , Triiodothyronine/pharmacology , Triiodothyronine/therapeutic use , Cuprizone/toxicity , Mice, Inbred C57BL , Myelin Sheath/pathology , Disease Models, Animal
18.
Biomed Pharmacother ; 173: 116297, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38394854

ABSTRACT

Hericium erinaceus mycelium extract (HEM), containing erinacine A (HeA) and erinacine S (HeS), has shown promise in promoting the differentiation of oligodendrocyte precursor cells (OPCs) into mature oligodendrocytes (OLs), crucial for myelin production in the central nervous system (CNS). The main aim of this study was to characterize the protective effects of HEM and its components on OLs and myelin in demyelinating rodents by exposure to cuprizone (CPZ), a copper chelating agent commonly used to induce demyelination in the corpus callosum of the brain. Rats were fed by CPZ-containing diet and simultaneously orally administered HEM, HeA, or HeS on a daily basis for three weeks. We found that HEM and HeS preserved myelin and OLs in the corpus callosum of CPZ-fed rats, along with reduced microglia and astrocyte activation, and downregulated IL-1ß expression. Furthermore, post-treatment with HeS, in mouse models with acute (6 weeks) or chronic (12 weeks) CPZ-induced demyelination demonstrated oral administration during the final 4 weeks (HeS4/6 or HeS4/12) effectively preserved myelin in the corpus callosum. Additionally, HeS4/6 and HeS4/12 inhibited anxious and depressive-like behaviors in CPZ-fed mice. In summary, simultaneous administration of HEM and HeS in rats during short-term CPZ intoxication preserved OLs and myelin. Furthermore, post-administration of HeS not only inhibited demyelination and gliosis but also alleviated anxiety and depression in both acute and chronic CPZ-fed mice. This study presents compelling evidence supporting the potential of HeS as a promising small active compound for protecting OLs and preserving myelin in demyelinating diseases associated with emotional disorders.


Subject(s)
Cuprizone , Demyelinating Diseases , Hericium , Rats , Mice , Animals , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Demyelinating Diseases/drug therapy , Demyelinating Diseases/prevention & control , Rodentia , Oligodendroglia , Myelin Sheath/metabolism , Mice, Inbred C57BL , Disease Models, Animal
19.
Glia ; 72(5): 960-981, 2024 05.
Article in English | MEDLINE | ID: mdl-38363046

ABSTRACT

In the adult brain, activity-dependent myelin plasticity is required for proper learning and memory consolidation. Myelin loss, alteration, or even subtle structural modifications can therefore compromise the network activity, leading to functional impairment. In multiple sclerosis, spontaneous myelin repair process is possible, but it is heterogeneous among patients, sometimes leading to functional recovery, often more visible at the motor level than at the cognitive level. In cuprizone-treated mouse model, massive brain demyelination is followed by spontaneous and robust remyelination. However, reformed myelin, although functional, may not exhibit the same morphological characteristics as developmental myelin, which can have an impact on the activity of neural networks. In this context, we used the cuprizone-treated mouse model to analyze the structural, functional, and cognitive long-term effects of transient demyelination. Our results show that an episode of demyelination induces despite remyelination long-term cognitive impairment, such as deficits in spatial working memory, social memory, cognitive flexibility, and hyperactivity. These deficits were associated with a reduction in myelin content in the medial prefrontal cortex (mPFC) and hippocampus (HPC), as well as structural myelin modifications, suggesting that the remyelination process may be imperfect in these structures. In vivo electrophysiological recordings showed that the demyelination episode altered the synchronization of HPC-mPFC activity, which is crucial for memory processes. Altogether, our data indicate that the myelin repair process following transient demyelination does not allow the complete recovery of the initial myelin properties in cortical structures. These subtle modifications alter network features, leading to prolonged cognitive deficits in mice.


Subject(s)
Cognitive Dysfunction , Demyelinating Diseases , Humans , Animals , Mice , Myelin Sheath , Demyelinating Diseases/chemically induced , Cuprizone/toxicity , Brain , Disease Models, Animal , Cognitive Dysfunction/chemically induced , Mice, Inbred C57BL , Oligodendroglia/physiology
20.
Mol Neurobiol ; 61(9): 6822-6841, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38353925

ABSTRACT

Demyelination is the loss of myelin in CNS, resulting in damaged myelin sheath. Oxidative stress and neuroinflammation play a key role in inducing demyelinating diseases like MS; hence, controlling oxidative stress and neuroinflammation is important. Cuprizone (CPZ), a copper chelator, generates oxidative stress and neuroinflammation, thereby inducing demyelination. Therefore, the CPZ-induced demyelinating mouse model (CPZ model) is widely used in research. The present study was intended to unravel a mechanism of inhibition of demyelination by arsenic in a CPZ model, which is otherwise known for its toxicity. We investigated an alternative mechanism of inhibition of demyelination by arsenic through the reversal of SOD1 activity employing in silico analysis, analytical chemistry techniques, and in vitro and in vivo experiments. In vivo experiments showed protection of body weight, survivability, and myelination of the corpus callosum in CPZ and arsenic-co-exposed animals, where neuroinflammation was apparently not involved. In vitro experiments revealed that arsenic-mediated reversal of impaired SOD1 activity leads to reduced cellular ROS levels and better viability of primary oligodendrocytes. Reversal of SOD1 activity was also observed in the corpus callosum tissue isolated from experimental animals. In silico and analytical chemistry studies revealed that similar to copper, arsenic can potentially bind to CPZ and thereby make the copper freely available for SOD1 activity. Suitable neurobehavior tests further validated the protective effect of arsenic. Taken together, the present study revealed that arsenic protects oligodendrocytes and demyelination of corpus callosum by reversing CPZ-induced impaired SOD1 activity.


Subject(s)
Arsenic , Corpus Callosum , Cuprizone , Demyelinating Diseases , Disease Models, Animal , Microglia , Animals , Cuprizone/toxicity , Corpus Callosum/pathology , Corpus Callosum/drug effects , Corpus Callosum/metabolism , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Microglia/drug effects , Microglia/pathology , Microglia/metabolism , Arsenic/toxicity , Mice, Inbred C57BL , Mice , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Male , Superoxide Dismutase-1/metabolism , Oligodendroglia/drug effects , Oligodendroglia/pathology , Oligodendroglia/metabolism , Myelin Sheath/metabolism , Myelin Sheath/drug effects , Myelin Sheath/pathology , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL