Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 8 de 8
Filtrer
1.
Aging Cell ; 20(12): e13515, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34821024

RÉSUMÉ

Alzheimer's disease (AD) is characterized by the progressive accumulation of ß-amyloid (Aß)-containing amyloid plaques, and microglia play a critical role in mediating Aß clearance. Mounting evidence has confirmed that the ability of microglia in clearing Aß decreased with aging and AD progress, but the underlying mechanisms are unclear. Previously, we have demonstrated that Nogo receptor (NgR), a receptor for three axon growth inhibitors associated with myelin, can decrease adhesion and migration of microglia to fibrils Aß with aging. However, whether NgR expressed on microglia affect microglia phagocytosis of fibrils Aß with aging remains unclear. Here, we found that aged but not young microglia showed increased NgR expression and decreased Aß phagocytosis in APP/PS1 transgenic mice. NgR knockdown APP/PS1 mice showed simultaneous reduced amyloid burden and improved spatial learning and memory, which were associated with increased Aß clearance. Importantly, Nogo-P4, an agonist of NgR, enhanced the protein level of p-Smad2/3, leading to a significant transcriptional inhibition of CD36 gene expression, which in turn decreased the microglial phagocytosis of Aß. Moreover, ROCK accounted for Nogo-P4-induced activation of Smad2/3 signaling. Finally, the decreasing effect of NgR on microglial Aß uptake was confirmed in a mouse model of intra-hippocampal fAß injection. Our findings suggest that NgR may play an important role in the regulation of Aß homeostasis, and has potential as a therapeutic target for AD.


Sujet(s)
Maladie d'Alzheimer/génétique , Peptides bêta-amyloïdes/métabolisme , Microglie/métabolisme , Récepteurs Nogo/génétique , Maladie d'Alzheimer/physiopathologie , Animaux , Modèles animaux de maladie humaine , Évolution de la maladie , Souris , Transfection
2.
J Tradit Chin Med ; 41(1): 59-67, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33522198

RÉSUMÉ

OBJECTIVE: To evaluate the anti-apoptotic efficacy of Qingnao Yizhi formula (,QNYZ) in cultured cerebral cortical neuronal cells (CNCs) and the regulation of the NogoA-Nogo receptor (NgR)/Rho-Rho kinase (ROCK) signaling pathway. METHODS: Primary cultured CNCs were randomly divided into the following groups: normal control group (N-C), hypoxia-reoxygenation group (H/R), high-dose QNYZ group (Q-H), low-dose QNYZ group (Q-L) butylphthalide (NBP) group, and Y-27632 (a selective ROCK transduction pathway inhibiter) group. Except those in the N-C group, CNCs were placed in hypoxic conditions for 24 h and then in reoxygenation conditions for 24 h. Cell media was changed every 48 h, and various assays were performed on the 7th day. Cell viability was evaluated by measuring mitochondrial dehydrogenase activity, using a CCK-8 assay, in triplicate. Synapsin (SYN) protein concentrations were evaluated by enzyme-linked immunosorbent assay. NogoA and RhoA protein expression were evaluated through Western blotting. The gene expression of NogoA, NgR, RhoA, and ROCK was evaluated by reverse transcription-polymerase chain reaction. Cell apoptosis was measured using a terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling assay. RESULTS: Compared with the N-C group, the cell viability of the H/R group decreased significantly (P < 0.05). The cell viability values for the Q-H and Q-L groups increased compared with that for the H/R group, and the difference was significant for the Q-H group (P < 0.05). The NogoA and RhoA protein levels and the NogoA, NgR, RhoA, and ROCK mRNA expression levels increased in the H/R group, compared with the N-C group, and decreased significantly in the Q-H and Q-L groups (P < 0.05) and in the Y-27632 group (P < 0.05) compared with the H/R group. The SYN levels in the Q-H, Q-L, and NBP groups significantly increased compared with that in the H/R group (P < 0.05). Compared with the H/R group, the numbers of apoptotic cells in the Q-H, Q-L, and NBP groups significantly decreased (P < 0.05). CONCLUSION: The presented study demonstrated that QNYZ exerted anti-apoptotic effects on H/R-induced CNCs, possibly through the modulation of the NogoA-NgR/Rho-ROCK signaling pathway and the promotion of synaptic plasticity in H/R CNCs.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Médicaments issus de plantes chinoises/pharmacologie , Hypoxie/métabolisme , Neurones/effets des médicaments et des substances chimiques , Protéines Nogo/métabolisme , Récepteurs Nogo/métabolisme , Oxygène/métabolisme , rho-Associated Kinases/métabolisme , Alpinia , Animaux , Cellules cultivées , Cortex cérébral/cytologie , Cortex cérébral/effets des médicaments et des substances chimiques , Femelle , Humains , Hypoxie/traitement médicamenteux , Hypoxie/génétique , Mâle , Neurones/cytologie , Neurones/métabolisme , Protéines Nogo/génétique , Récepteurs Nogo/génétique , Extraits de plantes , Rats , Rat Wistar , Transduction du signal/effets des médicaments et des substances chimiques , rho-Associated Kinases/génétique
3.
Proc Natl Acad Sci U S A ; 117(21): 11744-11752, 2020 05 26.
Article de Anglais | MEDLINE | ID: mdl-32404418

RÉSUMÉ

Auditory experience drives neural circuit refinement during windows of heightened brain plasticity, but little is known about the genetic regulation of this developmental process. The primary auditory cortex (A1) of mice exhibits a critical period for thalamocortical connectivity between postnatal days P12 and P15, during which tone exposure alters the tonotopic topography of A1. We hypothesized that a coordinated, multicellular transcriptional program governs this window for patterning of the auditory cortex. To generate a robust multicellular map of gene expression, we performed droplet-based, single-nucleus RNA sequencing (snRNA-seq) of A1 across three developmental time points (P10, P15, and P20) spanning the tonotopic critical period. We also tone-reared mice (7 kHz pips) during the 3-d critical period and collected A1 at P15 and P20. We identified and profiled both neuronal (glutamatergic and GABAergic) and nonneuronal (oligodendrocytes, microglia, astrocytes, and endothelial) cell types. By comparing normal- and tone-reared mice, we found hundreds of genes across cell types showing altered expression as a result of sensory manipulation during the critical period. Functional voltage-sensitive dye imaging confirmed GABA circuit function determines critical period onset, while Nogo receptor signaling is required for its closure. We further uncovered previously unknown effects of developmental tone exposure on trajectories of gene expression in interneurons, as well as candidate genes that might execute tonotopic plasticity. Our single-nucleus transcriptomic resource of developing auditory cortex is thus a powerful discovery platform with which to identify mediators of tonotopic plasticity.


Sujet(s)
Cortex auditif , Noyau de la cellule/métabolisme , ARN , Analyse sur cellule unique/méthodes , Transcriptome/génétique , Animaux , Cortex auditif/croissance et développement , Cortex auditif/métabolisme , Régulation de l'expression des gènes au cours du développement/génétique , Glutamate decarboxylase/génétique , Glutamate decarboxylase/métabolisme , Souris , Récepteurs Nogo/génétique , Récepteurs Nogo/métabolisme , ARN/analyse , ARN/génétique , ARN/métabolisme , Analyse de séquence d'ARN/méthodes
4.
Neuropediatrics ; 50(6): 387-390, 2019 12.
Article de Anglais | MEDLINE | ID: mdl-31370080

RÉSUMÉ

BACKGROUND: Leukoencephalopathy associated with dysmorphic features may be attributed to chromosomal abnormalities such as 17p13.3 microdeletion syndrome. CASE: A 19-year-old female patient was referred to our hospital for diagnostic evaluation of her leukoencephalopathy. She demonstrated moderate intellectual disability, minor dysmorphic features, and short stature. Serial brain magnetic resonance images obtained within a 16-year interval revealed prolonged T2 signals in the deep cerebral white matter with enlarged Virchow-Robin spaces. A nonsymptomatic atlas anomaly was also noted. Using microarray-based comparative genomic hybridization, we identified a 2.2-Mb terminal deletion at 17p13.3, encompassing YWHAE, CRK, and RTN4RL1 but not PAFAH1B1. CONCLUSION: Except for atlas anomaly, the patient's clinical and imaging findings were compatible with the diagnosis of 17p13.3 microdeletion syndrome. The white matter abnormality was static and nonprogressive. The association between the atlas abnormality and this deletion remains elusive. We note the importance of exploring submicroscopic chromosomal imbalance when patients show prominent but static white matter abnormalities with discrepantly mild and stable neurological signs.


Sujet(s)
Délétion de segment de chromosome , Chromosomes humains de la paire 17/génétique , Leucoencéphalopathies/génétique , Protéines 14-3-3/génétique , Taille , Atlas (anatomie)/malformations , Atlas (anatomie)/imagerie diagnostique , Femelle , Humains , Déficience intellectuelle/étiologie , Déficience intellectuelle/génétique , Leucoencéphalopathies/imagerie diagnostique , Imagerie par résonance magnétique , Récepteurs Nogo/génétique , Protéines proto-oncogènes c-crk/génétique , Substance blanche/imagerie diagnostique , Jeune adulte
5.
BMC Complement Altern Med ; 17(1): 346, 2017 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-28668079

RÉSUMÉ

BACKGROUND: Axon growth inhibitory factors NogoA/Nogo receptor (NgR) and its signaling pathways RhoA/Rho kinase (ROCK) play a critical role in the repair of nerve damage in multiple sclerosis (MS). Bu Shen Yi Sui Capsule (BSYSC) is an effective Chinese formula utilized to treat MS in clinical setting and noted for its potent neuroprotective effects. In this study, we focus on the effects of BSYSC on promoting nerve repair and the underlying mechanisms in mice with experimental autoimmune encephalomyelitis (EAE), an animal model of MS. METHODS: The EAE mouse model was induced by injecting subcutaneously with myelin oligodendrocyte glycoprotein (MOG) 35-55 supplemented with pertussis toxin. BSYSC was orally administrated at dose of 3.0 g/kg once a day for 40 days. The levels of protein gene product (PGP) 9.5, p-Tau, growth associated protein (GAP) -43, KI67 and Nestin in the brain or spinal cord on 20 and 40 day post-induction (dpi) were detected via immunofluorescence and Western blot analysis. Furthermore, NogoA/NgR and RhoA/ROCK signaling molecules were studied by qRT-PCR and Western blot analysis. RESULTS: Twenty or 40 days of treatment with BSYSC increased markedly PGP9.5 and GAP-43 levels, reduced p-Tau in the brain or spinal cord of mice with EAE. In addition, BSYSC elevated significantly the expression of KI67 and Nestin in the spinal cord 40 dpi. Further study showed that the activation of NogoA/NgR and RhoA/ROCK were suppressed by the presence of BSYSC. CONCLUSIONS: BSYSC could attenuate axonal injury and promote repair of axonal damage in EAE mice in part through the down-regulation of NogoA/NgR and RhoA/ROCK signaling pathways.


Sujet(s)
Médicaments issus de plantes chinoises/administration et posologie , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Protéines Nogo/métabolisme , Récepteurs Nogo/métabolisme , rho-Associated Kinases/métabolisme , Protéine G RhoA/métabolisme , Animaux , Encéphalomyélite auto-immune expérimentale/génétique , Encéphalomyélite auto-immune expérimentale/métabolisme , Femelle , Humains , Souris , Souris de lignée C57BL , Protéines Nogo/génétique , Récepteurs Nogo/génétique , Transduction du signal , rho-Associated Kinases/génétique , Protéine G RhoA/génétique
6.
Neuroreport ; 28(9): 533-539, 2017 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-28489665

RÉSUMÉ

Neuronal regeneration and axonal regrowth mechanisms in the injured mammalian central nervous system are largely unknown. As part of a major pathway for inhibiting axonal regeneration, activated neuronal glycosylphosphatidylinositol-anchored Nogo receptor (NgR) interacts with LINGO-1 and p75NTR to form a complex at the cell surface. However, it was found in our previous report that upregulation of NgR stimulated by injury plays a key role in neuronal regeneration in the neonatal cortex freeze-lesion model, but its downstream signalling remains elusive. In the present study, the novel regulatory role of NgR in a serine-threonine kinase WNK1 was identified. NgR's transcriptional regulation of WNK1 was identified by RT-qPCR and semiquantitative western blot after the overexpression or knockdown of NgR, and the regulation is specific to WNK1, which is not the same for its family members, WNK2, WNK3 and WNK4. Furthermore, NgR inhibition by NEP fails to affect WNK1, which indicates that WNK1 functions outside of the Nogo-A/NgR pathway. By performing a proliferation, migration and axonal extension assay, we also identified that overexpressed NgR critically regulated these processes and impairment by overexpressing NgR was rescued with coexpression of WNK1, indicating the partial role of WNK1 in NgR-mediated morphological regulation. Our study identifies a separation of functions for the NgR-regulated WNK1 in mediating proliferation, migration and axonal extension in PC12 cells as well as a specific regulatory role between NgR and WNK1 that is important for recovery from central nervous system injury.


Sujet(s)
Axones/physiologie , Mouvement cellulaire/physiologie , Prolifération cellulaire/physiologie , Régulation de l'expression des gènes/physiologie , Récepteurs Nogo/métabolisme , Protéine kinase déficiente en lysine WNK-1/métabolisme , Animaux , Axones/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/génétique , Facteur de croissance nerveuse/pharmacologie , Récepteurs Nogo/génétique , Cellules PC12/cytologie , ARN messager/métabolisme , Rats , Sincalide/métabolisme , Transfection , Protéine kinase déficiente en lysine WNK-1/génétique
7.
Int J Biol Sci ; 13(2): 135-144, 2017.
Article de Anglais | MEDLINE | ID: mdl-28255266

RÉSUMÉ

Endoplasmic reticulum (ER) stress, a feature of many conditions associated with pulmonary hypertension (PH), is increasingly recognized as a common response to promote proliferation in the walls of pulmonary arteries. Increased expression of Lipocalin-2 in PH led us to test the hypothesis that Lipocalin-2, a protein known to sequester iron and regulate it intracellularly, might facilitate the ER stress and proliferation in pulmonary arterial smooth muscle cells (PASMCs). In this study, we observed greatly increased Lcn2 expression accompanied with increased ATF6 cleavage in a standard rat model of pulmonary hypertension induced by monocrotaline. In cultured human PASMCs, Lcn2 significantly promoted ER stress (determined by augmented cleavage and nuclear localization of ATF6, up-regulated transcription of GRP78 and NOGO, increased expression of SOD2, and mild augmented mitochondrial membrane potential) and proliferation (assessed by Ki67 staining and BrdU incorporation). Lcn2 promoted ER stress accompanied with augmented intracellular iron levels in human PASMCs. Treatment human PASMCs with FeSO4 induced the similar ER stress and proliferation response and iron chelator (deferoxamine) abrogated the ER stress and proliferation induced by Lcn2 in cultured human PASMCs. In conclusion, Lcn2 significantly promoted human PASMC ER stress and proliferation by augmenting intracellular iron. The up-regulation of Lcn2 probably involved in the pathogenesis and progression of PH.


Sujet(s)
Stress du réticulum endoplasmique/physiologie , Fer/métabolisme , Lipocaline-2/métabolisme , Myocytes du muscle lisse/cytologie , Myocytes du muscle lisse/métabolisme , Artère pulmonaire/cytologie , Facteur de transcription ATF-6/génétique , Facteur de transcription ATF-6/métabolisme , Animaux , Technique de Western , Prolifération cellulaire/génétique , Prolifération cellulaire/physiologie , Cellules cultivées , Chaperonne BiP du réticulum endoplasmique , Stress du réticulum endoplasmique/génétique , Protéines du choc thermique/génétique , Protéines du choc thermique/métabolisme , Humains , Immunohistochimie , Lipocaline-2/génétique , Mâle , Potentiel de membrane mitochondriale/génétique , Potentiel de membrane mitochondriale/physiologie , Récepteurs Nogo/génétique , Récepteurs Nogo/métabolisme , Rats , Rat Sprague-Dawley , RT-PCR , Transduction du signal/génétique , Transduction du signal/physiologie , Superoxide dismutase/génétique , Superoxide dismutase/métabolisme
8.
J Neuroinflammation ; 13(1): 265, 2016 10 11.
Article de Anglais | MEDLINE | ID: mdl-27724971

RÉSUMÉ

BACKGROUND: Nogo-A and its putative receptor NgR are considered to be among the inhibitors of axonal regeneration in the CNS. However, few studies so far have addressed the issue of local NgR complex multilateral localization within inflammation in an MS mouse model of autoimmune demyelination. METHODS: Chronic experimental autoimmune encephalomyelitis (EAE) was induced in C57BL/6 mice. Analyses were performed on acute (days 18-22) and chronic (day 50) time points and compared to controls. The temporal and spatial expression of the Nogo receptor complex (NgR and coreceptors) was studied at the spinal cord using epifluorescent and confocal microscopy or real-time PCR. Data are expressed as cells/mm2, as mean % ± SEM, or as arbitrary units of integrated density. RESULTS: Animals developed a moderate to severe EAE without mortality, followed by a progressive, chronic clinical course. NgR complex spatial expression varied during the main time points of EAE. NgR with coreceptors LINGO-1 and TROY was increased in the spinal cord in the acute phase whereas LINGO-1 and p75 signal seemed to be dominant in the chronic phase, respectively. NgR was detected on gray matter NeuN+ neurons of the spinal cord, within the white matter inflammatory foci (14.2 ± 4.3 % NgR+ inflammatory cells), and found to be colocalized with GAP-43+ axonal growth cones while no ß-TubIII+, SMI-32+, or APP+ axons were found as NgR+. Among the NgR+ inflammatory cells, 75.6 ± 9.0 % were microglial/macrophages (lectin+), 49.6 ± 14.2 % expressed CD68 (phagocytic ED1+ cells), and no cells were Mac-3+. Of these macrophages/monocytes, only Arginase-1+/NgR+ but not iNOS+/NgR+ were present in lesions both in acute and chronic phases. CONCLUSIONS: Our data describe in detail the expression of the Nogo receptor complex within the autoimmune inflammatory foci and suggest a possible immune action for NgR apart from the established inhibitory one on axonal growth. Its expression by inflammatory macrophages/monocytes could signify a possible role of these cells on axonal guidance and clearance of the lesioned area during inflammatory demyelination.


Sujet(s)
Système nerveux central/métabolisme , Système nerveux central/anatomopathologie , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Régulation de l'expression des gènes/immunologie , Récepteurs Nogo/métabolisme , Transduction du signal/physiologie , Animaux , Antigènes de différenciation/métabolisme , Arginase/métabolisme , Modèles animaux de maladie humaine , Encéphalomyélite auto-immune expérimentale/induit chimiquement , Encéphalomyélite auto-immune expérimentale/complications , Encéphalomyélite auto-immune expérimentale/immunologie , Femelle , Adjuvant Freund/immunologie , Adjuvant Freund/toxicité , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Souris , Souris de lignée C57BL , Glycoprotéine MOG/immunologie , Glycoprotéine MOG/toxicité , Protéines de tissu nerveux/métabolisme , Protéines Nogo/génétique , Protéines Nogo/métabolisme , Récepteurs Nogo/génétique , Fragments peptidiques/immunologie , Fragments peptidiques/toxicité , Récepteurs facteur croissance nerf/génétique , Récepteurs facteur croissance nerf/métabolisme , Récepteurs aux facteurs de nécrose tumorale/génétique , Récepteurs aux facteurs de nécrose tumorale/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/immunologie , Statistique non paramétrique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...