Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 158
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35064078

RESUMO

Prostate-specific membrane antigen (PSMA) is highly overexpressed in most prostate cancers and is clinically visualized using PSMA-specific probes incorporating glutamate-ureido-lysine (GUL). PSMA is effectively absent from certain high-mortality, treatment-resistant subsets of prostate cancers, such as neuroendocrine prostate cancer (NEPC); however, GUL-based PSMA tracers are still reported to have the potential to identify NEPC metastatic tumors. These probes may bind unknown proteins associated with PSMA-suppressed cancers. We have identified the up-regulation of PSMA-like aminopeptidase NAALADaseL and the metabotropic glutamate receptors (mGluRs) in PSMA-suppressed prostate cancers and find that their expression levels inversely correlate with PSMA expression and are associated with GUL-based radiotracer uptake. Furthermore, we identify that NAALADaseL and mGluR expression correlates with a unique cell cycle signature. This provides an opportunity for the future study of the biology of NEPC and potential therapeutic directions. Computationally predicting that GUL-based probes bind well to these targets, we designed and synthesized a fluorescent PSMA tracer to investigate these proteins in vitro, where it shows excellent affinity for PSMA, NAALADaseL, and specific mGluRs associated with poor prognosis.


Assuntos
Antígenos de Superfície/metabolismo , Glutamato Carboxipeptidase II/metabolismo , Glutamatos , Lisina , Sondas Moleculares , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/metabolismo , Ureia , Animais , Antígenos de Superfície/química , Sítios de Ligação , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Imunofluorescência , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/química , Expressão Gênica , Glutamato Carboxipeptidase II/química , Glutamatos/química , Humanos , Imuno-Histoquímica , Lisina/química , Masculino , Camundongos , Modelos Moleculares , Conformação Molecular , Imagem Molecular/métodos , Sondas Moleculares/química , Neoplasias da Próstata/genética , Ligação Proteica , Receptores de Ácido Caínico/genética , Receptores de Ácido Caínico/metabolismo , Relação Estrutura-Atividade , Ureia/análogos & derivados , Ureia/química
2.
Nat Commun ; 12(1): 5330, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34504093

RESUMO

Most autosomal genes are thought to be expressed from both alleles, with some notable exceptions, including imprinted genes and genes showing random monoallelic expression (RME). The extent and nature of RME has been the subject of debate. Here we investigate the expression of several candidate RME genes in F1 hybrid mouse cells before and after differentiation, to define how they become persistently, monoallelically expressed. Clonal monoallelic expression is not present in embryonic stem cells, but we observe high frequencies of monoallelism in neuronal progenitor cells by assessing expression status in more than 200 clones. We uncover unforeseen modes of allelic expression that appear to be gene-specific and epigenetically regulated. This non-canonical allelic regulation has important implications for development and disease, including autosomal dominant disorders and opens up therapeutic perspectives.


Assuntos
Alelos , Desequilíbrio Alélico , Epigênese Genética , Doenças Musculares/genética , Células-Tronco Neurais/metabolismo , Doenças Neurodegenerativas/genética , Hidrolases Anidrido Ácido/genética , Hidrolases Anidrido Ácido/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Diferenciação Celular , Quimera , Células Clonais , Metilação de DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Feminino , Dosagem de Genes , Frequência do Gene , Loci Gênicos , Impressão Genômica , Masculino , Camundongos , Doenças Musculares/metabolismo , Doenças Musculares/patologia , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Células-Tronco Neurais/patologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Osteoporose/genética , Osteoporose/metabolismo , Osteoporose/patologia , Receptores de Ácido Caínico/genética , Receptores de Ácido Caínico/metabolismo , Receptor de GluK2 Cainato
3.
Epilepsia ; 62(7): 1559-1568, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34060082

RESUMO

OBJECTIVE: Previous positron emission tomography (PET) studies using [11 C]ABP688 show reduced metabotropic glutamate receptor type 5 (mGluR5) allosteric binding site availability in the epileptogenic hippocampus of mesial temporal lobe epilepsy (MTLE) patients. However, the link between mGluR5 abnormalities and postsurgical outcomes remains unclear. Here, we test whether reduced PET [11 C]ABP688 binding in cornu ammonis (CA) sectors more vulnerable to glutamatergic excitotoxicity relates to surgical outcomes. METHODS: We obtained magnetic resonance imaging (MRI) and [11 C]ABP688-PET from 31 unilateral MTLE patients and 30 healthy controls. MRI hippocampal subfields were segmented using FreeSurfer. To respect the lower PET special resolution, MRI-derived anatomical subfields were combined into CA1-3, CA4/dentate gyrus, and Subiculum. Partial volume corrected [11 C]ABP688 nondisplaceable binding potential (BPND ) values were averaged across each subfield, and Z-scores were calculated. Subfield [11 C]ABP688-BPND was compared between seizure-free and non-seizure-free patients. In addition, we also assessed subfield volumes and [18 F]fluorodeoxyglucose (FDG) uptake in each clinical group. RESULTS: MTLE [11 C]ABP688-BPND was reduced in ipsilateral (epileptogenic) CA1-3 and CA4/dentate-gyrus (p < .001, 95% confidence interval [CI] = .29-.51) compared to controls, with no difference in Subiculum. [11 C]ABP688-BPND and subfield volumes were compared between seizure-free (Engel IA, n = 13) and non-seizure-free patients (Engel IC-III, n = 10). In ipsilateral CA1-3 only, [11 C]ABP688-BPND was lower in seizure-free patients than in non-seizure-free patients (p = .012, 95% CI = 1.46-11.0) independently of volume. A subset analysis of 12 patients with [11 C]ABP688-PET+[18 F]FDG-PET showed no between-group significant difference in [18 F]FDG uptake, whereas CA1-3 [11 C]ABP688-BPND remained significantly lower in the seven of 12 seizure-free patients (p = .03, 95% CI = -3.13 to -.21). SIGNIFICANCE: Reduced mGluR5 allosteric site availability in hippocampal CA1-3, measured in vivo by [11 C]ABP688-PET, is associated with postsurgery seizure freedom independent of atrophy or hypometabolism. Information derived from hippocampal CA1-3 [11 C]ABP688-PET is a promising imaging biomarker potentially impactful in surgical decisions for MRI-negative/PET-negative MTLE patients.


Assuntos
Epilepsia do Lobo Temporal/genética , Epilepsia do Lobo Temporal/cirurgia , Ácido Glutâmico/genética , Hipocampo/metabolismo , Procedimentos Neurocirúrgicos , Receptores de Ácido Caínico/genética , Adolescente , Adulto , Idoso , Região CA1 Hipocampal/metabolismo , Região CA3 Hipocampal , Epilepsia do Lobo Temporal/diagnóstico por imagem , Feminino , Fluordesoxiglucose F18 , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Oximas , Tomografia por Emissão de Pósitrons , Piridinas , Compostos Radiofarmacêuticos , Receptores de Ácido Caínico/metabolismo , Resultado do Tratamento , Adulto Jovem
4.
Biosci Rep ; 41(5)2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-33969375

RESUMO

Synapse and synapse-associated proteins (SAPs) play critical roles in various neurodegeneration diseases and brain tumors. However, in lower-grade gliomas (LGG), SAPs have not been explored systematically. Herein, we are going to explore SAPs expression profile and its clinicopathological significance in LGG which can offer new insights to glioma therapy. In the present study, we integrate a list of SAPs that covered 231 proteins with synaptogenesis activity and post synapse formation. The LGG RNA-seq data were downloaded from GEO, TCGA and CGGA database. The prognosis associated SAPs in key modules of PPI (protein-protein interaction networks) was regarded as hub SAPs. Western blot, quantitative reverse transcription PCR (qRT-PCR) and immunochemistry results from HPA database were used to verify the expression of hub SAPs. There were 68 up-regulated SAPs and 44 down-regulated SAPs in LGG tissue compared with normal brain tissue. Data from function enrichment analysis revealed functions of differentially expressed SAPs in synapse organization and glutamatergic receptor pathway in LGGs. Survival analysis revealed that four SAPs, GRIK2, GABRD, GRID2 and ARC were correlate with the prognosis of LGG patients. Interestingly, we found that GABRD were up-regulated in LGG patients with seizures, indicating that SAPs may link to the pathogenesis of seizures in glioma patients. The four-SAPs signature was revealed as an independent prognostic factor in gliomas. Our study presented a novel strategy to assess the prognostic risks of LGGs, based on the expression of SAPs.


Assuntos
Biomarcadores Tumorais/normas , Neoplasias Encefálicas/genética , Glioma/genética , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Redes Reguladoras de Genes , Glioma/metabolismo , Glioma/patologia , Humanos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Valor Preditivo dos Testes , Mapas de Interação de Proteínas , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Receptores de Glutamato/genética , Receptores de Glutamato/metabolismo , Receptores de Ácido Caínico/genética , Receptores de Ácido Caínico/metabolismo , Receptor de GluK2 Cainato
5.
Neurochem Res ; 46(7): 1771-1780, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33847855

RESUMO

The mechanisms underlying postoperative pain differ from the inflammatory or neuropathic pain. Previous studies have demonstrated that intrathecal α-amino-3-hydroxy-5-methy-4-isoxazole propionate (AMPA) -kainate (KA) receptor antagonist inhibits the guarding pain behavior and mechanical hyperalgesia, indicating a critical role of spinal KA receptors in postoperative pain hypersensitivity. However, how the functional regulations of spinal KA receptor subunits are involved in the postoperative pain hypersensitivity remains elusive. Therefore, in the current study, we investigated the synaptic delivery of spinal KA receptor subunits and the interaction between KA receptor subunits and glutamate receptor-interacting protein (GRIP) during the postoperative pain. Our data indicated that plantar incision induced the synaptic delivery of GluK2, but not GluK1 or GluK3 in ipsilateral spinal cord dorsal horns. The co-immunoprecipitation showed an increased GluK2 -GRIP interaction in ipsilateral dorsal horn neurons at 6 h post-incision. Interestingly, Intrathecal pretreatment of GRIP siRNA increased the paw withdrawal thresholds to mechanical stimuli and decreased the cumulative pain scores in the paws ipsilateral to the incision at 6 h post-incision. Additionally, Intrathecal pretreatment of GRIP siRNA reduced the synaptic abundance of GluK2 in ipsilateral spinal dorsal horn at 6 h after plantar incision. In general, our data have demonstrated that the GluK2- GRIP interaction-mediated synaptic abundance of GluK2 in dorsal horn neurons plays an important role in the postoperative pain hypersensitivity. Disrupting the GluK2- GRIP interaction may provide a new approach for relieving postoperative pain.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Dor Pós-Operatória/tratamento farmacológico , RNA Interferente Pequeno/uso terapêutico , Receptores de Ácido Caínico/metabolismo , Corno Dorsal da Medula Espinal/metabolismo , Sinapses/efeitos dos fármacos , Animais , Procedimentos Cirúrgicos Dermatológicos , Regulação para Baixo/efeitos dos fármacos , Pé/cirurgia , Membro Anterior/cirurgia , Injeções Espinhais , Células do Corno Posterior/efeitos dos fármacos , Células do Corno Posterior/metabolismo , RNA Interferente Pequeno/administração & dosagem , Ratos , Pele/efeitos dos fármacos , Corno Dorsal da Medula Espinal/citologia , Sinapses/metabolismo , Receptor de GluK2 Cainato
6.
Mol Med Rep ; 23(4)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33537798

RESUMO

Ozone is widely used to relieve chronic pain clinically, but the precise mechanisms governing its action have yet to be elucidated. The present study aimed to investigate the mechanisms underlying the pain­alleviating effect of ozone in the chronic constriction injury (CCI) model of sciatic nerve in rats. Pain behaviours of rats were assessed by mechanical allodynia and thermal hyperalgesia. The expression of spinal glutamate receptor 6 (GluR6) and NF­κB/p65 was detected by western blotting and reverse transcription­quantitative PCR. Meanwhile, the expression of spinal IL­1ß, IL­6 and TNF­α was detected by ELISA. GluR6 short interfering (si)RNAs were used intrathecally immediately following CCI once per day. Ozone (10, 20 or 30 µg/ml) or oxygen was injected intrathecally on day 7 after CCI. The expression level of spinal GluR6 increased on day 3 and reached a peak on day 7 after CCI. The expression level of spinal IL­1ß, IL­6, TNF­α and NF­κB/p65 also increased on day 7 after CCI. In addition, pre­intrathecal injection of GluR6 siRNAs inhibited pain behaviours and suppressed the expression of spinal GluR6, IL­1ß, IL­6, TNF­α and NF­κB/p65 in CCI rats on day 7. Intrathecal injection of ozone was also observed to inhibit pain behaviours and suppress the expression of spinal GluR6, IL­1ß, IL­6, TNF­α and NF­κB/p65 in CCI rats on day 7. The present study suggested that GluR6 served a pivotal role in neuropathic pain and that intrathecal injection of ozone may alleviate neuropathic pain via the GluR6­NF­κB/p65 signalling pathway.


Assuntos
Neuralgia/tratamento farmacológico , Ozônio/farmacologia , Receptores de Ácido Caínico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo , Animais , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Injeções Espinhais , Masculino , Neuralgia/genética , Neuralgia/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Ácido Caínico/genética , Transdução de Sinais/genética , Fator de Transcrição RelA/genética , Receptor de GluK2 Cainato
7.
Cancer Genomics Proteomics ; 17(6): 707-714, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33099472

RESUMO

BACKGROUND/AIM: Glutamate receptor GRIK2, previously designated as GluR6, is best described in neuronal cells. However, its biological relevance in non-neuronal cells is not well understood. We have investigated the expression of this important protein in normal human fibroblasts as a function of cell proliferation. MATERIALS AND METHODS: We introduced expression constructs of all five isoforms (A-E) of GRIK2 in normal human fibroblasts and investigated the cells for the presence and localization of GRIK2, as well as for cell proliferation and senescence over a period of 24 days. RESULTS: The expression of GRIK2-A isoform led to immediate cessation of cell proliferation. However, the cell numbers increased by 1.5- to 9.0-fold in 24 days upon transfection with B, C, D and E isoforms, after which they entered a state of senescence. The decreased proliferation was reflected by incorporation of BrdU in only 2-8% of transfected cells even after culturing them for 16 days. CONCLUSION: Our results are indicative of an association between GRIK2 and aging of fibroblasts.


Assuntos
Proliferação de Células , Senescência Celular , Replicação do DNA , Fibroblastos/citologia , Receptores de Ácido Caínico/metabolismo , Apoptose , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Receptores de Ácido Caínico/genética , Receptor de GluK2 Cainato
8.
Int J Mol Sci ; 21(17)2020 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-32825350

RESUMO

Melatonin, a pineal gland secretion, is an amphiphilic neurohormone involved in the biological and physiologic regulation of bodily functions. Numerous studies have shown the effects of melatonin on the release of gonadotropins and their actions at one or several levels of the hypothalamic-pituitary-gonadal axis. However, direct melatonin action on gonadotropin-releasing hormone (GnRH) neurons and its mechanism of action remain unclear. Here, plasma melatonin levels were measured and the effect of melatonin on GnRH neurons was assessed using brain slice patch clamp techniques. The plasma melatonin levels in prepubertal mice were higher than those in the adults. Melatonin itself did not change the firing activity of GnRH neurons. Interestingly, the kainate receptor-mediated responses but not the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)- and N-methyl-D-aspartic acid (NMDA)-induced responses were suppressed by melatonin in both the voltage clamp and current clamp modes. The inhibitory effects of the kainate-induced response by melatonin tended to increase with higher melatonin concentrations and persisted in the presence of tetrodotoxin, a voltage-sensitive Na+ channel blocker, or luzindole, a non-selective melatonin receptor antagonist. However, the response was completely abolished by pretreatment with pertussis toxin. These results suggest that melatonin can regulate GnRH neuronal activities in prepubertal mice by partially suppressing the excitatory signaling mediated by kainate receptors through pertussis toxin-sensitive G-protein-coupled receptors.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Melatonina/farmacologia , Neurônios/fisiologia , Receptores de Ácido Caínico/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Ácido Caínico/farmacologia , Masculino , Melatonina/sangue , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Toxina Pertussis/farmacologia , Puberdade , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
9.
Int J Mol Sci ; 20(17)2019 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-31450867

RESUMO

Kainate (KA) receptors (KAR) have important modulatory roles of synaptic transmission. In the cerebellum, the action mechanisms of KAR-mediated glutamatergic depression are unknown. We studied these mechanisms by recording evoked excitatory postsynaptic currents (eEPSCs) from cerebellar slices using the whole-cell configuration of the patch-clamp technique. We observed that 3 µM KA decreased the amplitude of eEPSCs and increased the number of failures at the synapses established between parallel fibers (PF) and Purkinje neurons, and the effect was antagonized by NBQX under the condition where AMPA receptors were previously blocked. The inhibition of protein kinase A (PKA) suppressed the effect of KAR activation on eEPSC, and effect was not prevented by protein kinase C inhibitors. Furthermore, in the presence of Pertussis toxin, the depression of glutamate release mediated by KAR activation was prevented, invoking the participation of a Gi/o protein in this modulation. Finally, the KAR-mediated depression of glutamate release was not prevented by blocking calcium-permeable KARs or by treatments that affect calcium release from intracellular stores. We conclude that KARs present at these synapses mediate an inhibition of glutamate release through a mechanism that involves the activation of G-protein and protein kinase A.


Assuntos
Cerebelo/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ácido Glutâmico/metabolismo , Receptores de Ácido Caínico/metabolismo , Animais , Cálcio/metabolismo , Fenômenos Eletrofisiológicos , Potenciais Pós-Sinápticos Excitadores , Masculino , Camundongos , Receptores de N-Metil-D-Aspartato/metabolismo , Sistemas do Segundo Mensageiro , Transmissão Sináptica
10.
Mol Carcinog ; 58(7): 1314-1323, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30977227

RESUMO

Glutamate Ionotropic Receptor Kainate Type Subunit 3 (GRIK3) is an important excitatory neurotransmitter receptor that plays a significant role in various neurodegenerative diseases. However, the biological functions of GRIK3 in malignancies are largely unknown because of limited related studies. Here, we primarily reported that the expression of GRIK3 was higher in breast cancer tissues than in adjacent noncancerous tissues. GRIK3 expression was also positively correlated with the prognosis of patients with breast cancer. GRIK3 promoted the proliferation and migration abilities of breast cancer cells and enhanced the growth of orthotopically implanted tumors. Mechanically, GRIK3 influenced a range of signaling pathways and key signal transducers, including two epithelial-mesenchymal transition regulators, SPDEF and CDH1. Heterogenous expression of SPDEF and CDH1 counteracted the migration and invasion abilities, respectively, of breast cancer cells induced by GRIK3. Moreover, overexpression of GRIK3 increased the expression of mesenchymal markers and decreased the expression of epithelial markers, resulting in the translocation of ß-catenin into the nucleus and the increased ß-catenin transcriptional activity. In conclusion, the present study reported a novel oncogenic role of GRIK3. Meanwhile, GRIK3, as a membrane receptor, may also serve as a potential therapeutic target for the treatment of breast cancer.


Assuntos
Antígenos CD/metabolismo , Neoplasias da Mama/patologia , Caderinas/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Proteínas Proto-Oncogênicas c-ets/metabolismo , Receptores de Ácido Caínico/metabolismo , Transporte Ativo do Núcleo Celular/genética , Animais , Neoplasias da Mama/mortalidade , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Transição Epitelial-Mesenquimal/genética , Feminino , Humanos , Células MCF-7 , Camundongos , Camundongos Nus , Prognóstico , Receptores de Ácido Caínico/genética , Transdução de Sinais , beta Catenina/metabolismo , Receptor de GluK3 Cainato
11.
Mol Neurobiol ; 56(8): 5917-5933, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30689196

RESUMO

As mitochondrial dysfunction is evident in neurodegenerative disorders that are accompanied by pain, we generated inducible mutant mice with disruption of mitochondrial respiratory chain complex IV, by COX10 deletion limited to sensory afferent neurons through the use of an Advillin Cre-reporter. COX10 deletion results in a selective energy-deficiency phenotype with minimal production of reactive oxygen species. Mutant mice showed reduced activity of mitochondrial respiratory chain complex IV in many sensory neurons, increased ADP/ATP ratios in dorsal root ganglia and dorsal spinal cord synaptoneurosomes, as well as impaired mitochondrial membrane potential, in these synaptoneurosome preparations. These changes were accompanied by marked pain hypersensitivity in mechanical and thermal (hot and cold) tests without altered motor function. To address the underlying basis, we measured Ca2+ fluorescence responses of dorsal spinal cord synaptoneurosomes to activation of the GluK1 (kainate) receptor, which we showed to be widely expressed in small but not large nociceptive afferents, and is minimally expressed elsewhere in the spinal cord. Synaptoneurosomes from mutant mice showed greatly increased responses to GluK1 agonist. To explore whether altered nucleotide levels may play a part in this hypersensitivity, we pharmacologically interrogated potential roles of AMP-kinase and ADP-sensitive purinergic receptors. The ADP-sensitive P2Y1 receptor was clearly implicated. Its expression in small nociceptive afferents was increased in mutants, whose in vivo pain hypersensitivity, in mechanical, thermal and cold tests, was reversed by a selective P2Y1 antagonist. Energy depletion and ADP elevation in sensory afferents, due to mitochondrial respiratory chain complex IV deficiency, appear sufficient to induce pain hypersensitivity, by ADP activation of P2Y1 receptors.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/genética , Hipersensibilidade/patologia , Mitocôndrias/metabolismo , Mutação/genética , Neurônios Aferentes/patologia , Dor/patologia , Receptores Purinérgicos P2Y1/metabolismo , Difosfato de Adenosina/metabolismo , Monofosfato de Adenosina/metabolismo , Alquil e Aril Transferases/metabolismo , Animais , Comportamento Animal , Cálcio/metabolismo , Células Cultivadas , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Fluorescência , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Hipersensibilidade/complicações , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/metabolismo , Nociceptividade/efeitos dos fármacos , Dor/complicações , Fenótipo , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Receptores de Ácido Caínico/metabolismo , Medula Espinal/patologia , Sinapses/efeitos dos fármacos , Sinapses/metabolismo
12.
Biochim Biophys Acta Mol Cell Res ; 1866(3): 459-473, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30339823

RESUMO

Classically, endoplasmic reticulum (ER) retention signals in secreted integral membrane proteins impose the requirement to assemble with other cognate subunits to form functional assemblies before they can exit the ER. We report that GluK5 has two ER retention signals in its cytoplasmic C-terminus: an arginine-based signal and a di-leucine motif previously thought to be an endocytic motif. GluK5 assembles with GluK2, but surprisingly GluK2 association does little to block the ER retention signals. We find instead that the ER retention signals are blocked by two proteins involved in intracellular trafficking, SAP97 and CASK. We show that SAP97, in the presence of CASK and the receptor complex, assumes an extended conformation. In the extended conformation, SAP97 makes its SH3 and GuK domains available to bind and sterically mask the ER retention signals in the GluK5 C-terminus. SAP97 and CASK are also necessary for sorting receptor cargoes into the local dendritic secretory pathway in neurons. We show that the ER retention signals of GluK5 play a vital role in sorting the receptor complex in the local dendritic secretory pathway in neurons. These data suggest a new role for ER retention signals in trafficking integral membrane proteins in neurons. SIGNIFICANCE: We present evidence that the ER retention signals in the kainate receptors containing GluK5 impose a requirement for sorting into local dendritic secretory pathways in neurons, as opposed to traversing the somatic Golgi apparatus. There are two ER retention signals in the C-terminus of GluK5. We show that both are blocked by physical association with SAP97 and CASK. The SH3 and GuK domains of SAP97, in the presence of CASK, bind directly to each ER retention signal and form a complex. These results support an entirely new function for ER retention signals in the C-termini of neuronal receptors, such as NMDA and kainate receptors, and define a mechanism for selective entry of receptors into local secretory pathways.


Assuntos
Retículo Endoplasmático/metabolismo , Guanilato Quinases/metabolismo , Receptores de Ácido Caínico/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Aminoácidos , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Proteína 1 Homóloga a Discs-Large , Complexo de Golgi/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Neurônios/metabolismo , Transporte Proteico
13.
J Gen Physiol ; 151(4): 435-451, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30498132

RESUMO

Kainate receptors are ligand-gated ion channels that have two major roles in the central nervous system: they mediate a postsynaptic component of excitatory neurotransmission at some glutamatergic synapses and modulate transmitter release at both excitatory and inhibitory synapses. Accumulating evidence implicates kainate receptors in a variety of neuropathologies, including epilepsy, psychiatric disorders, developmental delay, and cognitive impairment. Here, to gain a deeper understanding of the conformational changes associated with agonist binding and channel opening, we generate a series of Cys substitutions in the GluK2 kainate receptor subunit, focusing on the M3 helices that line the ion pore and form the bundle-crossing gate at the extracellular mouth of the channel. Exposure to 50 µM Cd produces direct activation of homomeric mutant channels bearing Cys substitutions in (A657C), or adjacent to (L659C), the conserved SYTANLAAF motif. Activation by Cd is occluded by modification with 2-aminoethyl MTS (MTSEA), indicating that Cd binds directly and specifically to the substituted cysteines. Cd potency for the A657C mutation (EC50 = 10 µM) suggests that binding involves at least two coordinating residues, whereas weaker Cd potency for L659C (EC50 = 2 mM) implies that activation does not require tight coordination by multiple side chains for this substitution. Experiments with heteromeric and chimeric channels indicate that activation by Cd requires Cys substitution at only two of the four subunits within a tetrameric receptor and that activation is similar for substitution within subunits in either the A/C or B/D conformations. We develop simple kinetic models for the A657C substitution that reproduce several features of Cd activation as well as the low-affinity inhibition observed at higher Cd concentrations (5-20 mM). Together, these results demonstrate rapid and reversible channel activation, independent of agonist site occupancy, upon Cd binding to Cys side chains at two specific locations along the GluK2 inner helix.


Assuntos
Cádmio/farmacologia , Cisteína/química , Receptores de Ácido Caínico/metabolismo , Substituição de Aminoácidos , Células HEK293 , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Subunidades Proteicas , Receptores de Ácido Caínico/genética , Receptor de GluK2 Cainato
14.
Mol Neurobiol ; 56(7): 5095-5110, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30484111

RESUMO

Kainic acid (KA) is an analogue of the excitatory neurotransmitter glutamate that, when injected systemically into adult rats, can trigger seizures and progressive neuronal loss in a manner that mirrors the neuropathology of human mesial temporal lobe epilepsy. However, biomolecular mechanisms responsible for the neuronal loss that occurs as a consequence of this treatment remains elusive. We have recently reported that toxicity induced by KA can partly be mediated by astrocyte-derived amyloid ß (Aß) peptides, which are critical in the development of Alzheimer's disease (AD). Nonetheless, little is known how KA can influence amyloid precursor protein (APP) levels and processing in astrocytes. Thus, in the present study using human U-373 astrocytoma and rat primary astrocytes, we evaluated the role of KA on APP metabolism. Our results revealed that KA treatment increased the levels of APP and its cleaved products (α-/ß-CTFs) in cultured U-373 astrocytoma and primary astrocytes, without altering the cell viability. The cellular and secretory levels of Aß1-40/Aß1-42 were markedly increased in KA-treated astrocytes. We also demonstrated that the steady-state levels of APP-secretases were not altered but the activity of γ-secretase is enhanced in KA-treated U-373 astrocytoma. Furthermore, using selective receptor antagonists, we showed that the effects of KA is mediated by activation of kainate receptors and not NMDA or AMPA receptors. These results suggest that KA can enhance amyloidogenic processing of APP by activating its own receptor leading to increased production/secretion of Aß-related peptides from activated astrocytes which may contribute to the pathogenesis of temporal lobe epilepsy.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Ácido Caínico/farmacologia , Receptores de Ácido Caínico/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Humanos , Ácido Caínico/toxicidade , Receptores de Ácido Caínico/agonistas
15.
Cancer Genomics Proteomics ; 16(1): 59-64, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30587499

RESUMO

BACKGROUND: The aberrant regulation of growth and proliferation is a key feature of carcinoma cells. In order to use molecular strategies to correct these defects toward therapeutic purposes, it is important to characterize the entire spectrum of causative molecules. MATERIALS AND METHODS: By using gene transfer technique, SKOV3 ovarian carcinoma cells were transduced with an expression construct of glutamate receptor 6 (glutamate ionotropic receptor kainate type subunit 2, GRIK2) in retroviral vector PQCXIP. The senescence of transduced cells was subsequently characterized. RESULTS: Our results demonstrated that retroviral transduction occurs with high frequency and transduced cells continue to proliferate, albeit at a significantly reduced rate, up to 39 days. Some transduced colonies stopped proliferating after 12 days, and none of the clones proliferated beyond 37 days. The doubling time for these transduced cells increased progressively until they reached a complete cell-cycle arrest. The proliferating cells were distinguished by bromodeoxyuridine incorporation and 3-(4, 5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide assay. The growth and cell cycle arrest in transduced cells accompanied activation of senescence-associated ß-galactosidase. Furthermore, we have demonstrated a decrease in the levels of active protein kinase B and increase in the abundance of inactive cyclin-dependent kinase 1. CONCLUSION: These results indicate involvement of GRIK2 in senescence and suggests GRIK2 as a potential target for therapeutic intervention of cancer cells.


Assuntos
Carcinoma/metabolismo , Senescência Celular , Receptores de Ácido Caínico/metabolismo , Carcinoma/genética , Linhagem Celular Tumoral , Sobrevivência Celular , Senescência Celular/genética , Imunofluorescência , Expressão Gênica , Técnicas de Transferência de Genes , Genes Reporter , Ácido Glutâmico/metabolismo , Humanos , Imuno-Histoquímica , Isoformas de Proteínas , Receptores de Ácido Caínico/química , Receptores de Ácido Caínico/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Receptor de GluK2 Cainato
16.
J Cell Sci ; 131(24)2018 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-30559217

RESUMO

Kainate receptors (KARs) regulate neuronal excitability and network function. Most KARs contain the subunit GluK2 (also known as GRIK2), and the properties of these receptors are determined in part by ADAR2 (also known as ADARB1)-mediated mRNA editing of GluK2, which changes a genomically encoded glutamine residue into an arginine residue (Q/R editing). Suppression of synaptic activity reduces ADAR2-dependent Q/R editing of GluK2 with a consequential increase in GluK2-containing KAR surface expression. However, the mechanism underlying this reduction in GluK2 editing has not been addressed. Here, we show that induction of KAR upscaling, a phenomenon in which surface expression of receptors is increased in response to a chronic decrease in synaptic activity, results in proteasomal degradation of ADAR2, which reduces GluK2 Q/R editing. Because KARs incorporating unedited GluK2(Q) assemble and exit the ER more efficiently, this leads to an upscaling of KAR surface expression. Consistent with this, we demonstrate that partial ADAR2 knockdown phenocopies and occludes KAR upscaling. Moreover, we show that although the AMPA receptor (AMPAR) subunit GluA2 (also known as GRIA2) also undergoes ADAR2-dependent Q/R editing, this process does not mediate AMPAR upscaling. These data demonstrate that activity-dependent regulation of ADAR2 proteostasis and GluK2 Q/R editing are key determinants of KAR, but not AMPAR, trafficking and upscaling.This article has an associated First Person interview with the first author of the paper.


Assuntos
Adenosina Desaminase/metabolismo , Edição de RNA/genética , Receptores de AMPA/metabolismo , Receptores de Ácido Caínico/metabolismo , Animais , Transporte Proteico/fisiologia , Proteínas de Ligação a RNA/metabolismo , Ratos Wistar , Receptor de GluK2 Cainato
17.
Brain Res ; 1699: 69-78, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30343685

RESUMO

A combination of a beneficial neuroprotectant, hypothermia, with targeted medication is a perspective therapeutic approach. Here, we analyzed both non-specific (deep and profound hypothermia, 27 °C and 17 °C, respectively) and targeted (anticonvulsant drug levetiracetam) modulation of l-[14C]glutamate release induced by activation of presynaptic NMDA, AMPA, and kainate receptors in rat brain nerve terminals (synaptosomes). Gradual dynamics of hypothermia-mediated decrease in synaptosomal l-[14C]glutamate release evoked by the receptor agonists NMDA-, AMPA-, and kainate (250 µM) has been demonstrated that can be of value for the justification of optimal temperature regimes in therapeutic hypothermia. 250 µM NMDA-induced l-[14C]glutamate release from nerve terminals was higher in the presence of levetiracetam (100 µM) as compared to that without the drug. Despite levetiracetam effects decreased in hypothermia, combined application of hypothermia and levetiracetam resulted in higher NMDA-induced l-[14C]glutamate release from nerve terminals as compared to that without the drug. These effects were not revealed for synaptosomal AMPA- and kainate-induced l-[14C]glutamate release in the presence of levetiracetam at the similar concentration. Therefore, levetiracetam administration significantly mitigated a hypothermia-induced decrease in NMDA receptor response at the presynaptic level and can be used for the targeted neurocorrection to reduce side effects of hypothermia in cardiac surgery. However, levetiracetam-mediated improvement of NMDA receptor response is not applicable in stroke, brain trauma and neonatal asphyxia therapies, where the main neuroprotective action of hypothermia is associated with prevention of damaging consequence of pre-existing acute glutamate exitotoxicity.


Assuntos
Ácido Glutâmico/metabolismo , Hipotermia/tratamento farmacológico , Levetiracetam/farmacologia , Neurotransmissores/farmacologia , Sinaptossomos/efeitos dos fármacos , Transativadores/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Hipotermia/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Ratos Wistar , Receptores de AMPA/metabolismo , Receptores de Ácido Caínico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinaptossomos/metabolismo
18.
Genome Biol ; 18(1): 195, 2017 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-29061182

RESUMO

BACKGROUND: Adenosine to inosine (A-to-I) RNA editing has been shown to be an essential event that plays a significant role in neuronal function, as well as innate immunity, in mammals. It requires a structure that is largely double-stranded for catalysis but little is known about what determines editing efficiency and specificity in vivo. We have previously shown that some editing sites require adjacent long stem loop structures acting as editing inducer elements (EIEs) for efficient editing. RESULTS: The glutamate receptor subunit A2 is edited at the Q/R site in almost 100% of all transcripts. We show that efficient editing at the Q/R site requires an EIE in the downstream intron, separated by an internal loop. Also, other efficiently edited sites are flanked by conserved, highly structured EIEs and we propose that this is a general requisite for efficient editing, while sites with low levels of editing lack EIEs. This phenomenon is not limited to mRNA, as non-coding primary miRNAs also use EIEs to recruit ADAR to specific sites. CONCLUSIONS: We propose a model where two regions of dsRNA are required for efficient editing: first, an RNA stem that recruits ADAR and increases the local concentration of the enzyme, then a shorter, less stable duplex that is ideal for efficient and specific catalysis. This discovery changes the way we define and determine a substrate for A-to-I editing. This will be important in the discovery of novel editing sites, as well as explaining cases of altered editing in relation to disease.


Assuntos
Adenosina/metabolismo , Inosina/metabolismo , Edição de RNA , Sequências Reguladoras de Ácido Ribonucleico , Adenosina Desaminase/metabolismo , Animais , Células HEK293 , Células HeLa , Humanos , Canal de Potássio Kv1.1/genética , Canal de Potássio Kv1.1/metabolismo , Camundongos , MicroRNAs/química , MicroRNAs/metabolismo , Conformação de Ácido Nucleico , Receptores de AMPA/genética , Receptores de AMPA/metabolismo , Receptores de Ácido Caínico/genética , Receptores de Ácido Caínico/metabolismo , Transcriptoma , Receptor de GluK2 Cainato
19.
Oncotarget ; 8(17): 28826-28839, 2017 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-28418868

RESUMO

Cancer stem-like cells (CSCs)/cancer-initiating cells (CICs) are small sub-population of cancer cells that are endowed with higher tumor-initiating ability, self-renewal ability and differentiation ability. CSCs/CICs could be isolated as high aldehyde dehydrogenase 1 activity cells (ALDH1high) from various cancer samples. In this study, we isolated urothelial carcinoma CSCs/CICs as ALDHhigh cells and investigated the molecular aspects. ALDH1high cells showed greater sphere-forming ability and higher tumor-initiating ability in immune-deficient mice than those of ALDH1low cells, indicating that CSCs/CICs were enriched in ALDH1high cells. cDNA microarray analysis revealed that an ionotropic glutamate receptor glutamate receptor, ionotropic, kainate 2 (GRIK2) was expressed in ALDH1high cells at a higher level than that in ALDH1low cells. GRIK2 gene knockdown by siRNAs decreased the sphere-forming ability and invasion ability, whereas GRIK2 overexpression increased the sphere-forming ability, invasion ability and tumorigenicity, indicating that GRIK2 has a role in the maintenance of CSCs/CICs. Immunohistochemical staining revealed that higher levels of GRIK2 and ALDH1 expression were related to poorer prognosis in urinary tract carcinoma cases. The findings indicate that GRIK2 has a role in the maintenance of urothelial CSCs/CICs and that GRIK2 and ALDH1 can be prognosis prediction markers for urinary tract carcinomas.


Assuntos
Isoenzimas/metabolismo , Células-Tronco Neoplásicas/patologia , Receptores de Ácido Caínico/metabolismo , Retinal Desidrogenase/metabolismo , Neoplasias Urológicas/genética , Urotélio/patologia , Família Aldeído Desidrogenase 1 , Animais , Carcinogênese , Linhagem Celular Tumoral , Autorrenovação Celular/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos SCID , Células-Tronco Neoplásicas/metabolismo , Prognóstico , RNA Interferente Pequeno/genética , Receptores de Ácido Caínico/genética , Neoplasias Urológicas/diagnóstico , Neoplasias Urológicas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Receptor de GluK2 Cainato
20.
Neuroscience ; 340: 153-165, 2017 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-27984177

RESUMO

Hydrogen sulfide (H2S) is an endogenous gasotransmitter with neuroprotective properties that participates in the regulation of transmitter release and neuronal excitability in various brain structures. The role of H2S in the growth and maturation of neural networks however remains unclear. The aim of the present study is to reveal the effects of H2S on neuronal spontaneous activity relevant to neuronal maturation in hippocampal slices of neonatal rats. Sodium hydrosulfide (NaHS) (100µM), a classical donor of H2S produced a biphasic effect with initial activation and subsequent concentration-dependent suppression of network-driven giant depolarizing potentials (GDPs) and neuronal spiking activity. Likewise, the substrate of H2S synthesis l-cysteine (1mM) induced an initial increase followed by an inhibition of GDPs and spiking activity. Our experiments indicate that the increase in initial discharge activity by NaHS is evoked by neuronal depolarization which is partially mediated by a reduction of outward K+ currents. The subsequent decrease in the neuronal activity by H2S appears to be due to the rightward shift of activation and inactivation of voltage-gated Na+ currents, thus preventing network activity. NaHS also reduced N-methyl-d-aspartate (NMDA)-mediated currents, without essential effect on AMPA/kainate or GABAA-mediated currents. Finally, H2S abolished the interictal-like events induced by bicuculline. In summary, our results suggest that through the inhibitory action on voltage-gated Na+ channels and NMDA receptors, H2S prevents the enhanced neuronal excitability typical to early hippocampal networks.


Assuntos
Anticonvulsivantes/farmacologia , Epilepsia/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Sulfeto de Hidrogênio/farmacologia , Animais , Animais Recém-Nascidos , Cátions Monovalentes/metabolismo , Epilepsia/fisiopatologia , Hipocampo/crescimento & desenvolvimento , Hipocampo/fisiopatologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Fármacos Neuroprotetores/farmacologia , Técnicas de Patch-Clamp , Potássio/metabolismo , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiologia , Ratos Wistar , Receptores de AMPA/metabolismo , Receptores de GABA-A/metabolismo , Receptores de Ácido Caínico/metabolismo , Receptores de N-Metil-D-Aspartato , Sódio/metabolismo , Tetrodotoxina/farmacologia , Técnicas de Cultura de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA