Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biomed Res Int ; 2017: 5071786, 2017.
Article in English | MEDLINE | ID: mdl-29181395

ABSTRACT

Neuroinflammation is recognised as one of the potential mechanisms mediating the onset of a broad range of psychiatric disorders and may contribute to nonresponsiveness to current therapies. Both preclinical and clinical studies have indicated that aberrant inflammatory responses can result in altered behavioral responses and cognitive deficits. In this review, we discuss the role of inflammation in the pathogenesis of neuropsychiatric disorders and ask the question if certain genetic copy-number variants (CNVs) associated with psychiatric disorders might play a role in modulating inflammation. Furthermore, we detail some of the potential treatment strategies for psychiatric disorders that may operate by altering inflammatory responses.


Subject(s)
Cognitive Dysfunction , Genetic Variation , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/therapy , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/physiopathology , Inflammation/therapy
3.
Acta Neuropathol Commun ; 4(1): 89, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27552807

ABSTRACT

Cervical spondylotic myelopathy (CSM) is the most common spinal cord disorder and a major cause of disability in adults. Improvements following surgical decompression are limited and patients often remain severely disabled. Post mortem studies indicate that CSM is associated with profound axonal loss. However, our understanding of the pathophysiology of CSM remains limited.To investigate the hypothesis that axonal plasticity plays a role in the recovery following surgical decompression, we adopted a novel preclinical model of mild to moderate CSM. Spinal cord compression resulted in significant locomotor deterioration, increased expression of the axonal injury marker APP, and loss of serotonergic fibres. Surgical decompression partially reversed the deficits and attenuated APP expression. Decompression was also associated with axonal sprouting, reflected in the restoration of serotonergic fibres and an increase of GAP43 expression. The re-expression of synaptophysin indicated the restoration of functional synapses following decompression. Promoting axonal plasticity may therefore be a therapeutic strategy for promoting neurological recovery in CSM.


Subject(s)
Axons/physiology , Decompression, Surgical , Neuronal Plasticity/physiology , Recovery of Function/physiology , Spinal Cord Compression/surgery , Spondylosis/surgery , Animals , Apoptosis/physiology , Axons/pathology , Disease Models, Animal , Immunohistochemistry , Male , Motor Activity/physiology , Neuroglia/pathology , Neuroglia/physiology , Random Allocation , Rats, Sprague-Dawley , Severity of Illness Index , Spinal Cord Compression/pathology , Spinal Cord Compression/physiopathology , Spondylosis/pathology , Spondylosis/physiopathology , Synapses/pathology , Synapses/physiology , Synaptophysin/metabolism , Treatment Outcome
4.
Sci Rep ; 6: 31599, 2016 08 24.
Article in English | MEDLINE | ID: mdl-27554391

ABSTRACT

Enhancing central nervous system (CNS) myelin regeneration is recognized as an important strategy to ameliorate the devastating consequences of demyelinating diseases such as multiple sclerosis. Previous findings have indicated that myelin proteins, which accumulate following demyelination, inhibit remyelination by blocking the differentiation of rat oligodendrocyte progenitor cells (OPCs) via modulation of PKCα. We therefore screened drugs for their potential to overcome this differentiation block. From our screening, tamoxifen emerges as a potent inducer of OPC differentiation in vitro. We show that the effects of tamoxifen rely on modulation of the estrogen receptors ERα, ERß, and GPR30. Furthermore, we demonstrate that administration of tamoxifen to demyelinated rats in vivo accelerates remyelination. Tamoxifen is a well-established drug and is thus a promising candidate for a drug to regenerate myelin, as it will not require extensive safety testing. In addition, Tamoxifen plays an important role in biomedical research as an activator of inducible genetic models. Our results highlight the importance of appropriate controls when using such models.


Subject(s)
Cell Differentiation/drug effects , Demyelinating Diseases , Neural Stem Cells , Oligodendroglia , Tamoxifen/pharmacology , Animals , Demyelinating Diseases/drug therapy , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Rats , Rats, Sprague-Dawley
6.
Acta Neuropathol ; 131(2): 281-298, 2016 02.
Article in English | MEDLINE | ID: mdl-26687980

ABSTRACT

Remyelination in multiple sclerosis (MS) lesions often remains incomplete despite the presence of oligodendrocyte progenitor cells (OPCs). Amongst other factors, successful remyelination depends on the phagocytic clearance of myelin debris. However, the proteins in myelin debris that act as potent and selective inhibitors on OPC differentiation and inhibit CNS remyelination remain unknown. Here, we identify the transmembrane signalling protein EphrinB3 as important mediator of this inhibition, using a protein analytical approach in combination with a primary rodent OPC assay. In the presence of EphrinB3, OPCs fail to differentiate. In a rat model of remyelination, infusion of EphrinB3 inhibits remyelination. In contrast, masking EphrinB3 epitopes using antibodies promotes remyelination. Finally, we identify EphrinB3 in MS lesions and demonstrate that MS lesion extracts inhibit OPC differentiation while antibody-mediated masking of EphrinB3 epitopes promotes it. Our findings suggest that EphrinB3 could be a target for therapies aiming at promoting remyelination in demyelinating disease.


Subject(s)
Ephrin-B3/metabolism , Multiple Sclerosis/metabolism , Myelin Sheath/metabolism , Neural Stem Cells/metabolism , Oligodendroglia/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Ephrin-B3/genetics , Epitopes/metabolism , Female , Humans , Macrophages/metabolism , Macrophages/pathology , Mice, Knockout , Multiple Sclerosis/pathology , Myelin Sheath/pathology , Nerve Regeneration/physiology , Neural Stem Cells/pathology , Neurogenesis/physiology , Oligodendroglia/pathology , Random Allocation , Rats, Sprague-Dawley , Receptor, EphA4/metabolism
7.
Stem Cell Reports ; 1(4): 293-306, 2013.
Article in English | MEDLINE | ID: mdl-24319665

ABSTRACT

Human pluripotent stem cells (hPSCs) could provide an infinite source of clinically relevant cells with potential applications in regenerative medicine. However, hPSC lines vary in their capacity to generate specialized cells, and the development of universal protocols for the production of tissue-specific cells remains a major challenge. Here, we have addressed this limitation for the endodermal lineage by developing a defined culture system to expand and differentiate human foregut stem cells (hFSCs) derived from hPSCs. hFSCs can self-renew while maintaining their capacity to differentiate into pancreatic and hepatic cells. Furthermore, near-homogenous populations of hFSCs can be obtained from hPSC lines which are normally refractory to endodermal differentiation. Therefore, hFSCs provide a unique approach to bypass variability between pluripotent lines in order to obtain a sustainable source of multipotent endoderm stem cells for basic studies and to produce a diversity of endodermal derivatives with a clinical value.


Subject(s)
Cell Line , Gastrula/cytology , Multipotent Stem Cells/cytology , Pluripotent Stem Cells/cytology , Biomarkers/metabolism , Cell Culture Techniques , Cell Differentiation , Humans , Multipotent Stem Cells/metabolism , Pluripotent Stem Cells/metabolism
8.
EMBO Mol Med ; 5(12): 1918-34, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24293318

ABSTRACT

The increasing effectiveness of new disease-modifying drugs that suppress disease activity in multiple sclerosis has opened up opportunities for regenerative medicines that enhance remyelination and potentially slow disease progression. Although several new targets for therapeutic enhancement of remyelination have emerged, few lend themselves readily to conventional drug development. Here, we used transcription profiling to identify mitogen-activated protein kinase (Mapk) signalling as an important regulator involved in the differentiation of oligodendrocyte progenitor cells (OPCs) into oligodendrocytes. We show in tissue culture that activation of Mapk signalling by elevation of intracellular levels of cyclic adenosine monophosphate (cAMP) using administration of either dibutyryl-cAMP or inhibitors of the cAMP-hydrolysing enzyme phosphodiesterase-4 (Pde4) enhances OPC differentiation. Finally, we demonstrate that systemic delivery of a Pde4 inhibitor leads to enhanced differentiation of OPCs within focal areas of toxin-induced demyelination and a consequent acceleration of remyelination. These data reveal a novel approach to therapeutic enhancement of remyelination amenable to pharmacological intervention and hence with significant potential for translation.


Subject(s)
Cell Differentiation , Central Nervous System/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Myelin Sheath/metabolism , Animals , Bucladesine/chemistry , Bucladesine/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Humans , Immunity, Innate/drug effects , Mitogen-Activated Protein Kinases/metabolism , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Myelin Sheath/chemistry , Oligodendroglia/cytology , Oligodendroglia/metabolism , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Transcriptome
9.
J Neurosci ; 31(10): 3719-28, 2011 Mar 09.
Article in English | MEDLINE | ID: mdl-21389227

ABSTRACT

Failure of oligodendrocyte precursor cell (OPC) differentiation has been recognized as the leading cause for the failure of myelin regeneration in diseases such as multiple sclerosis (MS). One explanation for the failure of OPC differentiation in MS is the presence of inhibitory molecules in demyelinated lesions. So far only a few inhibitory substrates have been identified in MS lesions. Semaphorin 3A (Sema3A), a secreted member of the semaphorin family, can act as repulsive guidance cue for neuronal and glial cells in the CNS. Recent studies suggest that Sema3A is also expressed in active MS lesions. However, the implication of Sema3A expression in MS lesions remains unclear as OPCs are commonly present in chronic demyelinated lesions. In the present study we identify Sema3A as a potent, selective, and reversible inhibitor of OPC differentiation in vitro. Furthermore, we show that administration of Sema3A into demyelinating lesions in the rat CNS results in a failure of remyelination. Our results imply an important role for Sema3A in the differentiation block occurring in MS lesions.


Subject(s)
Cell Differentiation/physiology , Myelin Sheath/metabolism , Nerve Regeneration/physiology , Oligodendroglia/metabolism , Semaphorin-3A/metabolism , Analysis of Variance , Animals , Animals, Newborn , Cell Differentiation/drug effects , Female , Immunohistochemistry , In Situ Hybridization , Myelin Sheath/drug effects , Myelin Sheath/pathology , Oligodendroglia/drug effects , Oligodendroglia/pathology , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Semaphorin-3A/pharmacology
10.
Brain ; 132(Pt 2): 465-81, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19208690

ABSTRACT

Failure of oligodendrocyte precursor cell (OPC) differentiation contributes significantly to failed myelin sheath regeneration (remyelination) in chronic demyelinating diseases. Although the reasons for this failure are not completely understood, several lines of evidence point to factors present following demyelination that specifically inhibit differentiation of cells capable of generating remyelinating oligodendrocytes. We have previously demonstrated that myelin debris generated by demyelination inhibits remyelination by inhibiting OPC differentiation and that the inhibitory effects are associated with myelin proteins. In the present study, we narrow down the spectrum of potential protein candidates by proteomic analysis of inhibitory protein fractions prepared by CM and HighQ column chromatography followed by BN/SDS/SDS-PAGE gel separation using Nano-HPLC-ESI-Q-TOF mass spectrometry. We show that the inhibitory effects on OPC differentiation mediated by myelin are regulated by Fyn-RhoA-ROCK signalling as well as by modulation of protein kinase C (PKC) signalling. We demonstrate that pharmacological or siRNA-mediated inhibition of RhoA-ROCK-II and/or PKC signalling can induce OPC differentiation in the presence of myelin. Our results, which provide a mechanistic link between myelin, a mediator of OPC differentiation inhibition associated with demyelinating pathologies and specific signalling pathways amenable to pharmacological manipulation, are therefore of significant potential value for future strategies aimed at enhancing CNS remyelination.


Subject(s)
Demyelinating Diseases/pathology , Myelin Sheath/metabolism , Oligodendroglia/pathology , Proto-Oncogene Proteins c-fyn/metabolism , Stem Cells/pathology , rhoA GTP-Binding Protein/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Animals, Newborn , Carbazoles/pharmacology , Cell Differentiation , Demyelinating Diseases/metabolism , Electrophoresis, Polyacrylamide Gel , Indoles/pharmacology , Maleimides/pharmacology , Nerve Regeneration , Oligodendroglia/metabolism , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/genetics , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stem Cells/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/genetics
11.
Neurosurg Focus ; 24(3-4): E5, 2008.
Article in English | MEDLINE | ID: mdl-18341408

ABSTRACT

OBJECT: Promoting repair of central nervous system (CNS) white matter represents an important approach to easing the course of a number of tragic neurological diseases. For this purpose, strategies are currently being evaluated for transplanting cells capable of generating new oligodendrocytes into areas of demyelination and/or enhancing the potential of endogenous stem/precursor cells to give rise to new oligodendrocytes. Emerging evidence, however, indicates that increasing the presence of cells capable of forming new myelin sheaths is not sufficient to promote repair because of unknown inhibitors that accumulate in lesions as a consequence of myelin degeneration and impair the generation of new oligodendrocytes. The aim of the present study was to characterize the nature of the inhibitory molecules present in myelin. METHODS: Differentiation of primary rat oligodendrocyte precursor cells (OPCs) in the presence of CNS and peripheral nervous system myelin was assessed by immunocytochemical methods. The authors further characterized the nature of the inhibitors by submitting myelin membrane preparations to biochemical precipitation and digestion. Finally, OPCs were grown on purified Nogo-A, oligodendrocyte myelin glycoprotein, and myelin-associated glycoprotein, the most prominent inhibitors of axon regeneration. RESULTS: Myelin membrane preparations induced a differentiation block in OPCs that was associated with down-regulation of expression of the transcription factor Nkx2.2. The inhibitory activity in myelin was restricted to the CNS and was predominantly associated with white matter. Furthermore, the results demonstrate that myelin proteins that are distinct from the most prominent inhibitors of axon outgrowth are specific inhibitors of OPC differentiation. CONCLUSIONS: The inhibitory effect of unknown myelin-associated proteins should be considered in future treatment strategies aimed at enhancing CNS repair.


Subject(s)
Myelin Basic Protein/metabolism , Oligodendroglia/physiology , Stem Cells/physiology , Analysis of Variance , Animals , Animals, Newborn , Axons/metabolism , CHO Cells , Cell Differentiation , Cells, Cultured , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Myelin Basic Protein/pharmacology , Oligodendroglia/drug effects , Prosencephalon/cytology , Rats , Rats, Sprague-Dawley , Stem Cells/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection/methods , Zebrafish Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...