Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Genes Brain Behav ; 18(1): e12475, 2019 01.
Article in English | MEDLINE | ID: mdl-29566304

ABSTRACT

Oligodendrocyte gene expression is downregulated in stress-related neuropsychiatric disorders, including depression. In mice, chronic social stress (CSS) leads to depression-relevant changes in brain and emotional behavior, and the present study shows the involvement of oligodendrocytes in this model. In C57BL/6 (BL/6) mice, RNA-sequencing (RNA-Seq) was conducted with prefrontal cortex, amygdala and hippocampus from CSS and controls; a gene enrichment database for neurons, astrocytes and oligodendrocytes was used to identify cell origin of deregulated genes, and cell deconvolution was applied. To assess the potential causal contribution of reduced oligodendrocyte gene expression to CSS effects, mice heterozygous for the oligodendrocyte gene cyclic nucleotide phosphodiesterase (Cnp1) on a BL/6 background were studied; a 2 genotype (wildtype, Cnp1+/- ) × 2 environment (control, CSS) design was used to investigate effects on emotional behavior and amygdala microglia. In BL/6 mice, in prefrontal cortex and amygdala tissue comprising gray and white matter, CSS downregulated expression of multiple oligodendroycte genes encoding myelin and myelin-axon-integrity proteins, and cell deconvolution identified a lower proportion of oligodendrocytes in amygdala. Quantification of oligodendrocyte proteins in amygdala gray matter did not yield evidence for reduced translation, suggesting that CSS impacts primarily on white matter oligodendrocytes or the myelin transcriptome. In Cnp1 mice, social interaction was reduced by CSS in Cnp1+/- mice specifically; using ionized calcium-binding adaptor molecule 1 (IBA1) expression, microglia activity was increased additively by Cnp1+/- and CSS in amygdala gray and white matter. This study provides back-translational evidence that oligodendrocyte changes are relevant to the pathophysiology and potentially the treatment of stress-related neuropsychiatric disorders.


Subject(s)
Oligodendroglia/metabolism , Social Behavior , Stress, Psychological/genetics , Transcriptome , Amygdala/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Prefrontal Cortex/metabolism , Stress, Psychological/metabolism
2.
Nat Commun ; 9(1): 3025, 2018 08 02.
Article in English | MEDLINE | ID: mdl-30072689

ABSTRACT

In patients with Charcot-Marie-Tooth disease 1A (CMT1A), peripheral nerves display aberrant myelination during postnatal development, followed by slowly progressive demyelination and axonal loss during adult life. Here, we show that myelinating Schwann cells in a rat model of CMT1A exhibit a developmental defect that includes reduced transcription of genes required for myelin lipid biosynthesis. Consequently, lipid incorporation into myelin is reduced, leading to an overall distorted stoichiometry of myelin proteins and lipids with ultrastructural changes of the myelin sheath. Substitution of phosphatidylcholine and phosphatidylethanolamine in the diet is sufficient to overcome the myelination deficit of affected Schwann cells in vivo. This treatment rescues the number of myelinated axons in the peripheral nerves of the CMT rats and leads to a marked amelioration of neuropathic symptoms. We propose that lipid supplementation is an easily translatable potential therapeutic approach in CMT1A and possibly other dysmyelinating neuropathies.


Subject(s)
Charcot-Marie-Tooth Disease/therapy , Lipid Metabolism , Myelin Sheath/metabolism , Animals , Axons/metabolism , Axons/ultrastructure , Dietary Fats/pharmacology , Lipid Metabolism/drug effects , Lipids/biosynthesis , Myelin Sheath/ultrastructure , Phospholipids/metabolism , Rats, Transgenic , Schwann Cells/drug effects , Schwann Cells/metabolism , Schwann Cells/pathology
3.
Mol Psychiatry ; 23(2): 476-486, 2018 02.
Article in English | MEDLINE | ID: mdl-27922606

ABSTRACT

Despite high heritability of schizophrenia, genome-wide association studies (GWAS) have not yet revealed distinct combinations of single-nucleotide polymorphisms (SNPs), relevant for mental disease-related, quantifiable behavioral phenotypes. Here we propose an individual-based model to use genome-wide significant markers for extracting first genetic signatures of such behavioral continua. 'OTTO' (old Germanic=heritage) marks an individual characterized by a prominent phenotype and a particular load of phenotype-associated risk SNPs derived from GWAS that likely contributed to the development of his personal mental illness. This load of risk SNPs is shared by a small squad of 'similars' scattered under the genetically and phenotypically extremely heterogeneous umbrella of a schizophrenia end point diagnosis and to a variable degree also by healthy subjects. In a discovery sample of >1000 deeply phenotyped schizophrenia patients and several independent replication samples, including the general population, a gradual increase in the severity of 'OTTO's phenotype' expression is observed with an increasing share of 'OTTO's risk SNPs', as exemplified here by autistic and affective phenotypes. These data suggest a model in which the genetic contribution to dimensional behavioral traits can be extracted from combinations of GWAS SNPs derived from individuals with prominent phenotypes. Even though still in the 'model phase' owing to a world-wide lack of sufficiently powered, deeply phenotyped replication samples, the OTTO approach constitutes a conceptually novel strategy to delineate biological subcategories of mental diseases starting from GWAS findings and individual subjects.


Subject(s)
Genome-Wide Association Study/methods , Schizophrenia/genetics , Adult , Aged , Female , Genetic Heterogeneity , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Phenotype , Polymorphism, Single Nucleotide/genetics , Psychotic Disorders/genetics , Risk Factors
4.
Hippocampus ; 26(10): 1250-64, 2016 10.
Article in English | MEDLINE | ID: mdl-27101945

ABSTRACT

Expression of the lacZ-sequence is a widely used reporter-tool to assess the transgenic and/or transfection efficacy of a target gene in mice. Once activated, lacZ is permanently expressed. However, protein accumulation is one of the hallmarks of neurodegenerative diseases. Furthermore, the protein product of the bacterial lacZ gene is ß-galactosidase, an analog to the mammalian senescence-associated ß-galactosidase, a molecular marker for aging. Therefore we studied the behavioral, structural and molecular consequences of lacZ expression in distinct neuronal sub-populations. lacZ expression in cortical glutamatergic neurons resulted in severe impairments in hippocampus-dependent memory accompanied by marked structural alterations throughout the CNS. In contrast, GFP expression or the expression of the ChR2/YFP fusion product in the same cell populations did not result in either cognitive or structural deficits. GABAergic lacZ expression caused significantly decreased hyper-arousal and mild cognitive deficits. Attenuated structural and behavioral consequences of lacZ expression could also be induced in adulthood, and lacZ transfection in neuronal cell cultures significantly decreased their viability. Our findings provide a strong caveat against the use of lacZ reporter mice for phenotyping studies and point to a particular sensitivity of the hippocampus formation to detrimental consequences of lacZ expression. © 2016 Wiley Periodicals, Inc.


Subject(s)
Hippocampus/metabolism , Lac Operon , Memory/physiology , Neurons/metabolism , beta-Galactosidase/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Survival/physiology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Gene Expression , Glutamic Acid/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/diagnostic imaging , Hippocampus/pathology , Integrases/genetics , Integrases/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Neurons/pathology , Recombinant Fusion Proteins/metabolism , gamma-Aminobutyric Acid/metabolism
5.
Mol Psychiatry ; 21(12): 1752-1767, 2016 12.
Article in English | MEDLINE | ID: mdl-26809838

ABSTRACT

Recombinant human erythropoietin (EPO) improves cognitive performance in neuropsychiatric diseases ranging from schizophrenia and multiple sclerosis to major depression and bipolar disease. This consistent EPO effect on cognition is independent of its role in hematopoiesis. The cellular mechanisms of action in brain, however, have remained unclear. Here we studied healthy young mice and observed that 3-week EPO administration was associated with an increased number of pyramidal neurons and oligodendrocytes in the hippocampus of ~20%. Under constant cognitive challenge, neuron numbers remained elevated until >6 months of age. Surprisingly, this increase occurred in absence of altered cell proliferation or apoptosis. After feeding a 15N-leucine diet, we used nanoscopic secondary ion mass spectrometry, and found that in EPO-treated mice, an equivalent number of neurons was defined by elevated 15N-leucine incorporation. In EPO-treated NG2-Cre-ERT2 mice, we confirmed enhanced differentiation of preexisting oligodendrocyte precursors in the absence of elevated DNA synthesis. A corresponding analysis of the neuronal lineage awaits the identification of suitable neuronal markers. In cultured neurospheres, EPO reduced Sox9 and stimulated miR124, associated with advanced neuronal differentiation. We are discussing a resulting working model in which EPO drives the differentiation of non-dividing precursors in both (NG2+) oligodendroglial and neuronal lineages. As endogenous EPO expression is induced by brain injury, such a mechanism of adult neurogenesis may be relevant for central nervous system regeneration.


Subject(s)
Erythropoietin/metabolism , Neurogenesis/drug effects , Oligodendroglia/drug effects , Animals , Brain/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Central Nervous System/metabolism , Cognition/drug effects , Hippocampus/metabolism , Hippocampus/physiology , Male , Mice , Mice, Inbred C57BL , Neurogenesis/physiology , Neurons/metabolism , Oligodendroglia/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Recombinant Proteins/metabolism
8.
Mol Psychiatry ; 19(10): 1143-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23999527

ABSTRACT

In 2007, a multifaceted syndrome, associated with anti-NMDA receptor autoantibodies (NMDAR-AB) of immunoglobulin-G isotype, has been described, which variably consists of psychosis, epilepsy, cognitive decline and extrapyramidal symptoms. Prevalence and significance of NMDAR-AB in complex neuropsychiatric disease versus health, however, have remained unclear. We tested sera of 2817 subjects (1325 healthy, 1081 schizophrenic, 263 Parkinson and 148 affective-disorder subjects) for presence of NMDAR-AB, conducted a genome-wide genetic association study, comparing AB carriers versus non-carriers, and assessed their influenza AB status. For mechanistic insight and documentation of AB functionality, in vivo experiments involving mice with deficient blood-brain barrier (ApoE(-/-)) and in vitro endocytosis assays in primary cortical neurons were performed. In 10.5% of subjects, NMDAR-AB (NR1 subunit) of any immunoglobulin isotype were detected, with no difference in seroprevalence, titer or in vitro functionality between patients and healthy controls. Administration of extracted human serum to mice influenced basal and MK-801-induced activity in the open field only in ApoE(-/-) mice injected with NMDAR-AB-positive serum but not in respective controls. Seropositive schizophrenic patients with a history of neurotrauma or birth complications, indicating an at least temporarily compromised blood-brain barrier, had more neurological abnormalities than seronegative patients with comparable history. A common genetic variant (rs524991, P=6.15E-08) as well as past influenza A (P=0.024) or B (P=0.006) infection were identified as predisposing factors for NMDAR-AB seropositivity. The >10% overall seroprevalence of NMDAR-AB of both healthy individuals and patients is unexpectedly high. Clinical significance, however, apparently depends on association with past or present perturbations of blood-brain barrier function.


Subject(s)
Autoantibodies/blood , Blood-Brain Barrier/metabolism , Mood Disorders/metabolism , Parkinson Disease/metabolism , Receptors, N-Methyl-D-Aspartate/immunology , Schizophrenia/metabolism , Adult , Aged , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Cerebral Cortex/metabolism , Endocytosis/physiology , Female , Genome-Wide Association Study , Humans , Influenza, Human/genetics , Influenza, Human/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mood Disorders/genetics , Neurons/metabolism , Parkinson Disease/genetics , Polymorphism, Single Nucleotide , Receptors, N-Methyl-D-Aspartate/genetics , Schizophrenia/genetics
9.
Transl Psychiatry ; 3: e254, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23632458

ABSTRACT

Claustrophobia, the well-known fear of being trapped in narrow/closed spaces, is often considered a conditioned response to traumatic experience. Surprisingly, we found that mutations affecting a single gene, encoding a stress-regulated neuronal protein, can cause claustrophobia. Gpm6a-deficient mice develop normally and lack obvious behavioral abnormalities. However, when mildly stressed by single-housing, these mice develop a striking claustrophobia-like phenotype, which is not inducible in wild-type controls, even by severe stress. The human GPM6A gene is located on chromosome 4q32-q34, a region linked to panic disorder. Sequence analysis of 115 claustrophobic and non-claustrophobic subjects identified nine variants in the noncoding region of the gene that are more frequent in affected individuals (P=0.028). One variant in the 3'untranslated region was linked to claustrophobia in two small pedigrees. This mutant mRNA is functional but cannot be silenced by neuronal miR124 derived itself from a stress-regulated transcript. We suggest that loosing dynamic regulation of neuronal GPM6A expression poses a genetic risk for claustrophobia.


Subject(s)
Membrane Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Phobic Disorders/genetics , Adult , Amygdala/chemistry , Animals , Behavior, Animal , Electroretinography , Female , Genetic Engineering/methods , Heterozygote , Humans , Male , Mice , Mice, Inbred C57BL , Psychological Tests , Reflex, Startle/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stress, Psychological/genetics
10.
Mol Psychiatry ; 16(1): 26-36, 1, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20479759

ABSTRACT

Neurodevelopmental abnormalities together with neurodegenerative processes contribute to schizophrenia, an etiologically heterogeneous, complex disease phenotype that has been difficult to model in animals. The neurodegenerative component of schizophrenia is best documented by magnetic resonance imaging (MRI), demonstrating progressive cortical gray matter loss over time. No treatment exists to counteract this slowly proceeding atrophy. The hematopoietic growth factor erythropoietin (EPO) is neuroprotective in animals. Here, we show by voxel-based morphometry in 32 human subjects in a placebo-controlled study that weekly high-dose EPO for as little as 3 months halts the progressive atrophy in brain areas typically affected in schizophrenia, including hippocampus, amygdala, nucleus accumbens, and several neocortical areas. Specifically, gray matter protection is highly associated with improvement in attention and memory functions. These findings suggest that a neuroprotective strategy is effective against common pathophysiological features of schizophrenic patients, and strongly encourage follow-up studies to optimize EPO treatment dose and duration.


Subject(s)
Brain/pathology , Erythropoietin/administration & dosage , Neuroprotective Agents/administration & dosage , Schizophrenia/pathology , Adult , Analysis of Variance , Atrophy/drug therapy , Attention/drug effects , Brain/drug effects , Double-Blind Method , Humans , Male , Memory/drug effects , Middle Aged , Recombinant Proteins , Schizophrenia/drug therapy , Treatment Outcome
11.
Transl Psychiatry ; 1: e45, 2011 Oct 04.
Article in English | MEDLINE | ID: mdl-22833191

ABSTRACT

Genotype-phenotype correlations of common monogenic diseases revealed that the degree of deviation of mutant genes from wild-type structure and function often predicts disease onset and severity. In complex disorders such as schizophrenia, the overall genetic risk is still often >50% but genotype-phenotype relationships are unclear. Recent genome-wide association studies (GWAS) replicated a risk for several single-nucleotide polymorphisms (SNPs) regarding the endpoint diagnosis of schizophrenia. The biological relevance of these SNPs, however, for phenotypes or severity of schizophrenia has remained obscure. We hypothesized that the GWAS 'top-10' should as single markers, but even more so upon their accumulation, display associations with lead features of schizophrenia, namely positive and negative symptoms, cognitive deficits and neurological signs (including catatonia), and/or with age of onset of the disease prodrome as developmental readout and predictor of disease severity. For testing this hypothesis, we took an approach complementary to GWAS, and performed a phenotype-based genetic association study (PGAS). We utilized the to our knowledge worldwide largest phenotypical database of schizophrenic patients (n>1000), the GRAS (Göttingen Research Association for Schizophrenia) Data Collection. We found that the 'top-10' GWAS-identified risk SNPs neither as single markers nor when explored in the sense of a cumulative genetic risk, have any predictive value for disease onset or severity in the schizophrenic patients, as demonstrated across all core symptoms. We conclude that GWAS does not extract disease genes of general significance in schizophrenia, but may yield, on a hypothesis-free basis, candidate genes relevant for defining disease subgroups.


Subject(s)
Genetic Association Studies , Schizophrenia/diagnosis , Schizophrenia/genetics , Severity of Illness Index , Adult , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Genotype , Humans , Male , Polymorphism, Single Nucleotide/genetics , Risk
12.
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
13.
Mol Cell Neurosci ; 36(3): 416-24, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17889554

ABSTRACT

Tissue damage in the CNS is critically influenced by the adaptive immune system. Primary oligodendrocyte damage (by overexpression of PLP) leads to low-grade inflammation of high pathological impact, which is mediated by CD8+ T cells. To yield further insight into pathogenesis and nature of immune responses in myelin mutated mice, we here apply a detailed immunological characterization of CD8+ T cells in PLP-transgenic and aged wild type mice. We provide evidence that T effector cells accumulate in the CNS of PLP-transgenic and wild-type mice and show a higher level of activation in mutant mice, indicated by surface markers and clonal expansions, as demonstrated by T cell receptor CDR3-spectratype analysis. Vbeta-Jbeta similarities suggest specificity against a common antigen, albeit we could not find specific responses against myelin-antigen-derived peptides. The association of primary oligodendrocyte damage with secondary expansions of pathogenic cells underlines the role of adaptive immune reactions in neurodegenerative and neuroinflammatory diseases.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Central Nervous System/immunology , Demyelinating Diseases/immunology , Immune System/immunology , Myelin Sheath/immunology , Animals , Autoimmunity/genetics , Autoimmunity/immunology , CD8 Antigens/immunology , Cell Proliferation , Central Nervous System/metabolism , Central Nervous System/physiopathology , Clone Cells/immunology , Demyelinating Diseases/genetics , Demyelinating Diseases/physiopathology , Immune System/physiopathology , Immunity, Innate/genetics , Immunity, Innate/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Mice, Transgenic , Myelin Sheath/genetics , Myelin Sheath/metabolism , Oligodendroglia/immunology , Oligodendroglia/metabolism , Oligodendroglia/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
14.
J Neurosci ; 26(47): 12339-50, 2006 Nov 22.
Article in English | MEDLINE | ID: mdl-17122059

ABSTRACT

Fibroblast growth factor receptors (Fgfr) comprise a widely expressed family of developmental regulators implicated in oligodendrocyte (OL) maturation of the CNS. Fgfr2 is expressed by OLs in myelinated fiber tracks. In vitro, Fgfr2 is highly upregulated during OL terminal differentiation, and its activation leads to enhanced growth of OL processes and the formation of myelin-like membranes. To investigate the in vivo function of Fgfr2 signaling by myelinating glial cells, we inactivated the floxed Fgfr2 gene in mice that coexpress Cre recombinase (cre) as a knock-in gene into the OL-specific 2',3'-cyclic nucleotide phosphodiesterase (Cnp1) locus. Surprisingly, no obvious defects were detected in brain development of these conditional mutants, including the number of OLs, the onset and extent of myelination, the ultrastructure of myelin, and the expression level of myelin proteins. However, unexpectedly, a subset of these conditional Fgfr2 knock-out mice that are homozygous for cre and therefore are also Cnp1 null, displayed a dramatic hyperactive behavior starting at approximately 2 weeks of age. This hyperactivity was abolished by treatment with dopamine receptor antagonists or catecholamine biosynthesis inhibitors, suggesting that the symptoms involve a dysregulation of the dopaminergic system. Although the molecular mechanisms are presently unknown, this novel mouse model of hyperactivity demonstrates the potential involvement of OLs in neuropsychiatric disorders, as well as the nonpredictable role of genetic interactions in the behavioral phenotype of mice.


Subject(s)
2',3'-Cyclic-Nucleotide Phosphodiesterases/physiology , Fibroblast Growth Factor 2/physiology , Hyperkinesis/genetics , Hyperkinesis/physiopathology , Oligodendroglia/metabolism , 2',3'-Cyclic-Nucleotide Phosphodiesterases/deficiency , 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism , Animals , Animals, Newborn , Behavior, Animal , Blotting, Western/methods , Brain/cytology , Cell Differentiation/genetics , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Fibroblast Growth Factor 2/deficiency , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Immunohistochemistry/methods , In Situ Hybridization/methods , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Motor Activity/drug effects , Motor Activity/physiology , Myelin Basic Protein/metabolism , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Oligodendroglia/ultrastructure , Tyrosine 3-Monooxygenase/metabolism
15.
Neuromolecular Med ; 8(1-2): 205-16, 2006.
Article in English | MEDLINE | ID: mdl-16775377

ABSTRACT

The most frequent genetic subtype of Charcot-Marie-Tooth disease is CMT1A, linked to chromosome 17p11.2. In the majority of cases, CMT1A is a gene dosage disease associated with a 1.5 Mb large genomic duplication. Transgenic models with extra copies of the Pmp22 gene have provided formal proof that overexpression of only this candidate gene is sufficent to cause peripheral demyelination, onion bulb formation, secondary axonal loss, and progressive muscle atrophy, the pathological hallmarks of CMT1A. The transgenic CMT rat with about 1.6-fold PMP22 overexpression exhibits clinical abnormalities, such as reduced nerve conduction velocity and lower grip strength that mimick findings in CMT1A patients. Also transgenic mice, carrying yeast artifical chromosomes as Pmp22 transgenes, demonstrate the variability of disease expression as a function of increased gene dosage. Recently, the first rational experimental therapies of CMT1A were tested, using transgenic animal models. In one proof-of-principle study with the CMT rat, a synthetic antagonist of the nuclear progesterone receptor was shown to reduce PMP22 overexpression and to ameliorate the clinical severity. In another study, administration of ascorbic acid, an essential factor of in vitro myelination, prolonged the survival and restored myelination of a dysmyelinated mouse model. Application of gene expression analysis to nerve biopsies that are readily available from such CMT1A animal models might identify additional pharmacological targets.


Subject(s)
Charcot-Marie-Tooth Disease/physiopathology , Disease Models, Animal , Animals , Animals, Genetically Modified , Antioxidants/therapeutic use , Ascorbic Acid/therapeutic use , Charcot-Marie-Tooth Disease/pathology , Charcot-Marie-Tooth Disease/therapy , Chromosomes, Human, Pair 17 , Gene Dosage , Humans , Myelin Proteins/genetics , Myelin Proteins/metabolism , Neuroprotective Agents/therapeutic use , Progesterone/antagonists & inhibitors , Progesterone/therapeutic use
16.
J Neurosci Res ; 84(1): 68-77, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16625661

ABSTRACT

In vivo and in vitro studies have shown that alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA)-receptor-mediated excitotoxicity causes cytoskeletal damage to axons. AMPA/kainate receptors are present on oligodendrocytes and myelin, but currently there is no evidence to suggest that axon cylinders contain AMPA receptors. Proteolipid protein (PLP) and DM20 are integral membrane proteins expressed by CNS oligodendrocytes and located in compact myelin. Humans and mice lacking normal PLP/DM20 develop axonal swellings and degeneration, suggesting that local interactions between axons and the oligodendrocyte/myelin unit are important for the normal functioning of axons and that PLP/DM20 is involved in this process. To determine whether perturbed glial-axonal interaction affects AMPA-receptor-mediated axonal damage, AMPA (1.5 nmol) was injected into the caudate nucleus of anesthetized Plp knockout and wild-type male mice (n = 13). Twenty-four hours later, axonal damage was detected by using neurofilament 200 (NF 200) immunohistochemistry and neuronal damage detected via histology. AMPA-induced axonal damage, assessed with NF 200 immunohistochemistry, was significantly reduced in Plp knockout mice compared with wild-type mice (P = 0.015). There was no significant difference in the levels of neuronal perikaryal damage between the Plp knockout and wild-type mice. In addition, there was no significant difference in the levels of glutamate receptor subunits GluR1-4 or KA2 in Plp knockout compared with wild-type littermates. The present study suggests that PLP-mediated interactions among oligodendrocytes, myelin, and axons may be involved in AMPA-mediated axonal damage.


Subject(s)
Axons/drug effects , Brain Injuries/chemically induced , Excitatory Amino Acid Agonists/toxicity , Myelin Proteolipid Protein/deficiency , Nerve Tissue Proteins/deficiency , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/toxicity , Animals , Axons/metabolism , Axons/pathology , Blotting, Western/methods , Brain Injuries/pathology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Immunohistochemistry/methods , Male , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism
17.
Cell Death Differ ; 11(12): 1247-57, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15375385

ABSTRACT

Mutations of the myelin proteolipid protein gene (Plp) are associated with excessive programmed cell death (PCD) of oligodendrocytes. We show for the first time that PLP is a molecule ubiquitously expressed in non-neural tissues during normal development, and that the level of native PLP modulates the level of PCD. We analyze three non-neural tissues, and show that native PLP is expressed in trophoblasts, spermatogonia, and cells of interdigital webbing. The non-neural cells that express high levels of native PLP also undergo PCD. The level of PLP expression modulates the level of PCD because mice that overexpress native PLP have increased PCD and mice deficient in PLP have decreased PCD. We show that overexpression of native PLP causes a dramatic acidification of extracellular fluid that, in turn, causes increased PCD. These studies show that the level of native PLP modulates the amount of PCD during normal development via a pH-dependent mechanism.


Subject(s)
Apoptosis/genetics , Extracellular Fluid/metabolism , Gene Expression Regulation/genetics , Myelin Proteolipid Protein/genetics , Animals , Caspase 3 , Caspases/metabolism , Central Nervous System/cytology , Central Nervous System/metabolism , Female , Hydrogen-Ion Concentration , Male , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Neurologic Mutants , Mice, Transgenic , Protons , Skin/cytology , Skin/metabolism , Spermatogonia/metabolism , Trophoblasts/metabolism
18.
J Neurosci Res ; 72(1): 12-24, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12645075

ABSTRACT

The rumpshaker mutation of the proteolipid protein (Plp) gene causes dysmyelination in man and mouse. We show that the phenotype in the mouse depends critically on the genetic background in which the mutation is expressed. On the C3H background there is normal longevity whereas changing to a C57BL/6 strain results in seizures and death at around postnatal day 30. The more severe phenotype is associated with less myelin and reduced levels of major myelin proteins. There are also more apoptotic cells, including oligodendrocytes, increased numbers of proliferating cells, increased numbers of NG2+ oligodendrocyte progenitors and increased microglia compared to the milder phenotype. The number of mature oligodendrocytes is similar to wild-type in both strains of mutant, however, suggesting that increased oligodendrocyte death is matched by increased generation from progenitors. The dichotomy of phenotype probably reflects the influence of modifying loci. The localization of these putative modifying genes and their mode of action remain to be determined.


Subject(s)
Mutation/physiology , Myelin Proteolipid Protein/deficiency , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/physiopathology , Phenotype , Animals , Brain/pathology , Female , Genotype , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Myelin Proteolipid Protein/genetics , Myelin Sheath/genetics , Myelin Sheath/pathology , Species Specificity
19.
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
20.
Glia ; 39(3): 256-67, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12203392

ABSTRACT

Myelin-associated oligodendrocytic basic protein (MOBP) is a recently identified major component of central nervous system (CNS) myelin. We previously reported a detailed characterization of the genomic region encompassing the Mobp gene, elucidating the complex series of transcript splicing responsible for the generation of its diverse family of protein isoforms. These basic, positively charged polypeptides display spatial and temporal expression patterns consistent with a potential role in the compaction and maintenance of the myelin sheath. MOBP isoforms have also been localized to the nucleus and the microtubular network of oligodendrocytes; transcript corresponding to one isoform is present during embryonic development. Recent reports have identified a role for this protein family in the pathogenesis of multiple sclerosis, but a clear function for the wild-type protein has remained unclear. We report a detailed analysis of a targeted mutation of Mobp, which results in the deletion of the translational start site and most of the coding sequence of MOBP, and the deletion of the entire coding sequence corresponding to a novel, putative MOBP isoform. Our analyses clearly demonstrate that MOBP-deficient mice develop normally, generate intact compact CNS myelin, and demonstrate no obvious clinical phenotype. Furthermore, in contrast with another recent study, we find that Mobp null mice demonstrate no significant influence on the axonal diameter of myelinated axons. Although MOBP is not essential for myelination, it appears that its absence is not simply compensated for by increased expression of the "classic" myelin basic protein (MBP).


Subject(s)
Myelin-Associated Glycoprotein/deficiency , Myelin-Associated Glycoprotein/genetics , Alternative Splicing/genetics , Animals , Axons/metabolism , Cerebellum/cytology , Cerebellum/metabolism , Female , Gene Dosage , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Neurologic Mutants , Multigene Family/genetics , Myelin Proteins , Myelin Sheath/metabolism , Myelin-Associated Glycoprotein/biosynthesis , Myelin-Oligodendrocyte Glycoprotein , Phenotype , Pregnancy , Protein Isoforms/biosynthesis , Protein Isoforms/deficiency , Protein Isoforms/genetics , Spinal Cord/cytology , Spinal Cord/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...