Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Brain ; 143(7): 2073-2088, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32577755

ABSTRACT

Multiple sclerosis is an immune-mediated chronic inflammatory disease of the CNS that leads to demyelinated lesions in the grey and white matter. Inflammatory, active demyelinating white matter lesions predominate in the relapsing-remitting disease stages, whereas in the progressive stage the so-called slowly expanding lesion is characteristic. These lesions show an accumulation of macrophages/microglia at their borders, mediating the ongoing myelin breakdown and axonal degeneration. The exact pathogenetic mechanisms of lesion progression in chronic multiple sclerosis are still not clear. In the present study, we performed a detailed immunological and molecular profiling of slowly expanding lesions (n = 21) from 13 patients aged between 30 to 74 years (five females and eight males), focusing on macrophage/microglia differentiation. By applying the microglia-specific marker TMEM119, we demonstrate that cells accumulating at the lesion edge almost exclusively belonged to the microglia lineage. Macrophages/microglia can be subdivided into the M1 type, which are associated with inflammatory and degenerative processes, and M2 type, with protective properties, whereby also intermediate polarization phenotypes can be observed. By using a panel of markers characterizing M1- or M2-type macrophages/microglia, we observed a preferential accumulation of M1-type differentiated cells at the lesion edge, indicating a crucial role of these cells in lesion progression. Additionally, unbiased RNA microarray analyses of macrodissected lesion edges from slowly expanding and chronic inactive lesions as well as normal-appearing white matter were performed. In slowly expanding lesions, we identified a total of 165 genes that were upregulated and 35 genes that were downregulated. The upregulated genes included macrophage/microglia-associated genes involved in immune defence and inflammatory processes. Among the upregulated genes were ALOX15B, MME and TNFRSF25. We confirmed increased expression of ALOX15B by quantitative PCR, and of all three genes on the protein level by immunohistochemistry. In conclusion, the present study characterized in detail slowly expanding lesions in progressive multiple sclerosis and demonstrated a preferential accumulation of resident microglia with M1 differentiation at the lesion edge. Microarray analysis showed an increased expression of genes related to immune function, metabolic processes as well as transcription/translation. Thus, these genes may serve as future therapeutic targets to impede lesion progression.


Subject(s)
Brain/immunology , Brain/pathology , Microglia/pathology , Multiple Sclerosis, Chronic Progressive/immunology , Multiple Sclerosis, Chronic Progressive/pathology , Adult , Aged , Disease Progression , Female , Humans , Male , Middle Aged
2.
Neurobiol Dis ; 49: 221-31, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22940629

ABSTRACT

Charcot-Marie-Tooth disease type 1A (CMT1A) is a hereditary demyelinating peripheral neuropathy caused by the duplication of the PMP22 gene. Demyelination precedes the occurrence of clinical symptoms that correlate with axonal degeneration. It was postulated that a disturbed axon-glia interface contributes to altered myelination consequently leading to axonal degeneration. In this study, we examined the expression of MAG and Necl4, two critical adhesion molecules that are present at the axon-glia interface, in sural nerve biopsies of CMT1A patients and in peripheral nerves of mice overexpressing human PMP22, an animal model for CMT1A. We show an increase in the expression of MAG and a strong decrease of Necl4 in biopsies of CMT1A patients as well as in CMT1A mice. Expression analysis revealed that MAG is strongly upregulated during peripheral nerve maturation, whereas Necl4 expression remains very low. Ablating MAG in CMT1A mice results in separation of axons from their myelin sheath. Our data show that MAG is important for axon-glia contact in a model for CMT1A, and suggest that its increased expression in CMT1A disease has a compensatory role in the pathology of the disease. Thus, we demonstrate that MAG together with other adhesion molecules such as Necl4 is important in sustaining axonal integrity.


Subject(s)
Axons/metabolism , Charcot-Marie-Tooth Disease/metabolism , Myelin Sheath/metabolism , Myelin-Associated Glycoprotein/metabolism , Sural Nerve/metabolism , Adult , Aged, 80 and over , Animals , Axons/pathology , Cell Adhesion Molecules/metabolism , Charcot-Marie-Tooth Disease/pathology , Disease Models, Animal , Female , Humans , Immunoglobulins/metabolism , Male , Mice, Knockout , Mice, Transgenic , Middle Aged , Myelin Proteins/genetics , Myelin Proteins/metabolism , Myelin Sheath/pathology , Myelin-Associated Glycoprotein/genetics , Sural Nerve/pathology , Young Adult
3.
J Immunol ; 186(6): 3452-61, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21289301

ABSTRACT

Autoimmune encephalomyelitis is a disease of the CNS that can develop when an initial peripheral inflammatory stimulus is followed by infiltration and reactivation of T lymphocytes in the CNS. We report a crucial role for coronin 1, which is essential for maintenance of the naive T cell pool, for the development of murine experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis. In the absence of coronin 1, immunization with myelin oligoglycoprotein (MOG(35-55)) peptide largely failed to induce EAE symptoms, despite normal mobilization of leukocyte subsets in the blood, as well as effector cytokine expression comparable with wild-type T cells on polyclonal stimulation. Susceptibility of coronin 1-deficient mice to EAE induction was restored by transfer of wild-type CD4(+) T cells, suggesting that the observed resistance of coronin 1-deficient mice to EAE development is T cell intrinsic. Importantly, although coronin 1-deficient regulatory T cells (Tregs) showed a suppressor activity comparable with wild-type Tregs, Treg depletion failed to restore EAE development in coronin 1-deficient animals. These results suggest a hitherto unrecognized role of naive T cells in the development of autoimmune encephalomyelitis and reveal coronin 1 as a crucial modulator of EAE induction.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Microfilament Proteins/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Cell Survival/genetics , Cell Survival/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Myelin Basic Protein/administration & dosage , Myelin Basic Protein/immunology , Resting Phase, Cell Cycle/genetics , Resting Phase, Cell Cycle/immunology , T-Lymphocyte Subsets/cytology
4.
Nat Commun ; 11(1): 3420, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32647127

ABSTRACT

Remyelination of the peripheral and central nervous systems (PNS and CNS, respectively) is a prerequisite for functional recovery after lesion. However, this process is not always optimal and becomes inefficient in the course of multiple sclerosis. Here we show that, when acetylated, eukaryotic elongation factor 1A1 (eEF1A1) negatively regulates PNS and CNS remyelination. Acetylated eEF1A1 (Ac-eEF1A1) translocates into the nucleus of myelinating cells where it binds to Sox10, a key transcription factor for PNS and CNS myelination and remyelination, to drag Sox10 out of the nucleus. We show that the lysine acetyltransferase Tip60 acetylates eEF1A1, whereas the histone deacetylase HDAC2 deacetylates eEF1A1. Promoting eEF1A1 deacetylation maintains the activation of Sox10 target genes and increases PNS and CNS remyelination efficiency. Taken together, these data identify a major mechanism of Sox10 regulation, which appears promising for future translational studies on PNS and CNS remyelination.


Subject(s)
Peptide Elongation Factor 1/metabolism , Remyelination/genetics , Transcriptional Activation/genetics , Acetylation , Aging/metabolism , Animals , Cell Dedifferentiation/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Lysine Acetyltransferase 5/metabolism , Mice , Models, Biological , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Peripheral Nervous System/drug effects , Peripheral Nervous System/physiology , Recovery of Function/drug effects , Remyelination/drug effects , SOXE Transcription Factors/metabolism , STAT3 Transcription Factor/metabolism , Schwann Cells/drug effects , Schwann Cells/metabolism , Theophylline/pharmacology , Trans-Activators/metabolism , Transcriptional Activation/drug effects
5.
Article in English | MEDLINE | ID: mdl-31882398

ABSTRACT

OBJECTIVE: To investigate molecular changes in multiple sclerosis (MS) normal-appearing cortical gray matter (NAGM). METHODS: We performed a whole-genome gene expression microarray analysis of human brain autopsy tissues from 64 MS NAGM samples and 42 control gray matter samples. We further examined our cases by HLA genotyping and performed immunohistochemical and immunofluorescent analysis of all human brain tissues. RESULTS: HLA-DRB1 is the transcript with highest expression in MS NAGM with a bimodal distribution among the examined cases. Genotyping revealed that every case with the MS-associated HLA-DR15 haplotype also shows high HLA-DRB1 expression and also of the tightly linked HLA-DRB5 allele. Quantitative immunohistochemical analysis confirmed the higher expression of HLA-DRB1 in HLA-DRB1*15:01 cases at the protein level. Analysis of gray matter lesion size revealed a significant increase of cortical lesion size in cases with high HLA-DRB1 expression. CONCLUSIONS: Our data indicate that increased HLA-DRB1 and -DRB5 expression in the brain of patients with MS may be an important factor in how the HLA-DR15 haplotype contributes to MS pathomechanisms in the target organ.


Subject(s)
Gray Matter/metabolism , Gray Matter/pathology , HLA-DR Serological Subtypes/genetics , HLA-DRB1 Chains/metabolism , HLA-DRB5 Chains/metabolism , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Aged , Aged, 80 and over , Autopsy , Female , Gene Expression Profiling , HLA-DRB1 Chains/genetics , Haplotypes , Humans , Immunohistochemistry , Male , Middle Aged , Protein Array Analysis
6.
Brain ; 131(Pt 1): 288-303, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18056737

ABSTRACT

Multiple sclerosis is a chronic inflammatory disease of the CNS. Although progressive axonal injury and diffuse inflammatory damage has been shown in the chronic phase of the disease, little is known about the molecular mechanisms underlying these pathological processes. In order to identify these mechanisms, we have studied the gene expression profile in non-lesion containing tissue, the so-called normal-appearing white matter (NAWM). We performed differential gene expression analysis and quantitative RT-PCR on subcortical white matter from 11 multiple sclerosis and 8 control cases. Differentially expressed genes were further analysed in detail by in situ hybridization and immunofluorescence studies. We show that genes known to be involved in anti-inflammatory and protective mechanisms such as STAT6, JAK1, IL-4R, IL-10, Chromogranin C and Hif-1alpha are consistently upregulated in the multiple sclerosis NAWM. On the other hand, genes involved in pro-inflammatory mechanisms, such as STAT4, IL-1beta and MCSF, were also upregulated but less regularly. Immunofluorescence colocalization analysis revealed expression of STAT6, JAK1, IL-4R and IL-13R mainly in oligodendrocytes, whereas STAT4 expression was detected predominantly in microglia. In line with these data, in situ hybridization analysis showed an increased expression in multiple sclerosis NAWM of HIF-1alpha in oligodendrocytes and HLA-DRalpha in microglia cells. The consistency of the expression levels of STAT6, JAK1, JAK3 and IL-4R between the multiple sclerosis cases suggests an overall activation of the STAT6-signalling pathway in oligodendrocytes, whereas the expression of STAT4 and HLA-DRalpha indicates the activation of pro-inflammatory pathways in microglia. The upregulation of genes involved in anti-inflammatory mechanisms driven by oligodendrocytes may protect the CNS environment and thus limit lesion formation, whereas the activation of pro-inflammatory mechanisms in microglia may favour disease progression. Altogether, our data suggests an endogenous inflammatory reaction throughout the whole white matter of multiple sclerosis brain, in which oligodendrocytes actively participate. This reaction might further influence and to some extent facilitate lesion formation.


Subject(s)
Brain/pathology , Multiple Sclerosis/pathology , Aged , Brain/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Chemokines/biosynthesis , Chemokines/genetics , Cytokines/biosynthesis , Cytokines/genetics , Female , Gene Expression Profiling , HLA-DR Antigens/biosynthesis , HLA-DR Antigens/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Inflammation Mediators/metabolism , Male , Microglia/physiology , Middle Aged , Multiple Sclerosis/metabolism , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/genetics , Oligodendroglia/physiology , Oligonucleotide Array Sequence Analysis/methods , Receptors, Chemokine/biosynthesis , Receptors, Chemokine/genetics , Receptors, Cytokine/biosynthesis , Receptors, Cytokine/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , STAT6 Transcription Factor/metabolism , Signal Transduction , Up-Regulation
7.
Cells ; 8(11)2019 11 12.
Article in English | MEDLINE | ID: mdl-31726669

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease of the central nervous system. Neurological deficits are attributed to inflammatory demyelination, which compromises axonal function and survival. These are mitigated in experimental models by rapid and often complete remyelination of affected axons, but in MS this endogenous repair mechanism frequently fails, leaving axons increasingly vulnerable to the detrimental effects of inflammatory and metabolic stress. Understanding the molecular basis of remyelination and remyelination failure is essential to develop improved therapies for this devastating disease. However, recent studies suggest that this is not due to a single dominant mechanism, but rather represents the biological outcome of multiple changes in the lesion microenvironment that combine to disrupt oligodendrocyte differentiation. This identifies a pressing need to develop technical platforms to investigate combinatory and/or synergistic effects of factors differentially expressed in MS lesions on oligodendrocyte proliferation and differentiation. Here we describe protocols using primary oligodendrocyte cultures from Bl6 mice on 384-well nanofiber plates to model changes affecting oligodendrogenesis and differentiation in the complex signaling environment associated with multiple sclerosis lesions. Using platelet-derived growth factor (PDGF-AA), fibroblast growth factor 2 (FGF2), bone morphogenetic protein 2 (BMP2) and bone morphogenetic protein 4 (BMP4) as representative targets, we demonstrate that we can assess their combinatory effects across a wide range of concentrations in a single experiment. This in vitro model is ideal for assessing the combinatory effects of changes in availability of multiple factors, thus more closely modelling the situation in vivo and furthering high-throughput screening possibilities.


Subject(s)
Multiple Sclerosis/metabolism , Myelin Sheath/metabolism , Oligodendroglia/cytology , Primary Cell Culture/instrumentation , Animals , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Fibroblast Growth Factor 2/pharmacology , Humans , Mice , Models, Theoretical , Multiple Sclerosis/therapy , Nanofibers , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Platelet-Derived Growth Factor/pharmacology , Primary Cell Culture/methods
8.
Acta Neuropathol Commun ; 7(1): 212, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31856924

ABSTRACT

Fibroblast growth factor (FGF) signaling contributes to failure of remyelination in multiple sclerosis, but targeting this therapeutically is complicated by its functional pleiotropy. We now identify FGF2 as a factor up-regulated by astrocytes in active inflammatory lesions that disrupts myelination via FGF receptor 2 (FGFR2) mediated activation of Wingless (Wnt) signaling; pharmacological inhibition of Wnt being sufficient to abrogate inhibition of myelination by FGF2 in tissue culture. Using a novel FGFR1-selective agonist (F2 V2) generated by deleting the N-terminal 26 amino acids of FGF2 we demonstrate polarizing signal transduction to favor FGFR1 abrogates FGF mediated inhibition of myelination but retains its ability to induce expression of pro-myelinating and immunomodulatory factors that include Cd93, Lif, Il11, Hbegf, Cxcl1 and Timp1. Our data provide new insights into the mechanistic basis of remyelination failure in MS and identify selective activation of FGFR1 as a novel strategy to induce a neuroprotective signaling environment in multiple sclerosis and other neurological diseases.


Subject(s)
Astrocytes/metabolism , Fibroblast Growth Factor 2/biosynthesis , Multiple Sclerosis/metabolism , Nerve Fibers, Myelinated/metabolism , Neuroprotection/physiology , Receptor, Fibroblast Growth Factor, Type 1/biosynthesis , Animals , Astrocytes/chemistry , Astrocytes/pathology , Fibroblast Growth Factor 2/analysis , Fibroblast Growth Factor 2/genetics , Humans , Mice , Mice, Inbred C57BL , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Nerve Fibers, Myelinated/pathology , Rats , Rats, Sprague-Dawley
9.
Front Immunol ; 10: 540, 2019.
Article in English | MEDLINE | ID: mdl-31024521

ABSTRACT

Immune responses to citrullinated peptides have been described in autoimmune diseases like rheumatoid arthritis (RA) and multiple sclerosis (MS). We investigated the post-translational modification (PTM), arginine to citrulline, in brain tissue of MS patients and controls (C) by proteomics and subsequently the cellular immune response of cerebrospinal fluid (CSF)-infiltrating T cells to citrullinated and unmodified peptides of myelin basic protein (MBP). Using specifically adapted tissue extraction- and combined data interpretation protocols we could establish a map of citrullinated proteins by identifying more than 80 proteins with two or more citrullinated peptides in human brain tissue. We report many of them for the first time. For the already described citrullinated proteins MBP, GFAP, and vimentin, we could identify additional citrullinated sites. The number of modified proteins in MS white matter was higher than control tissue. Citrullinated peptides are considered neoepitopes that may trigger autoreactivity. We used newly identified epitopes and previously reported immunodominant myelin peptides in their citrullinated and non-citrullinated form to address the recognition of CSF-infiltrating CD4+ T cells from 22 MS patients by measuring proliferation and IFN-γ secretion. We did not detect marked responses to citrullinated peptides, but slightly more strongly to the non-modified version. Based on these data, we conclude that citrullination does not appear to be an important activating factor of a T cell response, but could be the consequence of an immune- or inflammatory response. Our approach allowed us to perform a deep proteome analysis and opens new technical possibilities to analyze complex PTM patterns on minute quantities of rare tissue samples.


Subject(s)
Brain/immunology , Multiple Sclerosis/immunology , Myelin Basic Protein/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Cerebrospinal Fluid/immunology , Citrullination , Female , Humans , Male , Middle Aged , Peptides/immunology , Young Adult
10.
J Cereb Blood Flow Metab ; 37(8): 2894-2909, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28273717

ABSTRACT

Activated leukocyte cell adhesion molecule (ALCAM) has been proposed to mediate leukocyte migration across the blood-brain barrier (BBB) in multiple sclerosis or experimental autoimmune encephalomyelitis (EAE). Here, we confirmed vascular ALCAM expression in human brain tissue samples in situ and on two different human in vitro BBB models. Antibody-mediated inhibition of ALCAM reduced diapedesis of human CD4+ Th1 but not of Th17 cells across the human BBB in vitro. In accordance to human Th1 cells, mouse Th1 cells showed reduced diapedesis across an ALCAM-/- in vitro BBB model under static but no longer under flow conditions. In contrast to the limited role of ALCAM in T cell extravasation across the BBB, we found a contribution of ALCAM to rolling, adhesion, and diapedesis of human CD14+ monocytes across the human BBB under flow and static conditions. Taken together, our study highlights the potential differences in the CNS expression of ALCAM in mouse and human and supports a prominent role for ALCAM in the multi-step extravasation of monocytes across the BBB.


Subject(s)
Antigens, CD/metabolism , Blood-Brain Barrier/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Fetal Proteins/metabolism , Monocytes/immunology , T-Lymphocytes/immunology , Transendothelial and Transepithelial Migration/immunology , Animals , Antigens, CD/genetics , Blood-Brain Barrier/immunology , Cell Adhesion Molecules, Neuronal/genetics , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Fetal Proteins/genetics , Humans , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , T-Lymphocytes/metabolism , Transendothelial and Transepithelial Migration/physiology
11.
Brain Res ; 1641(Pt A): 139-148, 2016 06 15.
Article in English | MEDLINE | ID: mdl-26423932

ABSTRACT

Oligodendrocytes, the myelinating glial cells of the central nervous system (CNS), are due to their high specialization and metabolic needs highly vulnerable to various insults. This led to a general view that oligodendrocytes are defenseless victims during brain damage such as occurs in acute and chronic CNS inflammation. However, this view is challenged by increasing evidence that oligodendrocytes are capable of expressing a wide range of immunomodulatory molecules. They express various cytokines and chemokines (e.g. Il-1ß, Il17A, CCL2, CXCL10), antigen presenting molecules (MHC class I and II) and co-stimulatory molecules (e.g. CD9, CD81), complement and complement receptor molecules (e.g. C1s, C2 and C3, C1R), complement regulatory molecules (e.g. CD46, CD55, CD59), tetraspanins (e.g. TSPAN2), neuroimmune regulatory proteins (e.g. CD200, CD47) as well as extracellular matrix proteins (e.g. VCAN) and many others. Their potential immunomodulatory properties can, at specific times and locations, influence ongoing immune processes as shown by numerous publications. Therefore, oligodendrocytes are well capable of immunomodulation, especially during the initiation or resolution of immune processes in which subtle signaling might tip the scale. A better understanding of the immunomodulatory oligodendrocyte can help to invent new, innovative therapeutic interventions in various diseases such as Multiple Sclerosis. This article is part of a Special Issue entitled SI: Myelin Evolution.


Subject(s)
Neuroimmunomodulation/physiology , Oligodendroglia/metabolism , Animals , Brain/immunology , Humans
12.
Curr Neurovasc Res ; 13(1): 58-67, 2016.
Article in English | MEDLINE | ID: mdl-26503025

ABSTRACT

Cerebral small-vessel disease (SVD) is characterized by periventricular white matter (WM) changes and general brain atrophy. SVD is prevalent in elderly individuals and is frequently associated with the development of vascular dementia (VaD). Studies of the molecular basis of SVD are sparse. We have to gain further insight into the pathogenic mechanisms of SVD. Therefore, we compared gene expression patterns in the brains of SVD and control patients, in order to identify cellular pathways changed in diseased brains. We compared the expression of mRNA transcripts in postmortem, macroscopically normal-appearing human brain tissues isolated from frontal, temporal and occipital cortical and subcortical regions in 5 SVD and 5 non-SVD control patients. Significant expression changes were determined by fold change F>1.2 in either direction, and p<0.05. We identified 228 genes differentially expressed in cortex (89 up-, 139 down-regulated) and 555 genes in WM (223 up-, 332 down-regulated) in SVD patients. Pathway analyses revealed that upregulated genes were associated with inflammation and apoptosis in WM, suggesting active cell death. Downregulated genes were associated with coagulation and fatty and amino acids metabolisms. In the cortex, down-regulated genes were principally associated with neuronal functions. Our data revealed widespread changes in the transcriptome profiles in the cortex and WM of human SVD brains, with a predominance of changes in WM. We provide for the first time a comprehensive view of the molecular alterations in human SVD brains that seem to contribute to the neuropathogenesis of SVD.


Subject(s)
Brain/metabolism , Cerebral Small Vessel Diseases/pathology , Gene Expression Regulation/physiology , Inflammation/diagnosis , Metabolic Diseases/diagnosis , Signal Transduction/physiology , Aged , Aged, 80 and over , Cell Survival/physiology , Cerebral Small Vessel Diseases/complications , Chromosome Mapping , Computational Biology , Cytokines/genetics , Cytokines/metabolism , Diagnosis , Female , Humans , Inflammation/etiology , Male , Metabolic Diseases/etiology , Middle Aged , Oligonucleotide Array Sequence Analysis , Principal Component Analysis , RNA, Messenger/metabolism , Signal Transduction/genetics , Transcriptome
13.
Exp Neurol ; 265: 142-51, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25625851

ABSTRACT

Cerebral inflammation plays a crucial role in the pathophysiology of ischemic stroke and is involved in all stages of the ischemic cascade. Fullerene derivatives, such as fullerenol (OH-F) are radical scavengers acting as neuroprotective agents while glucosamine (GlcN) attenuates cerebral inflammation after stroke. We created novel glucosamine-fullerene conjugates (GlcN-F) to combine their protective effects and compared them to OH-F regarding stroke-induced cerebral inflammation and cellular damage. Fullerene derivatives or vehicle was administered intravenously in normotensive Wistar-Kyoto (WKY) rats and spontaneously hypertensive rats (SHR) immediately after transient middle cerebral artery occlusion (tMCAO). Infarct size was determined at day 5 and neurological outcome at days 1 and 5 after tMCAO. CD68- and NeuN-staining were performed to determine immunoreactivity and neuronal survival respectively. Cytokine and toll like receptor 4 (TLR-4) expression was assessed using quantitative real-time PCR. Magnetic resonance imaging revealed a significant reduction of infarct volume in both, WKY and SHR that were treated with fullerene derivatives. Treated rats showed an amelioration of neurological symptoms as both OH-F and GlcN-F prevented neuronal loss in the perilesional area. Cerebral immunoreactivity was reduced in treated WKY and SHR. Expression of IL-1ß and TLR-4 was attenuated in OH-F-treated WKY rats. In conclusion, OH-F and GlcN-F lead to a reduction of cellular damage and inflammation after stroke, rendering these compounds attractive therapeutics for stroke.


Subject(s)
Cerebral Infarction/drug therapy , Fullerenes/administration & dosage , Glucosamine/administration & dosage , Hypertension/drug therapy , Stroke/drug therapy , Animals , Brain Ischemia/drug therapy , Brain Ischemia/etiology , Brain Ischemia/pathology , Cerebral Infarction/etiology , Cerebral Infarction/pathology , Hypertension/pathology , Injections, Intravenous , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Stroke/complications , Stroke/pathology
14.
ASN Neuro ; 6(3): 137-57, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24641305

ABSTRACT

In peripheral nerves, Schwann cell development is regulated by a variety of signals. Some of the aspects of Schwann cell differentiation can be reproduced in vitro in response to forskolin, an adenylyl cyclase activator elevating intracellular cAMP levels. Herein, the effect of forskolin treatment was investigated by a comprehensive genome-wide expression study on primary mouse Schwann cell cultures. Additional to myelin-related genes, many so far unconsidered genes were ascertained to be modulated by forskolin. One of the strongest differentially regulated gene transcripts was the transcription factor Olig1 (oligodendrocyte transcription factor 1), whose mRNA expression levels were reduced in treated Schwann cells. Olig1 protein was localized in myelinating and nonmyelinating Schwann cells within the sciatic nerve as well as in primary Schwann cells, proposing it as a novel transcription factor of the Schwann cell lineage. Data analysis further revealed that a number of differentially expressed genes in forskolin-treated Schwann cells were associated with the ECM (extracellular matrix), underlining its importance during Schwann cell differentiation in vitro. Comparison of samples derived from postnatal sciatic nerves and from both treated and untreated Schwann cell cultures showed considerable differences in gene expression between in vivo and in vitro, allowing us to separate Schwann cell autonomous from tissue-related changes. The whole data set of the cell culture microarray study is provided to offer an interactive search tool for genes of interest.


Subject(s)
Cyclic AMP/pharmacology , Gene Expression Regulation/drug effects , Schwann Cells/drug effects , Transcription Factors/metabolism , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/metabolism , Genome/drug effects , Intermediate Filaments/metabolism , Mice , Mice, Inbred C57BL , Myelin Basic Protein/metabolism , Receptors, Nerve Growth Factor/metabolism , S100 Calcium Binding Protein beta Subunit/metabolism , Sciatic Nerve/cytology , Signal Transduction/drug effects , Transcription Factors/genetics , Vasodilator Agents/pharmacology
15.
ASN Neuro ; 6(5)2014.
Article in English | MEDLINE | ID: mdl-25290060

ABSTRACT

In the developing peripheral nervous system, a coordinated reciprocal signaling between Schwann cells and axons is crucial for accurate myelination. The myelin and lymphocyte protein MAL is a component of lipid rafts that is important for targeting proteins and lipids to distinct domains. MAL overexpression impedes peripheral myelinogenesis, which is evident by a delayed onset of myelination and reduced expression of the myelin protein zero (Mpz/P0) and the low-affinity neurotrophin receptor p75(NTR). This study shows that MAL overexpression leads to a significant reduction of Mpz and p75(NTR) expression in primary mouse Schwann cell cultures, which was already evident before differentiation, implicating an effect of MAL in early Schwann cell development. Their transcription was robustly reduced, despite normal expression of essential transcription factors and receptors. Further, the cAMP response element-binding protein (CREB) and phosphoinositide 3-kinase signaling pathways important for Schwann cell differentiation were correctly induced, highlighting that other so far unknown rate limiting factors do exist. We identified novel genes expressed by Schwann cells in a MAL-dependent manner in vivo and in vitro. A number of those, including S100a4, RhoU and Krt23, are implicated in cytoskeletal organization and plasma membrane dynamics. We showed that S100a4 is predominantly expressed by nonmyelinating Schwann cells, whereas RhoU was localized within myelin membranes, and Krt23 was detected in nonmyelinating as well as in myelinating Schwann cells. Their differential expression during early peripheral nerve development further underlines their possible role in influencing Schwann cell differentiation and myelination.


Subject(s)
Cell Differentiation/genetics , Cytoskeleton/metabolism , Myelin and Lymphocyte-Associated Proteolipid Proteins/metabolism , Schwann Cells/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cells, Cultured , Colforsin/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin P0 Protein/genetics , Myelin P0 Protein/metabolism , Myelin and Lymphocyte-Associated Proteolipid Proteins/genetics , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , S100 Calcium-Binding Protein A4 , S100 Proteins/genetics , S100 Proteins/metabolism , Schwann Cells/drug effects , Sciatic Nerve/cytology , Signal Transduction/drug effects , Signal Transduction/genetics
16.
Brain Pathol ; 19(3): 459-66, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19016740

ABSTRACT

A stereotactic biopsy of a 17-year-old woman revealed an active inflammatory demyelinating lesion compatible with pattern III multiple sclerosis (MS) according to Lucchinetti et al. The biopsy included a white matter region distant from the active inflammatory demyelinating lesion with abnormal MRI signal, lacking histopathological signs of demyelination and/or oligodendrocyte apoptosis. Expression analysis of this area revealed a strong up-regulation of neuronal nitric oxide synthase (nNOS). Furthermore, detection of nitrotyrosine provided evidence for reactive nitrogen species (RNS)-mediated damage to oligodendrocytes. Concomitantly, genes involved in neuroprotection against oxidative stress such as heme oxygenase 1 were up-regulated. Even though a single case report, this study shows earliest molecular changes in white matter surrounding an actively demyelinating lesion during the first manifestation of MS, pointing toward a more widespread pathological process. Therapeutic targeting of the identified mechanisms of tissue injury might be crucial to prevent further lesion formation or secondary tissue damage.


Subject(s)
Demyelinating Diseases/metabolism , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Nitric Oxide Synthase Type I/biosynthesis , Adolescent , Adult , Aged , Brain , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Female , Gene Expression , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Multiple Sclerosis/genetics , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL