Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 256
Filter
Add more filters











Publication year range
1.
Nat Commun ; 12(1): 5091, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34429415

ABSTRACT

Ten-eleven translocation (TET) proteins, the dioxygenase for DNA hydroxymethylation, are important players in nervous system development and diseases. However, their role in myelination and remyelination after injury remains elusive. Here, we identify a genome-wide and locus-specific DNA hydroxymethylation landscape shift during differentiation of oligodendrocyte-progenitor cells (OPC). Ablation of Tet1 results in stage-dependent defects in oligodendrocyte (OL) development and myelination in the mouse brain. The mice lacking Tet1 in the oligodendrocyte lineage develop behavioral deficiency. We also show that TET1 is required for remyelination in adulthood. Transcriptomic, genomic occupancy, and 5-hydroxymethylcytosine (5hmC) profiling reveal a critical TET1-regulated epigenetic program for oligodendrocyte differentiation that includes genes associated with myelination, cell division, and calcium transport. Tet1-deficient OPCs exhibit reduced calcium activity, increasing calcium activity rescues the differentiation defects in vitro. Deletion of a TET1-5hmC target gene, Itpr2, impairs the onset of OPC differentiation. Together, our results suggest that stage-specific TET1-mediated epigenetic programming and intracellular signaling are important for proper myelination and remyelination in mice.


Subject(s)
Brain/metabolism , DNA-Binding Proteins/metabolism , DNA/metabolism , Mice, Neurologic Mutants/metabolism , Proto-Oncogene Proteins/metabolism , Remyelination/physiology , 5-Methylcytosine/analogs & derivatives , Animals , Cell Cycle , Cell Differentiation , DNA Methylation , DNA-Binding Proteins/genetics , Genome , Mice , Mice, Knockout , Oligodendroglia/metabolism , Organogenesis , Proto-Oncogene Proteins/genetics
2.
Brain Struct Funct ; 216(3): 201-18, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21442415

ABSTRACT

Mapping of multiple receptors of neurotransmitters provides insight into the spatial distribution of neurotransmission-relevant molecules in the cerebral cortex. During development, lack of reelin leads to impaired migration, disturbed lamination of the hippocampus and inverted neocortical layering. In the adult, reelin may regulate synaptic plasticity by modulating neurotransmitter receptor function. Using quantitative in vitro receptor autoradiography, different receptors, in particular, the binding site densities and laminar distribution of various glutamate, GABA, muscarinic and nicotinic acetylcholine, serotonin, dopamine and adenosine receptors, were analyzed in cortical and subcortical structures of reeler and wild-type brains. Differential changes in the laminar distribution, maximum binding capacity (B (max)) and regional density of neurotransmitter receptors were found in the reeler brain. A decrease of whole brain B (max) was found for adenosine A(1) and GABA(A) receptors. In the forebrain, several binding sites were differentially up- or down-regulated (kainate, A(1), benzodiazepine, 5-HT(1), M(2), α(1) and α(2)). In the hippocampus, a significant decrease of GABA(B), 5-HT(1) and A'1 receptors were observed. The density of M(2) receptors increased, while other receptors remained unchanged. In the neocortex, some receptors demonstrated an obviously inverted laminar distribution (AMPA, kainate, NMDA, GABA(B), 5-HT(1), M(1), M(3), nAch), while the distribution of others (A(1), GABA(A), benzodiazepine, 5-HT(2), muscarinic M(2), adrenergic α(1), α(2)) seemed to be less affected. Thus, the laminar receptor distribution is modulated by the developmental impairment and suggests and reflects partially the laminar inversion in reeler mice.


Subject(s)
Brain Mapping/methods , Brain/metabolism , Cell Adhesion Molecules, Neuronal/deficiency , Extracellular Matrix Proteins/deficiency , Mice, Neurologic Mutants/metabolism , Nerve Tissue Proteins/deficiency , Receptors, Neurotransmitter/metabolism , Serine Endopeptidases/deficiency , Synaptic Transmission/physiology , Analysis of Variance , Animals , Autoradiography , Brain/anatomy & histology , Densitometry , Image Processing, Computer-Assisted , Male , Mice , Mice, Neurologic Mutants/anatomy & histology , Radioligand Assay , Reelin Protein , Regression Analysis
3.
Brain Res ; 1326: 40-50, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20219442

ABSTRACT

The weaver mouse represents the only genetic animal model of gradual nigrostriatal dopaminergic neurodegeneration which is proposed as a pathophysiological phenotype of Parkinson's disease. The aim of the present study was to analyze the nitric oxide and dopaminergic systems in selected brain regions of homozygous weaver mice at different postnatal ages corresponding to specific stages of the dopamine loss. Structural deficits were evaluated by quantification of tyrosine hydroxylase and neuronal nitric oxide synthase-immunostaining in the cortex, striatum, accumbens nuclei, subthalamic nuclei, ventral tegmental area, and substantia nigra compacta of 10-day, 1- and 2-month-old wild-type and weaver mutant mice. The results confirmed the progressive loss of dopamine during the postnatal development in the adult weaver mainly affecting the substantia nigra pars compacta, striatum, and subthalamic nucleus and slightly affecting the accumbens nuclei and ventral tegmental area. A general decrease in neuronal nitric oxide synthase-immunostaining with age was revealed in both the weaver and wild-type mice, with the decrease being most pronounced in the weaver. In contrast, there was an increase in the substantia nigra pars compacta nitric oxide synthase-immunostaining and a decrease mainly in the subthalamic and accumbens nuclei of the 2-month-old weaver mutant. The decrease in the expression of nNOS may bear functional significance related to the process of aging. DA neurons from the substantia nigra directly modulate the activity of subthalamic nucleus neurons, and their loss may contribute to the abnormal activity of subthalamic nucleus neurons. Although the functional significance of these changes is not clear, it may represent plastic compensating adjustments resulting from the loss of dopamine innervation, highlighting a possible role of nitric oxide in this process.


Subject(s)
Brain/enzymology , Gene Expression Regulation, Developmental/physiology , Mice, Neurologic Mutants/metabolism , Nitric Oxide Synthase Type I/metabolism , Analysis of Variance , Animals , Animals, Newborn , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Gene Expression Regulation, Developmental/genetics , Genotype , Mice , Nitric Oxide Synthase Type I/genetics , Tyrosine 3-Monooxygenase/metabolism
4.
J Comp Neurol ; 517(4): 524-38, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19795496

ABSTRACT

Somatostatin 28 immunoreactivity (Sst28-ir) identifies a specific subset of mossy fiber terminals in the adult mouse cerebellum. By using double-labeling immunohistochemistry, we determined that Sst28-ir is associated with presynaptic mossy fiber terminal rosettes, and not Purkinje cells, Golgi cells, or unipolar brush cells. Sst28-ir mossy fibers are restricted to the central zone (lobules VI/VII) and nodular zone (lobules IX, X) of the vermis, and the paraflocculus and flocculus. Within each transverse zone the mossy fiber terminal fields form a reproducible array of parasagittal stripes. The boundaries of Sst28-ir stripes align with a specific array of Purkinje cell stripes revealed by using immunocytochemistry for the small heat shock protein HSP25. In the cerebellum of the homozygous weaver mouse, in which a subpopulation of HSP25-ir Purkinje cells are located ectopically, the corresponding Sst28-ir mossy fiber projection is also ectopic, suggesting a role for a specific Purkinje cell subset in afferent pattern formation. Likewise, in the scrambler mutant mouse, Sst28-ir mossy fibers show a very close association with HSP25-ir Purkinje cell clusters. HSP25 itself does not appear to be critical for normal patterning, however: in the KJR mouse, which does not express cerebellar HSP25, Sst28 expression appears to be normal. Likewise, the Purkinje cell patterning antigens zebrin II and HSP25 are expressed normally in both Sst- and Sst-receptor knockout mice, suggesting that somatostatinergic transmission is not necessary for Purkinje cell stripe formation.


Subject(s)
Cerebellum/cytology , Heat-Shock Proteins/metabolism , Neoplasm Proteins/metabolism , Nerve Fibers/metabolism , Purkinje Cells/metabolism , Somatostatin-28/metabolism , Animals , CD57 Antigens/metabolism , Choline O-Acetyltransferase/metabolism , Corticotropin-Releasing Hormone/metabolism , Gene Expression Regulation/genetics , Mice , Mice, Knockout , Mice, Neurologic Mutants/metabolism , Molecular Chaperones , Nerve Tissue Proteins/metabolism , Purkinje Cells/cytology , Receptors, Somatostatin/deficiency , Somatostatin-28/genetics , gamma-Aminobutyric Acid/metabolism
5.
Dev Growth Differ ; 51(3): 271-86, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19379278

ABSTRACT

The reeler mouse is one of the most famous spontaneously occurring mutants in the research field of neuroscience, and this mutant has been used as a model animal to understand mammalian brain development. The classical observations emphasized that laminar structures of the reeler brain are highly disrupted. Molecular cloning of Reelin, the gene responsible for reeler mutant provided insights into biochemistry of Reelin signal, and some models had been proposed to explain the function of Reelin signal in brain development. However, recent reports of reeler found that non-laminated structures in the central nervous system are also affected by the mutation, making function of Reelin signal more controversial. In this review, we summarized reported morphological and histological abnormalities throughout the central nervous system of the reeler comparing to those of the normal mouse. Based on this overview of the reeler abnormalities, we discuss possible function of Reelin signal in the neuronal migration and other morphological events in mouse development.


Subject(s)
Mice, Neurologic Mutants/anatomy & histology , Mice, Neurologic Mutants/metabolism , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Cell Movement/genetics , Cell Movement/physiology , Extracellular Matrix Proteins/metabolism , Mice , Mice, Neurologic Mutants/genetics , Models, Biological , Nerve Tissue Proteins/metabolism , Neurons/cytology , Reelin Protein , Serine Endopeptidases/metabolism , Signal Transduction/physiology
6.
Prog Neuropsychopharmacol Biol Psychiatry ; 32(5): 1269-76, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18501492

ABSTRACT

OBJECTIVE AND METHODS: The reeler heterozygous (HZ) mice have provided a model for studying the relationship between reelin (a protein of extracellular matrix) haploinsufficiency and the emergence of neuropsychiatric diseases. In a neurodevelopmental framework, the enduring consequences of early maternal separation (5 h/day during the first postnatal week, or handling controls, H) were studied in reeler HZ and wild type (WT) mice at adulthood. The modulatory effects of a chronic treatment with the atypical antipsychotic olanzapine (OLZ, 1.5 mg/kg for 40 days) were also investigated. RESULTS: Early maternal separation had long-term effects on brain plasticity, with a reduction of brain- and glial- derived neurotrophic factor (BDNF and GDNF) in several brain areas of mice, but such a consequence was less marked in the HZ genotype. On the other hand, treatment with OLZ did not affect at all the GDNF but led to an increase of BDNF levels in maternally separated (SEP) mice, an effect which was far more marked in the HZ genotype. Brain levels of serotonin (5-HT) were markedly increased, striatal dopamine (DA) was increased, whereas metabolites and turnover were decreased, in SEP mice of both genotypes. The spontaneous home-cage activity was generally lower in HZ than WT mice, and OLZ treatment contrasted this hypoactivity profile. Maternal separation also decreased the interest toward an unknown mouse proposed as a social stimulus, but only in WT mice. CONCLUSION: We investigated the interplay between genetic vulnerability (reelin haploinsufficiency), the outcome of early stressful experiences, and the efficacy of the antipsychotic drug therapy. The reeler HZ genotype exhibited a slightly lower sensitivity to the environmental insult as well as an enhanced response to the atypical antipsychotic treatment.


Subject(s)
Biogenic Monoamines/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain/metabolism , Heterozygote , Maternal Deprivation , Mice, Neurologic Mutants/metabolism , Animals , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Benzodiazepines/pharmacology , Brain/anatomy & histology , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/genetics , Naphthalenes , Olanzapine , Oxepins , Reelin Protein , Social Behavior
7.
Neurochem Int ; 51(2-4): 105-11, 2007.
Article in English | MEDLINE | ID: mdl-17586089

ABSTRACT

Parkinson's disease (PD) and Alzheimer's disease (AD), the most common neurodegenerative diseases, are caused by both genetic and environmental factors. Ubiquitin carboxy-terminal hydrolase L1 (UCH-L1) is a deubiquitinating enzyme that is involved in the pathogenesis of both of these neurodegenerative diseases. Several functions of UCH-L1, other than as an ubiquitin hydrolase, have been proposed; these include acting as an ubiquitin ligase and stabilizing mono-ubiquitin. This review focuses on recent findings on the functions and the regulation of UCH-L1, in particular those that relate to PD and AD.


Subject(s)
Alzheimer Disease/metabolism , Parkinson Disease/metabolism , Ubiquitin Thiolesterase/physiology , Alzheimer Disease/genetics , Animals , Humans , Mice , Mice, Neurologic Mutants/genetics , Mice, Neurologic Mutants/metabolism , Mutation/genetics , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/metabolism , Oxidative Stress/physiology , Parkinson Disease/genetics , Ubiquitin/metabolism , Ubiquitin Thiolesterase/genetics
8.
Int J Exp Pathol ; 88(3): 165-73, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17504446

ABSTRACT

Reelin is a neuronal glycoprotein that plays a crucial role in brain layer formation during prenatal development. The reeler mutant mouse lacks Reelin, leading to abnormalities in the neuronal layering of cerebral cortex and cerebellum, producing ataxia, tremor and abnormal locomotion. Reeler mice are reported to have growth retardation and most of them are sterile or unable to bring up their newborns. Since the brain is one of the main regulator of pituitary hormone secretion and no information was reported regarding pituitary function and structure in these mutant mice, we studied pituitary endocrine activity and morphology in reeler mice. Mice were classified in three groups as reeler homozygote (RHM), reeler heterozygote (RHT) or control (CO). Pituitary hormone blood levels were assessed by enzyme immunoassay (EIA) and immunoradiometric assay (IRMA). Animals and their pituitaries were weighted and pituitaries were studied by histology, immunohistochemistry and electron microscopy. Results showed statistically significant differences in body weight and in adrenocorticotropic hormone (ACTH) and luteinizing hormone (LH) blood levels between the three groups. In contrast, growth hormone (GH) blood levels showed a high individual variation and no decrease in reeler groups compared with CO. Morphological studies revealed no differences in pituitary cell types except that somatotrophs appeared to be slightly smaller in RHM and RHT. Although it seems that pituitary hypofunction is not responsible for growth retardation, more studies are needed to obtain a deeper insight into the endocrine status of these mutant mice to elucidate the cause of their low body weight and reproductive behaviour.


Subject(s)
Mice, Neurologic Mutants/metabolism , Pituitary Gland, Anterior/metabolism , Pituitary Gland, Anterior/pathology , Pituitary Hormones/analysis , Adrenocorticotropic Hormone/analysis , Animals , Follicle Stimulating Hormone/analysis , Growth Disorders/metabolism , Growth Disorders/pathology , Growth Hormone/analysis , Heterozygote , Homozygote , Immunohistochemistry , Luteinizing Hormone/analysis , Mice , Mice, Neurologic Mutants/genetics , Microscopy, Electron, Transmission , Prolactin/analysis , Reelin Protein , Thyrotropin/analysis
9.
Neurosci Res ; 58(4): 378-85, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17499872

ABSTRACT

Despite preserved cell differentiation, the Reln(rl-Orl) phenotype comprises laminar abnormalities of cell position in auditory cortex and dorsal cochlear nucleus. The metabolic consequences of the cell ectopias were determined by estimating cytochrome oxidase (CO) activity, a marker of neuronal activity. CO activity increased in the granular cell layer of dorsal cochlear nucleus, trapezoid body nucleus, intermediate lateral lemniscus, central and external inferior colliculus, and pyramidal cell layer of primary auditory cortex. On the contrary, CO activity decreased in the superficial molecular layer of dorsal cochlear nucleus as well as in the medioventral periolivary nucleus. These metabolic variations are discussed in terms of their possible relation to morphologic anomalies observed in the mutant.


Subject(s)
Auditory Cortex/anatomy & histology , Auditory Cortex/enzymology , Cochlear Nucleus/anatomy & histology , Cochlear Nucleus/enzymology , Electron Transport Complex IV/metabolism , Mice, Neurologic Mutants/metabolism , Animals , Mice , Mice, Inbred BALB C , Mice, Neurologic Mutants/anatomy & histology , Reelin Protein
10.
Brain Res ; 1140: 120-31, 2007 Apr 06.
Article in English | MEDLINE | ID: mdl-17447264

ABSTRACT

Conventional kinesin is a motor protein complex including two heavy chains and two light chains (KLC). Junco et al. (Junco, A., Bhullar, B., Tarnasky, H.A. and van der Hoorn, F.A., 2001. Kinesin light-chain KLC3 expression in testis is restricted to spermatids. Biol. Reprod. 64, 1320-1330). recently reported the isolation of a novel KLC gene, klc3. In the present report, immunohistochemistry has been used to characterize the expression of KLC3 in the cerebella of normal and scrambler (scm) mutant mice. In cryostat sections through the cerebellum of the normal adult mouse immunoperoxidase stained for KLC3, reaction product is deposited in the nuclei and somata of deep cerebellar nuclear neurons. No other structures are stained in the cerebellum. Strong and specific KLC3 expression is observed in the adult cerebellum in all three major cerebellar nuclei--medial, interposed, and lateral. Double immunofluorescence studies reveal that KLC3 immunoreactivity is colocalized with both endosomes and GW bodies. KLC3 immunohistochemistry has been exploited to study the organization of the cerebellar nuclei in scrambler mice, in which disruption of the mdab1 gene results in severe foliation defects due to Purkinje cell ectopia, with most Purkinje cells clumped in centrally located clusters. Despite the severe failure of Purkinje cell migration, the cerebellar nuclei appear normal in scrambler mutant mice, suggesting that their topography is dependent neither on normal Purkinje cell positioning nor the Reelin signaling pathway.


Subject(s)
Cerebellar Nuclei/metabolism , Gene Expression/physiology , Mice, Neurologic Mutants/anatomy & histology , Microtubule-Associated Proteins/metabolism , Age Factors , Animals , Gene Expression/genetics , Kinesins , Mice , Mice, Neurologic Mutants/metabolism , Microtubule-Associated Proteins/genetics , Reelin Protein
11.
Endocrinology ; 148(4): 1745-53, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17218417

ABSTRACT

The mutant mouse staggerer (sg) harbors a deletion within the gene encoding the retinoic acid receptor-related orphan receptor-alpha (RORalpha). Homozygotes show aberrant cerebellar development. However, the mechanisms responsible for the cerebellar defect are still poorly understood. In the present study, the involvement of neurotropins (NTs), including nerve growth factor, brain-derived neurotropic factor, NT-3 and NT-4/5, and their receptors, which play a crucial role in brain development, on the cerebellar defects of sg mice was studied by semiquantitative RT-PCR and in situ hybridization histochemistry. An evident alteration of these mRNA levels was observed in both heterozygotes and homozygotes. Such difference was most evident in the internal granule cell layer. Because the changes in NT expression as well as morphological alterations in sg cerebellum are similar to those in hypothyroid animals, the effect of mutant RORalpha (RORsg) on transcriptional regulation through the thyroid hormone (TH) response element or the ROR response element (RORE) was then studied. RORsg neither activated the transcription through RORE nor suppressed RORalpha-induced transcription, indicating that it does not function as a dominant negative inhibitor. On the other hand, although wild-type RORalpha augmented TH receptor (TR)alpha1/beta1-mediated transcription through various TH response elements, RORsg was not effective in augmenting TR action. These results suggest that the cerebellar defect of the sg mouse is partly caused by the altered expression of NTs and the lack of augmentation of TR-mediated transcription by RORalpha as well as the absence of RORalpha action through RORE.


Subject(s)
Cerebellum/metabolism , Mice, Neurologic Mutants/genetics , Polysaccharides/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Thyroid Hormone/metabolism , Trans-Activators/genetics , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Cerebellum/anatomy & histology , Cerebellum/growth & development , Gene Expression Regulation, Developmental/genetics , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants/metabolism , Mutation , Nerve Growth Factors/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1 , Polysaccharides/metabolism , RNA, Messenger/metabolism , Receptor, trkB/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Trans-Activators/physiology , Transcription, Genetic
12.
Brain Res ; 1140: 96-104, 2007 Apr 06.
Article in English | MEDLINE | ID: mdl-16359645

ABSTRACT

Nitric oxide (NO) is a diffusible messenger molecule produced primarily by neuronal nitric oxide synthase (nNOS) in the central nervous system. Both nNOS expression and NO production are regulated by calcium ions. Leaner and tottering mice carry a mutation in the pore forming subunit (alpha1A) of P/Q-type voltage-gated calcium ion channels, which decreases calcium ion current through the affected channels and disrupts calcium homeostasis. We have previously shown that nNOS expression is altered in adult leaner and tottering cerebella. In addition, leaner and tottering mice have been shown to have abnormal cerebellar granule cell-Purkinje cell synapses and leaner cerebellar granule cells undergo abnormal apoptosis during early postnatal development. Since NO production has been linked to several developmental roles including neuronal cell death, synaptogenesis and neuronal cell survival, our objective was to evaluate the expression of nNOS in developing leaner and tottering cerebella. Our results show that nNOS is differentially expressed in leaner and tottering cerebella compared to wild type cerebella and compared to each other. In whole cerebella, Western blotting revealed a significant increase in nNOS expression at postnatal day 12 in tottering but not leaner or wild type cerebella. At the cellular level the NADPH-diaphorase marker for nNOS revealed a significant increase in nNOS expression in basket cell interneurons in both mutant mice. nNOS expression in granule cells in the internal granule cell layer in tottering mice was increased at P12, while granule cells of leaner mice exhibited decreased nNOS expression at P20. The changes in nNOS expression at P12 did not correlate with a change in overall NO production, but rather maintained wild type NO concentrations. These findings suggest that changes in nNOS expression in the leaner and tottering cerebella are compensatory in nature with NO most likely functioning as a calcium-regulated neuroprotective/neurotrophic factor in postnatal cerebellar development.


Subject(s)
Calcium Channels, P-Type/genetics , Calcium Channels, Q-Type/genetics , Cerebellum/enzymology , Gene Expression Regulation, Developmental/physiology , Mice, Neurologic Mutants/metabolism , Nitric Oxide Synthase Type I/metabolism , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Cerebellum/cytology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NADPH Dehydrogenase/metabolism , Neurons/enzymology , Nitric Oxide Synthase Type I/genetics , Phenothiazines
13.
Neurosci Lett ; 413(3): 196-201, 2007 Feb 21.
Article in English | MEDLINE | ID: mdl-17174474

ABSTRACT

Recently, the waddles (wdl) mouse was identified as a carbonic anhydrase VIII (Car8) mutant. The mutation is associated with marked deficiency of Car8, an inositol triphosphate receptor 1-binding protein expressed at high levels in cerebellar Purkinje cells. To help unravel the molecular aberrations contributing to motor dysfunction in wdl mice, cerebellar gene expression profiles were examined in the mutants and their wild-type littermates. Genes involved in signaling, cell division, zinc ion-binding, synapse integrity and plasticity were downregulated in wdl mice. Several of the upregulated genes encode proteins that function in the Golgi apparatus which suggests that Car8 deficiency has important effects on synaptic vesicle formation and transport.


Subject(s)
Carbonic Anhydrase III/deficiency , Cerebellum/metabolism , Gene Expression Profiling , Gene Expression Regulation/physiology , Animals , Mice , Mice, Neurologic Mutants/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods
14.
J Neurosci ; 26(33): 8600-8, 2006 Aug 16.
Article in English | MEDLINE | ID: mdl-16914686

ABSTRACT

Stargazer (stg) mutant mice fail to express stargazin [transmembrane AMPA receptor regulatory protein gamma2 (TARPgamma2)] and consequently experience absence seizure-like thalamocortical spike-wave discharges that pervade the hippocampal formation via the dentate gyrus (DG). As in other seizure models, the dentate granule cells of stg develop elaborate reentrant axon collaterals and transiently overexpress brain-derived neurotrophic factor. We investigated whether GABAergic parameters were affected by the stg mutation in this brain region. GABA(A) receptor (GABAR) alpha4 and beta3 subunits were consistently upregulated, GABAR delta expression appeared to be variably reduced, whereas GABAR alpha1, beta2, and gamma2 subunits and the GABAR synaptic anchoring protein gephyrin were essentially unaffected. We established that the alpha4 betagamma2 subunit-containing, flunitrazepam-insensitive subtype of GABARs, not normally a significant GABAR in DG neurons, was strongly upregulated in stg DG, apparently arising at the expense of extrasynaptic alpha4 betadelta-containing receptors. This change was associated with a reduction in neurosteroid-sensitive GABAR-mediated tonic current. This switch in GABAR subtypes was not reciprocated in the tottering mouse model of absence epilepsy implicating a unique, intrinsic adaptation of GABAergic networks in stg. Contrary to previous reports that suggested that TARPgamma2 is expressed in the dentate, we find that TARPgamma2 was neither detected in stg nor control DG. We report that TARPgamma8 is the principal TARP isoform found in the DG and that its expression is compromised by the stargazer mutation. These effects on GABAergic parameters and TARPgamma8 expression are likely to arise as a consequence of failed expression of TARPgamma2 elsewhere in the brain, resulting in hyperexcitable inputs to the dentate.


Subject(s)
Dentate Gyrus/metabolism , Epilepsy, Absence/genetics , Epilepsy, Absence/metabolism , Mice, Neurologic Mutants/metabolism , Receptors, GABA-A/metabolism , Animals , Calcium Channels/deficiency , Cells, Cultured , Dentate Gyrus/physiopathology , Electrophysiology , Epilepsy, Absence/physiopathology , Mice , Neuronal Plasticity , Protein Isoforms/metabolism , Synapses/metabolism , Tissue Distribution , gamma-Aminobutyric Acid/metabolism
15.
BMC Neurosci ; 7: 59, 2006 Jul 27.
Article in English | MEDLINE | ID: mdl-16872511

ABSTRACT

BACKGROUND: Lurcher mice suffer from a complete Purkinje cell (PC) loss in the first four postnatal weeks. Parallel to this degeneration, GABAergic synapses in the deep cerebellar nuclei (DCN), the major recipient of the inhibitory PC projection, increase synaptic conductance. Here, we further investigated this phenomenon, using real-time RT-PCR to assess GABAA receptor subunit gene expression during PC degeneration. RESULTS: We observed a specific reduction in gamma2 subunit gene expression, while alpha1-5, beta1-2, gamma1,3 and delta subunits were unaffected. We made two further specific findings. First, the difference in gene expression was shown in tissue from DCN only. Neither the hippocampus nor coronal sections through the forebrain showed such effects. Furthermore, the involvement of different levels of corticosterone, a possible humeral trigger for differences in gene expression, could be excluded. Second, like the known potentiation of GABAergic synapses, the gamma2 down-regulation was present only after the onset of degeneration at p14. The difference in gamma2 mRNA expression, however, appeared transient, since it was no longer detectable in adult Lurcher mice. CONCLUSION: In conclusion, the down-regulation of gamma2 subunits may be related to differences in synaptic efficacy and, as such, may reflect the initial phase of adaptive responses of DCN tissue to massive GABAergic deafferentation. Its transient course, however, does not support the idea that modulations in GABAergic transmission are at the basis of the well-known DCN-based functional benefit of Lurcher mice present throughout their life.


Subject(s)
Cerebellar Nuclei/metabolism , Mice, Neurologic Mutants/metabolism , Nerve Degeneration/metabolism , Purkinje Cells/metabolism , RNA, Messenger/metabolism , Receptors, GABA-A/genetics , Aging/metabolism , Animals , Animals, Newborn , Computer Systems , Down-Regulation , Gene Expression , Mice , Protein Isoforms/genetics , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
16.
Brain Res ; 1070(1): 1-14, 2006 Jan 27.
Article in English | MEDLINE | ID: mdl-16405874

ABSTRACT

TPO1 is a member of the AIGP family, a unique group of proteins that contains 11 putative transmembrane domains. Expression of the rat TPO1 gene is upregulated in cultured oligodendrocytes (OLs) during development from pro-oligodendroblasts to postmitotic OLs. However, the distribution of native TPO1 protein in cultured OLs and in the brain has not been elucidated. We investigated the distribution and cellular function of TPO1 in myelinating cells of the nervous system. In mice, TPO1 gene expression was detected in the central (CNS) and peripheral (PNS) nervous systems and was markedly upregulated at postnatal days 10-20, an early phase of myelination in the mouse brain. To investigate TPO1 localization, we generated affinity-purified antibodies to synthetic peptides derived from mouse TPO1. Immunohistochemical analysis showed that TPO1 was expressed in OLs and Schwann cells but not in neurons and astrocytes. Schwann cells from trembler mice, which lack PNS myelin, had significantly decreased TPO1 expression and an altered localization pattern, suggesting that TPO1 is a functional myelin membrane protein. In OL lineage cell cultures, TPO1 was detected in A2B5+ bipolar early progenitors, A2B5+ multipolar Pro-OLs, GalC+ immature OLs and MBP+ mature OLs. The subcellular localization of TPO1 in OL lineage cells was mapped to the GM130+ Golgi in cell bodies and Fyn+ cell processes and myelin-like sheets. Furthermore, TPO1 selectively colocalized with non-phosphorylated Fyn and promoted Fyn autophosphorylation in COS7 cells, suggesting that TPO1 may play a role in myelin formation via Fyn kinase activation in the PNS and CNS.


Subject(s)
Membrane Proteins/physiology , Myelin Sheath/physiology , Nerve Tissue Proteins/physiology , Neurons/physiology , Aging/metabolism , Animals , Animals, Newborn , Antibodies, Monoclonal/pharmacology , Brain/growth & development , Brain/metabolism , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cross-Linking Reagents/pharmacology , Golgi Apparatus/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants/metabolism , Myelin-Associated Glycoprotein/immunology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Peripheral Nerves/growth & development , Peripheral Nerves/metabolism , Phosphorylation , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-fyn/metabolism , Rats , Schwann Cells/metabolism , Tissue Distribution
17.
Neurosci Behav Physiol ; 35(9): 887-90, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16270167

ABSTRACT

The transmitter organization of anomalously formed neocortex was studied in reeler mutant mice by immunohistochemical studies of GABA- and serotoninergic structures and Ca(2+)-binding protein. GABAergic structures were identified in terms of the localization of glutamate decarboxylase (GDC) within them, this being an enzyme involved in GABA synthesis. The neocortex of reeler mutant mice was found to contain an unusual distribution of cells morphologically and chemically identical to Cajal-Retzius cells - beneath layer I rather than in its upper third, as seen in normal animals. GDC-immunoreactive label accumulated in the neuropil of the intermediate and deep layers, layer I containing only occasional granules. Serotonin-immunoreactive fibers did not form superficial or deep plexuses, as seen in normal animals, though they did reach their innervation targets. Thus, the anomalously formed neocortex which lacks the typical cytoarchitectonic organization, showed abnormalities in the structure of both intrinsic and projectional transmitter systems.


Subject(s)
Mice, Neurologic Mutants/metabolism , Neocortex/metabolism , S100 Calcium Binding Protein G/metabolism , Serotonin/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Calbindins , Glutamate Decarboxylase/metabolism , Immunohistochemistry/methods , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants/anatomy & histology
18.
J Comp Neurol ; 491(1): 69-78, 2005 Oct 10.
Article in English | MEDLINE | ID: mdl-16127699

ABSTRACT

Four transverse zones can be distinguished in the adult mouse cerebellar cortex based on differential expression of cell-specific antigens, termination patterns of mossy fiber afferents, and phenotypes of mouse mutants with cerebellar defects: the anterior zone (AZ), central zone (CZ), posterior zone (PZ), and nodular zone (NZ). In the heterozygous Lurcher (Lc/+) mouse a zonally restricted abnormality in Purkinje cell development is seen. The Purkinje cell-specific antigen zebrin II is normally differentially expressed in all four zones of the adult cerebellum, but in the Lc/+ mutant is confined to the PZ and NZ, caudal to a transverse boundary in the dorsal aspect of lobule VIII. In this study we wanted to understand why zebrin II expression is arrested at this boundary and whether the Lc mutation affects the differentiation of additional Purkinje cell antigens in a similar manner. To determine this, we took advantage of the dynamic developmental timetable of another Purkinje cell antigen, the small heat shock protein Hsp25. Using immunohistochemistry we demonstrate that cerebellar maturation anterior to the CZ/PZ transverse boundary appears to be unaffected by the Lc allele, in that initial progression of Hsp25 expression in the Lc/+ cerebellum was similar to controls. Double-labeling experiments with anti-Hsp25 and anti-calbindin suggest that characteristic banding patterns of Hsp25 in Lc/+ cerebellum develop and are preserved despite cell loss. Thus, since simple temporal or spatial models cannot account for the zonal restriction seen during Lc/+ cerebellar development, the abnormality may be zebrin II-specific.


Subject(s)
Cerebellum/growth & development , Heat-Shock Proteins/metabolism , Mice, Neurologic Mutants/metabolism , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Purkinje Cells/metabolism , Animals , Calbindins , Cell Differentiation/genetics , Cell Differentiation/physiology , Cerebellum/cytology , Cerebellum/metabolism , Heterozygote , Immunohistochemistry , Mice , Molecular Chaperones , Purkinje Cells/cytology , S100 Calcium Binding Protein G/metabolism
19.
Cerebellum ; 4(2): 105-11, 2005.
Article in English | MEDLINE | ID: mdl-16035192

ABSTRACT

In the central nervous system, dopamine is known to play a critical role in motor and cognitive functions. Although the cerebellum plays a role in the control of movement and posture and in cognitive functions, it has not been considered to be a dopaminergic region and the dopamine present was thought to represent a precursor of noradrenaline. However, recent evidence suggests that in the cerebellum there is a small dopaminergic element, whose properties are similar to the well characterized system of striatum. In order to better understand the functional role of this system and to delineate its specific interactions within the cerebellum, the distribution and properties of dopamine transporter (DAT) in the cerebellum of reeler and Purkinje cell degeneration (Nna1pcd) mutant mice, which are characterized by severe loss of different cell populations and abnormalities in synapse formation, have been studied. Kinetic studies revealed that [3H]dopamine is transported into cerebellar synaptosomes prepared from normal mice with affinities similar to that into striatal synaptosomes but with much lower maximal velocities. In reeler cerebellar synaptosomes the number of transport sites is significantly reduced. In Nna1pcd cerebellar synaptosomes the kinetic properties of transport of [3H]dopamine are similar to the normal. However, in vitro quantitative DAT autoradiography revealed a significantly increased binding in cerebellar nuclei, a decreased binding in molecular layer and an unaltered binding in the granule cell layer. These observations confirm a dopaminergic innervation of the cerebellum and contribute to our understanding of the intracerebellar distribution of the dopaminergic system.


Subject(s)
Cerebellum/physiology , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/metabolism , Mice, Neurologic Mutants/physiology , Nerve Tissue Proteins/metabolism , Animals , Autoradiography/methods , Cerebellum/cytology , Dopamine Plasma Membrane Transport Proteins , GTP-Binding Proteins/genetics , Mice , Mice, Neurologic Mutants/metabolism , Purkinje Cells/metabolism , Serine-Type D-Ala-D-Ala Carboxypeptidase/genetics
20.
Endocrine ; 26(2): 117-25, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15888923

ABSTRACT

IGF-I is an anabolic growth factor essential for growth and development, both as a mediator of growth hormone (GH) action and as a local stimulator of cell proliferation and differentiation. Although the importance of IGF-I in postnatal growth has been studied for several decades, its functions in pathological states are not fully understood. The weaver (wv) mutant mouse is a commonly used model for studying hereditary cerebellar ataxia and provides us with an opportunity to study the function of IGF-I in postnatal growth during neurodegeneration. In prepubertal wv mice, we found a parallel decrease in body weight and serum IGF-I. This parallel relationship was maintained in females, but not in males, as wv mice entered puberty. Interestingly, we found an increase in the levels of circulating IGF-I and hepatic mRNA preceded the catch-up of body weight of pubertal male wv mice. The increase in IGF-I levels coincided with a surge of circulating androgen at the onset of male puberty, suggesting that androgen might trigger the increase in IGF-I production in the pubertal and adult male wv mice. Overall, our results support the concept that IGF-I plays an important role in postnatal growth during and after neurodegeneration of wv mice. In addition, IGF-I's regulation of systemic growth during and after puberty is likely modulated by androgen in male wv mice.


Subject(s)
Cerebellar Ataxia/metabolism , Insulin-Like Growth Factor Binding Protein 3/biosynthesis , Insulin-Like Growth Factor I/physiology , Mice, Neurologic Mutants/growth & development , Animals , Blotting, Northern , Blotting, Western , Cerebellar Ataxia/physiopathology , Female , Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/genetics , Liver/metabolism , Male , Mice , Mice, Neurologic Mutants/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors , Sexual Maturation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL