Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Neurosci Res ; 193: 41-51, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36804599

ABSTRACT

Mislocalization and aggregate formation of TAR DNA-biding protein of 43kD (TDP-43) in the cytoplasm are signatures of amyotrophic lateral sclerosis(ALS) and frontotemporal lobar degeneration (FTLD). However, the role of two cytopathologies in ALS/FTLD pathogenesis is unclear. This study aims to elucidate the difference in their causality of TDP-43 in ALS/FTLD in vivo, using transgenic mice expressing human TDP-43 with defective nuclear localizing signals in neurons (Cyto-TDP) and those with aggregation propensity (Cyto-aggTDP). The expression levels of both proteins are less than half of endogenous TDP-43. Despite the low amount of Cyto-aggTDP, the TDP-43 phosphorylation is more evident than Cyto-TDP. Histopathological study showed accelerated astrogliosis in the anterior cerebral cortex of both mice. Cyto-aggTDP mice demonstrated significant but faint loss of neurons in the perirhinal(PERI) and ectorhinal(ECT) areas and higher Iba1-staining in the spinal cord than aged control. Despite the lack of locomotor dysfunctions in both mice, the open-field test showed enhanced exploratory behavior, indicating that the perpetual mislocalization of TDP-43 may suffice to trigger FTLD behavior. Besides, the aggregation propensity of TDP-43 promotes phosphorylation, but its role in the clinicopathological phenotype may not be primary.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Lobar Degeneration , Humans , Mice , Animals , Amyotrophic Lateral Sclerosis/genetics , Frontotemporal Lobar Degeneration/genetics , DNA-Binding Proteins/metabolism , Neurons/metabolism , Cerebral Cortex/metabolism , Mice, Transgenic
2.
Sci Rep ; 12(1): 16030, 2022 09 26.
Article in English | MEDLINE | ID: mdl-36163369

ABSTRACT

Genetic mutations in fused in sarcoma (FUS) cause amyotrophic lateral sclerosis (ALS). Although mitochondrial dysfunction and stress granule have been crucially implicated in FUS proteinopathy, the molecular basis remains unclear. Here, we show that DHX30, a component of mitochondrial RNA granules required for mitochondrial ribosome assembly, interacts with FUS, and plays a crucial role in ALS-FUS. WT FUS did not affect mitochondrial localization of DHX30, but the mutant FUS lowered the signal of mitochondrial DHX30 and promoted the colocalization of cytosolic FUS aggregates and stress granule markers. The immunohistochemistry of the spinal cord from an ALS-FUS patient also confirmed the colocalization, and the immunoelectron microscope demonstrated decreased mitochondrial DHX30 signal in the spinal motor neurons. Subcellular fractionation by the detergent-solubility and density-gradient ultracentrifugation revealed that mutant FUS also promoted cytosolic mislocalization of DHX30 and aggregate formation. Interestingly, the mutant FUS disrupted the DHX30 conformation with aberrant disulfide formation, leading to impaired mitochondrial translation. Moreover, blue-native gel electrophoresis revealed an OXPHOS assembly defect caused by the FUS mutant, which was similar to that caused by DHX30 knockdown. Collectively, our study proposes DHX30 as a pivotal molecule in which disulfide-mediated conformational change mediates mitochondrial dysfunction and cytosolic aggregate formation in ALS-FUS.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Amyotrophic Lateral Sclerosis/genetics , Detergents , Disulfides , Humans , Mitochondria/genetics , Mutation , RNA , RNA Helicases/genetics , RNA-Binding Protein FUS/chemistry , RNA-Binding Protein FUS/genetics
3.
Commun Biol ; 5(1): 358, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35418705

ABSTRACT

DNA double-strand break (DSB) is the most severe form of DNA damage and accumulates with age, in which cytoskeletal proteins are polymerized to repair DSB in dividing cells. Since tau is a microtubule-associated protein, we investigate whether DSB is involved in tau pathologies in Alzheimer's disease (AD). First, immunohistochemistry reveals the frequent coexistence of DSB and phosphorylated tau in the cortex of AD patients. In vitro studies using primary mouse cortical neurons show that non-p-tau accumulates perinuclearly together with the tubulin after DSB induction with etoposide, followed by the accumulation of phosphorylated tau. Moreover, the knockdown of endogenous tau exacerbates DSB in neurons, suggesting the protective role of tau on DNA repair. Interestingly, synergistic exposure of neurons to microtubule disassembly and the DSB strikingly augments aberrant p-tau aggregation and apoptosis. These data suggest that DSB plays a pivotal role in AD-tau pathology and that the failure of DSB repair leads to tauopathy.


Subject(s)
Alzheimer Disease , Tauopathies , Alzheimer Disease/metabolism , Animals , DNA , DNA Repair , Humans , Mice , Tauopathies/metabolism , Tubulin/metabolism
4.
Int J Mol Sci ; 21(10)2020 May 21.
Article in English | MEDLINE | ID: mdl-32455741

ABSTRACT

Aberrant angiogenesis is a pathological feature of a number of diseases and arises from the uncoordinated expression of angiogenic factors as response to different cellular stresses. Age-related macular degeneration (AMD), a leading cause of vision loss, can result from pathological angiogenesis. As a mutation in the mitochondrial ferritin (FTMT) gene has been associated with AMD, its possible role in modulating angiogenic factors and angiogenesis was investigated. FTMT is an iron-sequestering protein primarily expressed in metabolically active cells and tissues with high oxygen demand, including retina. In this study, we utilized the human retinal pigment epithelial cell line ARPE-19, both as undifferentiated and differentiated cells. The effects of proinflammatory cytokines, FTMT knockdown, and transient and stable overexpression of FTMT were investigated on expression of pro-angiogenic vascular endothelial growth factor (VEGF) and anti-angiogenic pigment epithelial-derived factor (PEDF). Proinflammatory cytokines induced FTMT and VEGF expression, while NF-κB inhibition significantly reduced FTMT expression. VEGF protein and mRNA expression were significantly increased in FTMT-silenced ARPE-19 cells. Using an in vitro angiogenesis assay with endothelial cells, we showed that conditioned media from FTMT-overexpressing cells had significant antiangiogenic effects. Collectively, our findings indicate that increased levels of FTMT inhibit angiogenesis, possibly by reducing levels of VEGF and increasing PEDF expression. The cellular models developed can be used to investigate if increased FTMT may be protective in angiogenic diseases, such as AMD.


Subject(s)
Ferritins/metabolism , Mitochondrial Proteins/metabolism , Neovascularization, Physiologic , Retinal Pigment Epithelium/metabolism , Cell Line , Cytokines/genetics , Cytokines/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Ferritins/genetics , Humans , Mitochondrial Proteins/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Retinal Pigment Epithelium/cytology , Serpins/genetics , Serpins/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
5.
J Alzheimers Dis ; 75(1): 45-60, 2020.
Article in English | MEDLINE | ID: mdl-32250299

ABSTRACT

Alzheimer's disease (AD) is the most common cause of dementia and understanding its pathogenesis should lead to improved therapeutic and diagnostic methods. Although several groups have developed transgenic mouse models overexpressing the human amyloid-ß precursor protein (APP) gene with AD mutations, with and without presenilin mutations, as well as APP gene knock-in mouse models, these animals display amyloid pathology but do not show neurofibrillary tangles or neuronal loss. This presumably is due to differences between the etiology of the aged-related human disease and the mouse models. Here we report the generation of two transgenic cynomolgus monkeys overexpressing the human gene for APP with Swedish, Artic, and Iberian mutations, and demonstrated expression of gene tagged green fluorescent protein marker in the placenta, amnion, hair follicles, and peripheral blood. We believe that these nonhuman primate models will be very useful to study the pathogenesis of dementia and AD. However, generated Tg monkeys still have some limitations. We employed the CAG promoter, which will promote gene expression in a non-tissue specific manner. Moreover, we used transgenic models but not knock-in models. Thus, the inserted transgene destroys endogenous gene(s) and may affect the phenotype(s). Nevertheless, it will be of great interest to determine whether these Tg monkeys will develop tauopathy and neurodegeneration similar to human AD.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Brain/metabolism , Macaca fascicularis/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Animals, Genetically Modified , Disease Models, Animal , Promoter Regions, Genetic
6.
Sci Rep ; 8(1): 6030, 2018 04 16.
Article in English | MEDLINE | ID: mdl-29662239

ABSTRACT

Aggregation of TAR DNA-binding protein of 43 kDa (TDP-43) is implicated in the pathogenesis of sporadic and certain familial forms of amyotrophic lateral sclerosis (ALS), suggesting elimination of TDP-43 aggregates as a possible therapeutic strategy. Here we generated and investigated a single-chain variable fragment (scFv) derived from the 3B12A monoclonal antibody (MAb) that recognises D247 of the TDP-43 nuclear export signal, an epitope masked in the physiological state. In transfected HEK293A cells, 3B12A scFv recapitulated the affinity of the full-length MAb to mislocalised TDP-43 with a defective nuclear localising signal and to a TDP-43 inclusion mimic with cysteine-to-serine substitution at RRM1. Moreover, 3B12A scFv accelerated proteasome-mediated degradation of aggregated TDP-43, likely due to an endogenous PEST-like proteolytic signal sequence in the VH domain CDR2 region. Addition of the chaperone-mediated autophagy (CMA)-related signal to 3B12A scFv induced HSP70 transcription, further enhancing TDP-43 aggregate clearance and cell viability. The 3B12A scFv also reduced TDP-43 aggregates in embryonic mouse brain following in utero electroporation while causing no overt postnatal brain pathology or developmental anomalies. These results suggest that a misfolding-specific intrabody prone to synergistic proteolysis by proteasomal and autophagic pathways is a promising strategy for mitigation of TDP-43 proteinopathy in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Antibodies, Monoclonal/pharmacology , DNA-Binding Proteins/metabolism , Protein Aggregation, Pathological/drug therapy , Protein Folding/drug effects , Single-Chain Antibodies/pharmacology , Amino Acid Sequence , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Antibodies, Monoclonal/therapeutic use , Autophagy/drug effects , DNA-Binding Proteins/analysis , HEK293 Cells , HSP70 Heat-Shock Proteins/metabolism , Humans , Inclusion Bodies/drug effects , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Mice, Inbred ICR , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Proteolysis , Proteostasis Deficiencies/drug therapy , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology , Single-Chain Antibodies/therapeutic use
7.
Sci Rep ; 6: 19118, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26751167

ABSTRACT

The molecular machinery responsible for cytosolic accumulation of misfolded TDP-43 in amyotrophic lateral sclerosis (ALS) remains elusive. Here we identified a cullin-2 (CUL2) RING complex as a novel ubiquitin ligase for fragmented forms of TDP-43. The von Hippel Lindau protein (VHL), a substrate binding component of the complex, preferentially recognized misfolded TDP-43 at Glu246 in RNA-recognition motif 2. Recombinant full-length TDP-43 was structurally fragile and readily cleaved, suggesting that misfolded TDP-43 is cleared by VHL/CUL2 in a step-wise manner via fragmentation. Surprisingly, excess VHL stabilized and led to inclusion formation of TDP-43, as well as mutant SOD1, at the juxtanuclear protein quality control center. Moreover, TDP-43 knockdown elevated VHL expression in cultured cells, implying an aberrant interaction between VHL and mislocalized TDP-43 in ALS. Finally, cytoplasmic inclusions especially in oligodendrocytes in ALS spinal cords were immunoreactive to both phosphorylated TDP-43 and VHL. Thus, our results suggest that an imbalance in VHL and CUL2 may underlie oligodendrocyte dysfunction in ALS, and highlight CUL2 E3 ligase emerges as a novel therapeutic potential for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Cullin Proteins/metabolism , DNA-Binding Proteins/metabolism , Oligodendroglia/metabolism , Protein Folding , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , DNA-Binding Proteins/chemistry , Epitopes/metabolism , HEK293 Cells , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Inclusion Bodies/metabolism , Mice, Transgenic , Models, Biological , Mutant Proteins/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Aggregates , Protein Binding , Protein Domains , Protein Stability , Proteolysis , Rats , Superoxide Dismutase/metabolism , Ubiquitin/metabolism , Ubiquitination
8.
Exp Biol Med (Maywood) ; 239(4): 489-501, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24521562

ABSTRACT

Pelizaeus-Merzbacher disease (PMD) is a hypomyelinating disorder caused by the duplication and missense mutations of the proteolipid protein 1 (PLP1) gene. PLP1 missense proteins accumulate in the endoplasmic reticulum (ER) of premature oligodendrocytes and induce severe ER stress followed by apoptosis of the cells. Here, we demonstrate that an anti-malaria drug, chloroquine, decreases the amount of an ER-resident mutant PLP1 containing an alanine-243 to valine (A243V) substitution, which induces severe PMD in human. By preventing mutant PLP1 translation through enhancing the phosphorylation of eukaryotic initiation factor 2 alpha, chloroquine ameliorated the ER stress induced by the mutant protein in HeLa cells. Chroloquine also attenuated ER stress in the primary oligodendrocytes obtained from myelin synthesis deficit (msd) mice, which carry the same PLP1 mutation. In the spinal cords of msd mice, chloroquine inhibited ER stress and upregulated the expression of marker genes of mature oligodendrocytes. Chloroquine-mediated attenuation of ER stress was observed in HeLa cells treated with tunicamycin, an N-glycosylation inhibitor, but not with thapsigargin, a sarco/ER Ca(2+)ATPase inhibitor, which confirms its efficacy against ER stress caused by nascent proteins. These findings indicate that chloroquine is an ER stress attenuator with potential use in treating PMD and possibly other ER stress-related diseases.


Subject(s)
Antimalarials/pharmacology , Chloroquine/pharmacology , Endoplasmic Reticulum Stress/drug effects , Pelizaeus-Merzbacher Disease/drug therapy , Animals , Antimalarials/therapeutic use , Apoptosis/drug effects , Chloroquine/therapeutic use , HeLa Cells , Humans , Mice , Models, Biological , Mutation , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Pelizaeus-Merzbacher Disease/pathology , Spinal Cord/metabolism
9.
J Biol Chem ; 288(21): 14886-905, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23558684

ABSTRACT

Aggregation of TAR DNA-binding protein of 43 kDa (TDP-43) is a pathological signature of amyotrophic lateral sclerosis (ALS). Although accumulating evidence suggests the involvement of RNA recognition motifs (RRMs) in TDP-43 proteinopathy, it remains unclear how native TDP-43 is converted to pathogenic forms. To elucidate the role of homeostasis of RRM1 structure in ALS pathogenesis, conformations of RRM1 under high pressure were monitored by NMR. We first found that RRM1 was prone to aggregation and had three regions showing stable chemical shifts during misfolding. Moreover, mass spectrometric analysis of aggregated RRM1 revealed that one of the regions was located on protease-resistant ß-strands containing two cysteines (Cys-173 and Cys-175), indicating that this region served as a core assembly interface in RRM1 aggregation. Although a fraction of RRM1 aggregates comprised disulfide-bonded oligomers, the substitution of cysteine(s) to serine(s) (C/S) resulted in unexpected acceleration of amyloid fibrils of RRM1 and disulfide-independent aggregate formation of full-length TDP-43. Notably, TDP-43 aggregates with RRM1-C/S required the C terminus, and replicated cytopathologies of ALS, including mislocalization, impaired RNA splicing, ubiquitination, phosphorylation, and motor neuron toxicity. Furthermore, RRM1-C/S accentuated inclusions of familial ALS-linked TDP-43 mutants in the C terminus. The relevance of RRM1-C/S-induced TDP-43 aggregates in ALS pathogenesis was verified by immunolabeling of inclusions of ALS patients and cultured cells overexpressing the RRM1-C/S TDP-43 with antibody targeting misfolding-relevant regions. Our results indicate that cysteines in RRM1 crucially govern the conformation of TDP-43, and aberrant self-assembly of RRM1 at amyloidogenic regions contributes to pathogenic conversion of TDP-43 in ALS.


Subject(s)
Amyloid , Amyotrophic Lateral Sclerosis , Intranuclear Inclusion Bodies , Neurons , Protein Folding , Amino Acid Motifs , Amyloid/chemistry , Amyloid/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Female , HEK293 Cells , Humans , Intranuclear Inclusion Bodies/metabolism , Intranuclear Inclusion Bodies/pathology , Magnetic Resonance Spectroscopy , Male , Neurons/metabolism , Neurons/pathology , Protein Structure, Quaternary , Protein Structure, Tertiary , RNA Splicing , Ubiquitination
10.
J Biol Chem ; 288(11): 7451-7466, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23344956

ABSTRACT

Missense mutations in the proteolipid protein 1 (PLP1) gene cause a wide spectrum of hypomyelinating disorders, from mild spastic paraplegia type 2 to severe Pelizaeus-Merzbacher disease (PMD). Mutant PLP1 accumulates in the endoplasmic reticulum (ER) and induces ER stress. However, the link between the clinical severity of PMD and the cellular response induced by mutant PLP1 remains largely unknown. Accumulation of misfolded proteins in the ER generally leads to up-regulation of ER chaperones to alleviate ER stress. Here, we found that expression of the PLP1-A243V mutant, which causes severe disease, depletes some ER chaperones with a KDEL (Lys-Asp-Glu-Leu) motif, in HeLa cells, MO3.13 oligodendrocytic cells, and primary oligodendrocytes. The same PLP1 mutant also induces fragmentation of the Golgi apparatus (GA). These organelle changes are less prominent in cells with milder disease-associated PLP1 mutants. Similar changes are also observed in cells expressing another disease-causing gene that triggers ER stress, as well as in cells treated with brefeldin A, which induces ER stress and GA fragmentation by inhibiting GA to ER trafficking. We also found that mutant PLP1 disturbs localization of the KDEL receptor, which transports the chaperones with the KDEL motif from the GA to the ER. These data show that PLP1 mutants inhibit GA to ER trafficking, which reduces the supply of ER chaperones and induces GA fragmentation. We propose that depletion of ER chaperones and GA fragmentation induced by mutant misfolded proteins contribute to the pathogenesis of inherited ER stress-related diseases and affect the disease severity.


Subject(s)
Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Golgi Apparatus/metabolism , Molecular Chaperones/metabolism , Mutation , Myelin Proteolipid Protein/metabolism , Pelizaeus-Merzbacher Disease/metabolism , Amino Acid Motifs , Animals , Biotinylation , Disease Models, Animal , HeLa Cells , Humans , Mice , Mutation, Missense , Neuroglia/cytology , Oligodendroglia/cytology , Organelles/metabolism , Protein Transport , Protein Unfolding
11.
Mol Genet Metab ; 106(1): 108-14, 2012 May.
Article in English | MEDLINE | ID: mdl-22436581

ABSTRACT

PLP1 amino acid substitutions cause accumulation of misfolded protein and induce endoplasmic reticulum (ER) stress, causing Pelizaeus-Merzbacher disease (PMD), a hypomyelinating disorder of the central nerve system. Currently no effective therapy is available for PMD. Promoted by its curative effects in other genetic disease models caused by similar molecular mechanisms, we tested if curcumin, a dietary compound, can rescue the lethal phenotype of a PMD mouse model (myelin synthesis deficient, msd). Curcumin was administered orally to myelin synthesis deficit (msd) mice at 180 mg·kg(-1)·day(-1) from the postnatal day 3. We evaluated general and motor status, changes in myelination and apoptosis of oligodendrocytes by neuropathological and biochemical examination, and transcription levels for ER-related molecules. We also examined the pharmacological effect of curcumin in cell culture system. Oral curcumin treatment resulted in 25% longer survival (p<0.01). In addition, oligodendrocytes undergoing apoptosis were reduced in number (p<0.05). However, no apparent improvement in motor function, neurological phenotype, and myelin formation was observed. Curcumin treatment did not change the expression of ER stress markers and subcellular localization of the mutant protein in vitro and/or in vivo. Curcumin partially mitigated the clinical and pathological phenotype of msd mice, although molecular mechanisms underlying this curative effect are yet undetermined. Nonetheless, curcumin may serve as a potential therapeutic compound for PMD caused by PLP1 point mutations.


Subject(s)
Central Nervous System , Curcumin/administration & dosage , Myelin Proteolipid Protein/metabolism , Pelizaeus-Merzbacher Disease/genetics , Animals , Apoptosis/drug effects , Central Nervous System/metabolism , Central Nervous System/pathology , Disease Models, Animal , Gene Expression/drug effects , HeLa Cells , Humans , Mice , Myelin Proteolipid Protein/genetics , Myelin Sheath/metabolism , Oligodendroglia/drug effects , Optic Nerve/metabolism , Optic Nerve/pathology , Pelizaeus-Merzbacher Disease/drug therapy , Pelizaeus-Merzbacher Disease/pathology
12.
PLoS One ; 7(12): e52776, 2012.
Article in English | MEDLINE | ID: mdl-23300771

ABSTRACT

Accumulating evidence suggests that pathogenic TAR DNA-binding protein (TDP)-43 fragments contain a partial RNA-recognition motif domain 2 (RRM2) in amyotrophic lateral sclerosis (ALS)/frontotemporal lobar degeneration. However, the molecular basis for how this domain links to the conformation and function of TDP-43 is unclear. Previous crystal analyses have documented that the RRM2-DNA complex dimerizes under acidic and high salt conditions, mediated by the intermolecular hydrogen bonds of Glu246-Ile249 and Asp247-Asp247. The aims of this study were to investigate the roles of Glu246 and Asp247 in the molecular assembly of RRM2 under physiological conditions, and to evaluate their potential use as markers for TDP-43 misfolding due to the aberrantly exposed dimer interface. Unexpectedly, gel filtration analyses showed that, regardless of DNA interaction, the RRM2 domain remained as a stable monomer in phosphate-buffered saline. Studies using substitution mutants revealed that Glu246 and, especially, Asp247 played a crucial role in preserving the functional RRM2 monomers. Substitution to glycine at Glu246 or Asp247 induced the formation of fibrillar oligomers of RRM2 accompanied by the loss of DNA-binding affinity, which also affected the conformation and the RNA splicing function of full-length TDP-43. A novel monoclonal antibody against peptides containing Asp247 was found to react with TDP-43 inclusions of ALS patients and mislocalized cytosolic TDP-43 in cultured cells, but not with nuclear wild-type TDP-43. Our findings indicate that Glu246 and Asp247 play pivotal roles in the proper conformation and function of TDP-43. In particular, Asp247 should be studied as a molecular target with an aberrant conformation related to TDP-43 proteinopathy.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA-Binding Proteins/chemistry , Aged , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Amyotrophic Lateral Sclerosis/pathology , Animals , Antibodies, Monoclonal, Murine-Derived/chemistry , Conserved Sequence , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , HEK293 Cells , Humans , Hydrogen Bonding , Inclusion Bodies/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Localization Signals , Protein Binding , Protein Folding , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary , Protein Transport , RNA Splicing , Solubility , Spinal Cord/metabolism
13.
Brain Res ; 1304: 26-37, 2009 Dec 22.
Article in English | MEDLINE | ID: mdl-19796633

ABSTRACT

Reelin regulates radial migration of the projection neurons in the developing cerebral cortex by inducing tyrosine phosphorylation of an intracellular adaptor protein, Disabled-1 (Dab1), through activation of Src family tyrosine kinases (SFKs). Five tyrosine residues of Dab1 (Y185, Y198, Y200, Y220, and Y232) are capable of being phosphorylated by SFKs. Among them, phosphorylation of Y198, Y220, and Y232 has been demonstrated after Reelin stimulation, and Y185 has been suggested to be an additional Reelin-induced phosphorylation site. In this study we established a reconstitution system in which a migratory defect in the cortex of Dab1-deficient mice is rescued by transfection with a wild-type Dab1 gene. The transfected neurons in the mutant cortex migrated radially and split the superficial preplate into the marginal zone and subplate by a mechanism that depended on interaction between Dab1 and Reelin receptors. Although this migration rescue was also observed in the mutant cortex transfected with a Dab1 gene containing a single substitution at Y198 by phenylalanine (Y198F), Y220F, Y232F, both of the Y185F and Y200F (Y185F/Y200F), Y185F/Y220F, Y185F/Y232F, Y198F/Y220F, or Y198F/Y232F, it was never observed in the mutant cortex transfected with a Dab1 gene containing the Y185F/Y198F or Y220F/Y232F. These findings suggest that Reelin induces phosphorylation at Y185 of Dab1, and that there are two Reelin signaling pathways, one mediated by the Y185/Y198 phosphorylation of Dab1 and the other mediated by the Y220/Y232 phosphorylation of Dab1. The results also suggest that phosphorylation of either one of the residues in each pair is sufficient for the transmission of Reelin signaling.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cell Movement/physiology , Extracellular Matrix Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/physiology , Serine Endopeptidases/metabolism , Tyrosine/metabolism , Animals , Cerebral Cortex/embryology , Cerebral Cortex/physiology , Electroporation , Gene Transfer Techniques , Immunohistochemistry , Mice , Mice, Knockout , Phosphorylation , Reelin Protein , Repressor Proteins/metabolism , Signal Transduction , Time Factors , Tumor Suppressor Proteins/metabolism , src-Family Kinases/metabolism
14.
Cell ; 132(3): 487-98, 2008 Feb 08.
Article in English | MEDLINE | ID: mdl-18267078

ABSTRACT

The cell-cycle transition from G1 to S phase has been difficult to visualize. We have harnessed antiphase oscillating proteins that mark cell-cycle transitions in order to develop genetically encoded fluorescent probes for this purpose. These probes effectively label individual G1 phase nuclei red and those in S/G2/M phases green. We were able to generate cultured cells and transgenic mice constitutively expressing the cell-cycle probes, in which every cell nucleus exhibits either red or green fluorescence. We performed time-lapse imaging to explore the spatiotemporal patterns of cell-cycle dynamics during the epithelial-mesenchymal transition of cultured cells, the migration and differentiation of neural progenitors in brain slices, and the development of tumors across blood vessels in live mice. These mice and cell lines will serve as model systems permitting unprecedented spatial and temporal resolution to help us better understand how the cell cycle is coordinated with various biological events.


Subject(s)
Cell Cycle , Cytological Techniques , Animals , COS Cells , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cells, Cultured , Chlorocebus aethiops , Fluorescence , Geminin , HeLa Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Confocal , Molecular Sequence Data , Morphogenesis , Neoplasms/pathology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ubiquitination
15.
Brain Res ; 1140: 84-95, 2007 Apr 06.
Article in English | MEDLINE | ID: mdl-16529723

ABSTRACT

The Reelin signaling and Cyclin-dependent kinase 5 (Cdk5) both regulate neuronal positioning in the developing brain. Using double-transgenic mice, we have previously shown that these two signaling pathways lie in parallel fashion and have a genetic interaction. Disabled-1 (Dab1), an adapter protein, mediates Reelin signaling and becomes tyrosine-phosphorylated on the binding of Reelin to its receptors. Several isoforms of Dab1 are expressed in embryonic mouse brain, and p80 [Dab1(555)] is the major protein translated. In the present study, we investigated whether Cdk5-mediated phosphorylation of Dab1 modulates Reelin signaling. Cdk5 phosphorylates p80 Dab1 at multiple sites in its carboxyl-terminal region, and tyrosine phosphorylation of p80 Dab1 by Fyn tyrosine kinase is attenuated by this Cdk5-mediated phosphorylation in vitro. Tyrosine phosphorylation of p80 Dab1 induced by exogenous Reelin is enhanced in Cdk5-deficient neurons, corroborating the inhibitory effect of Cdk5-mediated Ser/Thr phosphorylation on tyrosine phosphorylation of p80 Dab1. Another isoform, p45 Dab1 [Dab1(271)], however, is phosphorylated by Cdk5 at one serine residue within a unique carboxyl-terminal region, and its serine phosphorylation enhances tyrosine phosphorylation by Fyn and results in progressive degradation of p45 Dab1. These results indicate that Cdk5 modulates Reelin signaling through the Ser/Thr phosphorylation of Dab1 differently in an isoform-specific manner.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cyclin-Dependent Kinase 5/physiology , Extracellular Matrix Proteins/metabolism , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Signal Transduction/physiology , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cells, Cultured , Cerebral Cortex/cytology , Chlorocebus aethiops , Cyclin-Dependent Kinase 5/genetics , Embryo, Mammalian , Extracellular Matrix Proteins/genetics , Female , Humans , Immunoprecipitation/methods , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Weight , Mutagenesis/physiology , Nerve Tissue Proteins/genetics , Neurons/metabolism , Phosphorylation , Reelin Protein , Serine/metabolism , Serine Endopeptidases/genetics , Threonine/metabolism , Transfection
16.
Leuk Res ; 30(8): 987-92, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16448698

ABSTRACT

Several kinds of the p53 transcripts in which their open reading frames (ORFs) were truncated (ranging from 101 to 765 bp) were identified in Marek's disease (MD)-derived tumor cell lines as well as avian leukosis- and reticuloendotheliosis-derived ones, detected by nested RT-PCR and subsequent nucleotide sequence analysis. In these ORFs, regions encoding the proline-rich and DNA-binding domains of the p53 protein were frequently deleted, and many of these deletions were found to cause frame shift. Western blot analysis using anti-p53 monoclonal antibodies revealed that multiple p53 isoform proteins with various molecular weights including 45-46, 35 and 28 kDa were expressed in these tumor cell lines, though the p53 protein with a molecular weight of 49 kDa was detected in chicken embryo fibroblasts transformed by the SV40 T antigen as a control. Since no deletions were found in the p53 gene of these MD tumor cell lines, truncations in the p53 ORFs observed in this study might result from alternative splicing of the p53 gene.


Subject(s)
Marek Disease/genetics , Open Reading Frames/genetics , Sequence Deletion , Transcription, Genetic/genetics , Tumor Suppressor Protein p53/genetics , Alternative Splicing , Animals , Base Sequence , Birds , Cell Line, Tumor , Humans , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction/methods , Sensitivity and Specificity , Sequence Analysis, DNA/methods
17.
J Biol Chem ; 280(17): 16901-8, 2005 Apr 29.
Article in English | MEDLINE | ID: mdl-15718228

ABSTRACT

Reelin is a huge secreted protein that controls proper laminar formation in the developing brain. It is generally believed that tyrosine phosphorylation of Disabled1 (Dab1) by Src family tyrosine kinases is the most critical downstream event in Reelin signaling. The receptors for Reelin belong to the low density lipoprotein receptor family, most of whose members undergo regulated intracellular trafficking. In this study, we propose novel roles for Dab1 in Reelin signaling. We first demonstrated that cell surface expression of Reelin receptors was decreased in Dab1-deficient neurons. In heterologous cells, Dab1 enhanced cell surface expression of Reelin receptors, and this effect was mediated by direct interaction with the receptors. Moreover, Dab1 did not stably associate with the receptors at the plasma membrane in the resting state. When Reelin was added to primary cortical neurons, Dab1 was recruited to the receptors, and its tyrosine residues were phosphorylated. Although Reelin and Dab1 colocalized well shortly after the addition of Reelin, Dab1 was no longer associated with internalized Reelin. When Src family tyrosine kinases were inhibited, internalization of Reelin was severely abrogated, and Reelin colocalized with Dab1 near the plasma membrane for a prolonged period. Taken together, these results indicate that Dab1 regulates both cell surface expression and internalization of Reelin receptors, and these regulations may play a role in correct laminar formation in the developing brain.


Subject(s)
Brain/embryology , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Receptors, Cell Surface/metabolism , Serine Endopeptidases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Biotinylation , Brain/metabolism , COS Cells , Cell Membrane/metabolism , Genetic Vectors , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Transgenic , Microscopy, Confocal , Models, Biological , Neurons/metabolism , Phosphorylation , Protein Transport , Protein-Tyrosine Kinases/metabolism , Receptors, LDL/chemistry , Reelin Protein , Signal Transduction , Time Factors , Transfection , Tyrosine/chemistry , src-Family Kinases/metabolism
18.
Biochem J ; 384(Pt 1): 1-8, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15361067

ABSTRACT

Migration of cells is critical to development of the central nervous system. Reelin, which was identified from the reeler mutant mice having a defect in the multilamellar structure of the brain, is thought to be a key signalling molecule that functions as a cue for determination of cell position. mDab1 (mouse Disabled homologue 1) functions downstream of Reelin. However, the mechanism by which mDab1 regulates cell migration during brain development is unknown. In the present paper, we show that mDab1 associates with N-WASP (neuronal Wiskott-Aldrich syndrome protein) in vitro and in brains of embryonic mice. mDab1 activates N-WASP directly, and induces actin polymerization through the Arp2/3 (actin-related protein 2/3) complex. mDab1 induces formation of filopodia when it is overexpressed in COS-7 cells. This filopodium formation is dependent on N-WASP, because expression of an N-WASP mutant that cannot induce Arp2/3-complex-mediated actin polymerization suppressed filopodium formation. The PTB (phosphotyrosine-binding) domain of mDab1 binds to N-WASP via the NRFY (Asn-Arg-Phe-Tyr) sequence close to the CRIB (Cdc42/Rac-interactive binding) motif of N-WASP and activates N-WASP in vitro. When mDab1 is phosphorylated by Fyn kinase in COS-7 cells, mDab1 is ubiquitinated in a Cbl-dependent manner, and mDab1 does not induce filopodium in the presence of activated Fyn. These findings suggest that mDab1 regulates the actin cytoskeleton through N-WASP, which is negatively regulated by phosphorylation-mediated ubiquitination of mDab1.


Subject(s)
Actins/physiology , Cytoskeleton/physiology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Ubiquitin/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Insecta/cytology , Reelin Protein , Wiskott-Aldrich Syndrome Protein, Neuronal
SELECTION OF CITATIONS
SEARCH DETAIL
...