Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
J Neurosci ; 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830764

ABSTRACT

Human genetics and preclinical studies have identified key contributions of TREM2 to several neurodegenerative conditions, inspiring efforts to modulate TREM2 therapeutically. Here, we characterize the activities of three TREM2 agonist antibodies in multiple mixed-sex mouse models of Alzheimer's Disease (AD) pathology and remyelination. Receptor activation and downstream signaling are explored in vitro, and active dose ranges are determined in vivo based on pharmacodynamic responses from microglia. For mice bearing amyloid-ß (Aß) pathology (PS2APP) or combined Aß and tau pathology (TauPS2APP), chronic TREM2 agonist antibody treatment had limited impact on microglia engagement with pathology, overall pathology burden, or downstream neuronal damage. For mice with demyelinating injuries triggered acutely with lysolecithin, TREM2 agonist antibodies unexpectedly disrupted injury resolution. Likewise, TREM2 agonist antibodies limited myelin recovery for mice experiencing chronic demyelination from cuprizone. We highlight the contributions of dose timing and frequency across models. These results introduce important considerations for future TREM2-targeting approaches.Significance Statement Multiple TREM2 agonist antibodies are investigated in mouse models of Alzheimer's Disease and Multiple Sclerosis. Despite agonism in culture models and after acute dosing in mice, antibodies do not show benefit in overall AD pathology and worsen recovery after demyelination.

2.
Interv Neuroradiol ; : 15910199231217547, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38173241

ABSTRACT

OBJECTIVE: To report periprocedural thromboembolic complications of LEO Baby stent-assisted coiling of wide-necked intracranial aneurysms and to analyze the possible influencing factors. METHODS: We retrospectively identified 149 patients with aneurysms who underwent LEO Baby stent-assisted embolization between October 2018 and March 2022. Clinical and radiographic data of patients were reviewed to determine whether a thromboembolic event had occurred. Multivariate logistic analysis was performed to identify significant factors associated with thromboembolic events. RESULTS: Successful stent deployment of the stent was achieved in all patients in the target artery. There were 66 patients (44.3%) with acutely ruptured aneurysms and 83 patients (55.7%) with unruptured aneurysms. Fourteen (9.4%, 95% confidence interval: 4.7%-14.1%) patients were confirmed to have developed a thromboembolic event, including nine patients with acute intraoperative thrombosis and five patients with postoperative thromboembolic events. The rate of thromboembolic events was 6.0% (5/83) in patients with unruptured aneurysms and 13.6% (9/66) in patients with acutely ruptured aneurysms. There was a trend toward an increased rate of thromboembolic events in patients with acute ruptured aneurysms (p = 0.087). Thromboembolic events were significantly associated with the parent-artery diameter (p = 0.010). CONCLUSIONS: Our study demonstrates a low rate of thromboembolic complications in unruptured aneurysms treated with LEO Baby stent. Thromboembolic events appear to be more common in ruptured aneurysms. A small diameter of the parent artery is associated with an increased risk of thromboembolic complications, and more relevant studies are still needed.

3.
Glia ; 71(5): 1247-1258, 2023 05.
Article in English | MEDLINE | ID: mdl-36625077

ABSTRACT

Disability in multiple sclerosis (MS) is driven in part by the failure of remyelination and progressive neurodegeneration. Microglia, and specifically triggering receptor expressed on myeloid cells 2 (TREM2), a factor highly expressed in microglia, have been shown to play an important role in remyelination. Here, using a focal demyelination model in the brain, we demonstrate that demyelination is persistent in TREM2 knockout mice, lasting more than 6 weeks after lysolecithin injection and resulting in substantial neurodegeneration. We also find that TREM2 knockout mice exhibit an altered glial response following demyelination. TREM2 knockout microglia demonstrate defects in migration and phagocytosis of myelin debris. In addition, human monocyte-derived macrophages from subjects with a TREM2 mutation prevalent in human disease also show a defect in myelin debris phagocytosis. Together, we highlight the central role of TREM2 signaling in remyelination and neuroprotection. These findings provide insights into how chronic demyelination might lead to axonal damage and could help identify novel neuroprotective therapeutic targets for MS.


Subject(s)
Multiple Sclerosis , Remyelination , Animals , Mice , Humans , Microglia/physiology , Neuroprotection , Multiple Sclerosis/drug therapy , Myelin Sheath , Mice, Knockout , Mice, Inbred C57BL , Membrane Glycoproteins/genetics , Receptors, Immunologic/genetics
4.
Front Neurol ; 13: 990532, 2022.
Article in English | MEDLINE | ID: mdl-36176553

ABSTRACT

Background and purpose: Low-profile intracranial stents such as the LEO Baby stents are considered to be advantageous for the treatment of intracranial aneurysms originating from small arteries. This study aimed to evaluate the initial and mid-term clinical and angiographic results of LEO Baby stents in stent-assisted coiling of intracranial aneurysms with small parent arteries (<2.5 mm). Methods: We performed a retrospective study to identify 131 patients with aneurysms arising from small parent arteries treated with Leo Baby stent-assisted coiling in a single institution between October 2018 and June 2021. We assessed the immediate and progressive aneurysm occlusion rates, procedure-related complications, and clinical outcomes. Results: A total of 131 patients with 135 aneurysms were identified, including 65 (48.1%) cases of acutely ruptured aneurysms. Technical success was achieved in all cases (100%). The immediate angiography showed complete occlusion in 111 aneurysms (82.2%), neck remnants in 19 (14.1%), and residual sac in 5 (3.7%). Procedure-related complications occurred in 14 cases (10.3%), including 13 (9.6%) thromboembolic complications and 1 (0.7%) hemorrhagic complication. Six-month follow-up angiography was achieved in 106 (78.5%) aneurysms, which showed complete occlusion in 102 (96.2%) aneurysms, neck remnants in 2 (1.9%), and residual sac in 2 (1.9%). Clinical follow-up was available in all patients with a median duration of 6.8 months, and favorable clinical outcomes (modified Rankin Scale score: 0-2) reached 91.6%. The mortality rate was 4.6%. Conclusion: Our results demonstrate that stent-assisted coiling of intracranial aneurysms located on small arteries using LEO Baby stents is technically feasible, highly effective, and has midterm durability in aneurysmal occlusion.

5.
Cell Rep ; 40(8): 111189, 2022 08 23.
Article in English | MEDLINE | ID: mdl-36001972

ABSTRACT

Oligodendrocyte dysfunction has been implicated in the pathogenesis of neurodegenerative diseases, so understanding oligodendrocyte activation states would shed light on disease processes. We identify three distinct activation states of oligodendrocytes from single-cell RNA sequencing (RNA-seq) of mouse models of Alzheimer's disease (AD) and multiple sclerosis (MS): DA1 (disease-associated1, associated with immunogenic genes), DA2 (disease-associated2, associated with genes influencing survival), and IFN (associated with interferon response genes). Spatial analysis of disease-associated oligodendrocytes (DAOs) in the cuprizone model reveals that DA1 and DA2 are established outside of the lesion area during demyelination and that DA1 repopulates the lesion during remyelination. Independent meta-analysis of human single-nucleus RNA-seq datasets reveals that the transcriptional responses of MS oligodendrocytes share features with mouse models. In contrast, the oligodendrocyte activation signature observed in human AD is largely distinct from those observed in mice. This catalog of oligodendrocyte activation states (http://research-pub.gene.com/OligoLandscape/) will be important to understand disease progression and develop therapeutic interventions.


Subject(s)
Demyelinating Diseases , Multiple Sclerosis , Neurodegenerative Diseases , Animals , Cuprizone/therapeutic use , Demyelinating Diseases/pathology , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Oligodendroglia
6.
Cell Rep ; 34(10): 108835, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33691116

ABSTRACT

In multiple sclerosis (MS) and other neurological diseases, the failure to repair demyelinated lesions contributes to axonal damage and clinical disability. Here, we provide evidence that Mertk, a gene highly expressed by microglia that alters MS risk, is required for efficient remyelination. Compared to wild-type (WT) mice, Mertk-knockout (KO) mice show impaired clearance of myelin debris and remyelination following demyelination. Using single-cell RNA sequencing, we characterize Mertk-influenced responses to cuprizone-mediated demyelination and remyelination across different cell types. Mertk-KO brains show an attenuated microglial response to demyelination but an elevated proportion of interferon (IFN)-responsive microglia. In addition, we identify a transcriptionally distinct subtype of surviving oligodendrocytes specific to demyelinated lesions. The inhibitory effect of myelin debris on remyelination is mediated in part by IFNγ, which further impedes microglial clearance of myelin debris and inhibits oligodendrocyte differentiation. Together, our work establishes a role for Mertk in microglia activation, phagocytosis, and migration during remyelination.


Subject(s)
Microglia/metabolism , Multiple Sclerosis/pathology , c-Mer Tyrosine Kinase/metabolism , Animals , Cell Differentiation , Cell Movement , Cuprizone/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Demyelinating Diseases/chemically induced , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Disease Models, Animal , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Multiple Sclerosis/genetics , Myelin Sheath/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Phagocytosis , Remyelination/drug effects , c-Mer Tyrosine Kinase/deficiency , c-Mer Tyrosine Kinase/genetics
7.
J Neurosci ; 39(12): 2184-2194, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30696729

ABSTRACT

A significant unmet need for patients with multiple sclerosis (MS) is the lack of U.S. Food and Drug Administration (FDA)-approved remyelinating therapies. We have identified a compelling remyelinating agent, bazedoxifene (BZA), a European Medicines Agency (EMA)-approved (and FDA-approved in combination with conjugated estrogens) selective estrogen receptor (ER) modulator (SERM) that could move quickly from bench to bedside. This therapy stands out as a tolerable alternative to previously identified remyelinating agents and other candidates within this family. Using an unbiased high-throughput screen, with subsequent validation in both murine and human oligodendrocyte precursor cells (OPCs) and coculture systems, we find that BZA enhances differentiation of OPCs into functional oligodendrocytes. Using an in vivo murine model of focal demyelination, we find that BZA enhances OPC differentiation and remyelination. Of critical importance, we find that BZA acts independently of its presumed target, the ER, in both in vitro and in vivo systems. Using a massive computational data integration approach, we independently identify six possible candidate targets through which SERMs may mediate their effect on remyelination. Of particular interest, we identify EBP (encoding 3ß-hydroxysteroid-Δ8,Δ7-isomerase), a key enzyme in the cholesterol biosynthesis pathway, which was previously implicated as a target for remyelination. These findings provide valuable insights into the implications for SERMs in remyelination for MS and hormonal research at large.SIGNIFICANCE STATEMENT Therapeutics targeted at remyelination failure, which results in axonal degeneration and ultimately disease progression, represent a large unmet need in the multiple sclerosis (MS) population. Here, we have validated a tolerable European Medicines Agency-approved (U.S. Food and Drug Administration-approved in combination with conjugated estrogens) selective estrogen receptor (ER) modulator (SERM), bazedoxifene (BZA), as a potent agent of oligodendrocyte precursor cell (OPC) differentiation and remyelination. SERMs, which were developed as nuclear ER-α and ER-ß agonists/antagonists, have previously been implicated in remyelination and neuroprotection, following a heavy focus on estrogens with underwhelming and conflicting results. We show that nuclear ERs are not required for SERMs to mediate their potent effects on OPC differentiation and remyelination in vivo and highlight EBP, an enzyme in the cholesterol biosynthesis pathway that could potentially act as a target for SERMs.


Subject(s)
Indoles/administration & dosage , Oligodendrocyte Precursor Cells/drug effects , Oligodendroglia/drug effects , Receptors, Estrogen/physiology , Remyelination/drug effects , Selective Estrogen Receptor Modulators/administration & dosage , Animals , Cell Differentiation/drug effects , Disease Models, Animal , Female , Male , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Oligodendrocyte Precursor Cells/physiology , Oligodendroglia/physiology
8.
Cell Rep ; 25(3): 544-550.e3, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30332636

ABSTRACT

Emerging evidence suggests that neuronal signaling is important for oligodendrocyte myelination; however, the necessity of this signaling during development is unclear. By eliminating dynamic neuronal signaling along the developing optic nerve, we find that oligodendrocyte differentiation is not dependent on neuronal signaling and that the initiation of myelination is dependent on a permissive substrate, namely supra-threshold axon caliber. Furthermore, we show that loss of dynamic neuronal signaling results in hypermyelination of axons. We propose that oligodendrocyte differentiation is regulated by non-neuronal factors during optic nerve development, whereas myelination is sensitive to the biophysical properties of axonal diameter.


Subject(s)
Axons/physiology , Brain/physiology , Myelin Sheath/physiology , Neurogenesis , Oligodendroglia/physiology , Optic Nerve/physiology , Animals , Axons/chemistry , Brain/cytology , Cell Differentiation , Cell Proliferation , Female , Male , Mice , Mice, Knockout , Oligodendroglia/cytology , Optic Nerve/cytology , PTEN Phosphohydrolase/physiology , Signal Transduction
9.
Methods Mol Biol ; 1791: 169-177, 2018.
Article in English | MEDLINE | ID: mdl-30006709

ABSTRACT

Myelination necessitates axons to initiate concentric membrane wrapping by oligodendroglia in the CNS. Here, we describe an in vitro system that models CNS myelination with a minimally permissive environment, termed Binary Indicant for myelination using Micropillar Arrays (BIMA). Engineered with conical micropillar arrays, BIMA allows for rapid translation of oligodendroglial wrapping and differentiation into binary readout under confocal microscopy. Fabricated into 96- or 384-well plates, BIMA serves as a high-throughput screening platform for compounds that may promote oligodendroglial differentiation and myelination. BIMA is also amenable for examining molecular signals and pathways that regulate axon-glia interaction and recognition.


Subject(s)
Central Nervous System/physiology , Models, Biological , Myelin Sheath/metabolism , Neurogenesis , Protein Array Analysis , Animals , Biomarkers , Cell Culture Techniques , Cells, Cultured , Central Nervous System/cytology , Fluorescent Antibody Technique , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Protein Array Analysis/methods , Rats
10.
Brain ; 141(1): 85-98, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29244098

ABSTRACT

Hypoxia can injure brain white matter tracts, comprised of axons and myelinating oligodendrocytes, leading to cerebral palsy in neonates and delayed post-hypoxic leukoencephalopathy (DPHL) in adults. In these conditions, white matter injury can be followed by myelin regeneration, but myelination often fails and is a significant contributor to fixed demyelinated lesions, with ensuing permanent neurological injury. Non-myelinating oligodendrocyte precursor cells are often found in lesions in plentiful numbers, but fail to mature, suggesting oligodendrocyte precursor cell differentiation arrest as a critical contributor to failed myelination in hypoxia. We report a case of an adult patient who developed the rare condition DPHL and made a nearly complete recovery in the setting of treatment with clemastine, a widely available antihistamine that in preclinical models promotes oligodendrocyte precursor cell differentiation. This suggested possible therapeutic benefit in the more clinically prevalent hypoxic injury of newborns, and we demonstrate in murine neonatal hypoxic injury that clemastine dramatically promotes oligodendrocyte precursor cell differentiation, myelination, and improves functional recovery. We show that its effect in hypoxia is oligodendroglial specific via an effect on the M1 muscarinic receptor on oligodendrocyte precursor cells. We propose clemastine as a potential therapy for hypoxic brain injuries associated with white matter injury and oligodendrocyte precursor cell maturation arrest.


Subject(s)
Clemastine/therapeutic use , Demyelinating Diseases/drug therapy , Demyelinating Diseases/etiology , Histamine H1 Antagonists/therapeutic use , Hypoxia, Brain/complications , Recovery of Function/drug effects , Action Potentials/drug effects , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cells, Cultured , Cerebellum/drug effects , Cerebellum/metabolism , Cerebellum/ultrastructure , Demyelinating Diseases/diagnostic imaging , Demyelinating Diseases/pathology , Disease Models, Animal , Gene Expression Regulation, Developmental/drug effects , Humans , Hypoxia, Brain/diagnostic imaging , Male , Mice , Mice, Knockout , Middle Aged , Myelin Sheath/drug effects , Myelin Sheath/ultrastructure , Oligodendrocyte Precursor Cells/drug effects , Optic Nerve/physiopathology , Oxygen/pharmacology , Receptor, Muscarinic M1/genetics , Receptor, Muscarinic M1/metabolism
11.
Elife ; 52016 Sep 27.
Article in English | MEDLINE | ID: mdl-27671734

ABSTRACT

Demyelination in MS disrupts nerve signals and contributes to axon degeneration. While remyelination promises to restore lost function, it remains unclear whether remyelination will prevent axonal loss. Inflammatory demyelination is accompanied by significant neuronal loss in the experimental autoimmune encephalomyelitis (EAE) mouse model and evidence for remyelination in this model is complicated by ongoing inflammation, degeneration and possible remyelination. Demonstrating the functional significance of remyelination necessitates selectively altering the timing of remyelination relative to inflammation and degeneration. We demonstrate accelerated remyelination after EAE induction by direct lineage analysis and hypothesize that newly formed myelin remains stable at the height of inflammation due in part to the absence of MOG expression in immature myelin. Oligodendroglial-specific genetic ablation of the M1 muscarinic receptor, a potent negative regulator of oligodendrocyte differentiation and myelination, results in accelerated remyelination, preventing axonal loss and improving functional recovery. Together our findings demonstrate that accelerated remyelination supports axonal integrity and neuronal function after inflammatory demyelination.

12.
Neuron ; 91(4): 824-836, 2016 Aug 17.
Article in English | MEDLINE | ID: mdl-27499083

ABSTRACT

Myelination occurs selectively around neuronal axons to increase the efficiency and velocity of action potentials. While oligodendrocytes are capable of myelinating permissive structures in the absence of molecular cues, structurally permissive neuronal somata and dendrites remain unmyelinated. Utilizing a purified spinal cord neuron-oligodendrocyte myelinating co-culture system, we demonstrate that disruption of dynamic neuron-oligodendrocyte signaling by chemical cross-linking results in aberrant myelination of the somatodendritic compartment of neurons. We hypothesize that an inhibitory somatodendritic cue is necessary to prevent non-axonal myelination. Using next-generation sequencing and candidate profiling, we identify neuronal junction adhesion molecule 2 (JAM2) as an inhibitory myelin-guidance molecule. Taken together, our results demonstrate that the somatodendritic compartment directly inhibits myelination and suggest a model in which broadly indiscriminate myelination is tailored by inhibitory signaling to meet local myelination requirements.


Subject(s)
Junctional Adhesion Molecule B/physiology , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Animals , Coculture Techniques , Junctional Adhesion Molecule B/biosynthesis , Junctional Adhesion Molecule B/genetics , Mice , Mice, Knockout , Myelin Sheath/ultrastructure , Oligodendroglia/ultrastructure , Primary Cell Culture , Rats , Spinal Cord/physiology , Spinal Cord/ultrastructure
13.
J Cell Biol ; 210(6): 891-8, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26347141

ABSTRACT

Myelin oligodendrocyte glycoprotein (MOG) is a central nervous system myelin-specific molecule expressed on the outer lamellae of myelin. To date, the exact function of MOG has remained unknown, with MOG knockout mice displaying normal myelin ultrastructure and no apparent specific phenotype. In this paper, we identify nerve growth factor (NGF) as a binding partner for MOG and demonstrate that this interaction is capable of sequestering NGF from TrkA-expressing neurons to modulate axon growth and survival. Deletion of MOG results in aberrant sprouting of nociceptive neurons in the spinal cord. Binding of NGF to MOG may offer widespread implications into mechanisms that underlie pain pathways.


Subject(s)
Axons/metabolism , Ganglia, Spinal/metabolism , Myelin-Oligodendrocyte Glycoprotein/metabolism , Nerve Growth Factor/metabolism , Oligodendroglia/metabolism , Spinal Cord/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cell Survival , Coculture Techniques , Cricetulus , Ganglia, Spinal/pathology , Genotype , Mice, Knockout , Molecular Sequence Data , Myelin-Oligodendrocyte Glycoprotein/deficiency , Myelin-Oligodendrocyte Glycoprotein/genetics , Phenotype , Protein Binding , Rats, Sprague-Dawley , Receptor, trkA/metabolism , Signal Transduction , Spinal Cord/pathology , Transfection
14.
Nat Commun ; 5: 4991, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25255972

ABSTRACT

The Schwann cell (SC)-axon interface represents a membrane specialization that integrates axonal signals to coordinate cytoskeletal dynamics resulting in myelination. Here we show that LKB1/Par-4 is asymmetrically localized to the SC-axon interface and co-localizes with the polarity protein Par-3. Using purified SCs and myelinating cocultures, we demonstrate that localization is dependent on the phosphorylation of LKB1 at serine-431. SC-specific deletion of LKB1 significantly attenuates developmental myelination, delaying the initiation and altering the myelin extent into adulthood, resulting in a 30% reduction in the conduction velocity along the adult sciatic nerves. Phosphorylation of LKB1 by protein kinase A is essential to establish the asymmetric localization of LKB1 and Par-3 and rescues the delay in myelination observed in the SC-specific knockout of LKB1. Our findings suggest that SC polarity may coordinate multiple signalling complexes that couple SC-axon contact to the redistribution of specific membrane components necessary to initiate and control myelin extent.


Subject(s)
Cell Polarity , Myelin Sheath/metabolism , Protein Serine-Threonine Kinases/metabolism , Schwann Cells/cytology , Schwann Cells/enzymology , AMP-Activated Protein Kinases , Adaptor Proteins, Signal Transducing , Amino Acid Motifs , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Cycle Proteins , Cells, Cultured , Mice , Mice, Knockout , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Rats , Schwann Cells/metabolism
15.
Nat Med ; 20(8): 954-960, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24997607

ABSTRACT

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


Subject(s)
High-Throughput Screening Assays/methods , Multiple Sclerosis/drug therapy , Muscarinic Antagonists/isolation & purification , Nerve Fibers, Myelinated/drug effects , Animals , Cell Differentiation/drug effects , Cells, Cultured , Clemastine/pharmacology , Drug Evaluation, Preclinical/methods , Female , Histamine H1 Antagonists/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscarinic Antagonists/pharmacology , Nanostructures , Oligodendroglia/cytology , Oligodendroglia/drug effects , Oligodendroglia/physiology , Rats , Rats, Sprague-Dawley , Regeneration/drug effects
16.
Proc Natl Acad Sci U S A ; 109(4): 1299-304, 2012 Jan 24.
Article in English | MEDLINE | ID: mdl-22160722

ABSTRACT

A requisite component of nervous system development is the achievement of cellular recognition and spatial segregation through competition-based refinement mechanisms. Competition for available axon space by myelinating oligodendrocytes ensures that all relevant CNS axons are myelinated properly. To ascertain the nature of this competition, we generated a transgenic mouse with sparsely labeled oligodendrocytes and establish that individual oligodendrocytes occupying similar axon tracts can greatly vary the number and lengths of their myelin internodes. Here we show that intercellular interactions between competing oligodendroglia influence the number and length of myelin internodes, referred to as myelinogenic potential, and identify the amino-terminal region of Nogo-A, expressed by oligodendroglia, as necessary and sufficient to inhibit this process. Exuberant and expansive myelination/remyelination is detected in the absence of Nogo during development and after demyelination, suggesting that spatial segregation and myelin extent is limited by microenvironmental inhibition. We demonstrate a unique physiological role for Nogo-A in the precise myelination of the developing CNS. Maximizing the myelinogenic potential of oligodendrocytes may offer an effective strategy for repair in future therapies for demyelination.


Subject(s)
Central Nervous System/pathology , Demyelinating Diseases/physiopathology , Myelin Proteins/metabolism , Myelin Sheath/physiology , Oligodendroglia/physiology , Animals , Blotting, Western , Central Nervous System/cytology , Gene Knockdown Techniques , Histological Techniques , Mice , Mice, Transgenic , Microscopy, Electron , Microspheres , Myelin Proteins/genetics , Nogo Proteins , Oligodendroglia/metabolism , Oligodendroglia/ultrastructure , Polystyrenes , RNA, Small Interfering/genetics , Ultracentrifugation
17.
J Neurosci ; 31(8): 3032-43, 2011 Feb 23.
Article in English | MEDLINE | ID: mdl-21414924

ABSTRACT

Myelination is dependent on complex reciprocal interactions between the Schwann cell (SC) and axon. Recent evidence suggests that the SC-axon interface represents a membrane specialization essential for myelination; however, the manner in which this polarized-apical domain is generated remains a mystery. The cell adhesion molecule N-cadherin is enriched at the SC-axon interface and colocalizes with the polarity protein Par-3. The asymmetric localization is induced on SC-SC and SC-axon contact. Knockdown of N-cadherin in SCs cocultured with DRG neurons disrupts Par-3 localization and delays the initiation of myelination. However, knockdown or overexpression of neuronal N-cadherin does not influence the distribution of Par-3 or myelination, suggesting that homotypic interactions between SC and axonal N-cadherin are not essential for the events surrounding myelination. To further investigate the role of N-cadherin, mice displaying SC-specific gene ablation of N-cadherin were generated and characterized. Surprisingly, myelination is only slightly delayed, and mice are viable without any detectable myelination defects. ß-Catenin, a downstream effector of N-cadherin, colocalizes and coimmunoprecipitates with N-cadherin on the initiation of myelination. To determine whether ß-catenin mediates compensation on N-cadherin deletion, SC-specific gene ablation of ß-catenin was generated and characterized. Consistent with our hypothesis, myelination is more severely delayed than when manipulating N-cadherin alone, but without any defect to the myelin sheath. Together, our results suggest that N-cadherin interacts with ß-catenin in establishing SC polarity and the timely initiation of myelination, but they are nonessential components for the formation and maturation of the myelin sheath.


Subject(s)
Axons/physiology , Cadherins/physiology , Ganglia, Spinal/embryology , Myelin Sheath/physiology , Schwann Cells/metabolism , beta Catenin/physiology , Animals , Animals, Newborn , Cadherins/genetics , Cell Polarity/physiology , Cells, Cultured , Coculture Techniques , Focal Adhesions/physiology , Ganglia, Spinal/cytology , Ganglia, Spinal/growth & development , Mice , Mice, Knockout , Rats , Schwann Cells/cytology , Schwann Cells/physiology , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , beta Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...