Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Genet ; 59(11): 1044-1057, 2022 11.
Article in English | MEDLINE | ID: mdl-35149592

ABSTRACT

BACKGROUND: Heterozygous loss of X-linked genes like CASK and MeCP2 (Rett syndrome) causes developmental delay in girls, while in boys, loss of the only allele of these genes leads to epileptic encephalopathy. The mechanism for these disorders remains unknown. CASK-linked cerebellar hypoplasia is presumed to result from defects in Tbr1-reelin-mediated neuronal migration. METHOD: Here we report clinical and histopathological analyses of a deceased 2-month-old boy with a CASK-null mutation. We next generated a mouse line where CASK is completely deleted (hemizygous and homozygous) from postmigratory neurons in the cerebellum. RESULT: The CASK-null human brain was smaller in size but exhibited normal lamination without defective neuronal differentiation, migration or axonal guidance. The hypoplastic cerebellum instead displayed astrogliosis and microgliosis, which are markers for neuronal loss. We therefore hypothesise that CASK loss-induced cerebellar hypoplasia is the result of early neurodegeneration. Data from the murine model confirmed that in CASK loss, a small cerebellum results from postdevelopmental degeneration of cerebellar granule neurons. Furthermore, at least in the cerebellum, functional loss from CASK deletion is secondary to degeneration of granule cells and not due to an acute molecular functional loss of CASK. Intriguingly, female mice with heterozygous deletion of CASK in the cerebellum do not display neurodegeneration. CONCLUSION: We suggest that X-linked neurodevelopmental disorders like CASK mutation and Rett syndrome are pathologically neurodegenerative; random X-chromosome inactivation in heterozygous mutant girls, however, results in 50% of cells expressing the functional gene, resulting in a non-progressive pathology, whereas complete loss of the only allele in boys leads to unconstrained degeneration and encephalopathy.


Subject(s)
Cerebellar Diseases , Neurodegenerative Diseases , Rett Syndrome , Male , Humans , Animals , Female , Mice , Infant , Genes, X-Linked/genetics , Guanylate Kinases/genetics , Rett Syndrome/genetics , Cerebellar Diseases/genetics , Neurodegenerative Diseases/genetics
2.
Cells ; 10(7)2021 06 22.
Article in English | MEDLINE | ID: mdl-34206213

ABSTRACT

The purpose of this study was to characterize the miRNA profile of purified retinal ganglion cells (RGC) from healthy and diseased rat retina. Diseased retina includes those after a traumatic optic nerve crush (ONC), and after ocular hypertension/glaucoma. Rats were separated into four groups: healthy/intact, 7 days after laser-induced ocular hypertension, 2 days after traumatic ONC, and 7 days after ONC. RGC were purified from rat retina using microbeads conjugated to CD90.1/Thy1. RNA were sequenced using Next Generation Sequencing. Over 100 miRNA were identified that were significantly different in diseased retina compared to healthy retina. Considerable differences were seen in the miRNA expression of RGC 7 days after ONC, whereas after 2 days, few changes were seen. The miRNA profiles of RGC 7 days after ONC and 7 days after ocular hypertension were similar, but discrete miRNA differences were still seen. Candidate mRNA showing different levels of expression after retinal injury were manipulated in RGC cultures using mimics/AntagomiRs. Of the five candidate miRNA identified and subsequently tested for therapeutic efficacy, miR-194 inhibitor and miR-664-2 inhibitor elicited significant RGC neuroprotection, whereas miR-181a mimic and miR-181d-5p mimic elicited significant RGC neuritogenesis.


Subject(s)
Glaucoma/genetics , Glaucoma/pathology , MicroRNAs/metabolism , Nerve Crush , Optic Nerve/pathology , Retinal Ganglion Cells/metabolism , Animals , Female , Gene Expression Regulation , Intraocular Pressure/genetics , Mice, Inbred C57BL , MicroRNAs/genetics , Neurites/metabolism , Neurogenesis/genetics , Neuroprotection/genetics , Ocular Hypertension/genetics , Ocular Hypertension/physiopathology , Rats, Sprague-Dawley
3.
Invest Ophthalmol Vis Sci ; 60(10): 3584-3594, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31425583

ABSTRACT

Purpose: Heterozygous mutations in the essential X-linked gene CASK associate with optic nerve hypoplasia (ONH) and other retinal disorders in girls. CASK+/- heterozygous knockout mice with mosaic CASK expression exhibit ONH with a loss of retinal ganglion cells (RGCs) but no changes in retinal morphology. It remains unclear if CASK deficiency selectively affects RGCs or also affects other retinal cells. Furthermore, it is not known if CASK expression in RGCs is critical for optic nerve (ON) development and maintenance. Methods: The visual behavior of CASK+/- mice was assessed and electroretinography (ERG) was performed. Using a mouse line with a floxed CASK gene that expresses approximately 40% CASK globally in all cells (hypomorph) under hemizygous and homozygous conditions, we investigated effects of CASK reduction on the retina and ON. CASK then was completely deleted from RGCs to examine its cell-autonomous role. Finally, for the first time to our knowledge, we describe a hemizygous CASK missense mutation in a boy with ONH. Results: CASK+/- heterozygous mutant mice display reduced visual contrast sensitivity, but ERG is indistinguishable from wildtype. CASK hypomorph mice exhibit ONH, but deletion of CASK from RGCs in this background does not exacerbate the condition. The boy with ONH harbors a missense mutation (p.Pro673Leu) that destabilizes CASK and weakens the crucial CASK-neurexin interaction. Conclusions: Our results demonstrate that mosaic or global reduction in CASK expression and/or function disproportionately affects RGCs. CASK expression in RGCs does not appear critical for cell survival, indicating a noncell autonomous role for CASK in the development of ON.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Guanylate Kinases/genetics , Optic Nerve Hypoplasia/genetics , Animals , Cell Survival , Child, Preschool , Contrast Sensitivity/physiology , Electroretinography , Female , Humans , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mutation, Missense , Optic Nerve Hypoplasia/physiopathology , Retina/physiopathology , Retinal Ganglion Cells/enzymology
4.
Elife ; 72018 02 09.
Article in English | MEDLINE | ID: mdl-29424692

ABSTRACT

It has long been thought that the mammalian visual system is organized into parallel pathways, with incoming visual signals being parsed in the retina based on feature (e.g. color, contrast and motion) and then transmitted to the brain in unmixed, feature-specific channels. To faithfully convey feature-specific information from retina to cortex, thalamic relay cells must receive inputs from only a small number of functionally similar retinal ganglion cells. However, recent studies challenged this by revealing substantial levels of retinal convergence onto relay cells. Here, we sought to identify mechanisms responsible for the assembly of such convergence. Using an unbiased transcriptomics approach and targeted mutant mice, we discovered a critical role for the synaptic adhesion molecule Leucine Rich Repeat Transmembrane Neuronal 1 (LRRTM1) in the emergence of retinothalamic convergence. Importantly, LRRTM1 mutant mice display impairment in visual behaviors, suggesting a functional role of retinothalamic convergence in vision.


Subject(s)
Neural Cell Adhesion Molecules/metabolism , Retina/anatomy & histology , Retina/physiology , Thalamus/anatomy & histology , Thalamus/physiology , Visual Pathways/anatomy & histology , Visual Pathways/physiology , Animals , Gene Expression Profiling , Membrane Proteins , Mice , Nerve Tissue Proteins , Neural Cell Adhesion Molecules/genetics , Retinal Ganglion Cells/physiology
5.
Invest Ophthalmol Vis Sci ; 58(12): 5485-5496, 2017 10 01.
Article in English | MEDLINE | ID: mdl-29067402

ABSTRACT

Purpose: Optic nerve hypoplasia (ONH) is the most common cause of childhood congenital blindness in developed nations, yet the fundamental pathobiology of ONH remains unknown. The objective of this study was to employ a 'face validated' murine model to determine the timing of onset and the pathologic characteristics of ONH. Methods: Based on the robust linkage between X-linked CASK haploinsufficiency and clinically diagnosed ONH, we hypothesized that heterozygous deletion of CASK (CASK(+/-)) in rodents will produce an optic nerve pathology closely recapitulating ONH. We quantitatively analyzed the entire subcortical visual system in female CASK(+/-) mice using immunohistochemistry, anterograde axonal tracing, toluidine blue staining, transmission electron microscopy, and serial block-face scanning electron microscopy. Results: CASK haploinsuffiency in mice phenocopies human ONH with complete penetrance, thus satisfying the 'face validity'. We demonstrate that the optic nerve in CASK(+/-) mice is not only thin, but is comprised of atrophic retinal axons and displays reactive astrogliosis. Myelination of the optic nerve axons remains unchanged. Moreover, we demonstrate a significant decrease in retinal ganglion cell (RGC) numbers and perturbation in retinothalamic connectivity. Finally, we used this mouse model to define the onset and progression of ONH pathology, demonstrating for the first time that optic nerve defects arise at neonatally in CASK(+/-)mice. Conclusions: Optic nerve hypoplasia is a complex neuropathology of the subcortical visual system involving RGC loss, axonopathy, and synaptopathy and originates at a developmental stage in mice that corresponds to the late third trimester development in humans.


Subject(s)
Optic Nerve Diseases/congenital , Optic Nerve/pathology , Animals , Axons/pathology , Disease Models, Animal , Disease Progression , Guanylate Kinases/genetics , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mutation , Optic Nerve Diseases/genetics , Optic Nerve Diseases/pathology , Retinal Ganglion Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...