Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 6880, 2022 11 12.
Article in English | MEDLINE | ID: mdl-36371400

ABSTRACT

Parkinson's disease is a progressive neurodegenerative disorder characterized by the preferential loss of tyrosine hydroxylase (TH)-expressing dopaminergic neurons in the substantia nigra. Although the abnormal accumulation and aggregation of α-synuclein have been implicated in the pathogenesis of Parkinson's disease, the underlying mechanisms remain largely elusive. Here, we found that TH converts Tyr136 in α-synuclein into dihydroxyphenylalanine (DOPA; Y136DOPA) through mass spectrometric analysis. Y136DOPA modification was clearly detected by a specific antibody in the dopaminergic neurons of α-synuclein-overexpressing mice as well as human α-synucleinopathies. Furthermore, dopanized α-synuclein tended to form oligomers rather than large fibril aggregates and significantly enhanced neurotoxicity. Our findings suggest that the dopanization of α-synuclein by TH may contribute to oligomer and/or seed formation causing neurodegeneration with the potential to shed light on the pathogenesis of Parkinson's disease.


Subject(s)
Parkinson Disease , alpha-Synuclein , Mice , Humans , Animals , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Tyrosine , Substantia Nigra/metabolism , Dopaminergic Neurons/metabolism
2.
J Neurosci ; 42(37): 7031-7046, 2022 09 14.
Article in English | MEDLINE | ID: mdl-35906071

ABSTRACT

Alpha-synuclein (αSyn) and tau are abundant multifunctional neuronal proteins, and their intracellular deposits have been linked to many neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Despite the disease relevance, their physiological roles remain elusive, as mice with knock-out of either of these genes do not exhibit overt phenotypes. To reveal functional cooperation, we generated αSyn-/-tau-/- double-knock-out mice and characterized the functional cross talk between these proteins during brain development. Intriguingly, deletion of αSyn and tau reduced Notch signaling and accelerated interkinetic nuclear migration of G2 phase at early embryonic stage. This significantly altered the balance between the proliferative and neurogenic divisions of progenitor cells, resulting in an overproduction of early born neurons and enhanced neurogenesis, by which the brain size was enlarged during the embryonic stage in both sexes. On the other hand, a reduction in the number of neural progenitor cells in the middle stage of corticogenesis diminished subsequent gliogenesis in the αSyn-/-tau-/- cortex. Additionally, the expansion and maturation of macroglial cells (astrocytes and oligodendrocytes) were suppressed in the αSyn-/-tau-/- postnatal brain, which in turn reduced the male αSyn-/-tau-/- brain size and cortical thickness to less than the control values. Our study identifies important functional cooperation of αSyn and tau during corticogenesis.SIGNIFICANCE STATEMENT Correct understanding of the physiological functions of αSyn and tau in CNS is critical to elucidate pathogenesis involved in the etiology of neurodegenerative diseases including Alzheimer's disease and Parkinson's disease. We show here that αSyn and tau are cooperatively involved in brain development via maintenance of progenitor cells. αSyn and tau double-knock-out mice exhibited an overproduction of early born neurons and accelerated neurogenesis at early corticogenesis. Furthermore, loss of αSyn and tau also perturbed gliogenesis at later embryonic stage, as well as the subsequent glial expansion and maturation at postnatal brain. Our findings provide new mechanistic insights and extend therapeutic opportunities for neurodegenerative diseases caused by aberrant αSyn and tau.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Parkinson Disease , Alzheimer Disease/metabolism , Animals , Female , Male , Mice , Mice, Knockout , Neurodegenerative Diseases/metabolism , Parkinson Disease/pathology , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
3.
Hum Mol Genet ; 31(6): 942-957, 2022 03 21.
Article in English | MEDLINE | ID: mdl-34635911

ABSTRACT

Human cerebral cortical malformations are associated with progenitor proliferation and neuronal migration abnormalities. Progenitor cells include apical radial glia, intermediate progenitors and basal (or outer) radial glia (bRGs or oRGs). bRGs are few in number in lissencephalic species (e.g. the mouse) but abundant in gyrencephalic brains. The LIS1 gene coding for a dynein regulator, is mutated in human lissencephaly, associated also in some cases with microcephaly. LIS1 was shown to be important during cell division and neuronal migration. Here, we generated bRG-like cells in the mouse embryonic brain, investigating the role of Lis1 in their formation. This was achieved by in utero electroporation of a hominoid-specific gene TBC1D3 (coding for a RAB-GAP protein) at mouse embryonic day (E) 14.5. We first confirmed that TBC1D3 expression in wild-type (WT) brain generates numerous Pax6+ bRG-like cells that are basally localized. Second, using the same approach, we assessed the formation of these cells in heterozygote Lis1 mutant brains. Our novel results show that Lis1 depletion in the forebrain from E9.5 prevented subsequent TBC1D3-induced bRG-like cell amplification. Indeed, we observe perturbation of the ventricular zone (VZ) in the mutant. Lis1 depletion altered adhesion proteins and mitotic spindle orientations at the ventricular surface and increased the proportion of abventricular mitoses. Progenitor outcome could not be further altered by TBC1D3. We conclude that disruption of Lis1/LIS1 dosage is likely to be detrimental for appropriate progenitor number and position, contributing to lissencephaly pathogenesis.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Lissencephaly , Microtubule-Associated Proteins/genetics , Nervous System Malformations , Animals , Dyneins/genetics , Ependymoglial Cells/metabolism , GTPase-Activating Proteins/genetics , Lissencephaly/genetics , Mice , Mitosis , Mutation , Nervous System Malformations/genetics
4.
Sci Rep ; 10(1): 2518, 2020 02 13.
Article in English | MEDLINE | ID: mdl-32054870

ABSTRACT

The field of genome editing was founded on the establishment of methods, such as the clustered regularly interspaced short palindromic repeat (CRISPR) and CRISPR-associated protein (CRISPR/Cas) system, used to target DNA double-strand breaks (DSBs). However, the efficiency of genome editing also largely depends on the endogenous cellular repair machinery. Here, we report that the specific modulation of targeting vectors to provide 3' overhangs at both ends increased the efficiency of homology-directed repair (HDR) in embryonic stem cells. We applied the modulated targeting vectors to produce homologous recombinant mice directly by pronuclear injection, but the frequency of HDR was low. Furthermore, we combined our method with the CRISPR/Cas9 system, resulting in a significant increase in HDR frequency. Thus, our HDR-based method, enhanced homologous recombination for genome targeting (eHOT), is a new and powerful method for genome engineering.


Subject(s)
CRISPR-Cas Systems , DNA Breaks, Double-Stranded , Gene Editing , Gene Targeting , Homologous Recombination , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , Female , Gene Editing/methods , Gene Targeting/methods , Genetic Vectors/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Recombinational DNA Repair
5.
Sci Rep ; 8(1): 8019, 2018 May 17.
Article in English | MEDLINE | ID: mdl-29769584

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

6.
Sci Rep ; 7(1): 16386, 2017 11 27.
Article in English | MEDLINE | ID: mdl-29180624

ABSTRACT

Although α-synuclein (αSyn) has been linked to Parkinson's disease (PD), the mechanisms underlying the causative role in PD remain unclear. We previously proposed a model for a transportable microtubule (tMT), in which dynein is anchored to a short tMT by LIS1 followed by the kinesin-dependent anterograde transport; however the mechanisms that produce tMTs have not been determined. Our in vitro investigations of microtubule (MT) dynamics revealed that αSyn facilitates the formation of short MTs and preferentially binds to MTs carrying 14 protofilaments (pfs). Live-cell imaging showed that αSyn co-transported with dynein and mobile ßIII-tubulin fragments in the anterograde transport. Furthermore, bi-directional axonal transports are severely affected in αSyn and γSyn depleted dorsal root ganglion neurons. SR-PALM analyses further revealed the fibrous co-localization of αSyn, dynein and ßIII-tubulin in axons. More importantly, 14-pfs MTs have been found in rat femoral nerve tissue, and they increased approximately 19 fold the control in quantify upon nerve ligation, indicating the unconventional MTs are mobile. Our findings indicate that αSyn facilitates to form short, mobile tMTs that play an important role in the axonal transport. This unexpected and intriguing discovery related to axonal transport provides new insight on the pathogenesis of PD.


Subject(s)
Axonal Transport , Axons/metabolism , Microtubules/metabolism , alpha-Synuclein/metabolism , Animals , Axons/ultrastructure , Chromatography, Liquid , Femoral Nerve/metabolism , Femoral Nerve/ultrastructure , Gas Chromatography-Mass Spectrometry , Male , Microtubules/chemistry , Neurons/metabolism , Protein Binding , Protein Multimerization , Protein Transport , Proteome , Proteomics/methods , Rats , Recombinant Proteins/metabolism , Tubulin/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/genetics
7.
Biol Open ; 6(7): 1041-1055, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28630356

ABSTRACT

The robust axonal growth and regenerative capacities of young neurons decrease substantially with age. This developmental downregulation of axonal growth may facilitate axonal pruning and neural circuit formation but limits functional recovery following nerve damage. While external factors influencing axonal growth have been extensively investigated, relatively little is known about the intrinsic molecular changes underlying the age-dependent reduction in regeneration capacity. We report that developmental downregulation of LIS1 is responsible for the decreased axonal extension capacity of mature dorsal root ganglion (DRG) neurons. In contrast, exogenous LIS1 expression or endogenous LIS1 augmentation by calpain inhibition restored axonal extension capacity in mature DRG neurons and facilitated regeneration of the damaged sciatic nerve. The insulator protein CTCF suppressed LIS1 expression in mature DRG neurons, and this reduction resulted in excessive accumulation of phosphoactivated GSK-3ß at the axon tip, causing failure of the axonal extension. Conversely, sustained LIS1 expression inhibited developmental axon pruning in the mammillary body. Thus, LIS1 regulation may coordinate the balance between axonal growth and pruning during maturation of neuronal circuits.

8.
Sci Rep ; 7: 39902, 2017 01 12.
Article in English | MEDLINE | ID: mdl-28079116

ABSTRACT

Human mutations in KATNB1 (p80) cause severe congenital cortical malformations, which encompass the clinical features of both microcephaly and lissencephaly. Although p80 plays critical roles during brain development, the underlying mechanisms remain predominately unknown. Here, we demonstrate that p80 regulates microtubule (MT) remodeling in combination with NuMA (nuclear mitotic apparatus protein) and cytoplasmic dynein. We show that p80 shuttles between the nucleus and spindle pole in synchrony with the cell cycle. Interestingly, this striking feature is shared with NuMA. Importantly, p80 is essential for aster formation and maintenance in vitro. siRNA-mediated depletion of p80 and/or NuMA induced abnormal mitotic phenotypes in cultured mouse embryonic fibroblasts and aberrant neurogenesis and neuronal migration in the mouse embryonic brain. Importantly, these results were confirmed in p80-mutant harboring patient-derived induced pluripotent stem cells and brain organoids. Taken together, our findings provide valuable insights into the pathogenesis of severe microlissencephaly, in which p80 and NuMA delineate a common pathway for neurogenesis and neuronal migration via MT organization at the centrosome/spindle pole.


Subject(s)
Adenosine Triphosphatases/metabolism , Fibroblasts/physiology , Induced Pluripotent Stem Cells/physiology , Katanin/metabolism , Microtubules/metabolism , Nervous System Malformations/metabolism , Neurons/physiology , Nuclear Proteins/metabolism , Adenosine Triphosphatases/genetics , Animals , Cell Cycle Proteins , Dyneins/metabolism , HeLa Cells , Humans , Katanin/genetics , Mice , Mice, Inbred Strains , Mitosis/genetics , Mutation/genetics , Nervous System Malformations/genetics , Neurogenesis/genetics , Nuclear Proteins/genetics , RNA, Small Interfering/genetics
9.
Cereb Cortex ; 27(8): 3918-3929, 2017 08 01.
Article in English | MEDLINE | ID: mdl-27371763

ABSTRACT

Neuronal nitric oxide synthase is involved in diverse signaling cascades that regulate neuronal development and functions via S-Nitrosylation-mediated mechanism or the soluble guanylate cyclase (sGC)/cyclic guanosine monophosphate (cGMP) pathway activated by nitric oxide. Although it has been studied extensively in vitro and in invertebrate animals, effects on mammalian brain development and underlying mechanisms remain poorly understood. Here we report that genetic deletion of "Nos1" disrupts dendritic development, whereas pharmacological inhibition of the sGC/cGMP pathway does not alter dendritic growth during cerebral cortex development. Instead, nuclear distribution element-like (NDEL1), a protein that regulates dendritic development, is specifically S-nitrosylated at cysteine 203, thereby accelerating dendritic arborization. This post-translational modification is enhanced by N-methyl-D-aspartate receptor-mediated neuronal activity, the main regulator of dendritic formation. Notably, we found that disruption of S-Nitrosylation of NDEL1 mediates impaired dendritic maturation caused by developmental alcohol exposure, a model of developmental brain abnormalities resulting from maternal alcohol use. These results highlight S-Nitrosylation as a key activity-dependent mechanism underlying neonatal brain maturation and suggest that reduction of S-Nitrosylation of NDEL1 acts as a pathological factor mediating neurodevelopmental abnormalities caused by maternal alcohol exposure.


Subject(s)
Carrier Proteins/metabolism , Dendrites/metabolism , Fetal Alcohol Spectrum Disorders/metabolism , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism , Synaptic Transmission/physiology , Animals , Carrier Proteins/genetics , Dendrites/drug effects , Dendrites/pathology , Disease Models, Animal , Fetal Alcohol Spectrum Disorders/pathology , Humans , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Nitric Oxide Synthase Type I/deficiency , Nitric Oxide Synthase Type I/genetics , Prefrontal Cortex/drug effects , Prefrontal Cortex/growth & development , Prefrontal Cortex/pathology , Pyramidal Cells/drug effects , Pyramidal Cells/pathology
10.
J Cell Biol ; 212(4): 409-23, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26880200

ABSTRACT

Primary cilia protrude from the surface of quiescent cells and disassemble at cell cycle reentry. We previously showed that ciliary reassembly is suppressed by trichoplein-mediated Aurora A activation pathway in growing cells. Here, we report that Ndel1, a well-known modulator of dynein activity, localizes at the subdistal appendage of the mother centriole, which nucleates a primary cilium. In the presence of serum, Ndel1 depletion reduces trichoplein at the mother centriole and induces unscheduled primary cilia formation, which is reverted by forced trichoplein expression or coknockdown of KCTD17 (an E3 ligase component protein for trichoplein). Serum starvation induced transient Ndel1 degradation, subsequent to the disappearance of trichoplein at the mother centriole. Forced expression of Ndel1 suppressed trichoplein degradation and axonemal microtubule extension during ciliogenesis, similar to trichoplein induction or KCTD17 knockdown. Most importantly, the proportion of ciliated and quiescent cells was increased in the kidney tubular epithelia of newborn Ndel1-hypomorphic mice. Thus, Ndel1 acts as a novel upstream regulator of the trichoplein-Aurora A pathway to inhibit primary cilia assembly.


Subject(s)
Aurora Kinase A/metabolism , Carrier Proteins/metabolism , Cell Proliferation , Epithelial Cells/enzymology , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Animals, Newborn , Aurora Kinase A/genetics , Carrier Proteins/genetics , Cell Cycle Checkpoints , Centrioles/enzymology , Cilia/enzymology , Genotype , HeLa Cells , Humans , Kidney Tubules/cytology , Kidney Tubules/enzymology , Mice , Mice, Knockout , Microtubules/enzymology , Phenotype , Protein Stability , Proteolysis , RNA Interference , Swiss 3T3 Cells , Time Factors , Transfection
11.
Microscopy (Oxf) ; 64(6): 419-27, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26371280

ABSTRACT

Cytoplasmic dynein is a microtubule-based motor protein that transports intracellular cargo and performs various functions during cell division. We previously reported that Lis1 suppressed dynein motility on microtubules in an idling state. Recently, a model showed that Lis1 prevents the ATPase domain of dynein from transmitting a detachment signal to its microtubule-binding domain. However, conformational information on dynein is limited. We used electron microscopy to investigate the conformation of dynein and nucleotide-induced conformational changes on microtubules. The conformation of dynein differed depending on the presence or absence of a nucleotide. In the presence of the nucleotide ADP-vanadate, dynein displayed an extended form on microtubules (extended form), whereas in the absence of a nucleotide, dynein lay along microtubules (compact form). This conformational change reflects chemomechanical coupling in dynein walking on microtubules. We also found that Lis1 fixed the conformation of dynein in the compact form regardless of the nucleotide condition. Removal of the Lis1 dimerization motif abolished Lis1-dependent fixation of dynein in the compact form. This suggests that the idling state of dynein on microtubules induced by Lis1 occurs through the Lis1-dependent arrest of dynein chemomechanical coupling.


Subject(s)
Cryoelectron Microscopy/methods , Cytoplasmic Dyneins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Animals , Baculoviridae/genetics , Cells, Cultured , Microtubule-Associated Proteins/genetics , Nucleotides/metabolism , Protein Conformation , Protein Structure, Tertiary , Swine , Vanadates/metabolism
12.
J Neurosci ; 35(7): 2942-58, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25698733

ABSTRACT

Cell positioning and neuronal network formation are crucial for proper brain function. Disrupted-in-Schizophrenia 1 (DISC1) is anterogradely transported to the neurite tips, together with Lis1, and functions in neurite extension via suppression of GSK3ß activity. Then, transported Lis1 is retrogradely transported and functions in cell migration. Here, we show that DISC1-binding zinc finger protein (DBZ), together with DISC1, regulates mouse cortical cell positioning and neurite development in vivo. DBZ hindered Ndel1 phosphorylation at threonine 219 and serine 251. DBZ depletion or expression of a double-phosphorylated mimetic form of Ndel1 impaired the transport of Lis1 and DISC1 to the neurite tips and hampered microtubule elongation. Moreover, application of DISC1 or a GSK3ß inhibitor rescued the impairments caused by DBZ insufficiency or double-phosphorylated Ndel1 expression. We concluded that DBZ controls cell positioning and neurite development by interfering with Ndel1 from disproportionate phosphorylation, which is critical for appropriate anterograde transport of the DISC1-complex.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Carrier Proteins/metabolism , Cell Movement/physiology , Cerebral Cortex/cytology , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/physiology , Animals , Biological Transport , Cells, Cultured , Cerebral Cortex/embryology , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Developmental/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Neurogenesis , Phosphorylation , Pregnancy , Transfection
13.
Nat Commun ; 5: 5295, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25342295

ABSTRACT

Cytoplasmic dynein acts as a motor for the intracellular retrograde motility of vesicles and organelles along microtubules. However, the regulatory mechanism underlying release of dynactin bound cargoes from dynein motor remains largely unknown. Here we report that ADP-ribosylation factor-like 3 (Arl3) and dynein light chain LC8 induce dissociation of dynactin from dynein. Immunoprecipitation and microtubule pull-down assays revealed that Arl3(Q71L) and LC8 facilitated detachment of dynactin from dynein. We also demonstrated Arl3(Q71L) or LC8-mediated dynactin release from a dynein-dynactin complex through trace experiments using quantum dot (Qdot)-conjugated proteins. Furthermore, we disclosed interactions of Arl3 and LC8 with dynactin and dynein, respectively, by live-cell imaging. Finally, knockdown (KD) of Arl3 and LC8 by siRNA induced abnormal localizations of dynein, dynactin and related organelles. Our findings uncovered the surprising functional relevance of GTP-bound Arl3 and LC8 for the unloading regulation of dynactin-bound cargo from dynein motor.


Subject(s)
ADP-Ribosylation Factors/metabolism , Dyneins/metabolism , Microtubule-Associated Proteins/metabolism , Animals , Cytoplasmic Dyneins , Dynactin Complex , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Gene Knockdown Techniques , Green Fluorescent Proteins/metabolism , Humans , Mice , Microscopy, Fluorescence , Microtubules/metabolism , Mutant Proteins/metabolism , RNA, Small Interfering/metabolism , Recombinant Proteins/metabolism
14.
Neuron ; 79(2): 254-65, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23830831

ABSTRACT

In the developing neocortex, progenitor cells expand through symmetric division before they generate cortical neurons through multiple rounds of asymmetric cell division. Here, we show that the orientation of the mitotic spindle plays a crucial role in regulating the transition between those two division modes. We demonstrate that the protein phosphatase PP4c regulates spindle orientation in early cortical progenitor cells. Upon removing PP4c, mitotic spindles fail to orient in parallel to the neuroepithelial surface and progenitors divide with random orientation. As a result, their divisions become asymmetric and neurogenesis starts prematurely. Biochemical and genetic experiments show that PP4c acts by dephosphorylating the microtubule binding protein Ndel1, thereby enabling complex formation with Lis1 to form a functional spindle orientation complex. Our results identify a key regulator of cortical development and demonstrate that changes in the orientation of progenitor division are responsible for the transition between symmetric and asymmetric cell division.


Subject(s)
Cell Proliferation , Neocortex/embryology , Neocortex/enzymology , Neurogenesis/physiology , Phosphoprotein Phosphatases/physiology , Spindle Apparatus/enzymology , Animals , Cell Division/physiology , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neocortex/cytology , Neural Stem Cells/cytology , Neural Stem Cells/enzymology , Pregnancy
15.
Nat Commun ; 4: 2033, 2013.
Article in English | MEDLINE | ID: mdl-23783758

ABSTRACT

Cytoplasmic dynein drives the movement of a wide range of cargoes towards the minus ends of microtubules. We previously demonstrated that LIS1 forms an idling complex with dynein, which is transported to the plus ends of microtubules by kinesin motors. Here we report that the small GTPase Rab6a is essential for activation of idling dynein. Immunoprecipitation and microtubule pull-down assays reveal that the GTP bound mutant, Rab6a(Q72L), dissociates LIS1 from a LIS1-dynein complex, activating dynein movement in in vitro microtubule gliding assays. We monitor transient interaction between Rab6a(Q72L) and dynein in vivo using dual-colour fluorescence cross-correlation spectroscopy in dorsal root ganglion (DRG) neurons. Finally, we demonstrate that Rab6a(Q72L) mediates LIS1 release from a LIS1-dynein complex followed by dynein activation through an in vitro single-molecule assay using triple-colour quantum dots. Our findings reveal a surprising function for GTP bound Rab6a as an activator of idling dynein.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Dyneins/metabolism , Microtubule-Associated Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Down-Regulation , Ganglia, Spinal/metabolism , Gene Knockdown Techniques , Guanosine Triphosphate/metabolism , Mice , Microscopy, Fluorescence , Microtubules/metabolism , Mutant Proteins/metabolism , Neurons/metabolism , Protein Binding , Protein Transport , RNA, Small Interfering/metabolism , Spectrometry, Fluorescence
16.
Elife ; 2: e00133, 2013 Mar 06.
Article in English | MEDLINE | ID: mdl-23482306

ABSTRACT

In neurons, microtubules (MTs) span the length of both axons and dendrites, and the molecular motors use these intracellular 'highways' to transport diverse cargo to the appropriate subcellular locations. Whereas axonal MTs are organized such that the plus-end is oriented out from the cell body, dendrites exhibit a mixed MTs polarity containing both minus-end-out and plus-end-out MTs. The molecular mechanisms underlying this differential organization, as well as its functional significance, are unknown. Here, we show that kinesin-1 is critical in establishing the characteristic minus-end-out MT organization of the dendrite in vivo. In unc-116 (kinesin-1/kinesin heavy chain) mutants, the dendritic MTs adopt an axonal-like plus-end-out organization. Kinesin-1 protein is able to cross-link anti-paralleled MTs in vitro. We propose that kinesin-1 regulates the dendrite MT polarity through directly gliding the plus-end-out MTs out of the dendrite using both the motor domain and the C-terminal MT-binding domain. DOI:http://dx.doi.org/10.7554/eLife.00133.001.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Cell Cycle Proteins/metabolism , Cell Polarity , Dendrites/metabolism , Kinesins/metabolism , Microtubules/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Cell Cycle Proteins/genetics , Genotype , Kinesins/genetics , Mutation , Phenotype , Protein Binding , Protein Interaction Domains and Motifs , Signal Transduction , Synaptic Vesicles/metabolism
17.
Sci Rep ; 3: 1224, 2013.
Article in English | MEDLINE | ID: mdl-23390575

ABSTRACT

Toward a therapeutic intervention of lissencephaly, we applied a novel calpain inhibitor, SNJ1945. Peri-natal or post-natal treatment with SNJ1945 rescued defective neuronal migration in Lis1⁺/⁻ mice, impaired behavioral performance and improvement of ¹8F-FDG uptake. Furthermore, SNJ1945 improved the neural circuit formation and retrograde transport of NFG in Lis1⁺/⁻ mice. Thus, SNJ1945 is a potential drug for the treatment of human lissencephaly patients.


Subject(s)
Blood-Brain Barrier/metabolism , Calpain/antagonists & inhibitors , Carbamates/therapeutic use , Glycoproteins/therapeutic use , Lissencephaly/drug therapy , 1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Administration, Oral , Animals , Calpain/metabolism , Carbamates/chemistry , Carbamates/pharmacology , Cell Line , Fluorodeoxyglucose F18/chemistry , Fluorodeoxyglucose F18/metabolism , Glycoproteins/chemistry , Glycoproteins/pharmacology , Humans , Lissencephaly/physiopathology , Lissencephaly/prevention & control , Male , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Motor Activity/drug effects , Nerve Growth Factor/metabolism , Neurons/metabolism , Positron-Emission Tomography , Receptors, GABA/metabolism
18.
J Neurophysiol ; 109(2): 429-36, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23100132

ABSTRACT

LIS1 gene mutations lead to a rare neurological disorder, classical lissencephaly, characterized by brain malformations, mental retardation, seizures, and premature death. Mice heterozygous for Lis1 (Lis1(+/-)) exhibit cortical malformations, defects in neuronal migration, increased glutamate-mediated synaptic transmission, and spontaneous electrographic seizures. Recent work demonstrated that in utero treatment of Lis1(+/-) mutant dams with ALLN, a calpain inhibitor, partially rescues neuronal migration defects in the offspring. Given the challenges of in utero drug administration, we examined the therapeutic potential of ALLN on postnatal lissencephalic cells. Voltage- and current-clamp studies were performed with acute hippocampal slices obtained from Lis1 mutant mice and age-matched littermate control mice. Specifically, we determined whether postnatal ALLN treatment can reverse excitatory synaptic transmission deficits, namely, an increase in spontaneous and miniature excitatory postsynaptic current (EPSC) frequency, on CA1 pyramidal neurons observed in tissue slices from Lis1(+/-) mice. We found that acute application of ALLN restored spontaneous and miniature EPSC frequencies to wild-type levels without affecting inhibitory postsynaptic synaptic current. Furthermore, Western blot analysis of protein expression, including proteins involved in excitatory synaptic transmission, demonstrated that ALLN blocks the cleavage of the calpain substrate αII-spectrin but does not rescue Lis1 protein levels in Lis1(+/-) mutants.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Cysteine Proteinase Inhibitors/therapeutic use , Excitatory Postsynaptic Potentials/drug effects , Leupeptins/therapeutic use , Lissencephaly/drug therapy , Microtubule-Associated Proteins/genetics , Animals , Calpain/antagonists & inhibitors , Calpain/metabolism , Gene Expression , Heterozygote , Lissencephaly/genetics , Lissencephaly/physiopathology , Mice , Mice, Mutant Strains , Miniature Postsynaptic Potentials/drug effects , Mutation , Proteolysis , Pyramidal Cells/metabolism , Pyramidal Cells/physiopathology , Spectrin/metabolism
19.
J Neurosci ; 32(32): 11050-66, 2012 Aug 08.
Article in English | MEDLINE | ID: mdl-22875938

ABSTRACT

Neuronal migration is a critical feature to ensure proper location and wiring of neurons during cortical development. Postmitotic neurons migrate from the ventricular zone into the cortical plate to establish neuronal lamina in an "inside-out" gradient of maturation. Here, we report that the mitotic kinase Aurora-A is critical for the regulation of microtubule organization during neuronal migration via an Aurora-A-NDEL1 pathway in the mouse. Suppression of Aurora-A activity by inhibitors or siRNA resulted in severe impairment of neuronal migration of granular neurons. In addition, in utero injection of the Aurora-A kinase-dead mutant provoked defective migration of cortical neurons. Furthermore, we demonstrated that suppression of Aurora-A impaired microtubule modulation in migrating neurons. Interestingly, suppression of CDK5 by an inhibitor or siRNA reduced Aurora-A activity and NDEL1 phosphorylation by Aurora-A, which led to defective neuronal migration. We found that CDK5RAP2 is a key molecule that mediates functional interaction and is essential for centrosomal targeting of Aurora-A. Our observations demonstrated novel and surprising cross talk between Aurora-A and CDK5 during neuronal migration.


Subject(s)
Cell Movement/physiology , Gene Expression Regulation, Developmental/physiology , Microtubules/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Amiodarone , Analysis of Variance , Animals , Animals, Newborn , Aurora Kinase A , Aurora Kinases , Bromodeoxyuridine/metabolism , Calcium-Binding Proteins/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Movement/drug effects , Cells, Cultured , Cerebellum/cytology , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mutation/genetics , Neurons/drug effects , Phosphorylation/genetics , Piperazines/pharmacology , Pregnancy , Protein Serine-Threonine Kinases/genetics , Purines/pharmacology , RNA, Small Interfering/pharmacology , Roscovitine , Sex Factors , Signal Transduction/drug effects , Signal Transduction/genetics
20.
Biochem Biophys Res Commun ; 425(2): 212-8, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22835934

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD), the most common hereditary disease affecting the kidneys, is caused in 85% of cases by mutations in the PKD1 gene. The protein encoded by this gene, polycystin-1, is a renal epithelial cell membrane mechanoreceptor, sensing morphogenetic cues in the extracellular environment, which regulate the tissue architecture and differentiation. However, how such mutations result in the formation of cysts is still unclear. We performed a precise characterization of mesenchymal differentiation using PAX2, WNT4 and WT1 as a marker, which revealed that impairment of the differentiation process preceded the development of cysts in Pkd1(-/-) mice. We performed an in vitro organ culture and found that progesterone and a derivative thereof facilitated mesenchymal differentiation, and partially prevented the formation of cysts in Pkd1(-/-) kidneys. An injection of progesterone or this derivative into the intraperitoneal space of pregnant females also improved the survival of Pkd1(-/-) embryos. Our findings suggest that compounds which enhance mesenchymal differentiation in the nephrogenesis might be useful for the therapeutic approach to prevent the formation of cysts in ADPKD patients.


Subject(s)
Cell Differentiation/drug effects , Cysts/prevention & control , Kidney Tubules/abnormalities , Mesoderm/drug effects , Polycystic Kidney, Autosomal Dominant/prevention & control , Progesterone/administration & dosage , Animals , Cysts/embryology , Cysts/genetics , Dilatation, Pathologic/embryology , Dilatation, Pathologic/prevention & control , Female , Mesoderm/cytology , Mice , Mice, Mutant Strains , Polycystic Kidney, Autosomal Dominant/embryology , Polycystic Kidney, Autosomal Dominant/genetics , Pregnancy , TRPP Cation Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...