Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Exp Neurol ; 305: 76-88, 2018 07.
Article in English | MEDLINE | ID: mdl-29596844

ABSTRACT

Little is known about the decisive molecular factors that regulate lesion remyelination in Multiple Sclerosis. To identify such factors, we performed a differential gene expression analysis of normal appearing white matter (NAWM), active, remyelinating, and inactive demyelinated lesions. As expected, many genes involved in inflammatory processes were detected to be differentially regulated between these tissue types. Among them, we found an increased expression of members of the STAT6 pathway such as STAT6, IL4 and IL4R in active, remyelinated and inactive demyelinated lesions. This suggests that a protective, anti-inflammatory reaction, as already reported to be present in MS NAWM, is further enhanced in lesion tissues. Focusing on genes influencing oligodendrogliogenesis, we found a decreased expression of NKX2-2 in active, remyelinated and inactive demyelinated lesions, whereas SOX10 was downregulated in inactive demyelinated lesions, when compared to NAWM. Simultaneously, CXCL12 (SDF1) expression was strongly increased in active, remyelinated and inactive demyelinated lesions, but increased expression of the IGF1 and IGF2 genes was found in inactive demyelinated lesions. This demonstrates that, in principle, expression of genes promoting oligodendrogliogenesis occurs in MS lesion tissue - even in inactive demyelinated lesions. In contrast, oligodendrogenesis inhibiting genes such as JAG1 were also expressed at higher levels in inactive demyelinated lesions. Both, oligodendrogliogenesis promoting as well as inhibiting genes are expressed in all lesion tissues. However, no clear promoting or inhibiting expression pattern could be detected in any of the different types of lesioned tissues. This might reflect the heterogeneity of lesion development in MS patients, both in terms of mechanisms and temporal differences.


Subject(s)
Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Oligodendroglia/pathology , Oligodendroglia/physiology , Adult , Female , Gene Expression , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , Male , Middle Aged , Multiple Sclerosis/epidemiology , Nuclear Proteins , Tissue Banks/trends , Transcription Factors , United Kingdom/epidemiology
2.
Brain Behav Immun ; 48: 313-25, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25937052

ABSTRACT

Emerging as an important correlate of neurological dysfunction in Multiple Sclerosis (MS), extended focal and diffuse gray matter abnormalities have been found and linked to clinical manifestations such as seizures, fatigue and cognitive dysfunction. To investigate possible underlying mechanisms we analyzed the molecular alterations in histopathological normal appearing cortical gray matter (NAGM) in MS. By performing a differential gene expression analysis of NAGM of control and MS cases we identified reduced transcription of astrocyte specific genes involved in the astrocyte-neuron lactate shuttle (ANLS) and the glutamate-glutamine cycle (GGC). Additional quantitative immunohistochemical analysis demonstrating a CX43 loss in MS NAGM confirmed a crucial involvement of astrocytes and emphasizes their importance in MS pathogenesis. Concurrently, a Toll-like/IL-1ß signaling expression signature was detected in MS NAGM, indicating that immune-related signaling might be responsible for the downregulation of ANLS and GGC gene expression in MS NAGM. Indeed, challenging astrocytes with immune stimuli such as IL-1ß and LPS reduced their ANLS and GGC gene expression in vitro. The detected upregulation of IL1B in MS NAGM suggests inflammasome priming. For this reason, astrocyte cultures were treated with ATP and ATP/LPS as for inflammasome activation. This treatment led to a reduction of ANLS and GGC gene expression in a comparable manner. To investigate potential sources for ANLS and GGC downregulation in MS NAGM, we first performed an adjuvant-driven stimulation of the peripheral immune system in C57Bl/6 mice in vivo. This led to similar gene expression changes in spinal cord demonstrating that peripheral immune signals might be one source for astrocytic gene expression changes in the brain. IL1B upregulation in MS NAGM itself points to a possible endogenous signaling process leading to ANLS and GGC downregulation. This is supported by our findings that, among others, MS NAGM astrocytes express inflammasome components and that astrocytes are capable to release Il-1ß in-vitro. Altogether, our data suggests that immune signaling of immune- and/or central nervous system origin drives alterations in astrocytic ANLS and GGC gene regulation in the MS NAGM. Such a mechanism might underlie cortical brain dysfunctions frequently encountered in MS patients.


Subject(s)
Astrocytes/immunology , Cerebral Cortex/immunology , Gene Expression , Multiple Sclerosis/genetics , Signal Transduction/genetics , Adult , Aged , Aged, 80 and over , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Female , Humans , Male , Mice , Middle Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Signal Transduction/immunology , Young Adult
3.
Cell Death Dis ; 3: e325, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22717580

ABSTRACT

Recent studies identified a highly tumorigenic subpopulation of glioma stem cells (GSCs) within malignant gliomas. GSCs are proposed to originate from transformed neural stem cells (NSCs). Several pathways active in NSCs, including the Notch pathway, were shown to promote proliferation and tumorigenesis in GSCs. Notch2 is highly expressed in glioblastoma multiforme (GBM), a highly malignant astrocytoma. It is therefore conceivable that increased Notch2 signaling in NSCs contributes to the formation of GBM. Here, we demonstrate that mice constitutively expressing the activated intracellular domain of Notch2 in NSCs display a hyperplasia of the neurogenic niche and reduced neuronal lineage entry. Neurospheres derived from these mice show increased proliferation, survival and resistance to apoptosis. Moreover, they preferentially differentiate into astrocytes, which are the characteristic cellular population of astrocytoma. Likewise, we show that Notch2 signaling increases proliferation and resistance to apoptosis in human GBM cell lines. Gene expression profiling of GBM patient tumor samples reveals a positive correlation of Notch2 transcripts with gene transcripts controlling anti-apoptotic processes, stemness and astrocyte fate, and a negative correlation with gene transcripts controlling proapoptotic processes and oligodendrocyte fate. Our data show that Notch2 signaling in NSCs produces features of GSCs and induces astrocytic lineage entry, consistent with a possible role in astrocytoma formation.


Subject(s)
Astrocytes/metabolism , Cell Transformation, Neoplastic/pathology , Neural Stem Cells/metabolism , Receptor, Notch2/metabolism , Signal Transduction , Animals , Astrocytes/pathology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Lineage , Cell Transformation, Neoplastic/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Neural Stem Cells/pathology , Receptor, Notch2/genetics
4.
Eur J Neurosci ; 29(12): 2276-90, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19508690

ABSTRACT

The myelin and lymphocyte protein (MAL) is a raft-associated membrane protein predominantly expressed by oligodendrocytes and Schwann cells. Here we show that MAL regulates myelination in the peripheral nervous system. In mice overexpressing MAL, myelination was retarded and fibers were hypomyelinated, whereas myelination in MAL knockout mice was accelerated. This was not due to impaired Schwann cell proliferation, differentiation or axonal sorting. We found that the expression level of p75 neurotrophin receptor mRNA and protein was strongly reduced in developing sciatic nerves in MAL-overexpressing mice. This reduction is well correlated with the observed alterations in myelination initiation, speed of myelination and alterations in Remak bundle development. Our results suggest a functional role for MAL in peripheral myelination by influencing the expression of membrane components that mediate axon-glia interaction during ensheathment and myelin wrapping.


Subject(s)
Membrane Transport Proteins/metabolism , Myelin Proteins/metabolism , Myelin Sheath/metabolism , Nerve Fibers, Myelinated/metabolism , Peripheral Nerves/metabolism , Proteolipids/metabolism , Receptor, Nerve Growth Factor/metabolism , Animals , Cell Communication/physiology , Cell Differentiation/physiology , Membrane Microdomains/metabolism , Membrane Transport Proteins/genetics , Mice , Mice, Transgenic , Myelin Proteins/genetics , Myelin Sheath/ultrastructure , Myelin and Lymphocyte-Associated Proteolipid Proteins , Nerve Fibers, Myelinated/ultrastructure , Peripheral Nerves/growth & development , Peripheral Nerves/ultrastructure , Proteolipids/genetics , RNA, Messenger/metabolism , Receptor, Nerve Growth Factor/genetics , Schwann Cells/metabolism
5.
Mol Cell Neurosci ; 40(2): 156-66, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19026747

ABSTRACT

Myelin is organized in subdomains with distinct protein and lipid composition. How these domains are established and maintained is currently unknown. Cytoskeletal elements interacting with membrane components could generate and sustain such structural domains. Here, we demonstrate that the transmembrane myelin protein MAL interacts with the cytoskeleton protein septin 6. Septins represent a fourth filamentous system involved in membrane compartmentalization, vesicle transport and scaffold formation. We report that multiple septin complexes are associated with myelin, and that they display an overlapping but non-identical composition in the central and peripheral nervous system. The expression of distinct subsets of septins was upregulated during myelin formation in peripheral nerves and oligodendrocytes. In the PNS, septins were highly enriched in non-compact myelin compartments, particularly in the paranodal loops and the microvilli at the node of Ranvier. Importantly in myelin lacking Septin 6, the abundance of its closest homolog Sept11 was increased, suggesting a functional compensatory role. Our data demonstrate that the septin cytoskeleton is an integral component of the myelin sheath and interacts with distinct myelin constituents such as MAL. We suggest that septins are intriguing candidates for membrane compartmentalization in myelin internodes.


Subject(s)
Cytoskeleton/metabolism , GTP-Binding Proteins/metabolism , Myelin Sheath/metabolism , Neuroglia/physiology , Animals , Biomarkers/metabolism , Cell Differentiation/physiology , Cell Line , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/genetics , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Myelin Proteins/genetics , Myelin Proteins/metabolism , Myelin and Lymphocyte-Associated Proteolipid Proteins , Neuroglia/cytology , Peripheral Nervous System/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proteolipids/genetics , Proteolipids/metabolism , Rats , Septins , Two-Hybrid System Techniques
6.
J Neuroimmunol ; 205(1-2): 10-9, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18950873

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system (CNS). Recent studies suggest that, beside focal lesions, diffuse inflammatory and degenerative processes take place throughout the MS brain. Especially, molecular alterations in the so-called normal appearing white matter suggest the induction of neuroprotective mechanisms against oxidative stress preserving cellular homeostasis and function. In this study we investigated whether in an animal model for MS, namely in experimental autoimmune encephalomyelitis (EAE), similar changes occur. We isolated normal appearing white and grey matter from the corpus callosum and the above lying cerebral cortex from DA rats with rMOG-induced EAE and carried out a gene expression analysis. Examination of corpus callosum revealed only minor changes in EAE rats. In contrast, we identified a number of gene expression alterations in the cerebral cortex even though morphological and cellular alterations were not evident. One of the most striking observations was the downregulation of genes involved in mitochondrial function as well as a whole set of genes coding for different glutamate receptors. Our data imply that molecular alterations are present in neurons far distant to inflammatory demyelinating lesions. These alterations might reflect degenerative processes induced by lesion-mediated axonal injury in the spinal cord. Our results indicate that the MOG-induced EAE in DA rats is a valuable model to analyze neuronal alterations due to axonal impairment in an acute phase of a MS-like disease, and could be used for development of neuroprotective strategies.


Subject(s)
Brain/pathology , Gene Expression/physiology , Multiple Sclerosis/pathology , Neuroglia/metabolism , Spinal Cord/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Brain/metabolism , Disease Models, Animal , Down-Regulation/physiology , Female , Gene Expression Profiling/methods , Glial Fibrillary Acidic Protein/metabolism , Multiple Sclerosis/chemically induced , Multiple Sclerosis/physiopathology , Myelin Proteins , Myelin Proteolipid Protein/metabolism , Myelin-Associated Glycoprotein , Myelin-Oligodendrocyte Glycoprotein , Neuroglia/pathology , Oligonucleotide Array Sequence Analysis/methods , Rats , Spinal Cord/pathology , Statistics, Nonparametric
7.
Int MS J ; 15(2): 51-8, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18782500

ABSTRACT

Recently, the introduction of RNA profiling using microarray technology has helped to elucidate gene expression changes in diseased tissue samples from postmortem human brains. Especially, in the field of multiple sclerosis (MS) research, microarray-based RNA profiling has been applied in the hope to identify disease specific alterations. The lack of good biomarkers for diagnostic as well as for prognostic purposes, but also the need for new drug targets and for a better understanding of the pathophysiology, makes this technique a valuable tool. Different RNA profiling approaches have been used, addressing distinct scientific questions. MS brain tissue samples have been proven to be an appropriate source for RNA profiling to investigate molecular pathomechanisms. This work discusses the critical parameters for RNA profiling of MS brain tissues, and reviews the results obtained by microarray studies analyzing differential gene expression in MS brain tissues.


Subject(s)
Brain/metabolism , Gene Expression Profiling , Multiple Sclerosis/metabolism , Oligonucleotide Array Sequence Analysis , Proteins/metabolism , Humans , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Proteins/genetics , RNA, Messenger/metabolism
8.
J Mol Neurosci ; 35(1): 91-100, 2008 May.
Article in English | MEDLINE | ID: mdl-18278568

ABSTRACT

In the pathogenesis of multiple sclerosis (MS), oligodendrocytes and its myelin sheaths are thought to be the primary target of destruction. The mechanism leading to oligodendrocyte injury and demyelination is still elusive. Oligodendrocytes are maintaining up to 50 internodes of myelin, which is an extraordinary metabolic demand. This makes them one of the most vulnerable cell types in the central nervous system (CNS), and even small insults can lead to oligodendrocyte impairment, demyelination, and axonal dysfunction. For this reason, oligodendrocytes are viewed as more or less the "lame ducks" of the CNS who can easily become victims. However, recent data demonstrate that this perception possibly needs to be revised. The latest data suggest that oligodendrocytes may also act as "fierce creatures," influencing the surrounding cells in many ways to preserve its own, as well as their function, allowing sustained functionality of the CNS upon an attack. In this review, the concept of "reactive or activated oligodendrocyte" is introduced, describing alterations in oligodendrocytes which are either protective mechanisms allowing survival in otherwise lethal environment or influence and possibly modulate the ongoing inflammation. Although "harnessed", oligodendrocytes might actively modulate and shape their environment and be part of the immune privilege of the brain.


Subject(s)
Multiple Sclerosis , Myelin Sheath/physiology , Oligodendroglia/physiology , Animals , Antibodies/immunology , Astrocytes/metabolism , B-Lymphocytes/immunology , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Macrophages/immunology , Microglia/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Oligodendroglia/cytology , Oligodendroglia/pathology , T-Lymphocytes/immunology
10.
Neurobiol Dis ; 16(2): 396-406, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15193296

ABSTRACT

Metachromatic leukodystrophy (MLD) is a lysosomal lipid storage disease caused by arylsulfatase A deficiency. In MLD patients the sphingolipid sulfatide increasingly accumulates leading to progressive demyelination. We have analysed arylsulfatase A-deficient mice, a MLD mouse model, and we show that accumulation of sulfatide is not restricted to the lysosomal compartment but also occurs in myelin itself. Although, this sulfatide storage did not affect the overall composition of most myelin proteins, it specifically caused a severe reduction of MAL. This demonstrates a regulatory link between sulfatide accumulation and MAL expression and indicates the existence of regulatory mechanisms between lipid and myelin protein synthesis in oligodendrocytes. In addition, in cultured renal epithelial cells, sulfatide accumulation diverts MAL to the late endosomal/lysosomal compartment and thus also affects the intracellular distribution of MAL. The specific reduction and mistargeting of MAL protein as a reaction to sulfatide overload may contribute to the pathogenic mechanisms in metachromatic leukodystrophy.


Subject(s)
Cerebroside-Sulfatase/genetics , Leukodystrophy, Metachromatic/metabolism , Leukodystrophy, Metachromatic/physiopathology , Membrane Microdomains/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Myelin Proteins/genetics , Myelin Proteins/metabolism , Proteolipids/genetics , Proteolipids/metabolism , Animals , Biological Transport/physiology , Detergents , Down-Regulation , Kidney/cytology , Kidney/metabolism , Lysosomes/metabolism , Membrane Proteins/metabolism , Mice , Mice, Mutant Strains , Myelin Sheath/metabolism , Myelin and Lymphocyte-Associated Proteolipid Proteins , Sulfoglycosphingolipids/metabolism
11.
J Neurosci Res ; 71(3): 326-37, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12526022

ABSTRACT

Myelin-associated glycoprotein (MAG), an immunoglobulin-like cell signaling protein involved in axon-glial interactions, displays two intracellular C-termini as a result of alternative mRNA splicing. During brain development, the two MAG mRNAs that encode L-MAG and S-MAG differ in their relative abundance. We have investigated the differential expression of L- and S-MAG upon cAMP treatment in the oligodendroglial cell line Oli-neu, a cell line able to differentiate in vitro. We have engineered GFP and VSVG fusions by small insertions into the alternatively spliced exons of the cloned MAG gene and reintroduced them into Oli-neu cells. The individually tagged MAG isoforms were expressed under the control of the MAG promoter and regulatory region. In this system, L-MAG was the predominant isoform before the stimulation of cells with cAMP, whereas upon cAMP treatment the S-MAG isoform was predominantly expressed in cells with a high degree of morphological differentiation. We suggest that the regulation of the MAG alternative splicing and the morphological differentiation in oligodendrocytes are controlled both by the same cAMP-responsive differentiation step.


Subject(s)
Cyclic AMP/pharmacology , Myelin-Associated Glycoprotein/biosynthesis , Oligodendroglia/cytology , Oligodendroglia/metabolism , Animals , COS Cells , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Chlorocebus aethiops , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Mice , Mice, Inbred BALB C , Myelin-Associated Glycoprotein/genetics , Oligodendroglia/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
12.
Acta Paediatr Suppl ; 92(443): 74-9; discussion 45, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14989469

ABSTRACT

UNLABELLED: Metachromatic leukodystrophy is a lysosomal lipid storage disorder. It is caused by mutations in the gene for arylsulphatase A, an enzyme involved in the degradation of the sphingolipid 3'-O-sulphogalactosylceramide (sulphatide). This membrane lipid can be found in various cell types, but in particularly high concentrations in the myelin of the nervous system. Patients suffer from progressive, finally lethal, demyelination due to accumulation of sulphatide. In the nervous system, lipid storage not only affects oligodendrocytes but also neurons and, in addition, leads to astrogliosis and activation of microglia. At the cellular level, lysosomal sulphatide storage also affects the lipid composition of myelin itself and has consequences for the amount and localization of particular myelin membrane-associated proteins. Here we review data, largely based on an arylsulphatase A knock-out mouse model of metachromatic leukodystrophy. CONCLUSION: The knock-out mouse model of metachromatic leukodystrophy has provided insights into the histopathological and cellular consequences of sulphatide storage.


Subject(s)
Leukodystrophy, Metachromatic/metabolism , Animals , Arylsulfatases/deficiency , Arylsulfatases/genetics , Cell Membrane/metabolism , Cerebroside-Sulfatase , Disease Models, Animal , Immunohistochemistry , Leukodystrophy, Metachromatic/enzymology , Membrane Lipids/metabolism , Mice , Mice, Knockout , Myelin-Associated Glycoprotein/metabolism
13.
J Neurochem ; 82(3): 550-62, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12153479

ABSTRACT

The myelin and lymphocyte protein (MAL) proteolipid is localized in central and peripheral compact myelin membranes, as well as in apical membranes of particular polarized cells. In this study, we addressed the question whether MAL and other peripheral myelin proteins are sorted and targeted to myelin membranes using mechanisms similar to those observed in polarized epithelial cells. To investigate the presence of raft-mediated sorting pathways in Schwann cells, we have isolated and analysed their composition in myelin membranes. Here, we show that rafts are present in adult human and rat peripheral compact myelin membranes and contain MAL, the GPI-anchored protein CD59, and substantial amounts of the PMP22 and P0. Colocalization studies show that CD59, and MAL have an almost identical expression pattern within compact myelin. Moreover, immuno-electron microscopy revealed that MAL, besides its localization in compact myelin, is also localized to Schmidt-Lanterman incisures. Taken together, our results demonstrate the presence of detergent-insoluble glycolipid-enriched complexes (DIGs) in different compartments of myelin membranes and indicate an important role for DIG-mediated transport mechanisms in the maintenance of the adult myelin sheath.


Subject(s)
CD59 Antigens/analysis , Membrane Microdomains/chemistry , Membrane Transport Proteins , Myelin Proteins/analysis , Myelin Sheath/chemistry , Peripheral Nerves/chemistry , Proteolipids/analysis , Aged , Aged, 80 and over , Animals , Biomarkers/analysis , CD59 Antigens/biosynthesis , Cell Compartmentation , Detergents/chemistry , Humans , Macromolecular Substances , Membrane Microdomains/metabolism , Microscopy, Immunoelectron , Middle Aged , Myelin P0 Protein/analysis , Myelin P0 Protein/biosynthesis , Myelin Proteins/biosynthesis , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Myelin and Lymphocyte-Associated Proteolipid Proteins , Peripheral Nerves/metabolism , Peripheral Nerves/ultrastructure , Proteolipids/biosynthesis , Proteolipids/ultrastructure , Rats , Solubility , Spinal Nerve Roots/metabolism , Spinal Nerve Roots/ultrastructure
14.
J Neurocytol ; 31(1): 5-14, 2002 Jan.
Article in English | MEDLINE | ID: mdl-12652083

ABSTRACT

The functional importance of the basal lamina in Schwann cell development and in adult peripheral nerve fibers is well known. We have demonstrated previously by confocal microscopy that IgM deposits are present on the basal lamina of myelinating Schwann cells of nerve biopsies from patients with an anti-MAG IgM neuropathy. Therefore, the basal lamina was postulated to represent an early target for the uptake of autoantibodies on the surface of myelinated nerve fibers. In this study, the preparation of cell- and myelin-free basal lamina from human peripheral nerves, using a detergent-dependent method is described and characterized by immunohistochemical and biochemical analysis. Using these methods we demonstrated that an enrichment of basal lamina components of Schwann cells with extraction of myelin could be achieved. Western blot analysis and immunohistochemical characterization showed that anti-MAG IgM antibodies did not recognize an epitope on the basal lamina of normal nerves. The established method will allow in situ investigations of basal lamina components from human peripheral nerves in health and in disease, e.g. peripheral neuropathies of infectious or inflammatory origin.


Subject(s)
Antibodies, Anti-Idiotypic/metabolism , Immunoglobulin M/metabolism , Myelin-Associated Glycoprotein/metabolism , Nerve Fibers/metabolism , Nerve Fibers/ultrastructure , Aged , Basement Membrane/metabolism , Basement Membrane/ultrastructure , Humans , Male , Middle Aged , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Protein Binding/physiology , Sural Nerve/metabolism , Sural Nerve/ultrastructure
15.
J Neurosci ; 21(22): 8830-41, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11698595

ABSTRACT

Perturbation of the homeostasis between proteases and their inhibitors has been associated with lesion-induced or degenerative neuronal changes. Protease nexin-1 (PN-1), a secreted serine protease inhibitor, is constitutively expressed in distinct neuronal cell populations of the adult CNS. In an earlier study we showed that transgenic mice with ectopic or increased expression of PN-1 in postnatal neurons have altered synaptic transmission. Here these mice are used to examine the impact of an extracellular proteolytic imbalance on long-term neuronal function. These mice develop disturbances in motor behavior from 12 weeks on, with some of the histopathological changes described in early stages of human motor neuron disease, and neurogenic muscle atrophy in old age. In addition, sensorimotor integration, measured by epicranial multichannel recording of sensory evoked potentials, is impaired. Our results suggest that axonal dysfunction rather than cell death underlies these phenotypes. In particular, long projecting neurons, namely cortical layer V pyramidal and spinal motor neurons, show an age-dependent vulnerability to PN-1 overexpression. These mice can serve to study early stages of in vivo neuronal dysfunction not yet associated with cell loss.


Subject(s)
Carrier Proteins/biosynthesis , Motor Neuron Disease/enzymology , Motor Neuron Disease/genetics , Motor Neurons/metabolism , Pyramidal Cells/metabolism , Amyloid beta-Protein Precursor , Animals , Axons/pathology , Behavior, Animal , Brain/metabolism , Brain/pathology , Carrier Proteins/genetics , Disease Progression , Electroencephalography , Evoked Potentials/genetics , Female , Gliosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Mice, Transgenic , Motor Activity/genetics , Motor Neuron Disease/diagnosis , Motor Neuron Disease/pathology , Motor Neurons/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Protease Nexins , Pyramidal Cells/pathology , Receptor, Nerve Growth Factor , Receptors, Cell Surface , Receptors, Nerve Growth Factor/metabolism , Serine Proteinase Inhibitors/biosynthesis , Serine Proteinase Inhibitors/genetics , Serpin E2 , Spinal Cord/metabolism , Spinal Cord/pathology , Survival Rate , Weight Loss/genetics
16.
Neurology ; 56(10): 1371-7, 2001 May 22.
Article in English | MEDLINE | ID: mdl-11376190

ABSTRACT

OBJECTIVE: To determine whether the cytokine tumor necrosis factor alpha (TNF-alpha) acts as a pain mediator in neuropathic pain in humans. BACKGROUND: In animal models, inflammatory cytokines such as TNF-alpha have been shown to facilitate neuropathic pain. METHODS: The expression of TNF-alpha was analyzed immunohistochemically in 20 human nerve biopsy specimens of patients with painful (n = 10) and nonpainful (n = 10) neuropathies. Additionally, serum soluble TNF-alpha receptor I (sTNF-RI) levels were determined in 24 patients with neuropathies, 16 of which were painful and 8 that were painless. RESULTS: Colocalization studies by confocal fluorescence microscopy for S-100 and TNF-alpha showed expression of TNF-alpha in human Schwann cells. Patients with painful neuropathies showed a stronger TNF-alpha immunoreactivity in myelinating Schwann cells relative to the epineurial background staining compared with patients with nonpainful neuropathy (0.949 +/- 0.047 vs 1.010 +/- 0.053, p < 0.05). Although there was no difference in sTNF-RI levels between painful (n = 16) and nonpainful (n = 8) neuropathies (sTNF-RI: 1412 +/- 545 pg/mL vs 1,318 +/- 175 pg/mL), patients with a mechanical allodynia (n = 9) had elevated serum sTNF-RI (1627 +/- 645 pg/mL vs 1233 +/- 192 pg/mL, p < 0.05) compared with patients without allodynia (n = 15). CONCLUSIONS: TNF-alpha expression of human Schwann cells may be up-regulated in painful neuropathies. The elevation of sTNF-RI in patients with centrally mediated mechanical allodynia suggests that systemic sTNF-RI levels may influence central pain processing mechanisms.


Subject(s)
Biomarkers/blood , Pain/blood , Pain/physiopathology , Peripheral Nervous System Diseases/blood , Peripheral Nervous System Diseases/physiopathology , Tumor Necrosis Factor-alpha/metabolism , Adult , Aged , Aged, 80 and over , Biopsy , Blood Proteins/metabolism , Female , Humans , Immunohistochemistry , Inflammation/blood , Inflammation/pathology , Inflammation/physiopathology , Lymphocytes/pathology , Macrophages/pathology , Male , Middle Aged , Pain/pathology , Peripheral Nervous System Diseases/pathology , Sural Nerve/metabolism , Sural Nerve/pathology , Sural Nerve/physiopathology
17.
Microsc Res Tech ; 52(6): 645-55, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11276117

ABSTRACT

During myelin formation, membrane-associated proteins have to be sorted and transported in specified membrane regions such as compact and non-compact myelin membranes. One protein that may be involved in such a process is the Myelin and Lymphocyte protein MAL (VIP17/ MVP17). MAL was identified as a novel myelin membrane component expressed by oligodendrocytes and Schwann cells. Since MAL has been shown to be important in the apical sorting machinery of polarized cells, we have started to investigate the possible functional role of MAL in sorting myelin membrane-associated molecules. In this study, we have generated cDNA constructs with green fluorescent protein (GFP) either at the N- or C-terminus of MAL. Transfection experiments showed that GFP-MAL expression resembles that of normal MAL, whereas the MAL-GFP fusion construct was not properly transported within the cell. Furthermore, we could demonstrate that GFP-MAL is enriched in detergent insoluble glycolipid-enriched microdomains as already seen for untagged MAL. As a prerequisite for the generation of transgenic mice expressing GFP-MAL under the control of its own regulatory elements, we have generated a cDNA construct with an 8-kb MAL promotor fragment fused to GFP-MAL. Transfection experiments of the Oli-neu oligodendrocyte cell line showed that GFP-MAL was expressed, but only in cells, which were stimulated for differentiation with cAMP. In summary, the results confirm that the fusion protein GFP-MAL is incorporated into detergent-insoluble complexes and the 8-kb MAL promotor fragment is sufficient to be activated in oligodendrocytes.


Subject(s)
Membrane Microdomains/metabolism , Membrane Transport Proteins , Myelin Proteins , Oligodendroglia/metabolism , Proteolipids/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Fluorescent Antibody Technique , Green Fluorescent Proteins , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Myelin and Lymphocyte-Associated Proteolipid Proteins , Proteolipids/genetics , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
18.
J Neuroimmunol ; 104(2): 155-63, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10713355

ABSTRACT

To investigate the possibility that an autoimmune mechanism may play a role in the hereditary neuropathy Charcot-Marie-Tooth type 1A (CMT1A), sera were analysed by Western blot for anti-peripheral myelin protein 22 (PMP22) autoantibodies. These sera were compared with sera from patients with CMT type 2 (CMT2), acquired peripheral neuropathies such as chronic inflammatory demyelinating neuropathy (CIDP), anti-MAG IgM neuropathy, Miller-Fisher syndrome (MFS), diabetic neuropathy and with control blood donors. Anti-PMP22 positive sera were detected in 70% of patients with CMT1 and unexpectedly in 60% of patients with CMT2. Interestingly, 44% of the patients with other peripheral neuropathies and 23% of the apparently healthy controls showed also anti-PMP22 antibody reactivity. Immunohistochemical analysis of the human anti-PMP22 antisera on healthy sural nerve sections and on PMP22-expressing COS cells revealed that these sera did not recognise endogenous PMP22. Our results indicate that anti-PMP22 autoantibodies are found in sera of patients with different types of peripheral neuropathies, but their role in the pathogenesis of these diseases remains to be determined.


Subject(s)
Autoantibodies/blood , Charcot-Marie-Tooth Disease/immunology , Myelin Proteins/immunology , Polyneuropathies/immunology , Adult , Aged , Blotting, Western , Case-Control Studies , Diabetic Neuropathies/immunology , Female , Humans , Male , Middle Aged , Miller Fisher Syndrome/immunology , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/immunology , Recombinant Proteins/immunology
19.
Schweiz Med Wochenschr ; 129(46): 1764-8, 1999 Nov 20.
Article in German | MEDLINE | ID: mdl-10603650

ABSTRACT

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. It results from an autoimmune response against one of several antigens of cerebral white matter tissue. The pathogenesis of MS lesions consists in migration of auto-reactive T-cells and disruption of the blood-brain-barrier. B-cells and their products are, however, also important for demyelination together with various cytotoxic or pro-inflammatory cytokines. Recent advances in immunology and biotechnology and protein engineering techniques have opened the door for novel immunotherapies. Immune modulation with beta-interferon has shown beneficial effect, both in relapsing remitting and secondary progressive multiple sclerosis.


Subject(s)
Multiple Sclerosis/pathology , Multiple Sclerosis/therapy , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Humans , Immunotherapy , Interferon-beta/therapeutic use , Multiple Sclerosis/epidemiology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
20.
J Neurochem ; 73(2): 587-97, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10428054

ABSTRACT

The myelin and lymphocyte protein MAL is a small proteolipid of 17 kDa and is expressed by oligodendrocytes and Schwann cells. We have characterized the embryonic and postnatal expression of MAL in the rat nervous system by in situ hybridization, immunocytochemistry, and western blotting and compared it with that of other myelin constituents. In the CNS, MAL is expressed during late steps of myelination: MAL protein appears approximately 3-5 days later than myelin basic protein and proteolipid protein. In contrast, in the PNS, MAL transcript and protein expression is detected prior to the onset of myelination, as early as embryonic day 17. Our results demonstrate that MAL is differentially expressed in oligodendrocytes and Schwann cells, likely reflecting different functions of the MAL proteolipid: (1) The late expression of MAL protein in the CNS points to a role in the final steps of myelin sheath formation, such as stabilization of the compacted myelin membranes. (2) The early expression of MAL protein in immature Schwann cells suggests an important role of MAL in the terminal differentiation step of the Schwann cell lineage and in the onset of peripheral myelination.


Subject(s)
Gene Expression Regulation, Developmental , Myelin Proteolipid Protein/genetics , Myelin Sheath/physiology , Schwann Cells/physiology , Spinal Cord/cytology , Animals , Blotting, Western , Cellular Senescence/physiology , Female , Immunohistochemistry , In Situ Hybridization , Myelin Basic Protein/analysis , Myelin Basic Protein/genetics , Myelin Proteins , Myelin Proteolipid Protein/analysis , Myelin-Associated Glycoprotein/analysis , Myelin-Associated Glycoprotein/genetics , Myelin-Oligodendrocyte Glycoprotein , Oligodendroglia/chemistry , Oligodendroglia/physiology , Pregnancy , RNA, Messenger/analysis , Rabbits , Rats , Rats, Inbred Lew , Schwann Cells/chemistry , Spinal Cord/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...