Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Int J Mol Sci ; 22(6)2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33804256

ABSTRACT

Lysosomal acid phosphatase 2 (Acp2) mutant mice (naked-ataxia, nax) have a severe cerebellar cortex defect with a striking reduction in the number of granule cells. Using a combination of in vivo and in vitro immunohistochemistry, Western blotting, BrdU assays, and RT-qPCR, we show downregulation of MYCN and dysregulation of the SHH signaling pathway in the nax cerebellum. MYCN protein expression is significantly reduced at P10, but not at the peak of proliferation at around P6 when the number of granule cells is strikingly reduced in the nax cerebellum. Despite the significant role of the SHH-MycN pathway in granule cell proliferation, our study suggests that a broader molecular pathway and additional mechanisms regulating granule cell development during the clonal expansion period are impaired in the nax cerebellum. In particular, our results indicate that downregulation of the protein synthesis machinery may contribute to the reduced number of granule cells in the nax cerebellum.


Subject(s)
Acid Phosphatase/genetics , Cerebellar Ataxia/genetics , Cerebellar Cortex/metabolism , Hedgehog Proteins/genetics , N-Myc Proto-Oncogene Protein/genetics , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cerebellar Ataxia/metabolism , Cerebellar Ataxia/pathology , Cerebellar Cortex/abnormalities , Cerebellar Cortex/pathology , Cytoplasmic Granules/genetics , Cytoplasmic Granules/pathology , Disease Models, Animal , Gene Expression Regulation, Developmental , Humans , Lysosomes/genetics , Lysosomes/pathology , Mice , Mutation , Neurons/metabolism , Neurons/pathology , Purkinje Cells/metabolism , Purkinje Cells/pathology , Signal Transduction/genetics
2.
Cerebellum ; 19(5): 629-635, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32472476

ABSTRACT

Chromosomal microarray analysis is commonly used as screening test for children with neurodevelopmental issues, also in case of complex neurological phenotypes. Developmental delay/intellectual disability is a common presentation sign in pediatric ataxias, diseases with high clinical and genetic heterogeneity. In order to determine the diagnostic yield of Array-CGH in such conditions, all the tests performed in the last 10-year activity of a single referral center in children who present, besides the neurodevelopmental impairment, cerebellar abnormalities have been systematically gathered. The study demonstrates that, except for Dandy-Walker malformation or poly-malformative phenotypes, chromosomal microarray analysis should be discouraged as first-line diagnostic test in pediatric ataxias with neurodevelopmental disability.


Subject(s)
Cerebellar Cortex/abnormalities , Developmental Disabilities/genetics , Intellectual Disability/genetics , Nervous System Malformations/genetics , Child , Child, Preschool , Developmental Disabilities/diagnosis , Diagnostic Tests, Routine/methods , Female , Humans , Infant , Intellectual Disability/diagnosis , Male , Microarray Analysis/methods , Nervous System Malformations/diagnosis
4.
Am J Hum Genet ; 95(2): 227-34, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25105227

ABSTRACT

Cerebellar dysplasia with cysts (CDC) is an imaging finding typically seen in combination with cobblestone cortex and congenital muscular dystrophy in individuals with dystroglycanopathies. More recently, CDC was reported in seven children without neuromuscular involvement (Poretti-Boltshauser syndrome). Using a combination of homozygosity mapping and whole-exome sequencing, we identified biallelic mutations in LAMA1 as the cause of CDC in seven affected individuals (from five families) independent from those included in the phenotypic description of Poretti-Boltshauser syndrome. Most of these individuals also have high myopia, and some have retinal dystrophy and patchy increased T2-weighted fluid-attenuated inversion recovery (T2/FLAIR) signal in cortical white matter. In one additional family, we identified two siblings who have truncating LAMA1 mutations in combination with retinal dystrophy and mild cerebellar dysplasia without cysts, indicating that cysts are not an obligate feature associated with loss of LAMA1 function. This work expands the phenotypic spectrum associated with the lamininopathy disorders and highlights the tissue-specific roles played by different laminin-encoding genes.


Subject(s)
Cerebellar Cortex/abnormalities , Cerebellar Diseases/genetics , Cysts/genetics , Laminin/genetics , Retinal Dystrophies/genetics , Adult , Alleles , Base Sequence , Child , Child, Preschool , Exome/genetics , Female , Humans , Male , Muscular Dystrophies/genetics , Sequence Analysis, DNA , Young Adult
5.
Epilepsy Res ; 105(1-2): 245-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23375885

ABSTRACT

Traditionally, subcortical structures such as the cerebellum are supposed to exert a modulatory effect on epileptic seizures, rather than being the primary seizure generator. We report a 14-month old girl presenting, since birth, with seizures symptomatic of a right cerebellar dysplasia, manifested as paroxystic contralateral hemifacial spasm and ipsilateral facial weakness. Multimodal imaging was used to investigate both anatomical landmarks related to the cerebellar lesion and mechanisms underlying seizure generation. Electric source imaging (ESI) supported the hypothesis of a right cerebellar epileptogenic generator in concordance with nuclear imaging findings; subsequently validated by intra-operative intralesional recordings. Diffusion spectrum imaging-related tractography (DSI) showed severe cerebellar structural abnormalities confirmed by histological examination. We suggest that hemispheric cerebellar lesions in cases like this are likely to cause epilepsy via an effect on the facial nuclei through ipsilateral and contralateral aberrant connections.


Subject(s)
Cerebellar Cortex/abnormalities , Cerebellar Cortex/pathology , Cerebellar Diseases/diagnosis , Epilepsy/diagnosis , Hemifacial Spasm/diagnosis , Adolescent , Cerebellar Diseases/complications , Epilepsy/etiology , Female , Hemifacial Spasm/etiology , Humans
6.
Nucleic Acids Res ; 40(21): e166, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22879379

ABSTRACT

Methods for generating loss-of-function mutations, such as conventional or conditional gene knockout, are widely used in deciphering gene function in vivo. By contrast, inducible and reversible regulation of endogenous gene expression has not been well established. Using a mouse model, we demonstrate that a chimeric transcriptional repressor molecule (tTS) can reversibly inhibit the expression of an endogenous gene, Nmyc. In this system, a tetracycline response element (TRE) artificially inserted near the target gene's promoter region turns the gene on and off in a tetracycline-inducible manner. Nmyc(TRE) mice were generated by inserting a TRE into the first intron of Nmyc by the knockin technique. Nmyc(TRE) mice were crossed to tTS transgenic mice to produce Nmyc(TRE/TRE): tTS embryos. In these embryos, tTS blocked Nmyc expression, and embryonic lethality was observed at E11.5d. When the dam was exposed to drinking water containing doxycycline (dox), normal endogenous Nmyc expression was rescued, and the embryo survived to birth. This novel genetic modification strategy based on the tTS-dox system for inducible and reversible regulation of endogenous mouse genes will be a powerful tool to investigate target genes that cause embryonic lethality or other defects where reversible regulation or temporary shutdown of the target gene is needed.


Subject(s)
Gene Expression Regulation , Gene Targeting/methods , Proto-Oncogene Proteins c-myc/genetics , Animals , Cerebellar Cortex/abnormalities , Cerebellar Cortex/growth & development , Cerebellar Cortex/metabolism , Doxycycline/pharmacology , Gene Expression Regulation/drug effects , Gene Knock-In Techniques , Genes, Lethal , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-myc/metabolism , Repressor Proteins/genetics , Response Elements , Syndactyly/etiology
8.
Brain Res ; 1343: 46-53, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20462503

ABSTRACT

The present study examined the spatial organization of tyrosine hydroxylase (TH) immunopositive Purkinje cells in the cerebellum of rolling mouse Nagoya with reference to the distribution pattern of the cerebellar compartmentation antigen, heat shock protein 25 (HSP25). Whole-mount immunostaining revealed a striking pattern of parasagittal stripes of TH staining in the rolling mouse cerebellum but not in the control cerebellum. Although the TH stripes resembled the zebrin II stripes in the rolling cerebellum, these two distributions did not completely overlap. The TH stripes were present in the lobules VI and VII (central zone), the lobule X (nodular zone), and the paraflocculus, where zebrin II immunostaining was uniformly expressed. Double immunostaining revealed that TH stripes were aligned in an alternative fashion with HSP25 stripes within the caudal half of lobule VIb, lobules IXb and X, and paraflocculus. Some, but not all, TH stripes shared boundaries with HSP25 stripes. These results revealed an alternating array of TH immunopositive Purkinje cell subsets with HSP25 immunopositive Purkinje cells in the zebrin II-defined transverse zone of the rolling mouse cerebellum. The constitutive expression of HSP25 may prevent the ectopic expression of TH in zebrin II immunopositive Purkinje cell subsets.


Subject(s)
Cerebellar Cortex/abnormalities , Cerebellar Cortex/metabolism , Heat-Shock Proteins/metabolism , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Purkinje Cells/metabolism , Tyrosine 3-Monooxygenase/metabolism , Animals , Biomarkers/analysis , Biomarkers/metabolism , Brain Mapping/methods , Calcium Channels/genetics , Calcium Channels/metabolism , Catecholamines/biosynthesis , Cerebellar Cortex/enzymology , Gene Expression Regulation, Developmental/physiology , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/genetics , Mice , Mice, Neurologic Mutants , Molecular Chaperones , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Purkinje Cells/cytology , Purkinje Cells/enzymology , Tyrosine 3-Monooxygenase/biosynthesis , Tyrosine 3-Monooxygenase/genetics
9.
J Neurosci ; 30(8): 2880-7, 2010 Feb 24.
Article in English | MEDLINE | ID: mdl-20181585

ABSTRACT

Opitz G/BBB syndrome (OS) is a genetic disorder characterized by midline developmental defects. Male patients with the X-linked form of OS, caused by loss-of-function mutations in the MID1 gene, show high variability of the clinical signs. MID1 encodes a ubiquitin ligase that controls phosphatase 2A, but its role in the pathogenesis of the disease is still unclear. Here, we report a mouse line carrying a nonfunctional ortholog of the human MID1 gene, Mid1. Mid1-null mice show the brain anatomical defect observed in patients (i.e., hypoplasia of the anterior portion of the medial cerebellum, the vermis). We found that the presence of this defect correlates with motor coordination and procedural and nonassociative learning impairments. The defect is limited to the most anterior lobes of the vermis, the region of the developing cerebellum adjacent to the dorsal midbrain. Analyses at midgestation reveal that lack of Mid1 causes the shortening of the posterior dorsal midbrain, the rostralization of the midbrain/cerebellum boundary, and the downregulation of a key player in the development of this region, Fgf17. Thus, lack of Mid1 causes a misspecification of the midbrain/cerebellar boundary that results in an abnormal development of the most anterior cerebellar lobes. This animal model provides a tool for additional in vivo studies of the physiological and pathological role of the Mid1 gene and a system to investigate the development and function of anterior cerebellar domains.


Subject(s)
Cerebellar Cortex/abnormalities , Cerebellar Cortex/metabolism , Gene Expression Regulation, Developmental/genetics , Nervous System Malformations/genetics , Nervous System Malformations/metabolism , Proteins/genetics , Animals , Cerebellar Cortex/cytology , Cerebellar Diseases/genetics , Cerebellar Diseases/metabolism , Cerebellar Diseases/physiopathology , Female , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Learning Disabilities/genetics , Learning Disabilities/metabolism , Learning Disabilities/physiopathology , Male , Mesencephalon/abnormalities , Mice , Mice, Inbred C57BL , Mice, Knockout , Movement Disorders/genetics , Movement Disorders/metabolism , Movement Disorders/physiopathology , Nervous System Malformations/physiopathology , Syndrome , Ubiquitin-Protein Ligases
10.
J Comp Neurol ; 513(1): 98-112, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19107796

ABSTRACT

Transcription factors of the Nuclear Factor I (Nfi) family are important for the development of specific neuronal and glial populations in the nervous system. One such population, the neurons of the basilar pontine nuclei, expresses high levels of Nfi proteins, and the pontine nuclei are greatly reduced in mice lacking a functional Nfib gene. Pontine neurons, along with other precerebellar neurons that populate the hindbrain, arise from precursors in the lower rhombic lip and migrate anteroventrally to reach their final location. Using immunohistochemistry, we find that NFI-B expression is specific for mossy fiber populations of the precerebellar system. Analysis of the Nfib(-/-) hindbrain indicates that the development of the basilar pontine nuclei is delayed, with pontine neurons migrating 1-2 days later than in control animals, and that significantly fewer pontine neurons are produced. While the mossy fiber nuclei of the caudal medulla do form, they also exhibit a developmental delay. Nfia and Nfix null mice exhibit no apparent pontine phenotype, implying specificity in the action of NFI family members. Collectively, these data demonstrate that Nfib plays an important role in the generation of precerebellar mossy fiber neurons, and may do so at least in part by regulating neurogenesis.


Subject(s)
Cerebellar Cortex , NFI Transcription Factors/metabolism , Neural Pathways , Pons , Animals , Cerebellar Cortex/abnormalities , Cerebellar Cortex/anatomy & histology , Cerebellar Cortex/embryology , Eye Proteins/genetics , Eye Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NFI Transcription Factors/genetics , Neural Pathways/abnormalities , Neural Pathways/anatomy & histology , Neural Pathways/embryology , Neurons/cytology , Neurons/physiology , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Phenotype , Pons/abnormalities , Pons/anatomy & histology , Pons/embryology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Rhombencephalon/abnormalities , Rhombencephalon/anatomy & histology , Rhombencephalon/embryology , Rhombencephalon/metabolism
11.
Brain Res ; 1238: 181-8, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18755166

ABSTRACT

Ts65Dn mice are a genetic model for Down syndrome. Among others, these mice have cerebellar pathology features which parallel those seen in Down syndrome patients. Both individuals with Down syndrome and Ts65Dn mice have reduced cerebellar volume and numbers of granule and Purkinje cells. In this report, we describe morphological abnormalities of axons of Purkinje cells in the cerebellum of Ts65Dn mice, by using anti-calbindin immunocytochemistry. A consistent number of Purkinje cells shows axons bearing giant varicosities along their transit through the granular layer. The cerebellar arbor vitae made by fasciculated Purkinje cell axons has a patchy appearance, some tracks being devoid of calbindin staining. The infraganglionic plexus, formed by recurrent collaterals of Purkinje cell axons, has enormously increased density, which is evidence for a compensatory reaction to degeneration of distal segments of axons. These alterations are accompanied by strong glial reaction as evidenced by GFAP immunocytochemistry. Moreover, the alterations are more consistent in the anterior lobules of the vermis and intermediate cortex. The axonal pathology of Purkinje cells may explain the impairment in cerebellar functions observed in Ts65Dn mice at the adulthood.


Subject(s)
Axons/pathology , Cerebellar Cortex/abnormalities , Down Syndrome/pathology , Purkinje Cells/pathology , Animals , Disease Models, Animal , Female , Image Processing, Computer-Assisted , Immunohistochemistry , Mice
12.
Dev Neurobiol ; 67(7): 909-23, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17506494

ABSTRACT

Myosin Va is an actin-based molecular motor that is involved in organelle transport and membrane trafficking. Here, we explored the role of myosin Va in the formation of synaptic circuitry by examining climbing fiber (CF) innervation of Purkinje cells (PCs) in the cerebella of dilute-neurological (d-n) mice and dilute-opisthotonus (dop) rats that have mutations in dilute-encoded myosin Va. Anterograde labeling of CFs with biotinylated dextran amine (BDA) revealed that they arborized poorly and that their tips extended only half way through the thickness of the molecular layer (ML) in adult d-n mice. Using immunohistochemistry specific for vesicular glutamate transporter 2 (VGluT2) to visualize CF synaptic terminals, we found that during development and in adulthood, these terminals did not ascend as far along the proximal shaft dendrites of PCs in d-n mice and dop rats as they did in normal animals. An irregular distribution of BDA-labeled bulbous varicosities and VGluT2 spots along CF branches were also noted in these animals. Finally, VGluT2-positive CF terminals were occasionally localized on the PC somata of adult d-n cerebella. These phenotypes are consistent with our electrophysiological findings that CF-mediated excitatory postsynaptic currents (EPSCs) were significantly smaller in amplitude and faster in decay in adult d-n mice, and that the regression of multiple CFs was slightly delayed in developing d-n mice. Taken together, our results suggest that myosin Va is essential for terminal CF extension and for the establishment of CF synapses within the proper dendritic territories of PCs.


Subject(s)
Afferent Pathways/abnormalities , Cerebellar Cortex/abnormalities , Myosin Heavy Chains/genetics , Myosin Type V/genetics , Nervous System Malformations/metabolism , Olivary Nucleus/abnormalities , Purkinje Cells/metabolism , Afferent Pathways/cytology , Afferent Pathways/metabolism , Animals , Biotin/analogs & derivatives , Calcium Signaling/physiology , Cell Differentiation/genetics , Cerebellar Cortex/cytology , Cerebellar Cortex/metabolism , Dextrans , Excitatory Postsynaptic Potentials/genetics , Female , Male , Mice , Mice, Mutant Strains , Microscopy, Electron, Transmission , Nervous System Malformations/genetics , Nervous System Malformations/physiopathology , Olivary Nucleus/cytology , Olivary Nucleus/metabolism , Presynaptic Terminals/metabolism , Presynaptic Terminals/pathology , Protein Transport/physiology , Purkinje Cells/pathology , Rats , Rats, Mutant Strains , Synaptic Transmission/genetics
13.
J Physiol ; 582(Pt 2): 629-46, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17463037

ABSTRACT

TrkB, the cognate receptor for brain-derived neurotrophic factor and neurotrophin-4, has been implicated in regulating synapse formation in the central nervous system. Here we asked whether TrkB plays a role in the maturation of the climbing fibre-Purkinje cell (CF-PC) synapse. In rodent cerebellum, Purkinje cells are initially innervated by multiple climbing fibres that are subsequently culled to assume the mature mono-innervated state, and whose contacts translocate from the soma to the dendrites. By employing transgenic mice hypomorphic or null for TrkB expression, our results indicated that perturbation of TrkB in the immature cerebellum resulted in ataxia, that Purkinje cells remained multiply innervated by climbing fibres beyond the normal developmental time frame, and that synaptic transmission at the parallel fibre-Purkinje cell synapse remained functionally unaltered. Mechanistically, we present evidence that attributes the persistence of multiple climbing fibre innervation to an obscured discrimination of relative strengths among competing climbing fibres. Soma-to-dendrite translocation of climbing fibre terminals was unaffected. Thus, TrkB regulates pruning but not translocation of nascent CF-PC synaptic contacts.


Subject(s)
Animals, Newborn/physiology , Cerebellum/physiology , Nerve Fibers/physiology , Purkinje Cells/physiology , Receptor, trkB/physiology , Synapses/physiology , Animals , Ataxia/etiology , Ataxia/physiopathology , Cerebellar Cortex/abnormalities , In Vitro Techniques , Mice , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Motor Activity , Nerve Fibers/ultrastructure , Postural Balance , Receptor, trkB/deficiency , Signal Transduction/physiology , Synaptic Transmission/physiology
14.
FASEB J ; 21(9): 2215-25, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17371797

ABSTRACT

Medulloblastoma, the most common brain tumor in childhood, appears to originate from cerebellar granule cell precursors (GCPs), located in the external granular layer (EGL) of the cerebellum. The antiproliferative gene PC3 (Tis21/BTG2) promotes cerebellar neurogenesis by inducing GCPs to shift from proliferation to differentiation. To assess whether PC3 can prevent the neoplastic transformation of GCPs and medulloblastoma development, we crossed transgenic mice conditionally expressing PC3 (TgPC3) in GCPs with Patched1 heterozygous mice (Ptc(+/-)), a model of medulloblastoma pathogenesis characterized by hyperactivation of the Sonic Hedgehog pathway. Perinatal up-regulation of PC3 in Ptc(+/-)/TgPC3 mice results in a decrease of medulloblastoma incidence of approximately 40% and in a marked reduction of preneoplastic abnormalities, such as hyperplastic EGL areas and lesions. Moreover, overexpression of cyclin D1, hyperproliferation, and defective differentiation--observed in Ptc(+/-) GCPs--are restored to normality in Ptc(+/-)/TgPC3 mice. The PC3-mediated inhibition of cyclin D1 expression correlates with recruitment of PC3 to the cyclin D1 promoter, which is accompanied by histone deacetylation. Remarkably, down-regulation of PC3 is observed in preneoplastic lesions, as well as in human and murine medulloblastomas. As a whole, this indicates that PC3 may prevent medulloblastoma development by controlling cell cycle and promoting differentiation of GCPs.


Subject(s)
Cerebellar Neoplasms/prevention & control , Genes, Tumor Suppressor , Immediate-Early Proteins/physiology , Medulloblastoma/prevention & control , Acetylation , Animals , Basal Cell Nevus Syndrome/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Division/genetics , Cell Division/physiology , Cell Transformation, Neoplastic/genetics , Cerebellar Cortex/abnormalities , Cerebellar Cortex/embryology , Cerebellar Neoplasms/genetics , Choristoma/genetics , Chromatin Immunoprecipitation , Cyclin D , Cyclins/biosynthesis , Cyclins/genetics , Hedgehog Proteins/physiology , Heterozygote , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Immediate-Early Proteins/genetics , Medulloblastoma/genetics , Mice , Mice, Transgenic , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/prevention & control , Neurons/pathology , PC12 Cells/chemistry , Patched Receptors , Patched-1 Receptor , Precancerous Conditions/genetics , Promoter Regions, Genetic , Protein Processing, Post-Translational , RNA, Neoplasm/genetics , Rats , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/physiology , Tumor Suppressor Proteins
15.
Folia Neuropathol ; 44(4): 327-32, 2006.
Article in English | MEDLINE | ID: mdl-17183460

ABSTRACT

The object of our report is the presentation of the morphological picture of cerebellar cortex malformation as a sequel of disturbed neuronal migration. In the disarranged tissue, cavities with a network of meningeal tissue and embedded pathological vessels were noted. The external granule cells did not form a proper external granule layer, but moved deeper, forming irregular aggregates. Abnormally aggregated external granular cells invaded the cerebellar tissue. Abnormal Purkinje cell positioning and a disarranged molecular layer were observed. The normal layered pattern of the cerebellar cortex was disorganized. The presented cases represent a spectrum of morphological changes which are the consequence of aberrant migration. Against a background of vascular pathology affecting the meningoglial network the migration pathways were disrupted. The defective movement of neurons and their faulty maturation resulted in disturbances of cortical layering, and defects of cerebellar folia formation.


Subject(s)
Cell Movement , Cerebellar Cortex/abnormalities , Cerebellar Cortex/physiopathology , Neurons/pathology , Cerebellar Cortex/pathology , Congenital Abnormalities/pathology , Congenital Abnormalities/physiopathology , Female , Humans , Infant, Newborn , Male , Stillbirth
16.
Mol Cell Neurosci ; 33(2): 109-25, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16914328

ABSTRACT

We demonstrate here that integrin-linked kinase (ILK), a serine/threonine kinase that binds to the beta1 integrin cytoplasmic domain, regulates cerebellar development. Mice with a CNS-restricted knock-out of the Ilk gene show perturbations in the laminar structure of the cerebellar cortex that are associated with defects in Bergmann glial fibers and the formation of meningeal basement membranes. Similar defects have been observed in mice lacking beta1 integrins in the CNS. ILK and beta1 integrins are coexpressed in Bergmann glial cells, and studies with primary cells in culture demonstrate that ILK and CDC42 are required for beta1-integrin-dependent glial process outgrowth. Consistent with these findings, the amount of GTP-bound CDC42 is impaired in the cerebellum of Ilk-deficient mice. We conclude that beta1 integrin, ILK and CDC42 are components of the signaling machinery that regulates glial process outgrowth in the cerebellum. We also show that granule cell precursor proliferation is affected in ILK-deficient mice, but our findings provide strong evidence that proliferative defects are a secondary consequence of ILK function in glia.


Subject(s)
Cerebellar Cortex/cytology , Cerebellar Cortex/growth & development , Neuroglia/cytology , Neuroglia/enzymology , Protein Serine-Threonine Kinases/metabolism , Actins/metabolism , Animals , Basement Membrane/enzymology , Cell Differentiation/physiology , Cell Movement/physiology , Cerebellar Cortex/abnormalities , Extracellular Matrix/enzymology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Integrin beta1/metabolism , Meninges/abnormalities , Meninges/cytology , Meninges/growth & development , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Purkinje Cells/cytology , Purkinje Cells/enzymology , Stem Cells/cytology , Stem Cells/enzymology , cdc42 GTP-Binding Protein/metabolism
18.
J Comp Neurol ; 497(4): 622-35, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16739195

ABSTRACT

The Lurcher mutation in the Grid2 gene causes the cell autonomous death of virtually all cerebellar Purkinje cells and the target-related death of 90% of the granule cells and 60-75% of the olivary neurons. Inactivation of Bax, a pro-apoptotic gene of the Bcl-2 family, in heterozygous Lurcher mutants (Grid2Lc/+) rescues approximately 60% of the granule cells, but does not rescue Purkinje or olivary neurons. Given the larger size of the cerebellar molecular layer in Grid2Lc/+;Bax(-/-) double mutants compared to Grid2Lc/+ mutants, we analyzed the survival of the stellate and basket interneurons as well as the synaptic connectivity of parallel fibers originating from the surviving granule cells in the absence of their Purkinje cell targets in the Grid2Lc/+;Bax(-/-) cerebellum. Quantification showed a significantly higher density of interneurons ( approximately 60%) in the molecular layer of the Grid2Lc/+;Bax(-/-) mice compared to Grid2Lc/+, suggesting that interneurons are subject to a BAX-dependent target-related death in the Lurcher mutants. Furthermore, electron microscopy showed the normal ultrastructural aspect of a number of parallel fibers in the molecular layer of the Grid2Lc/+; Bax(-/-) double mutant mice and preserved their numerous synaptic contacts on interneurons, suggesting that interneurons could play a trophic role for axon terminals of surviving granule cells. Finally, parallel fibers varicosities in the double mutant established "pseudo-synapses" on glia as well as displayed autophagic profiles, suggesting that the connections established by the parallel fibers in the absence of their Purkinje cell targets were subject to a high turnover involving autophagy.


Subject(s)
Cerebellar Cortex/abnormalities , Interneurons/metabolism , Purkinje Cells/metabolism , Receptors, Glutamate/genetics , Synapses/metabolism , bcl-2-Associated X Protein/genetics , Animals , Apoptosis/genetics , Autophagy/genetics , Cell Communication/genetics , Cell Count , Cell Differentiation/genetics , Cell Proliferation , Cell Survival/genetics , Cerebellar Cortex/cytology , Cerebellar Cortex/metabolism , Female , Gene Expression Regulation, Developmental/genetics , Interneurons/ultrastructure , Male , Mice , Mice, Knockout , Mice, Neurologic Mutants , Microscopy, Electron, Transmission , Nerve Growth Factors/metabolism , Neuroglia/metabolism , Neuroglia/ultrastructure , Purkinje Cells/ultrastructure , Synapses/ultrastructure , Up-Regulation/genetics
19.
J Neurosci ; 26(12): 3245-55, 2006 Mar 22.
Article in English | MEDLINE | ID: mdl-16554475

ABSTRACT

Synapse formation and maintenance require extensive transsynaptic interactions involving multiple signal transduction pathways. In the cerebellum, Purkinje cells (PCs) receive GABAergic, axo-dendritic synapses from stellate cells and axo-somatic synapses from basket cells, both with GABAA receptors containing the alpha1 subunit. Here, we investigated the effects of a targeted deletion of the alpha1 subunit gene on GABAergic synaptogenesis in PCs, using electrophysiology and immunoelectron microscopy. Whole-cell patch-clamp recordings in acute slices revealed that PCs from alpha1(0/0) mice lack spontaneous and evoked IPSCs, demonstrating that assembly of functional GABAA receptors requires the alpha1 subunit. Ultrastructurally, stellate cell synapses on PC dendrites were reduced by 75%, whereas basket cell synapses on the soma were not affected, despite the lack of GABAA-mediated synaptic transmission. Most strikingly, GABAergic terminals were retained in the molecular layer of adult alpha1(0/0) mice and formed heterologous synapses with PC spines characterized by a well differentiated asymmetric postsynaptic density. These synapses lacked presynaptic glutamatergic markers and postsynaptic AMPA-type glutamate receptors but contained delta2-glutamate receptors. During postnatal development, initial steps of GABAergic synapse formation were qualitatively normal, and heterologous synapses appeared in parallel with maturation of dendritic spines. These results suggest that synapse formation in the cerebellum is governed by neurotransmitter-independent mechanisms. However, in the absence of GABAA-mediated transmission, GABAergic terminals in the molecular layer apparently become responsive to synaptogenic signals from PC spines and form stable heterologous synapses. In contrast, maintenance of axo-somatic GABAergic synapses does not depend on functional GABAA receptors, suggesting differential regulation in distinct subcellular compartments.


Subject(s)
Cerebellar Cortex/abnormalities , Dendrites/metabolism , Purkinje Cells/metabolism , Receptors, GABA-A/genetics , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Cell Compartmentation/drug effects , Cell Compartmentation/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cerebellar Cortex/metabolism , Cerebellar Cortex/ultrastructure , Dendrites/drug effects , Dendrites/ultrastructure , Excitatory Amino Acid Antagonists/pharmacology , Female , Fluorescent Antibody Technique , GABA Antagonists/pharmacology , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Neural Inhibition/drug effects , Neural Inhibition/genetics , Organ Culture Techniques , Patch-Clamp Techniques , Purkinje Cells/drug effects , Purkinje Cells/ultrastructure , Synapses/drug effects , Synapses/ultrastructure , Synaptic Membranes/drug effects , Synaptic Membranes/genetics , Synaptic Membranes/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/genetics
20.
J Neuropathol Exp Neurol ; 64(12): 1118-26, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16319722

ABSTRACT

Fukuyama-type congenital muscular dystrophy (FCMD) is characterized by muscular dystrophy and cortical dysgenesis of the cerebrum and cerebellum. We investigated the extent and nature of tauopathy in the brains of 7 postfetal (14-34 years of age) and 2 fetal (18- and 20-week gestational age) FCMD cases. In all postfetal cases, tauopathy was found in the areas of cortical dysgenesis in the cerebrum, in addition to predictable sites such as the hippocampus. In fetal cases, the neuropil of malformed cerebral cortex was diffusely immunostained with anti-aberrantly phosphorylated tau antibodies. By immunoelectron microscopy, the epitope of the antibodies was associated with microtubule-like bundles within cellular processes protruding through disrupted glia limitans. In Western blot analysis, a unique 50-kDa band of tau was detected in a fetal and a postfetal case. In addition, 3 to 4 tau bands of 60 to 68 kD, similar to tau in Alzheimer disease, were also detected in the latter. After dephosphorylation, the insoluble tau from the fetal and the postfetal cases showed highly similar immunoblotting patterns. This anomalous phosphorylation of tau may be related to the development of the cortical dysgenesis in FCMD and may shed light on the biologic function of tau in the development of the central nervous system.


Subject(s)
Cerebellar Cortex/abnormalities , Cerebral Cortex/abnormalities , Muscular Dystrophies/congenital , Muscular Dystrophies/pathology , Tauopathies/congenital , Tauopathies/pathology , Adolescent , Adult , Blotting, Western , Female , Fetus , Humans , Immunohistochemistry , Male , Microscopy, Electron , Microscopy, Immunoelectron , Muscular Dystrophies/complications , Muscular Dystrophies/metabolism , Tauopathies/metabolism , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...