Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 123
Filtrer
1.
Neurochem Res ; 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-39235580

RÉSUMÉ

Dr. Harish Chandra Pant was Chief of the Section on Neuronal Cytoskeletal Protein Regulation within the National Institute of Neurological Disorders and Stroke at the NIH. A main focus of his group was understanding the mechanisms regulating neuronal cytoskeletal phosphorylation. Phosphorylation of neurofilaments can increase filament stability and confer resistance to proteolysis, but aberrant hyperphosphorylation of neurofilaments can be found in the neurofibrillary tangles that are seen with neurodegenerative diseases like Alzheimer disease (AD). Through his work, Harish would inevitably come across cyclin dependent kinase 5 (Cdk5), a key kinase that can phosphorylate neurofilaments at KSPXK motifs. Cdk5 differs from other Cdks in that its activity is mainly in post-mitotic neurons rather than being involved in the cell cycle in dividing cells. With continued interest in Cdk5, Harish and his group were instrumental in identifying important roles for this neuronal kinase in not only neuronal cytoskeleton phosphorylation but also in neuronal development, synaptogenesis, and neuronal survival. Here, we review the accomplishments of Harish in characterizing the functions of Cdk5 and its involvement in neuronal health and disease.

2.
Commun Biol ; 7(1): 57, 2024 01 08.
Article de Anglais | MEDLINE | ID: mdl-38191649

RÉSUMÉ

The lemur family of protein kinases has gained much interest in recent years as they are involved in a variety of cellular processes including regulation of axonal transport and endosomal trafficking, modulation of synaptic functions, memory and learning, and they are centrally placed in several intracellular signalling pathways. Numerous studies have also implicated role of the lemur kinases in the development and progression of a wide range of cancers, cystic fibrosis, and neurodegenerative diseases. However, parallel discoveries and inaccurate prediction of their kinase activity have resulted in a confusing and misleading nomenclature of these proteins. Herein, a group of international scientists with expertise in lemur family of protein kinases set forth a novel nomenclature to rectify this problem and ultimately help the scientific community by providing consistent information about these molecules.


Sujet(s)
Mucoviscidose , Lemur , Animaux , Protein kinases , Phosphorylation , Transport axonal
4.
Neurochem Res ; 47(9): 2773-2779, 2022 Sep.
Article de Anglais | MEDLINE | ID: mdl-35674931

RÉSUMÉ

Valproic acid (VPA) is a drug used for the treatment of epilepsy, seizures, migraines, and bipolar disorders. Cyclin-dependent kinase 5 (Cdk5) is a Ser/Thr kinase activated by p35 or p39 in neurons and plays a role in a variety of neuronal functions, including psychiatric behaviors. We previously reported that VPA suppressed Cdk5 activity by reducing the expression of p35 in cultured cortical neurons, leaving p39 unchanged. In this study, we asked for the role of Cdk5 in VPA-induced anxiety and depression behaviors. Wild-type (WT) mice displayed increased anxiety and depression after chronic administration of VPA for 14 days, when the expression of p35 was decreased. To clarify their relationship, we used p39 knockout (KO) mice, in which p35 is the only Cdk5 activator. When p39 KO mice were treated chronically with VPA, unexpectedly, they exhibited fewer anxiety and depression behaviors than WT mice. The effects were p39 cdk5r2 gene-dosage dependent. Together, these results indicate that Cdk5-p39 plays a specific role in VPA-induced anxiety and depression behaviors.


Sujet(s)
Anticonvulsivants , Antimaniacodépressifs , Anxiété , Protéines du cytosquelette , Dépression , Protéines liées à des lipides , Acide valproïque , Animaux , Anticonvulsivants/effets indésirables , Anticonvulsivants/usage thérapeutique , Antimaniacodépressifs/effets indésirables , Antimaniacodépressifs/usage thérapeutique , Anxiété/induit chimiquement , Anxiété/génétique , Protéines du cytosquelette/génétique , Dépression/induit chimiquement , Dépression/génétique , Protéines liées à des lipides/génétique , Souris , Souris knockout , Acide valproïque/effets indésirables , Acide valproïque/usage thérapeutique
5.
BMC Biol ; 20(1): 115, 2022 05 17.
Article de Anglais | MEDLINE | ID: mdl-35581583

RÉSUMÉ

BACKGROUND: Activated Cdk5 regulates a number of processes during nervous system formation, including neuronal differentiation, growth cone stabilization, and axonal growth. Cdk5 phosphorylates its downstream substrates located in axonal growth cones, where the highly expressed c-Jun N-terminal kinase (JNK)-interacting protein1 (JIP1) has been implicated as another important regulator of axonal growth. In addition, stringent control of the level of intracellular domain of Notch1 (Notch1-IC) plays a regulatory role in axonal outgrowth during neuronal differentiation. However, whether Cdk5-JIP1-Notch1 cooperate to regulate axonal outgrowth, and the mechanism of such joint contribution to this pathway, is presently unknown, and here we explore their potential interaction. RESULTS: Our interactome screen identified JIP1 as an interactor of p35, a Cdk5 activator, and we sought to explore the relationship between Cdk5 and JIP1 on the regulation of axonal outgrowth. We demonstrate that JIP1 phosphorylated by Cdk5 at Thr205 enhances axonal outgrowth and a phosphomimic JIP1 rescues the axonal outgrowth defects in JIP1-/- and p35-/- neurons. Axonal outgrowth defects caused by the specific increase of Notch1 in JIP1-/- neurons are rescued by Numb-mediated inhibition of Notch1. Finally, we demonstrate that Cdk5 phosphorylation of JIP1 further amplifies the phosphorylation status of yet another Cdk5 substrate E3-ubiquitin ligase Itch, resulting in increased Notch1 ubiquitination. CONCLUSIONS: Our findings identify a potentially critical signaling axis involving Cdk5-JIP1-Itch-Notch1, which plays an important role in the regulation of CNS development. Future investigation into the way this pathway integrates with additional pathways regulating axonal growth will further our knowledge of normal central nervous system development and pathological conditions.


Sujet(s)
Neurones , Transduction du signal , Cellules cultivées , Neurones/métabolisme , Phosphorylation , Transduction du signal/physiologie
6.
Life Sci Alliance ; 5(8)2022 08.
Article de Anglais | MEDLINE | ID: mdl-35470240

RÉSUMÉ

Microtubule (MT) dynamics are modulated through the coordinated action of various MT-associated proteins (MAPs). However, the regulatory mechanisms underlying MT dynamics remain unclear. We show that the MAP7 family protein Map7D2 stabilizes MTs to control cell motility and neurite outgrowth. Map7D2 directly bound to MTs through its N-terminal half and stabilized MTs in vitro. Map7D2 localized prominently to the centrosome and partially on MTs in mouse N1-E115 neuronal cells, which expresses two of the four MAP7 family members, Map7D2 and Map7D1. Map7D2 loss decreased the resistance to the MT-destabilizing agent nocodazole without affecting acetylated/detyrosinated stable MTs, suggesting that Map7D2 stabilizes MTs via direct binding. In addition, Map7D2 loss increased the rate of random cell migration and neurite outgrowth, presumably by disturbing the balance between MT stabilization and destabilization. Map7D1 exhibited similar subcellular localization and gene knockdown phenotypes to Map7D2. However, in contrast to Map7D2, Map7D1 was required for the maintenance of acetylated stable MTs. Taken together, our data suggest that Map7D2 and Map7D1 facilitate MT stabilization through distinct mechanisms in cell motility and neurite outgrowth.


Sujet(s)
Microtubules , Neurones , Animaux , Mouvement cellulaire/génétique , Souris , Protéines associées aux microtubules/génétique , Protéines associées aux microtubules/métabolisme , Microtubules/métabolisme , Neurones/métabolisme , Nocodazole/métabolisme , Nocodazole/pharmacologie
7.
J Proteomics ; 262: 104591, 2022 06 30.
Article de Anglais | MEDLINE | ID: mdl-35430389

RÉSUMÉ

Phosphorylation is a posttranslational modification of proteins that regulates many cellular processes, such as communication between cells, cell proliferation, cell movements, and gene expression. Therefore, many studies have been conducted to determine the significance and function of phosphorylation. These studies involve the identification of phosphorylation site(s), kinases and phosphatases, and regulatory mechanisms. Recently, phosphorylation sites were identified using mass spectrometry and detected by immunoblotting with phosphorylation site-specific antibodies. However, the in vivo phosphorylation profile of the target protein is not easy to grasp, and the quantification of site-specific phosphorylation is challenging if the protein is phosphorylated at multiple sites. Phos-tag is a phospho-affinity SDS-PAGE approach in which phosphorylated proteins are separated depending on the number and sites of phosphorylation during electrophoresis, which overcomes the aforementioned problems. We applied this technique to perform an in vivo analysis of the phosphorylation of many proteins. In this article, we show our results for the phosphorylation of tau protein, p35 Cdk5 activator and GSK3ß to reveal the utility and power of this technique in protein phosphorylation analyses in vivo. SIGNIFICANT: We show the in vivo phosphorylation of tau and two tau kinases analysed by using Phos-tag SDS-PAGE. Tau represents about 12 different phosphoisotypes when expressed in cultured cells. Tau is differently phosphorylated in patients with different tauopathy. Phosphorylation of p35 Cdk5 activator, which suppress the abnormal activation of Cdk5 by cleavage with calpain, is regulated developmentally. The Ser9 phosphorylation is not a proper marker of the GSK3ß activity in vivo.


Sujet(s)
Protéines tau , Électrophorèse sur gel de polyacrylamide , Glycogen synthase kinase 3 beta/métabolisme , Humains , Phosphorylation , Pyridines , Protéines tau/composition chimique , Protéines tau/métabolisme
8.
J Biochem ; 170(6): 729-738, 2022 Jan 07.
Article de Anglais | MEDLINE | ID: mdl-34523681

RÉSUMÉ

Lemur tail kinase 1 (LMTK1), previously called apoptosis-associated tyrosine kinase (AATYK), is an endosomal Ser/Thr kinase. We recently reported that LMTK1 regulates axon outgrowth, dendrite arborization and spine formation via Rab11-mediated vesicle transport. Rab11, a small GTPase regulating recycling endosome trafficking, is shown to be associated with late-onset Alzheimer's disease (LOAD). In fact, genome-wide association studies identified many proteins regulating vesicle transport as risk factors for LOAD. Furthermore, LMTK1 has been reported to be a risk factor for frontotemporal dementia. Then, we hypothesized that LMTK1 contributes to AD development through vesicle transport and examined the effect of LMTK1 on the cellular localization of AD-related proteins, amyloid precursor protein (APP) and ß-site APP cleaving enzyme 1 (BACE1). The ß-cleavage of APP by BACE1 is the initial and rate-limiting step in Aß generation. We found that LMTK1 accumulated BACE1, but not APP, to the perinuclear endosomal compartment, whereas the kinase-negative(kn) mutant of LMTK1A did not. The ß-C-terminal fragment was prone to increase under overexpression of LMTK1A kn. Moreover, the expression level of LMTK1A was reduced in AD brains. These results suggest the possibility that LMTK1 is involved in AD development through the regulation of the proper endosomal localization of BACE1.


Sujet(s)
Maladie d'Alzheimer/enzymologie , Amyloid precursor protein secretases/métabolisme , Protéines régulatrices de l'apoptose/métabolisme , Aspartic acid endopeptidases/métabolisme , Endosomes/enzymologie , Protein-tyrosine kinases/métabolisme , Maladie d'Alzheimer/génétique , Amyloid precursor protein secretases/génétique , Précurseur de la protéine bêta-amyloïde/génétique , Précurseur de la protéine bêta-amyloïde/métabolisme , Animaux , Protéines régulatrices de l'apoptose/génétique , Aspartic acid endopeptidases/génétique , Cellules CHO , Cellules COS , Chlorocebus aethiops , Cricetulus , Endosomes/génétique , Cellules HEK293 , Humains , Protein-tyrosine kinases/génétique
9.
Neurobiol Aging ; 108: 72-79, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34536819

RÉSUMÉ

Tauopathies are neurodegenerative diseases that are characterized by pathological accumulation of tau protein. Tau is hyperphosphorylated in the brain of tauopathy patients, and this phosphorylation is proposed to play a role in disease development. However, it has been unclear whether phosphorylation is different among different tauopathies. Here, we investigated the phosphorylation states of tau in several tauopathies, including corticobasal degeneration, Pick's disease, progressive supranuclear palsy (PSP), argyrophilic grain dementia (AGD) and Alzheimer's disease (AD). Analysis of tau phosphorylation profiles using Phos-tag SDS-PAGE revealed distinct phosphorylation of tau in different tauopathies, whereas similar phosphorylation patterns were found within the same tauopathy. For PSP, we found 2 distinct phosphorylation patterns suggesting that PSP may consist of 2 different related diseases. Immunoblotting with anti-phospho-specific antibodies showed different site-specific phosphorylation in the temporal lobes of patients with different tauopathies. AD brains showed increased phosphorylation at Ser202, Thr231 and Ser235, Pick's disease brains showed increased phospho-Ser202, and AGD brains showed increased phospho-Ser396. The cis conformation of the peptide bond between phospho-Thr231 and Pro232 (cis ptau) was increased in AD and AGD. These results indicate that while tau is differently phosphorylated in tauopathies, a similar pathological mechanism may occur in AGD and AD patients. The present data provide useful information regarding tau pathology and diagnosis of tauopathies.


Sujet(s)
Encéphale/métabolisme , Tauopathies/métabolisme , Protéines tau/métabolisme , Maladie d'Alzheimer/diagnostic , Maladie d'Alzheimer/métabolisme , Marqueurs biologiques/métabolisme , Dégénérescence corticobasale/diagnostic , Dégénérescence corticobasale/métabolisme , Démence/diagnostic , Démence/métabolisme , Électrophorèse sur gel de polyacrylamide/méthodes , Humains , Immunotransfert/méthodes , Phosphorylation , Démence de Pick/diagnostic , Démence de Pick/métabolisme , Paralysie supranucléaire progressive/diagnostic , Paralysie supranucléaire progressive/métabolisme , Tauopathies/diagnostic , Lobe temporal/métabolisme
10.
Sci Rep ; 10(1): 15461, 2020 09 22.
Article de Anglais | MEDLINE | ID: mdl-32963255

RÉSUMÉ

Lemur tail kinase 1 (LMTK1), previously called Apoptosis-Associated Tyrosine Kinase (AATYK), remains an uncharacterized Ser/Thr protein kinase that is predominantly expressed in the brain. It is recently reported that LMTK1A, an isoform of LMTK1, binds to recycling endosomes through its palmitoylation and regulates endosomal trafficking by suppressing the activity of Rab11 small GTPase. In neurons, knockdown or knockout of LMTK1 results in longer axons, greater branching of dendrites and increased number of spines, suggesting that LMTK1 plays a role in neuronal circuit formation. However, its in vivo function remained to be investigated. Here, we examined the brain structures and behaviors of LMTK1 knockout (KO) mice. LMTK1 was expressed in most neurons throughout the brain. The overall brain structure appeared to be normal in LMTK1 KO mice, but the numbers of synapses were increased. LMTK1 KO mice had a slight impairment in memory formation and exhibited distinct psychiatric behaviors such as hyperactivity, impulsiveness and high motor coordination without social interaction deficits. Some of these abnormal behaviors represent core features of attention deficit hyperactive disorder (ADHD), suggesting the possible involvement of LMTK1 in the pathogenesis of ADHD.


Sujet(s)
Protéines régulatrices de l'apoptose/physiologie , Trouble déficitaire de l'attention avec hyperactivité/anatomopathologie , Comportement animal , Encéphale/physiopathologie , Comportement impulsif , Neurones/anatomopathologie , Protein-tyrosine kinases/physiologie , Animaux , Trouble déficitaire de l'attention avec hyperactivité/étiologie , Trouble déficitaire de l'attention avec hyperactivité/psychologie , Femelle , Souris , Souris de lignée C57BL , Souris de lignée ICR , Souris knockout , Neurones/métabolisme
11.
Elife ; 92020 07 22.
Article de Anglais | MEDLINE | ID: mdl-32697196

RÉSUMÉ

Abnormal α-synuclein aggregation has been implicated in several diseases and is known to spread in a prion-like manner. There is a relationship between protein aggregate structure (strain) and clinical phenotype in prion diseases, however, whether differences in the strains of α-synuclein aggregates account for the different pathologies remained unclear. Here, we generated two types of α-synuclein fibrils from identical monomer and investigated their seeding and propagation ability in mice and primary-cultured neurons. One α-synuclein fibril induced marked accumulation of phosphorylated α-synuclein and ubiquitinated protein aggregates, while the other did not, indicating the formation of α-synuclein two strains. Notably, the former α-synuclein strain inhibited proteasome activity and co-precipitated with 26S proteasome complex. Further examination indicated that structural differences in the C-terminal region of α-synuclein strains lead to different effects on proteasome activity. These results provide a possible molecular mechanism to account for the different pathologies induced by different α-synuclein strains.


Sujet(s)
Neurones/anatomopathologie , Proteasome endopeptidase complex/métabolisme , alpha-Synucléine/métabolisme , Animaux , Cellules cultivées , Protéines fongiques/métabolisme , Mâle , Souris , Souris de lignée C57BL , Saccharomyces cerevisiae/composition chimique
12.
Front Mol Neurosci ; 13: 112, 2020.
Article de Anglais | MEDLINE | ID: mdl-32714146

RÉSUMÉ

Neurons extend long processes known as axons and dendrites, through which they communicate with each other. The neuronal circuits formed by the axons and dendrites are the structural basis of higher brain functions. The formation and maintenance of these processes are essential for physiological brain activities. Membrane components, both lipids, and proteins, that are required for process formation are supplied by vesicle transport. Intracellular membrane trafficking is regulated by a family of Rab small GTPases. A group of Rabs regulating endosomal trafficking has been studied mainly in nonpolarized culture cell lines, and little is known about their regulation in polarized neurons with long processes. As shown in our recent study, lemur tail (former tyrosine) kinase 1 (LMTK1), an as yet uncharacterized Ser/Thr kinase associated with Rab11-positive recycling endosomes, modulates the formation of axons, dendrites, and spines in cultured primary neurons. LMTK1 knockdown or knockout (KO) or the expression of a kinase-negative mutant stimulates the transport of endosomal vesicles in neurons, leading to the overgrowth of axons, dendrites, and spines. More recently, we found that LMTK1 regulates TBC1D9B Rab11 GAP and proposed the Cdk5/p35-LMTK1-TBC1D9B-Rab11 pathway as a signaling cascade that regulates endosomal trafficking. Here, we summarize the biochemical, cell biological, and physiological properties of LMTK1.

13.
J Biochem ; 168(1): 23-32, 2020 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-32044995

RÉSUMÉ

Lemur kinase 1 (LMTK1) is a membrane-bound Ser/Thr kinase that is expressed in neurons. There are two splicing variants of LMTK1 with different membrane binding modes, viz., cytosolic LMTK1A that binds to membranes through palmitoylation at the N-terminal cysteines and LMTK1B, an integral membrane protein with transmembrane sequences. We recently reported that LMTK1A regulates axon outgrowth and spine formation in neurons. However, data about LMTK1B are scarce. We analysed the expression and cellular localization of LMTK1B along with its role in axon and spine formation. We found that both LMTK1B and LMTK1A were expressed equally in the cerebral cortex and cerebellum of the mouse brain. Similar to LMTK1A, the wild type of LMTK1B was localized to Rab11-positive pericentrosomal compartment. The kinase negative (kn) mutant of LMTK1B was found to be associated with an increase in the tubular form of endoplasmic reticulum (ER), which was not the case with LMTK1A kn. Furthermore, unlike LMTK1A kn, LMTK1B kn did not stimulate the axon outgrowth and spine formation. These results suggest that while LMTK1A and LMTK1B share a common function in recycling endosomal trafficking at the pericentrosomal compartment, LMTK1B has an additional unique function in vesicle transport in the ER region.


Sujet(s)
Protéines régulatrices de l'apoptose/métabolisme , Axones/physiologie , Encéphale/croissance et développement , Excroissance neuronale/physiologie , Protein-tyrosine kinases/métabolisme , Fractions subcellulaires/métabolisme , Animaux , Encéphale/métabolisme , Cellules cultivées , Cricetinae , Réticulum endoplasmique/métabolisme , Endosomes/métabolisme , Humains , Souris , Souris de lignée C57BL , Souris knockout , Isoformes de protéines
14.
J Neurosci ; 39(48): 9491-9502, 2019 11 27.
Article de Anglais | MEDLINE | ID: mdl-31628178

RÉSUMÉ

Dendritic spines are postsynaptic protrusions at excitatory synapses that are critical for proper neuronal synaptic transmission. While lipid and protein membrane components are necessary for spine formation, it is largely unknown how they are recruited to developing spines. Endosomal trafficking is one mechanism that may influence this development. We recently reported that Lemur kinase 1A (LMTK1A), a membrane-bound Ser/Thr kinase, regulates trafficking of endosomes in neurons. LMTK1 has been shown to be a p35 Cdk5 activator-binding protein and a substrate for Cdk5-p35; however, its neuronal function has not been sufficiently studied. Here, we investigate the role of LMTK1 in spine formation. Depletion of LMTK1 increases spine formation, maturation, and density in primary cultured neurons and in mouse brain of either sex. Additionally, expression of kinase-negative LMTK1 stimulates spine formation in primary neurons and in vivo LMTK1 controls spine formation through Rab11, a regulator of recycling endosome trafficking. We identify TBC1D9B, a Rab11A GTPase-activating protein (Rab11A GAP), as a LMTK1 binding protein, and find that TBC1D9B mediates LMTK1 activity on Rab11A. TBC1D9B inactivates Rab11A under the control of LMTK1A. Further, by analyzing the effect of decreased TBC1D9B expression in primary neurons, we demonstrate that TBC1D9B indeed regulates spine formation. This is the first demonstration of the biological function of TBC1D9B. Together, with the regulation of LMTK1 by Cdk5-p35, we propose the Cdk5-LMTK1-TBC1D9B-Rab11A cascade as a novel signaling mechanism regulating endosomal transport for synapse formation and function.SIGNIFICANCE STATEMENT Dendritic spines are postsynaptic specializations essential for synaptic transmission. However, it is not known how critical membrane components are recruited to spines for their formation. Endosomal trafficking is one such mechanism that may mediate this process. Here we investigate regulators of endosomal trafficking and their contribution to spine formation. We identify two novel factors, LMTK1 and TBC1D9B, which regulate spine formation upstream of Rab11A, a small GTPase. LMTK1 is a membrane bound Ser/Thr kinase regulated by Cdk5-p35, and TBC1D9B is a recently identified Rab11 GAP. LMTK1 controls the GAP activity of TBC1D9B on Rab11A, and TBC1D9B mediates the LMTK1 activity on Rab11A. We propose the Cdk5-LMTK1-TBC1D9B-Rab11A cascade as a novel mechanism controlling spine formation and function.


Sujet(s)
Protéines régulatrices de l'apoptose/métabolisme , Épines dendritiques/métabolisme , Endosomes/métabolisme , Protein-tyrosine kinases/métabolisme , Transduction du signal/physiologie , Protéines G rab/métabolisme , Animaux , Protéines régulatrices de l'apoptose/génétique , Cellules COS , Chlorocebus aethiops , Épines dendritiques/génétique , Endosomes/génétique , Femelle , Cellules HEK293 , Hippocampe/cytologie , Hippocampe/métabolisme , Humains , Souris , Souris de lignée ICR , Souris knockout , Grossesse , Transport des protéines/physiologie , Protein-tyrosine kinases/génétique , Protéines G rab/génétique
15.
Stem Cell Reports ; 13(4): 684-699, 2019 10 08.
Article de Anglais | MEDLINE | ID: mdl-31543469

RÉSUMÉ

Mutations in the microtubule-associated protein tau (MAPT) gene are known to cause familial frontotemporal dementia (FTD). The R406W tau mutation is a unique missense mutation whose patients have been reported to exhibit Alzheimer's disease (AD)-like phenotypes rather than the more typical FTD phenotypes. In this study, we established patient-derived induced pluripotent stem cell (iPSC) models to investigate the disease pathology induced by the R406W mutation. We generated iPSCs from patients and established isogenic lines using CRISPR/Cas9. The iPSCs were induced into cerebral organoids, which were dissociated into cortical neurons with high purity. In this neuronal culture, the mutant tau protein exhibited reduced phosphorylation levels and was increasingly fragmented by calpain. Furthermore, the mutant tau protein was mislocalized and the axons of the patient-derived neurons displayed morphological and functional abnormalities, which were rescued by microtubule stabilization. The findings of our study provide mechanistic insight into tau pathology and a potential for therapeutic intervention.


Sujet(s)
Allèles , Substitution d'acide aminé , Démence frontotemporale/étiologie , Cellules souches pluripotentes induites/métabolisme , Mutation , Protéines tau/génétique , Calpain/métabolisme , Évolution de la maladie , Prédisposition aux maladies , Démence frontotemporale/métabolisme , Démence frontotemporale/physiopathologie , Humains , Cellules souches pluripotentes induites/cytologie , Mitochondries/métabolisme , Neurones/métabolisme , Phosphorylation , Phosphotransferases/métabolisme , Protéines tau/métabolisme
16.
Front Neurosci ; 13: 595, 2019.
Article de Anglais | MEDLINE | ID: mdl-31258461

RÉSUMÉ

Parkinson's disease (PD) and related disorders are characterized by filamentous or fibrous structures consisting of abnormal α-synuclein in the brains of patients, and the distributions and spread of these pathologies are closely correlated with disease progression. L-DOPA (a dopamine precursor) is the most effective therapy for PD, but it remains unclear whether the drug has any effect on the formation and propagation of pathogenic abnormal α-synuclein in vivo. Here, we tested whether or not L-DOPA influences the prion-like spread of α-synuclein pathologies in a wild-type (WT) mouse model of α-synuclein propagation. To quantitative the pathological α-synuclein in mice, we prepared brain sections stained with an anti-phosphoSer129 (PS129) antibody after pretreatments with autoclaving and formic acid, and carefully analyzed positive aggregates on multiple sections covering the areas of interest using a microscope. Notably, a significant reduction in the accumulation of phosphorylated α-synuclein was detected in substantia nigra of L-DOPA/benserazide (a dopamine decarboxylase inhibitor)-treated mice, compared with control mice. These results suggest that L-DOPA may slow the progression of PD in vivo by suppressing the aggregation of α-synuclein in dopaminergic neurons and the cell-to-cell propagation of abnormal α-synuclein. This is the first report describing the suppressing effect of L-DOPA/benserazide on the propagation of pathological α-synuclein. The experimental protocols and detection methods in this study are expected to be useful for evaluation of drug candidates or new therapies targeting the propagation of α-synuclein.

17.
J Biol Chem ; 294(30): 11433-11444, 2019 07 26.
Article de Anglais | MEDLINE | ID: mdl-31171723

RÉSUMÉ

Tau is a microtubule-associated protein expressed in neuronal axons. Hyperphosphorylated tau is a major component of neurofibrillary tangles, a pathological hallmark of Alzheimer's disease (AD). Hyperphosphorylated tau aggregates are also found in many neurodegenerative diseases, collectively referred to as "tauopathies," and tau mutations are associated with familial frontotemporal lobar degeneration (FTLD). Previous studies have generated transgenic mice with mutant tau as tauopathy models, but nonhuman primates, which are more similar to humans, may be a better model to study tauopathies. For example, the common marmoset is poised as a nonhuman primate model for investigating the etiology of age-related neurodegenerative diseases. However, no biochemical studies of tau have been conducted in marmoset brains. Here, we investigated several important aspects of tau, including expression of different tau isoforms and its phosphorylation status, in the marmoset brain. We found that marmoset tau does not possess the "primate-unique motif" in its N-terminal domain. We also discovered that the tau isoform expression pattern in marmosets is more similar to that of mice than that of humans, with adult marmoset brains expressing only four-repeat tau isoforms as in adult mice but unlike in adult human brains. Of note, tau in brains of marmoset newborns was phosphorylated at several sites associated with AD pathology. However, in adult marmoset brains, much of this phosphorylation was lost, except for Ser-202 and Ser-404 phosphorylation. These results reveal key features of tau expression and phosphorylation in the marmoset brain, a potentially useful nonhuman primate model of neurodegenerative diseases.


Sujet(s)
Encéphale/métabolisme , Isoformes de protéines/métabolisme , Protéines tau/métabolisme , Maladie d'Alzheimer/métabolisme , Animaux , Callithrix , Cellules cultivées , ADN complémentaire/génétique , Humains , Souris , Souris transgéniques , Phosphorylation , Phylogenèse , Isoformes de protéines/génétique , Protéines tau/génétique
18.
J Biol Chem ; 294(28): 10886-10899, 2019 07 12.
Article de Anglais | MEDLINE | ID: mdl-31138646

RÉSUMÉ

A hallmark of Alzheimer's disease (AD) pathology is the appearance of senile plaques, which are composed of ß-amyloid (Aß) peptides. Aß is produced by sequential cleavages of amyloid precursor protein (APP) by ß- and γ-secretases. These cleavages take place in endosomes during intracellular trafficking of APP through the endocytic and recycling pathways. Genome-wide association studies have identified several risk factors for late-onset AD, one of which is CD2-associated protein (CD2AP), an adaptor molecule that regulates membrane trafficking. Although CD2AP's involvement in APP trafficking has recently been reported, how APP trafficking is regulated remains unclear. We sought to address this question by investigating the effect of CD2AP overexpression or knockdown on the intracellular APP distribution and degradation of APP in cultured COS-7 and HEK293 cells. We found that overexpression of CD2AP increases the localization of APP to Rab7-positive late endosomes, and decreases its localization to Rab5-positive early endosomes. CD2AP overexpression accelerated the onset of APP degradation without affecting its degradation rate. Furthermore, nutrient starvation increased the localization of APP to Rab7-positive late endosomes, and CD2AP overexpression stimulated starvation-induced lysosomal APP degradation. Moreover, the effect of CD2AP on the degradation of APP was confirmed by CD2AP overexpression and knockdown in primary cortical neurons from mice. We conclude that CD2AP accelerates the transfer of APP from early to late endosomes. This transfer in localization stimulates APP degradation by reducing the amount of time before degradation initiation. Taken together, these results may explain why impaired CD2AP function is a risk factor for AD.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Précurseur de la protéine bêta-amyloïde/métabolisme , Protéines du cytosquelette/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Maladie d'Alzheimer/métabolisme , Amyloid precursor protein secretases/métabolisme , Peptides bêta-amyloïdes/métabolisme , Précurseur de la protéine bêta-amyloïde/physiologie , Animaux , Cellules COS , Chlorocebus aethiops , Protéines du cytosquelette/génétique , Endosomes/métabolisme , Étude d'association pangénomique , Cellules HEK293 , Humains , Lysosomes/métabolisme , Neurones/métabolisme , Plaque amyloïde/métabolisme , Transport des protéines , Protéolyse , Vésicules de transport/métabolisme
19.
EMBO Rep ; 20(4)2019 04.
Article de Anglais | MEDLINE | ID: mdl-30804014

RÉSUMÉ

Rab family small GTPases are master regulators of distinct steps of intracellular vesicle trafficking in eukaryotic cells. GDP-bound cytoplasmic forms of Rab proteins are prone to aggregation due to the exposure of hydrophobic groups but the machinery that determines the fate of Rab species in the cytosol has not been elucidated in detail. In this study, we find that BAG6 (BAT3/Scythe) predominantly recognizes a cryptic portion of GDP-associated Rab8a, while its major GTP-bound active form is not recognized. The hydrophobic residues of the Switch I region of Rab8a are essential for its interaction with BAG6 and the degradation of GDP-Rab8a via the ubiquitin-proteasome system. BAG6 prevents the excess accumulation of inactive Rab8a, whose accumulation impairs intracellular membrane trafficking. BAG6 binds not only Rab8a but also a functionally distinct set of Rab family proteins, and is also required for the correct distribution of Golgi and endosomal markers. From these observations, we suggest that Rab proteins represent a novel set of substrates for BAG6, and the BAG6-mediated pathway is associated with the regulation of membrane vesicle trafficking events in mammalian cells.


Sujet(s)
Chaperons moléculaires/métabolisme , Protéines G rab/métabolisme , Séquence d'acides aminés , Animaux , Lignée cellulaire , Membrane cellulaire/métabolisme , Cytoplasme/métabolisme , Cytosol/métabolisme , Réticulum endoplasmique/métabolisme , Endosomes/métabolisme , Délétion de gène , Appareil de Golgi/métabolisme , Humains , Modèles biologiques , Chaperons moléculaires/génétique , Liaison aux protéines , Motifs et domaines d'intéraction protéique , Transport des protéines , Protéolyse , Petit ARN interférent/génétique , Ubiquitine/métabolisme , Protéines G rab/composition chimique , Protéines G rab/génétique
20.
Biochem Biophys Res Commun ; 510(3): 370-375, 2019 03 12.
Article de Anglais | MEDLINE | ID: mdl-30712943

RÉSUMÉ

Serotonin (5-HT) is a major neurotransmitter in mammalian brains and is involved in brain development and psychiatric disorders. The 5-HT1A receptor (5-HT1AR) is a G-protein-coupled receptor (GPCR) associated with an inhibitory G-protein (Gi) with the widest and most abundant expression. It is not known; however, how expression or activity of 5-HTlAR is regulated. We studied here phosphorylation of 5-HT1AR by cyclin-dependent kinase 5 (Cdk5), a neuron-specific membrane-bound Ser/Thr kinase that is activated by binding of the p35 Cdk5 activator. 5-HT1AR was phosphorylated by the Cdk5-p35 complex at Thr314 in the third cytoplasmic loop. The phosphorylation stimulated the degradation of 5-HT1AR by the proteasome, resulting in neutralization of the inhibitory action of 5-HT1AR on intracellular cAMP concentration. These results suggest that Cdk5-p35 modulates 5-HT signaling through phosphorylation-dependent degradation of 5-HTlAR.


Sujet(s)
Kinase-5 cycline-dépendante/métabolisme , Proteasome endopeptidase complex/métabolisme , Récepteur de la sérotonine de type 5-HT1A/métabolisme , Animaux , Cellules CHO , Cellules COS , Membrane cellulaire/métabolisme , Chlorocebus aethiops , Cricetulus , Régulation négative , Cellules HEK293 , Humains , Protéines de tissu nerveux/métabolisme , Phosphorylation , Sérotonine/pharmacologie , Transduction du signal
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE