Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Int J Mol Sci ; 24(1)2022 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-36613848

RESUMO

The N-methyl-D-aspartate receptor (NMDAR)-mediated structural plasticity of dendritic spines plays an important role in synaptic transmission in the brain during learning and memory formation. The Rho family of small GTPase RhoA and its downstream effector Rho-kinase/ROCK are considered as one of the major regulators of synaptic plasticity and dendritic spine formation, including long-term potentiation (LTP). However, the mechanism by which Rho-kinase regulates synaptic plasticity is not yet fully understood. Here, we found that Rho-kinase directly phosphorylated discs large MAGUK scaffold protein 2 (DLG2/PSD-93), a major postsynaptic scaffold protein that connects postsynaptic proteins with NMDARs; an ionotropic glutamate receptor, which plays a critical role in synaptic plasticity. Stimulation of striatal slices with an NMDAR agonist induced Rho-kinase-mediated phosphorylation of PSD-93 at Thr612. We also identified PSD-93-interacting proteins, including DLG4 (PSD-95), NMDARs, synaptic Ras GTPase-activating protein 1 (SynGAP1), ADAM metallopeptidase domain 22 (ADAM22), and leucine-rich glioma-inactivated 1 (LGI1), by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Among them, Rho-kinase increased the binding of PSD-93 to PSD-95 and NMDARs. Furthermore, we found that chemical-LTP induced by glycine, which activates NMDARs, increased PSD-93 phosphorylation at Thr612, spine size, and PSD-93 colocalization with PSD-95, while these events were blocked by pretreatment with a Rho-kinase inhibitor. These results indicate that Rho-kinase phosphorylates PSD-93 downstream of NMDARs, and suggest that Rho-kinase mediated phosphorylation of PSD-93 increases the association with PSD-95 and NMDARs to regulate structural synaptic plasticity.


Assuntos
Receptores de N-Metil-D-Aspartato , Quinases Associadas a rho , Quinases Associadas a rho/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Cromatografia Líquida , Espectrometria de Massas em Tandem , Plasticidade Neuronal/fisiologia , Transmissão Sináptica/fisiologia , Proteína 4 Homóloga a Disks-Large/metabolismo , Sinapses/metabolismo , Hipocampo/metabolismo
2.
J Neurochem ; 157(6): 2145-2157, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33599284

RESUMO

Post-synaptic density 93 (PSD-93) mediates glutamate excitotoxicity induced by ischemic brain injury, which then induces microglial inflammatory response. However, the underlying mechanisms of how PSD-93 mediates the crosstalk between neurons and microglia in the post-synaptic dense region remain elusive. CX3 chemokine ligand 1 (CX3CL1) is a chemokine specifically expressed in neurons while its receptor CX3CR1 is highly expressed in microglia. In this study, we examined the interaction of PSD-93 and CX3CL1 in the crosstalk between neurons and microglia in acute ischemic stroke. We utilized male C57BL/6 mice to establish the middle cerebral artery occlusion model (MCAO) and designed a fusion small peptide Tat-CX3CL1 (357-395aa) to inhibit PSD-93 and CX3CL1 interaction. The combination peaks of PSD-93 and CX3CL1 at 6 hr after I/R were observed. The binding sites were located at the 420-535 amino acid sequence of PSD-93 and 357-395 amino acid sequence of CX3CL1. Tat-CX3CL1 (357-395aa) could inhibit the interaction of PSD-93 and CX3CL1 and inhibited the pro-inflammatory cytokine IL-1ß and TNF-α expression and provided neuroprotection following reperfusion. Together, these data suggest that PSD-93 binds CX3CL1 to activate microglia and initiate neuroinflammation. Specific blockade of PSD-93-CX3CL1 interaction reduces I/R induced neuronal cell death, and provides a new therapeutic target for ischemic stroke.


Assuntos
Isquemia Encefálica/metabolismo , Quimiocina CX3CL1/metabolismo , Guanilato Quinases/metabolismo , AVC Isquêmico/metabolismo , Proteínas de Membrana/metabolismo , Microglia/metabolismo , Neurônios/metabolismo , Sequência de Aminoácidos , Animais , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Quimiocina CX3CL1/genética , Guanilato Quinases/genética , Células HEK293 , Humanos , AVC Isquêmico/genética , AVC Isquêmico/patologia , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Microglia/patologia , Neurônios/patologia , Ligação Proteica/fisiologia
3.
Acta Neuropathol ; 142(6): 1045-1064, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34536123

RESUMO

Since the discovery of ketamine anti-depressant effects in last decade, it has effectively revitalized interest in investigating excitatory synapses hypothesis in the pathogenesis of depression. In the present study, we aimed to reveal the excitatory synaptic regulation of corticotropin-releasing hormone (CRH) neuron in the hypothalamus, which is the driving force in hypothalamic-pituitary-adrenal (HPA) axis regulation. This study constitutes the first observation of an increased density of PSD-93-CRH co-localized neurons in the hypothalamic paraventricular nucleus (PVN) of patients with major depression. PSD-93 overexpression in CRH neurons in the PVN induced depression-like behaviors in mice, accompanied by increased serum corticosterone level. PSD-93 knockdown relieved the depression-like phenotypes in a lipopolysaccharide (LPS)-induced depression model. Electrophysiological data showed that PSD-93 overexpression increased CRH neurons synaptic activity, while PSD-93 knockdown decreased CRH neurons synaptic activity. Furthermore, we found that LPS induced increased the release of glutamate from microglia to CRH neurons resulted in depression-like behaviors using fiber photometry recordings. Together, these results show that PSD-93 is involved in the pathogenesis of depression via increasing the synaptic activity of CRH neurons in the PVN, leading to the hyperactivity of the HPA axis that underlies depression-like behaviors.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Depressão/metabolismo , Guanilato Quinases/metabolismo , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Feminino , Humanos , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Sistema Hipófise-Suprarrenal/metabolismo , Transmissão Sináptica/fisiologia , Regulação para Cima
4.
Genet Med ; 22(8): 1329-1337, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32341572

RESUMO

PURPOSE: Impaired function of gonadotropin-releasing hormone (GnRH) neurons can cause a phenotypic spectrum ranging from delayed puberty to isolated hypogonadotropic hypogonadism (IHH). We sought to identify a new genetic etiology for these conditions. METHODS: Exome sequencing was performed in an extended family with autosomal dominant, markedly delayed puberty. The effects of the variant were studied in a GnRH neuronal cell line. Variants in the same gene were sought in a large cohort of individuals with IHH. RESULTS: We identified a rare missense variant (F900V) in DLG2 (which encodes PSD-93) that cosegregated with the delayed puberty. The variant decreased GnRH expression in vitro. PSD-93 is an anchoring protein of NMDA receptors, a type of glutamate receptor that has been implicated in the control of puberty in laboratory animals. The F900V variant impaired the interaction between PSD-93 and a known binding partner, Fyn, which phosphorylates NMDA receptors. Variants in DLG2 that also decreased GnRH expression were identified in three unrelated families with IHH. CONCLUSION: The findings indicate that variants in DLG2/PSD-93 cause autosomal dominant delayed puberty and may also contribute to IHH. The findings also suggest that the pathogenesis involves impaired NMDA receptor signaling and consequently decreased GnRH secretion.


Assuntos
Hormônio Liberador de Gonadotropina , Hipogonadismo , Hormônio Liberador de Gonadotropina/genética , Guanilato Quinases , Humanos , Hipogonadismo/genética , Proteínas , Transdução de Sinais , Proteínas Supressoras de Tumor , Sequenciamento do Exoma
5.
Biochem Biophys Res Commun ; 517(1): 69-76, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31326116

RESUMO

MicroRNAs (miRNAs) have emerged as critical regulators of ischemic stroke, a condition that affects neuronal survival. However, the precise role of miRNAs in regulating neuronal injury during ischemic stroke remains largely unknown. In this study, we investigated the potential role of miR-152-3p in regulating oxygen-glucose-deprivation/reoxygenation (OGD/R)-induced neuronal injury in vitro. We found that OGD/R-exposed neurons expressed less miR-152-3p. Functional analysis revealed that miR-152-3p overexpression increased the viability and reduced the apoptosis and reactive oxygen species (ROS) production of OGD/R-exposed neurons. By contrast, miR-152-3p inhibition exacerbated OGD/R-induced injury. Notably, we identified postsynaptic density protein-93 (PSD-93), an important regulator of neuroprotection during ischemic stroke, as a miR-152-3p target gene. PSD-93 inhibition by small interfering RNA (siRNA) or miR-152-3p reinforced the activation of nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE) antioxidant signaling in OGD/R-exposed neurons. However, PSD-93 overexpression or Nrf2 silencing partially reversed miR-152-3p-mediated neuroprotection in OGD/R-exposed neurons. Overall, these results demonstrated that miR-152-3p protected neurons from OGD/R-induced apoptosis and ROS production by reinforcing Nrf2/ARE antioxidant signaling through targeting and inhibiting PSD-93, findings that suggest miR-152-3p is a potential target for neuroprotection during ischemic stroke.


Assuntos
Guanilato Quinases/genética , Proteínas de Membrana/genética , MicroRNAs/genética , Fator 2 Relacionado a NF-E2/genética , Neurônios/metabolismo , Traumatismo por Reperfusão/genética , Animais , Elementos de Resposta Antioxidante , Apoptose , Linhagem Celular , Glucose/metabolismo , Guanilato Quinases/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , MicroRNAs/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Neurônios/citologia , Neurônios/patologia , Neuroproteção , Oxigênio/metabolismo , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais , Regulação para Cima
6.
BMC Genomics ; 19(1): 194, 2018 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-29703139

RESUMO

BACKGROUND: The murine discs large homolog 2 (DLG2; post synaptic density 93 (PSD-93); Chapsyn-110) is a member of the membrane-associated guanylate kinase (MAGUK) protein family involved in receptor assembly and associated with signaling enzymes on cell membranes. In neurons, DLG2 protein isoforms derived from alternatively spliced transcripts have been described to bind to NMDA (N-methyl-aspartate) receptors and K channels and to mediate clustering of these channels in the postsynaptic membrane. In myeloid cells of the immune system, such as dendritic cells (DCs), a lack of data exists on the expression or function of DLG2. In cDNA microarray transcriptome analyses, we found Dlg2 highly expressed in a subpopulation of plasmacytoid DCs (pDCs) stimulated to produce type I interferons (IFNs) such as IFNß. RESULTS: Using RACE- and RT-PCR as well as immunoprecipitation followed by Western blotting we characterised the differential expression of the Dlg2 splice variants in IFNß-producing pDCs. Besides Dlg2É£ this cell population expressed a novel short Dlg2η transcript we termed Dlg2η3. Our expression data were integrated into information from genome databases to obtain a novel and comprehensive overview of the mouse Dlg2 gene architecture. To elucidate the intracellular localisation pattern of protein isoforms, ectopical expression analysis of fluorescently tagged DLG2 splice variants was performed. Here we found an enrichment of the larger isoform DLG2α1 at the plasma membrane while the newly identified shorter (DLG2η) isoform as well as DLG2É£ were equally distributed throughout the cytoplasm. Additionally, DLG2η was also found in the nucleus. Analysis of Dlg2-knockout mice previously generated by deleting exon 9 surprisingly revealed that the protein for the novel DLG2η isoform was still expressed in the brain and in bone marrow-derived pDCs from mice carrying the homozygous deletion (Dlg2 ΔE9/ΔE9 ). CONCLUSION: We describe a novel splice variant of the mouse Dlg2 gene termed Dlg2η and define the differential expression pattern of DLG2 isoforms in IFNß-producing pDCs. The presence of DLG2η protein in the CNS of Dlg2 ΔE9/ΔE9 mice might influence the phenotype of these mice and has to be taken into account in the interpretation of results regarding the functional role of DLG2 in neuronal postsynaptic membranes.


Assuntos
Processamento Alternativo , Células Dendríticas/metabolismo , Guanilato Quinases/genética , Guanilato Quinases/metabolismo , Interferon beta/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Animais , Medula Óssea/metabolismo , Encéfalo/metabolismo , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células Dendríticas/citologia , Perfilação da Expressão Gênica , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Camundongos , Células NIH 3T3 , Análise de Sequência com Séries de Oligonucleotídeos , Isoformas de Proteínas/metabolismo , Regulação para Cima
7.
Neurobiol Dis ; 68: 104-11, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24787897

RESUMO

Modification of N-methyl-d-aspartate receptor (NMDAR)-mediated excitotoxicity appears to be a potential target in the treatment of ischemic stroke. Postsynaptic density protein-93 (PSD-93) specifically binds the C-terminal tails of the NMDAR, which is critical to couple NMDAR activity to specific intracellular signaling. This study is to investigate whether PSD-93 disruption displays neuroprotection in a focal ischemic stroke model of adult mice and, if it does, to explore possible mechanisms. It was found that, following middle cerebral artery occlusion (MCAO), PSD-93 knockout (KO) mice manifested significant reductions in infarcted volume, neurological deficits and number of degenerated neurons. PSD-93 deletion also reduced cultured cortical neuronal death caused by glucose and oxygen deprivation (OGD). Ischemic long term potentiation (i-LTP) could not be induced in the PSD-93 KO group and wild type (WT) groups pretreated with either AP-5 (NMDAR inhibitor) or PP2 (Src family inhibitor). PSD-93 KO decreased the phosphorylation of the NR2B at Tyr1472 and the interaction between NR2B and Fyn after MCAO. Together, our study demonstrated that PSD-93 KO confers profound neuroprotection against ischemic brain injury, which probably links to the inhibitory effect on Fyn-mediated phosphorylation of NR2B caused by PSD-93 deletion. These findings may provide a novel avenue for the treatment of ischemic stroke.


Assuntos
Potenciais Pós-Sinápticos Excitadores/genética , Guanilato Quinases/deficiência , Infarto da Artéria Cerebral Média/metabolismo , Proteínas de Membrana/deficiência , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large , Antagonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Guanilato Quinases/genética , Hipocampo/citologia , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/prevenção & controle , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças do Sistema Nervoso/etiologia , Fatores de Tempo , Valina/análogos & derivados , Valina/farmacologia , Valina/uso terapêutico
8.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 4): 587-94, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23519667

RESUMO

PSD-93 (chapsyn-110, DLG2) is a member of the family of membrane-associated guanylate kinase (MAGUK) proteins. The MAGUK proteins are involved in receptor localization and signalling pathways. The best characterized MAGUK protein, PSD-95, is known to be involved in NMDA receptor signalling via its PDZ domains. The PDZ domains of PSD-95 and PSD-93 are structurally very similar, but relatively little is known about the function of PSD-93. PSD-93 has been suggested to interact with GluD2 from the family of ionotropic glutamate receptors. Here, the interactions of four residues (GTSI) representing the extreme C-terminus of GluD2 with PSD-93 PDZ1 have been investigated in the crystalline phase. Two different binding modes of these residues were observed, suggesting that the peptide is not tightly bound to PSD-93 PDZ1. In accordance, the two N-terminal PSD-93 PDZ domains show no appreciable binding affinity for a GluD2-derived C-terminal octapeptide, whereas micromolar affinity was observed for a GluN2B-derived C-terminal octapeptide. This indicates that if present, the interactions between GluD2 and PSD-93 involve more than the extreme terminus of the receptor. In contrast, the tumour-suppressor protein SCRIB PDZ3 shows low micromolar affinity towards the GluD2-derived octapeptide, which is in agreement with previous findings using high-throughput assays.


Assuntos
Guanilato Quinases/metabolismo , Mapeamento de Interação de Proteínas/métodos , Proteínas Supressoras de Tumor/metabolismo , Comunicação Celular/fisiologia , Cristalização , Cristalografia por Raios X , Polarização de Fluorescência , Guanilato Quinases/biossíntese , Guanilato Quinases/química , Humanos , Microscopia de Fluorescência por Excitação Multifotônica , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Estrutura Terciária de Proteína , Espectrometria de Fluorescência , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/química
9.
Genes Brain Behav ; 22(6): e12865, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37705179

RESUMO

Variations in the Dlg2 gene have been linked to increased risk for psychiatric disorders, including schizophrenia, autism spectrum disorders, intellectual disability, bipolar disorder, attention deficit hyperactivity disorder, and pubertal disorders. Recent studies have reported disrupted brain circuit function and behaviour in models of Dlg2 knockout and haploinsufficiency. Specifically, deficits in hippocampal synaptic plasticity were found in heterozygous Dlg2+/- rats suggesting impacts on hippocampal dependent learning and cognitive flexibility. Here, we tested these predicted effects with a behavioural characterisation of the heterozygous Dlg2+/- rat model. Dlg2+/- rats exhibited a specific, mild impairment in reversal learning in a substrate deterministic bowl-digging reversal learning task. The performance of Dlg2+/- rats in other bowl digging task, visual discrimination and reversal, novel object preference, novel location preference, spontaneous alternation, modified progressive ratio, and novelty-suppressed feeding test were not impaired. These findings suggest that despite altered brain circuit function, behaviour across different domains is relatively intact in Dlg2+/- rats, with the deficits being specific to only one test of cognitive flexibility. The specific behavioural phenotype seen in this Dlg2+/- model may capture features of the clinical presentation associated with variation in the Dlg2 gene.


Assuntos
Guanilato Quinases , Aprendizagem , Proteínas de Membrana , Transtornos Mentais , Humanos , Animais , Ratos , Proteínas de Membrana/genética , Guanilato Quinases/genética , Cognição , Masculino , Feminino , Animais não Endogâmicos , Heterozigoto , Transtornos Mentais/genética , Hipocampo/fisiopatologia
10.
Genes Brain Behav ; 21(4): e12799, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35118804

RESUMO

Mutations affecting DLG2 are emerging as a genetic risk factor associated with neurodevelopmental psychiatric disorders including schizophrenia, autism spectrum disorder, and bipolar disorder. Discs large homolog 2 (DLG2) is a member of the membrane-associated guanylate kinase protein superfamily of scaffold proteins, a component of the post-synaptic density in excitatory neurons and regulator of synaptic function and plasticity. It remains an important question whether and how haploinsuffiency of DLG2 contributes to impairments in basic behavioural and cognitive functions that may underlie symptomatic domains in patients that cross diagnostic boundaries. Using a heterozygous Dlg2 mouse model we examined the impact of reduced Dlg2 expression on functions commonly impaired in neurodevelopmental psychiatric disorders including motor co-ordination and learning, pre-pulse inhibition and habituation to novel stimuli. The heterozygous Dlg2 mice exhibited behavioural impairments in long-term motor learning and long-term habituation to a novel context, but not motor co-ordination, initial responses to a novel context, PPI of acoustic startle or anxiety. We additionally showed evidence for the reduced regulation of the synaptic plasticity-associated protein cFos in the motor cortex during motor learning. The sensitivity of selective behavioural and cognitive functions, particularly those dependent on synaptic plasticity, to reduced expression of DLG2 give further credence for DLG2 playing a critical role in specific brain functions but also a mechanistic understanding of symptom expression shared across psychiatric disorders.


Assuntos
Transtorno do Espectro Autista , Animais , Ansiedade/genética , Guanilato Quinases/genética , Guanilato Quinases/metabolismo , Heterozigoto , Humanos , Proteínas de Membrana , Camundongos , Plasticidade Neuronal , Proteínas Supressoras de Tumor
11.
Transl Stroke Res ; 11(5): 1137-1147, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32130656

RESUMO

Postsynaptic density protein-93 (PSD-93) plays an important role in ischemic brain injury through N-methyl-D-aspartate receptor (NMDAR)-triggered neurotoxicity. GTPase-activating protein for Ras (SynGAP) is a GAP specifically expressed in the central nervous system to regulate nerve development and synaptic plasticity. However, the link between PSD-93 and SynGAP and their role in ischemic brain injury remain elusive. Here, we showed that PSD-93 interacted with SynGAP and mediated SynGAP ubiquitination and degradation following ischemic brain injury. Proteasome inhibitor MG-132 could reverse the decrease of SynGAP protein level in wild-type mice following cerebral ischemia reperfusion through inhibiting SynGAP ubiquitination. Furthermore, NMDA receptor inhibitor MK801 could increase SynGAP protein level in wild-type mice following cerebral ischemia reperfusion. However, in PSD-93 knockout mice, MG-132 or NMDAR inhibitor had no significant effect on SynGAP expression. Both MG-132 and PSD-93 knockout reduced infarct volume and improved neurological deficit in mice at different time points after cerebral ischemia reperfusion. Furthermore, we identified that 670-685 amino acid sequence of SynGAP was essential to the binding of SynGAP to PSD-93, and designed a fusion peptide Tat-SynGAP (670-685aa) that could attenuate ischemic brain damage in wild-type mice. In conclusion, we provide the first evidence that PSD-93 directly interacts with SynGAP and mediates its ubiquitination and degradation to aggravate ischemic brain damage. Tat-SynGAP (670-685aa) may be considered as a candidate for treatment of acute ischemic stroke.


Assuntos
Isquemia Encefálica/metabolismo , Guanilato Quinases/metabolismo , Proteínas de Membrana/metabolismo , Acidente Vascular Cerebral/metabolismo , Ubiquitinação , Animais , Proteínas Ativadoras de GTPase/metabolismo , Guanilato Quinases/genética , Infarto da Artéria Cerebral Média/metabolismo , Masculino , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Plasticidade Neuronal/fisiologia
12.
Mol Autism ; 11(1): 19, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-32164788

RESUMO

BACKGROUND: DLG2, also known as postsynaptic density protein-93 (PSD-93) or chapsyn-110, is an excitatory postsynaptic scaffolding protein that interacts with synaptic surface receptors and signaling molecules. A recent study has demonstrated that mutations in the DLG2 promoter region are significantly associated with autism spectrum disorder (ASD). Although DLG2 is well known as a schizophrenia-susceptibility gene, the mechanisms that link DLG2 gene disruption with ASD-like behaviors remain unclear. METHODS: Mice lacking exon 14 of the Dlg2 gene (Dlg2-/- mice) were used to investigate whether Dlg2 deletion leads to ASD-like behavioral abnormalities. To this end, we performed a battery of behavioral tests assessing locomotion, anxiety, sociability, and repetitive behaviors. In situ hybridization was performed to determine expression levels of Dlg2 mRNA in different mouse brain regions during embryonic and postnatal brain development. We also measured excitatory and inhibitory synaptic currents to determine the impacts of Dlg2 deletion on synaptic transmission in the dorsolateral striatum. RESULTS: Dlg2-/- mice showed hypoactivity in a novel environment. They also exhibited decreased social approach, but normal social novelty recognition, compared with wild-type animals. In addition, Dlg2-/- mice displayed strong self-grooming, both in home cages and novel environments. Dlg2 mRNA levels in the striatum were heightened until postnatal day 7 in mice, implying potential roles of DLG2 in the development of striatal connectivity. In addition, the frequency of excitatory, but not inhibitory, spontaneous postsynaptic currents in the Dlg2-/- dorsolateral striatum was significantly reduced. CONCLUSION: These results suggest that homozygous Dlg2 deletion in mice leads to ASD-like behavioral phenotypes, including social deficits and increased repetitive behaviors, as well as reductions in excitatory synaptic input onto dorsolateral spiny projection neurons, implying that the dorsal striatum is one of the brain regions vulnerable to the developmental dysregulation of DLG2.


Assuntos
Corpo Estriado/fisiologia , Guanilato Quinases/fisiologia , Proteínas de Membrana/fisiologia , Comportamento Social , Animais , Transtorno do Espectro Autista , Comportamento Animal , Potenciais Pós-Sinápticos Excitadores , Guanilato Quinases/genética , Potenciais Pós-Sinápticos Inibidores , Masculino , Proteínas de Membrana/genética , Camundongos Transgênicos , Transmissão Sináptica
13.
Elife ; 92020 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-33185190

RESUMO

The palmitoyl acyltransferase (PAT) ZDHHC14 is highly expressed in the hippocampus and is the only PAT predicted to bind Type-I PDZ domain-containing proteins. However, ZDHHC14's neuronal roles are unknown. Here, we identify the PDZ domain-containing Membrane-associated Guanylate Kinase (MaGUK) PSD93 as a direct ZDHHC14 interactor and substrate. PSD93, but not other MaGUKs, localizes to the axon initial segment (AIS). Using lentiviral-mediated shRNA knockdown in rat hippocampal neurons, we find that ZDHHC14 controls palmitoylation and AIS clustering of PSD93 and also of Kv1 potassium channels, which directly bind PSD93. Neurodevelopmental expression of ZDHHC14 mirrors that of PSD93 and Kv1 channels and, consistent with ZDHHC14's importance for Kv1 channel clustering, loss of ZDHHC14 decreases outward currents and increases action potential firing in hippocampal neurons. To our knowledge, these findings identify the first neuronal roles and substrates for ZDHHC14 and reveal a previously unappreciated role for palmitoylation in control of neuronal excitability.


Assuntos
Aciltransferases/metabolismo , Axônios/enzimologia , Superfamília Shaker de Canais de Potássio/metabolismo , Aciltransferases/genética , Animais , Fenômenos Eletrofisiológicos , Regulação Enzimológica da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HEK293 , Hipocampo/citologia , Humanos , Camundongos , Ligação Proteica , Superfamília Shaker de Canais de Potássio/genética , Técnicas do Sistema de Duplo-Híbrido
14.
Biol Psychiatry ; 86(3): 171-184, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30292394

RESUMO

BACKGROUND: Although synaptic impairment is a prerequisite to cognitive deficiencies in Alzheimer's disease (AD), mechanisms underlying the dysregulation of essential synaptic scaffolding components and their integrity remain elusive. RPS23RG1 is a newly identified protein implicated in AD. However, the physiological function of RPS23RG1 has yet to be determined. METHODS: We investigated the role of RPS23RG1 in maintaining synaptic structure and function in cell cultures and in Rps23rg1 knockout mice and determined whether targeting RPS23RG1-mediated pathways has therapeutic potential in APP/PS1 AD model mice. RESULTS: Deletion of the Rps23rg1 gene resulted in severe memory deficits and impairment of postsynaptic structure and function, with marked reductions in postsynaptic densities-93 and -95 (PSD-93 and PSD-95) levels. RPS23RG1 interacted with PSD-93/PSD-95 through its intracellular domain, consequently sequestering PSD-93/PSD-95 from murine double minute 2-mediated ubiquitination and degradation, thereby maintaining synaptic function. Restoration of PSD-93/PS-D95 levels reversed synaptic and memory deficits in Rps23rg1 knockout mice. We further observed attenuated RPS23RG1 expression in human AD, which positively correlated with PSD-93/PSD-95 levels. Importantly, an RPS23RG1-derived peptide comprising a unique PSD-93/PSD-95 interaction motif rescued synaptic and cognitive defects in Rps23rg1 knockout and AD mouse models. CONCLUSIONS: Our results reveal a role for RPS23RG1 in maintaining synaptic integrity and function and provide a new mechanism for synaptic dysfunction in AD pathogenesis. This demonstrates that RPS23RG1-mediated pathways show good therapeutic potential in AD intervention.


Assuntos
Disfunção Cognitiva/metabolismo , Proteína 4 Homóloga a Disks-Large/metabolismo , Plasticidade Neuronal , Proteínas Ribossômicas/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Animais , Disfunção Cognitiva/genética , Disfunção Cognitiva/fisiopatologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Hipocampo/metabolismo , Humanos , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Ribossômicas/genética
15.
J Comp Neurol ; 525(7): 1707-1730, 2017 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-28035673

RESUMO

Dopamine- and tyrosine hydroxylase-immunopositive cells (TH cells) modulate visually driven signals as they flow through retinal photoreceptor, bipolar, and ganglion cells. Previous studies suggested that TH cells release dopamine from varicose axons arborizing in the inner and outer plexiform layers after glutamatergic synapses depolarize TH cell dendrites in the inner plexiform layer and these depolarizations propagate to the varicosities. Although it has been proposed that these excitatory synapses are formed onto appendages resembling dendritic spines, spines have not been found on TH cells of most species examined to date or on TH cell somata that release dopamine when exposed to glutamate receptor agonists. By use of protocols that preserve proximal retinal neuron morphology, we have examined the shape, distribution, and synapse-related immunoreactivity of adult rat TH cells. We report here that TH cell somata, tapering and varicose inner plexiform layer neurites, and varicose outer plexiform layer neurites all bear spines, that some of these spines are immunopositive for glutamate receptor and postsynaptic density proteins (viz., GluR1, GluR4, NR1, PSD-95, and PSD-93), that TH cell somata and tapering neurites are also immunopositive for a γ-aminobutyric acid (GABA) receptor subunit (GABAA Rα1 ), and that a synaptic ribbon-specific protein (RIBEYE) is found adjacent to some colocalizations of GluR1 and TH in the inner plexiform layer. These results identify previously undescribed sites at which glutamatergic and GABAergic inputs may stimulate and inhibit dopamine release, especially at somata and along varicose neurites that emerge from these somata and arborize in various levels of the retina. J. Comp. Neurol. 525:1707-1730, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Espinhas Dendríticas/ultraestrutura , Interneurônios/ultraestrutura , Retina/citologia , Animais , Feminino , Imuno-Histoquímica , Masculino , Microscopia Confocal , Ratos , Ratos Endogâmicos Lew , Ratos Long-Evans , Tirosina 3-Mono-Oxigenase
16.
Curr Neuropharmacol ; 4(3): 217-23, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18615145

RESUMO

Persistent pain, a common clinical condition, could be caused by inflammation, tissue injury secondary to trauma or surgery, and nerve injuries. It is often inadequately controlled by current treatments, such as opioids and nonsteroidal anti-inflammatory drugs. The PDZ (Postsynaptic density 95, Discs large, and Zonula occludens-1) domains are ubiquitous protein interaction modules often found among multi-protein signaling complexes at neuronal synapses. Recent preclinical research shows that targeted disruption of PDZ domain-mediated protein interaction among N-methyl-Daspartate (NMDA) receptor signaling complexes significantly attenuates the development and maintenance of persistent pain without affecting nociceptive responsiveness to acute pain. PDZ domains at excitatory synapses may be new molecular targets for prevention and treatment of persistent pain. Here, we illustrate expression and distribution of the PDZ domain-containing proteins associated with NMDA receptors in the pain-related regions of the central nervous system, review the evidence for their roles in persistent pain states, and discuss potential mechanisms by which these PDZ domain-containing proteins are involved in persistent pain.

17.
Neurotoxicology ; 53: 1-11, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26680505

RESUMO

Postsynaptic density protein-93 (PSD-93) is enriched in the postsynaptic density and is involved in N-methyl-d-aspartate receptor (NMDAR) triggered neurotoxicity through PSD-93/NMDAR/nNOS signaling pathway. In the present study, we found that PSD-93 deficiency reduced infarcted volume and neurological deficits induced by transient middle cerebral artery occlusion (tMCAO) in the mice. To identify novel targets of PSD-93 related neurotoxicity, we applied isobaric tags for relative and absolute quantitative (iTRAQ) labeling and combined this labeling with on-line two-dimensional LC/MS/MS technology to elucidate the changes in protein expression in PSD-93 knockout mice following tMCAO. The proteomic data set consisted of 1892 proteins. Compared to control group, differences in expression levels in ischemic group >1.5-fold and <0.66-fold were considered as differential expression. A total of 104 unique proteins with differential abundance levels were identified, among which 17 proteins were selected for further validation. Gene ontology analysis using UniProt database revealed that these differentially expressed proteins are involved in diverse function such as synaptic transmission, neuronal neurotransmitter and ion transport, modification of organelle membrane components. Moreover, network analysis revealed that the interacting proteins were involved in the transport of synaptic vesicles, the integrity of synaptic membranes and the activation of the ionotropic glutamate receptors NMDAR1 and NMDAR2B. Finally, RT-PCR and Western blot analysis showed that SynGAP, syntaxin-1A, protein kinase C ß, and voltage-dependent L-type calcium channels were inhibited by ischemia-reperfusion. Identification of these proteins provides valuable clues to elucidate the mechanisms underlying the actions of PSD-93 in ischemia-reperfusion induced neurotoxicity.


Assuntos
Regulação da Expressão Gênica/genética , Guanilato Quinases/deficiência , Infarto da Artéria Cerebral Média/metabolismo , Proteínas de Membrana/deficiência , Proteômica , Animais , Canais de Cálcio/metabolismo , Modelos Animais de Doenças , Ontologia Genética , Guanilato Quinases/genética , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/genética , Mapas de Interação de Proteínas/genética , Proteína Quinase C/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Estatística como Assunto , Sintaxina 1/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo
18.
Cell Biochem Biophys ; 73(3): 695-700, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27259312

RESUMO

The aim of the study was to explore the effect of PSD-93 deficiency on the expression of early inflammatory cytokines induced by cerebral ischemia/reperfusion injury. Ten- to twelve-week-old male PSD-93 knockout (PSD-93 KO) mice (C57BL/6 genetic background) and wild-type (WT) littermates were randomly divided into sham and ischemia/reperfusion (I/R) group. The focal cerebral I/R model was established by middle cerebral artery occlusion (MCAO) suture method. RT-PCR was used to detect the mRNA expression of IL-6, IL-10, Cox-2, iNOS, and TNF-α4h following reperfusion. Infarct volume at different time points after I/R was analyzed using 2,3,5-triphenyl tetrazolium staining, and neurological damage score (neurological severity scores, NSS) was used to evaluate the effect of PSD-93 gene knockout on the MCAO-induced neurological injury. In WT mice, early I/R injury led to the increase in the mRNA expression of proinflammatory cytokines IL-6, Cox-2, iNOS, and TNF-α that coincided with the decrease in the expression of anti-inflammatory cytokine IL-10, as compared to the sham group (P < 0.05). This effect was markedly attenuated by depleting PSD-93 levels by gene knockout. As compared to sham group, in PSD-93 KO mice I/R4h led to downregulation of Cox-2 and iNOS expression, and increase in the mRNA levels of IL-10 (P < 0.05). In addition, following MCAO, PSD-93 KO mice exhibited improved NSS and reduced infarct volumes, as compared with WT animals. PSD-93 knockout may play a neuroprotective role by mediating the early release of inflammatory cytokines induced by cerebral ischemia.


Assuntos
Citocinas/metabolismo , Guanilato Quinases/deficiência , Infarto da Artéria Cerebral Média/metabolismo , Proteínas de Membrana/deficiência , Animais , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Citocinas/genética , Guanilato Quinases/genética , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo
19.
Artigo em Chinês | WPRIM | ID: wpr-576970

RESUMO

Objective To study the expression and distribution of postsynaptic density protein-93(PSD-93) after spinal cord injury(SCI). Methods With improved Allen's method,acute rats SCI models were established.Real-time PCR and Western blotting were used to detect the changes of PSD-93 mRNA and protein expression after SCI.The changes of PSD-93 localization after injury were investigated by immunofluorescence double staining. Results The expression of PSD-93 mRNA had a gradually decrease trend after SCI.The minimal mRNA was present at 3days;whereas the protein levels up-regulated significantly and peaked at 1day after injury.Immunofluorescence double staining indictated that PSD-93 was highly expressed on the activated microglias and astrocytes except that it co-localized with nNOS in the impaired spinal cord.Conclusion The time course of PSD-93 and its mRNA is vary significantly after SCI,and colocalized with nNOS and activated glias.These results indicate that PSD-93 may participate in the following response after SCI.

20.
Artigo em Chinês | WPRIM | ID: wpr-584347

RESUMO

Objective To investigate whether PSD 93 reduce neurotoxity induced by platelet activating factor (PAF) and study on the mechanism of cell molecularbiology.Methods PAF treated the cultural cortex neurons in wild type and PSD 93 knockout mice and the cells were stained with PI/Calcein AM for apoptosis; neuronal viability was measured by MTT. The expression of PSD93 and PSD95 protein in neurons was tested by Western Blot.Results PSD 93 and PSD 95 expressed in wild type neurons and only PSD 95 expressed in PSD 93 knockout. Cortex neuronal apoptosis in PSD 93 knockout mice was lower then that in wild type ( P

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa