Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Acta Neuropathol ; 147(1): 13, 2024 Jan 09.
Article En | MEDLINE | ID: mdl-38194050

The development of the cerebral cortex involves a series of dynamic events, including cell proliferation and migration, which rely on the motor protein dynein and its regulators NDE1 and NDEL1. While the loss of function in NDE1 leads to microcephaly-related malformations of cortical development (MCDs), NDEL1 variants have not been detected in MCD patients. Here, we identified two patients with pachygyria, with or without subcortical band heterotopia (SBH), carrying the same de novo somatic mosaic NDEL1 variant, p.Arg105Pro (p.R105P). Through single-cell RNA sequencing and spatial transcriptomic analysis, we observed complementary expression of Nde1/NDE1 and Ndel1/NDEL1 in neural progenitors and post-mitotic neurons, respectively. Ndel1 knockdown by in utero electroporation resulted in impaired neuronal migration, a phenotype that could not be rescued by p.R105P. Remarkably, p.R105P expression alone strongly disrupted neuronal migration, increased the length of the leading process, and impaired nucleus-centrosome coupling, suggesting a failure in nucleokinesis. Mechanistically, p.R105P disrupted NDEL1 binding to the dynein regulator LIS1. This study identifies the first lissencephaly-associated NDEL1 variant and sheds light on the distinct roles of NDE1 and NDEL1 in nucleokinesis and MCD pathogenesis.


Lissencephaly , Humans , Lissencephaly/genetics , Cell Movement/genetics , Cell Proliferation , Cerebral Cortex , Dyneins/genetics , Carrier Proteins , Microtubule-Associated Proteins/genetics
2.
Development ; 151(2)2024 Jan 15.
Article En | MEDLINE | ID: mdl-38149472

Lissencephaly is a neurodevelopmental disorder characterized by a loss of brain surface convolutions caused by genetic variants that disrupt neuronal migration. However, the genetic origins of the disorder remain unidentified in nearly one-fifth of people with lissencephaly. Using whole-exome sequencing, we identified a de novo BAIAP2 variant, p.Arg29Trp, in an individual with lissencephaly with a posterior more severe than anterior (P>A) gradient, implicating BAIAP2 as a potential lissencephaly gene. Spatial transcriptome analysis in the developing mouse cortex revealed that Baiap2 is expressed in the cortical plate and intermediate zone in an anterior low to posterior high gradient. We next used in utero electroporation to explore the effects of the Baiap2 variant in the developing mouse cortex. We found that Baiap2 knockdown caused abnormalities in neuronal migration, morphogenesis and differentiation. Expression of the p.Arg29Trp variant failed to rescue the migration defect, suggesting a loss-of-function effect. Mechanistically, the variant interfered with the ability of BAIAP2 to localize to the cell membrane. These results suggest that the functions of BAIAP2 in the cytoskeleton, cell morphogenesis and migration are important for cortical development and for the pathogenesis of lissencephaly in humans.


Lissencephaly , Animals , Humans , Mice , Brain/metabolism , Cell Movement/genetics , Cytoskeleton/metabolism , Lissencephaly/genetics , Lissencephaly/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism
3.
Neuropathol Appl Neurobiol ; 49(2): e12890, 2023 04.
Article En | MEDLINE | ID: mdl-36765387

AIMS: Muscleblind-like 2 (MBNL2) plays a crucial role in regulating alternative splicing during development and mouse loss of MBNL2 recapitulates brain phenotypes in myotonic dystrophy (DM). However, the mechanisms underlying DM neuropathogenesis during brain development remain unclear. In this study, we aim to investigate the impact of MBNL2 elimination on neuronal development by Mbnl2 conditional knockout (CKO) mouse models. METHODS: To create Mbnl2 knockout neurons, cDNA encoding Cre-recombinase was delivered into neural progenitors of Mbnl2flox/flox mouse brains by in utero electroporation. The morphologies and dynamics of dendritic spines were monitored by confocal and two-photon microscopy in brain slices and live animals from the neonatal period into adulthood. To investigate the underlying molecular mechanism, we further detected the changes in the splicing and molecular interactions of proteins associated with spinogenesis. RESULTS: We found that Mbnl2 knockout in cortical neurons decreased dendritic spine density and dynamics in adolescent mice. Mbnl2 ablation caused the adducin 1 (ADD1) isoform to switch from adult to fetal with a frameshift, and the truncated ADD1 failed to interact with alpha-II spectrin (SPTAN1), a critical protein for spinogenesis. In addition, expression of ADD1 adult isoform compensated for the reduced dendritic spine density in cortical neurons deprived of MBNL2. CONCLUSION: MBNL2 plays a critical role in maintaining the dynamics and homeostasis of dendritic spines in the developing brain. Mis-splicing of downstream ADD1 may account for the alterations and contribute to the DM brain pathogenesis.


Dendritic Spines , Myotonic Dystrophy , Animals , Mice , Brain/pathology , Dendritic Spines/metabolism , Dendritic Spines/pathology , Myotonic Dystrophy/genetics , Protein Isoforms/metabolism
4.
Methods Mol Biol ; 2623: 61-71, 2023.
Article En | MEDLINE | ID: mdl-36602679

During development of the cerebral cortex, neuroepithelial and radial glial cells undergo an oscillatory nuclear movement throughout their cell cycle, termed interkinetic nuclear migration. The nucleus of postmitotic neurons derived from these neural stem cells also translocates in a saltatory manner to enable neuronal migration toward the cortical plate. In these processes, various molecular motors, including cytoplasmic dynein, myosin II, and kinesins, are the driving force for nuclear migration at different stages. Despite efforts made to understand the mechanism regulating cortical development over decades, novel gene mutations discovered in neurodevelopmental disorders indicate that missing pieces still remain. Gene manipulation by in utero electroporation combined with live microscopy of neural stem cells in brain slices provides a powerful method to capture their detailed behaviors during proliferation and migration. The procedures described in this chapter enable the monitoring of cell cycle progression, mitosis, morphological changes, and migratory patterns in situ. This approach facilitates the elucidation of gene functions in cortical development and neurodevelopmental disorders.


Dyneins , Neural Stem Cells , Dyneins/genetics , Dyneins/metabolism , Microscopy , Cerebral Cortex , Neurons/metabolism , Cell Movement/physiology , Electroporation/methods
5.
Front Neurosci ; 16: 844410, 2022.
Article En | MEDLINE | ID: mdl-35422684

Expansion of the neocortex is thought to pave the way toward acquisition of higher cognitive functions in mammals. The highly conserved Notch signaling pathway plays a crucial role in this process by regulating the size of the cortical progenitor pool, in part by controlling the balance between self-renewal and differentiation. In this review, we introduce the components of Notch signaling pathway as well as the different mode of molecular mechanisms, including trans- and cis-regulatory processes. We focused on the recent findings with regard to the expression pattern and levels in regulating neocortical formation in mammals and its interactions with other known signaling pathways, including Slit-Robo signaling and Shh signaling. Finally, we review the functions of Notch signaling pathway in different species as well as other developmental process, mainly somitogenesis, to discuss how modifications to the Notch signaling pathway can drive the evolution of the neocortex.

6.
Acta Neuropathol Commun ; 8(1): 106, 2020 07 14.
Article En | MEDLINE | ID: mdl-32665036

During brain development, the nucleus of migrating neurons follows the centrosome and translocates into the leading process. Defects in these migratory events, which affect neuronal migration, cause lissencephaly and other neurodevelopmental disorders. However, the mechanism of nuclear translocation remains elusive. Using whole exome sequencing (WES), we identified a novel nonsense BICD2 variant p.(Lys775Ter) (K775X) from a lissencephaly patient. Interestingly, most BICD2 missense variants have been associated with human spinal muscular atrophy (SMA) without obvious brain malformations. By in utero electroporation, we showed that BicD2 knockdown in mouse embryos inhibited neuronal migration. Surprisingly, we observed severe blockage of neuronal migration in cells overexpressing K775X but not in those expressing wild-type BicD2 or SMA-associated missense variants. The centrosome of the mutant was, on average, positioned farther away from the nucleus, indicating a failure in nuclear translocation without affecting the centrosome movement. Furthermore, BicD2 localized at the nuclear envelope (NE) through its interaction with NE protein Nesprin-2. K775X variant disrupted this interaction and further interrupted the NE recruitment of BicD2 and dynein. Remarkably, fusion of BicD2-K775X with NE-localizing domain KASH resumed neuronal migration. Our results underscore impaired nuclear translocation during neuronal migration as an important pathomechanism of lissencephaly.


Brain/abnormalities , Cell Movement/genetics , Lissencephaly/genetics , Microtubule-Associated Proteins/genetics , Neurogenesis/genetics , Neurons , Animals , Brain/embryology , Child, Preschool , Codon, Nonsense , Dyneins/metabolism , Humans , Male , Mice , Neurons/metabolism , Neurons/pathology
7.
Aging Cell ; 19(5): e13107, 2020 05.
Article En | MEDLINE | ID: mdl-32233025

In mammals, microRNAs can be actively secreted from cells to blood. miR-29b-3p has been shown to play a pivotal role in muscle atrophy, but its role in intercellular communication is largely unknown. Here, we showed that miR-29b-3p was upregulated in normal and premature aging mouse muscle and plasma. miR-29b-3p was also upregulated in the blood of aging individuals, and circulating levels of miR-29b-3p were negatively correlated with relative appendicular skeletal muscle. Consistently, miR-29b-3p was observed in exosomes isolated from long-term differentiated atrophic C2C12 cells. When C2C12-derived miR-29b-3p-containing exosomes were uptaken by neuronal SH-SY5Y cells, increased miR-29b-3p levels in recipient cells were observed. Moreover, miR-29b-3p overexpression led to downregulation of neuronal-related genes and inhibition of neuronal differentiation. Interestingly, we identified HIF1α-AS2 as a novel c-FOS targeting lncRNA that is induced by miR-29b-3p through down-modulation of c-FOS and is required for miR-29b-3p-mediated neuronal differentiation inhibition. Our results suggest that atrophy-associated circulating miR-29b-3p may mediate distal communication between muscle cells and neurons.


Exosomes/metabolism , MicroRNAs/genetics , Muscle Fibers, Skeletal/metabolism , Muscular Atrophy/metabolism , Neurons/metabolism , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Animals , Cell Differentiation , Cells, Cultured , Cellular Senescence , Humans , Mice
8.
Epilepsia ; 60(5): 807-817, 2019 05.
Article En | MEDLINE | ID: mdl-30980674

OBJECTIVE: Variants in human PRRT2 cause paroxysmal kinesigenic dyskinesia (PKD) and other neurological disorders. Most reported variants resulting in truncating proteins failed to localize to cytoplasmic membrane. The present study identifies novel PRRT2 variants in PKD and epilepsy patients and evaluates the functional consequences of PRRT2 missense variations. METHODS: We investigated two families with PKD and epilepsies using Sanger sequencing and a multiple gene panel. Subcellular localization of mutant proteins was investigated using confocal microscopy and cell surface biotinylation assay in Prrt2-transfected cells. RESULTS: Two novel PRRT2 variants, p.His232Glnfs*10 and p.Leu298Pro, were identified, and functional study revealed impaired localization of both mutant proteins to the plasma membrane. Further investigation of other reported missense variants revealed decreased protein targeting to the plasma membrane in eight of the 13 missense variants examined (p.Trp281Arg, p.Ala287Thr, p.Ala291Val, p.Arg295Gln, p.Leu298Pro, p.Ala306Asp, p.Gly324Glu, and p.Gly324Arg). In contrast, all benign variants we tested exhibited predominant localization to the plasma membrane similar to wild-type Prrt2. Most likely pathogenic variants were located at conserved amino acid residues near the C-terminus, whereas truncating variants spread throughout the gene. SIGNIFICANCE: PRRT2 missense variants clustering at the C-terminus often lead to protein mislocalization. Failure in protein targeting to the plasma membrane by PRRT2 variants may be a key mechanism in causing PKD and related neurological disorders.


Dystonia/genetics , Membrane Proteins/genetics , Mutation, Missense , Nerve Tissue Proteins/genetics , Adult , Amino Acid Sequence , Animals , Biotinylation , Cell Membrane/metabolism , Conserved Sequence , Dystonia/metabolism , Female , HEK293 Cells , Humans , Male , Membrane Proteins/metabolism , Microscopy, Confocal , Nerve Tissue Proteins/metabolism , Polymorphism, Genetic , Protein Domains , Protein Transport , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Subcellular Fractions/chemistry , Taiwan , Transfection , Vertebrates/genetics , Young Adult
9.
Mol Neurobiol ; 56(9): 6095-6105, 2019 Sep.
Article En | MEDLINE | ID: mdl-30721447

Mutations in RAB18, a member of small G protein, cause Warburg micro syndrome (WARBM), whose clinical features include vision impairment, postnatal microcephaly, and lower limb spasticity. Previously, our Rab18-/- mice exhibited hind limb weakness and spasticity as well as signs of axonal degeneration in the spinal cord and lumbar spinal nerves. However, the cellular and molecular function of RAB18 and its roles in the pathogenesis of WARBM are still not fully understood. Using immunofluorescence staining and expression of Rab18 and organelle markers, we find that Rab18 associates with lysosomes and actively traffics along neurites in cultured neurons. Interestingly, Rab18-/- neurons exhibit impaired lysosomal transport. Using autophagosome marker LC3-II, we show that Rab18 dysfunction leads to aberrant autophagy activities in neurons. Electron microscopy further reveals accumulation of lipofuscin-like granules in the dorsal root ganglion of Rab18-/- mice. Surprisingly, Rab18 colocalizes, cofractionates, and coprecipitates with the lysosomal regulator Rab7, mutations of which cause Charcot-Marie-Tooth (CMT) neuropathy type 2B. Moreover, Rab7 is upregulated in Rab18-deficient neurons, suggesting a compensatory effect. Together, our results suggest that the functions of RAB18 and RAB7 in lysosomal and autophagic activities may constitute an overlapping mechanism underlying WARBM and CMT pathogenesis in the nervous system.


Abnormalities, Multiple/metabolism , Autophagy , Cataract/congenital , Charcot-Marie-Tooth Disease/metabolism , Cornea/abnormalities , Hypogonadism/metabolism , Intellectual Disability/metabolism , Lysosomes/metabolism , Microcephaly/metabolism , Nervous System/metabolism , Optic Atrophy/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Cataract/metabolism , Cornea/metabolism , Epistasis, Genetic , HEK293 Cells , Humans , Laminopathies , Mice , Neurons/metabolism , PC12 Cells , Protein Binding , Rats , Rats, Sprague-Dawley
10.
Oncotarget ; 7(26): 39184-39196, 2016 Jun 28.
Article En | MEDLINE | ID: mdl-27172900

Mutations in the proline-rich transmembrane protein 2 (PRRT2) gene cause a wide spectrum of neurological diseases, ranging from paroxysmal kinesigenic dyskinesia (PKD) to mental retardation and epilepsy. Previously, seven PKD-related PRRT2 heterozygous mutations were identified in the Taiwanese population: P91QfsX, E199X, S202HfsX, R217PfsX, R217EfsX, R240X and R308C. This study aimed to investigate the disease-causing mechanisms of these PRRT2 mutations. We first documented that Prrt2 was localized at the pre- and post-synaptic membranes with a close spatial association with SNAP25 by synaptic membrane fractionation and immunostaining of the rat neurons. Our results then revealed that the six truncating Prrt2 mutants were accumulated in the cytoplasm and thus failed to target to the cell membrane; the R308C missense mutant had significantly reduced protein expression, suggesting loss-of function effects generated by these mutations. Using in utero electroporation of shRNA into cortical neurons, we further found that knocking down Prrt2 expression in vivo resulted in a delay in neuronal migration during embryonic development and a marked decrease in synaptic density after birth. These pathologic effects and novel disease-causing mechanisms may contribute to the severe clinical symptoms in PRRT2-related diseases.


Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Neurodegenerative Diseases/genetics , Neurons/metabolism , Animals , COS Cells , Chlorocebus aethiops , Cytoplasm/metabolism , Disease Models, Animal , Dystonia/genetics , Epilepsy/genetics , Genetic Predisposition to Disease , HEK293 Cells , Heterozygote , Hippocampus/metabolism , Humans , Intellectual Disability/genetics , Mice , Mice, Inbred ICR , Mutation , Mutation, Missense , Neurodegenerative Diseases/metabolism , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Taiwan
11.
Sci Rep ; 6: 19145, 2016 Jan 13.
Article En | MEDLINE | ID: mdl-26758390

Intracellular endogenous fluorescent co-enzymes, reduced nicotinamide adenine dinucleotide (NADH) and flavin adenine dinucleotide (FAD), play a pivotal role in cellular metabolism; quantitative assessment of their presence in living cells can be exploited to monitor cellular energetics in Parkinson's disease (PD), a neurodegenerative disorder. Here, we applied two-photon fluorescence lifetime imaging microscopy (2P-FLIM) to noninvasively measure the fluorescence lifetime components of NADH and FAD, and their relative contributions in MPP(+) (1-methyl-4-phenylpyridinium) treated neuronal cells, derived from PC12 cells treated with nerve growth factor (NGF), to mimic PD conditions. A systematic FLIM data analysis showed a statistically significant (p < 0.001) decrease in the fluorescence lifetime of both free and protein-bound NADH, as well as free and protein-bound FAD in MPP(+) treated cells. On the relative contributions of the free and protein-bound NADH and FAD to the life time, however, both the free NADH contribution and the corresponding protein-bound FAD contribution increase significantly (p < 0.001) in MPP(+) treated cells, compared to control cells. These results, which indicate a shift in energy production in the MPP(+) treated cells from oxidative phosphorylation towards anaerobic glycolysis, can potentially be used as cellular metabolic metrics to assess the condition of PD at the cellular level.


Energy Metabolism , Metabolomics , Microscopy, Fluorescence , Models, Biological , Animals , Energy Metabolism/drug effects , Flavin-Adenine Dinucleotide/metabolism , Metabolomics/methods , Microscopy, Fluorescence/methods , NAD/metabolism , Nerve Growth Factor/pharmacology , PC12 Cells , Parkinson Disease/metabolism , Rats
12.
Exp Neurol ; 267: 143-51, 2015 May.
Article En | MEDLINE | ID: mdl-25779931

Mutations in the gene of RAB18, a member of Ras superfamily of small G-proteins, cause Warburg Micro Syndrome (WARBM) which is characterized by defective neurodevelopmental and ophthalmological phenotypes. Despite loss of Rab18 had been reported to induce disruption of the endoplasmic reticulum structure and neuronal cytoskeleton organization, parts of the pathogenic mechanism caused by RAB18 mutation remain unclear. From the N-ethyl-N-nitrosourea (ENU)-induced mutagenesis library, we identified a mouse line whose Rab18 was knocked out. This Rab18(-/-) mouse exhibited stomping gait, smaller testis and eyes, mimicking several features of WARBM. Rab18(-/-) mice were obviously less sensitive to pain and touch than WT mice. Histological examinations on Rab18(-/-) mice revealed progressive axonal degeneration in the optic nerves, dorsal column of the spinal cord and sensory roots of the spinal nerves while the motor roots were spared. All the behavioral and pathological changes that resulted from abnormalities in the sensory axons were prevented by introducing an extra copy of Rab18 transgene in Rab18(-/-) mice. Our results reveal that sensory axonal degeneration is the primary cause of stomping gait and progressive weakness of the hind limbs in Rab18(-/-) mice, and optic nerve degeneration should be the major pathology of progressive optic atrophy in children with WARBM. Our results indicate that the sensory nervous system is more vulnerable to Rab18 deficiency and WARBM is not only a neurodevelopmental but also neurodegenerative disease.


Abnormalities, Multiple , Cataract/congenital , Cornea/abnormalities , Ethylnitrosourea/pharmacology , Hypogonadism , Intellectual Disability , Microcephaly , Mutagenesis/drug effects , Nerve Degeneration/etiology , Optic Atrophy , Sequence Deletion/genetics , rab GTP-Binding Proteins/deficiency , Abnormalities, Multiple/chemically induced , Abnormalities, Multiple/genetics , Age Factors , Animals , Axons/pathology , Axons/ultrastructure , Cataract/chemically induced , Cataract/complications , Cataract/genetics , Disease Models, Animal , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum/ultrastructure , Eye/pathology , Hypogonadism/chemically induced , Hypogonadism/complications , Hypogonadism/genetics , Intellectual Disability/chemically induced , Intellectual Disability/complications , Intellectual Disability/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microcephaly/chemically induced , Microcephaly/complications , Microcephaly/genetics , Microphthalmos/etiology , Microphthalmos/genetics , Nerve Degeneration/pathology , Optic Atrophy/chemically induced , Optic Atrophy/complications , Optic Atrophy/genetics , Optic Nerve Diseases/etiology , Optic Nerve Diseases/genetics , Psychomotor Performance/drug effects , Testis/pathology , Touch Perception/drug effects , Touch Perception/genetics , rab GTP-Binding Proteins/genetics
13.
PLoS One ; 7(5): e32984, 2012.
Article En | MEDLINE | ID: mdl-22666313

OBJECTIVES: The forced swim test (FST) is a commonly used model to predict antidepressant efficacy. Uncovering the genetic basis of the model may unravel the mechanism of antidepressant treatment. METHODS: FVB/NJ (FVB) and C57BL/6J (B6) were first identified as the response and non-response strains to fluoxetine (a serotonin-specific reuptake inhibitor antidepressant) treatment in the mouse FST. Simple-interval (SIM) and composite-interval (CIM) mappings were applied to map the quantitative trait loci (QTLs) of the anti-immobility effect of fluoxetine in FST (FST(FLX)) in 865 male B6×FVB-F2 mice. The brain mRNA expressions of the gene with the maximum QTL-linkage signal for FST(FLX) after the FST were compared between B6 and FVB mice and also compared between fluoxetine and saline treatment. The association of the variants in the human homologue of the mouse FST(FLX)-QTL gene with major depressive disorder (MDD) and antidepressant response were investigated in 1080 human subjects (MDD/control = 582/498). RESULTS: One linkage signal for FST(FLX)-QTL was detected at an intronic SNP (rs6215396) of the mouse Zfp326 gene (maximal CIM-LOD = 9.36). The Zfp326 mRNA expression in the FVB thalamus was significantly down-regulated by fluoxetine in the FST, and the higher FVB-to-B6 Zfp326 mRNA expressions in the frontal cortex, striatum and hypothalamus diminished after fluoxetine treatment. Two coding-synonymous SNPs (rs2816881 and rs10922744) in the human homologue of Zfp326, ZNF326, were significantly associated with the 8-week antidepressant treatment response in the MDD patients (Bonferroni-corrected p = 0.004-0.028). CONCLUSIONS: The findings suggest the involvement of the Zfp326 and ZNF326 genes in antidepressant treatment response.


Antidepressive Agents/pharmacology , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Adult , Aged , Animals , Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Case-Control Studies , DNA-Binding Proteins/metabolism , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/genetics , Exons/genetics , Female , Fluoxetine/pharmacology , Fluoxetine/therapeutic use , Gene Expression Regulation/drug effects , Humans , Male , Mice , Middle Aged , Nuclear Proteins/metabolism , Polymorphism, Genetic/genetics , Quantitative Trait Loci/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use , Species Specificity , Swimming , Transcription Factors/metabolism , Treatment Outcome , Young Adult
...