Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(6): e2312861121, 2024 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-38285939

RESUMO

The N6-methyladenosine (m6A) modification of RNA is an emerging epigenetic regulatory mechanism that has been shown to participate in various pathophysiological processes. However, its involvement in modulating neuropathic pain is still poorly understood. In this study, we elucidate a functional role of the m6A demethylase alkylation repair homolog 5 (ALKBH5) in modulating trigeminal-mediated neuropathic pain. Peripheral nerve injury selectively upregulated the expression level of ALKBH5 in the injured trigeminal ganglion (TG) of rats. Blocking this upregulation in injured TGs alleviated trigeminal neuropathic pain, while mimicking the upregulation of ALKBH5 in intact TG neurons sufficiently induced pain-related behaviors. Mechanistically, histone deacetylase 11 downregulation induced by nerve injury increases histone H3 lysine 27 acetylation (H3K27ac), facilitating the binding of the transcription factor forkhead box protein D3 (FOXD3) to the Alkbh5 promoter and promoting Alkbh5 transcription. The increased ALKBH5 erases m6A sites in Htr3a messenger RNA (mRNA), resulting in an inability of YT521-B homology domain 2 (YTHDF2) to bind to Htr3a mRNA, thus causing an increase in 5-HT3A protein expression and 5-HT3 channel currents. Conversely, blocking the increased expression of ALKBH5 in the injured TG destabilizes nerve injury-induced 5-HT3A upregulation and reverses mechanical allodynia, and the effect can be blocked by 5-HT3A knockdown. Together, FOXD3-mediated transactivation of ALKBH5 promotes neuropathic pain through m6A-dependent stabilization of Htr3a mRNA in TG neurons. This mechanistic understanding may advance the discovery of new therapeutic targets for neuropathic pain management.


Assuntos
Neuralgia , Neuralgia do Trigêmeo , Animais , Ratos , Homólogo AlkB 5 da RNA Desmetilase/genética , Homólogo AlkB 5 da RNA Desmetilase/metabolismo , Neuralgia/genética , Neuralgia/metabolismo , RNA Mensageiro/metabolismo , Células Receptoras Sensoriais/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional/genética , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Receptores 5-HT3 de Serotonina/genética
2.
Proc Natl Acad Sci U S A ; 119(14): e2117209119, 2022 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-35353623

RESUMO

microRNA (miRNA)­mediated gene regulation has been studied as a therapeutic approach, but its functional regulatory mechanism in neuropathic pain is not well understood. Here, we identify that miRNA-32-5p (miR-32-5p) is a functional RNA in regulating trigeminal-mediated neuropathic pain. High-throughput sequencing and qPCR analysis showed that miR-32-5p was the most down-regulated miRNA in the injured trigeminal ganglion (TG) of rats. Intra-TG injection of miR-32-5p agomir or overexpression of miR-32-5p by lentiviral delivery in neurons of the injured TG attenuated established trigeminal neuropathic pain. miR-32-5p overexpression did not affect acute physiological pain, while miR-32-5p down-regulation in intact rats was sufficient to cause pain-related behaviors. Nerve injury increased the methylated histone occupancy of binding sites for the transcription factor glucocorticoid receptor in the miR-32-5p promoter region. Inhibition of the enzymes that catalyze H3K9me2 and H3K27me3 restored the expression of miR-32-5p and markedly attenuated pain behaviors. Further, miR-32-5p­targeted Cav3.2 T-type Ca2+ channels and decreased miR-32-5p associated with neuropathic pain caused an increase in Cav3.2 protein expression and T-type channel currents. Conversely, miR-32-5p overexpression in injured TG suppressed the increased expression of Cav3.2 and reversed mechanical allodynia. Together, we conclude that histone methylation-mediated miR-32-5p down-regulation in TG neurons regulates trigeminal neuropathic pain by targeting Cav3.2 channels.


Assuntos
MicroRNAs , Neuralgia , Animais , Regulação para Baixo , Gânglios Espinais/metabolismo , Histonas/genética , Histonas/metabolismo , Metilação , MicroRNAs/genética , MicroRNAs/metabolismo , Neuralgia/metabolismo , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/metabolismo
3.
J Headache Pain ; 24(1): 49, 2023 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-37158881

RESUMO

BACKGROUND: Trace amines, such as tyramine, are endogenous amino acid metabolites that have been hypothesized to promote headache. However, the underlying cellular and molecular mechanisms remain unknown. METHODS: Using patch-clamp recording, immunostaining, molecular biological approaches and behaviour tests, we elucidated a critically functional role of tyramine in regulating membrane excitability and pain sensitivity by manipulating Kv1.4 channels in trigeminal ganglion (TG) neurons. RESULTS: Application of tyramine to TG neurons decreased the A-type K+ current (IA) in a manner dependent on trace amine-associated receptor 1 (TAAR1). Either siRNA knockdown of Gαo or chemical inhibition of ßγ subunit (Gßγ) signaling abrogated the response to tyramine. Antagonism of protein kinase C (PKC) prevented the tyramine-induced IA response, while inhibition of conventional PKC isoforms or protein kinase A elicited no such effect. Tyramine increased the membrane abundance of PKCθ in TG neurons, and either pharmacological or genetic inhibition of PKCθ blocked the TAAR1-mediated IA decrease. Furthermore, PKCθ-dependent IA suppression was mediated by Kv1.4 channels. Knockdown of Kv1.4 abrogated the TAAR1-induced IA decrease, neuronal hyperexcitability, and pain hypersensitivity. In a mouse model of migraine induced by electrical stimulation of the dura mater surrounding the superior sagittal sinus, blockade of TAAR1 signaling attenuated mechanical allodynia; this effect was occluded by lentiviral overexpression of Kv1.4 in TG neurons. CONCLUSION: These results suggest that tyramine induces Kv1.4-mediated IA suppression through stimulation of TAAR1 coupled to the Gßγ-dependent PKCθ signaling cascade, thereby enhancing TG neuronal excitability and mechanical pain sensitivity. Insight into TAAR1 signaling in sensory neurons provides attractive targets for the treatment of headache disorders such as migraine.


Assuntos
Nociceptividade , Gânglio Trigeminal , Animais , Camundongos , Neurônios , Dor
4.
Sensors (Basel) ; 22(16)2022 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-36015945

RESUMO

Structural health monitoring (SHM) is gradually replacing traditional manual detection and is becoming a focus of the research devoted to the operation and maintenance of tunnel structures. However, in the face of massive SHM data, the autonomous early warning method is still required to further reduce the burden of manual analysis. Thus, this study proposed a dynamic warning method for SHM data based on ARIMA and applied it to the concrete strain data of the Hong Kong-Zhuhai-Macao Bridge (HZMB) immersed tunnel. First, wavelet threshold denoising was applied to filter noise from the SHM data. Then, the feasibility and accuracy of establishing an ARIMA model were verified, and it was adopted to predict future time series of SHM data. After that, an anomaly detection scheme was proposed based on the dynamic model and dynamic threshold value, which set the confidence interval of detected anomalies based on the statistical characteristics of the historical series. Finally, a hierarchical warning system was defined to classify anomalies according to their detection threshold and enable hierarchical treatments. The illustrative example of the HZMB immersed tunnel verified that a three-level (5.5 σ, 6.5 σ, and 7.5 σ) dynamic warning schematic can give good results of anomalies detection and greatly improves the efficiency of SHM data management of the tunnel.


Assuntos
Projetos de Pesquisa , Previsões , Hong Kong , Macau
5.
J Biol Chem ; 294(14): 5496-5507, 2019 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-30745360

RESUMO

α-Melanocyte-stimulating hormone (α-MSH) has been shown to be involved in nociception, but the underlying molecular mechanisms remain largely unknown. In this study, we report that α-MSH suppresses the transient outward A-type K+ current (IA) in trigeminal ganglion (TG) neurons and thereby modulates neuronal excitability and peripheral pain sensitivity in rats. Exposing small-diameter TG neurons to α-MSH concentration-dependently decreased IA This α-MSH-induced IA decrease was dependent on the melanocortin type 4 receptor (MC4R) and associated with a hyperpolarizing shift in the voltage dependence of A-type K+ channel inactivation. Chemical inhibition of phosphatidylinositol 3-kinase (PI3K) with wortmannin or of class I PI3Ks with the selective inhibitor CH5132799 prevented the MC4R-mediated IA response. Blocking Gi/o-protein signaling with pertussis toxin or by dialysis of TG neurons with the Gßγ-blocking synthetic peptide QEHA abolished the α-MSH-mediated decrease in IA Further, α-MSH increased the expression levels of phospho-p38 mitogen-activated protein kinase, and pharmacological or genetic inhibition of p38α abrogated the α-MSH-induced IA response. Additionally, α-MSH significantly increased the action potential firing rate of TG neurons and increased the sensitivity of rats to mechanical stimuli applied to the buccal pad area, and both effects were abrogated by IA blockade. Taken together, our findings suggest that α-MSH suppresses IA by activating MC4R, which is coupled sequentially to the Gßγ complex of the Gi/o-protein and downstream class I PI3K-dependent p38α signaling, thereby increasing TG neuronal excitability and mechanical pain sensitivity in rats.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Dor/metabolismo , Canais de Potássio/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Células Receptoras Sensoriais/metabolismo , Transdução de Sinais/efeitos dos fármacos , Gânglio Trigeminal/metabolismo , alfa-MSH/farmacologia , Animais , Proteínas de Ligação ao GTP/metabolismo , Dor/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Ratos , Ratos Sprague-Dawley , Receptor Tipo 4 de Melanocortina/agonistas , Células Receptoras Sensoriais/patologia , Gânglio Trigeminal/patologia , Wortmanina/farmacologia
6.
Mol Pain ; 16: 1744806920931737, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32513089

RESUMO

Interleukin-33 (IL-33)/suppressor of tumorigenicity 2 (ST2) signaling is known to promote inflammation and the genesis and maintenance of neuropathic pain. However, it remained mostly unknown how IL-33/ST2 signaling can be enhanced by neuropathic stimulations. Here, we report that the chronic constriction nerve injury (CCI)-induced increases in the expression of IL-33 and ST2 and a decrease in microRNA (miRNA)-547-5p not only in the dorsal root ganglia (DRG) but also in spinal dorsal horn (SDH) ipsilateral to the CCI. We found that increasing endogenous miRNA-547-5p by the intrathecal (i.t.) infusion of agomir-miR-547-5p did not produce any effect in naive rats but blocked the CCI-induced increases in the IL-33 and ST2, and pain sensitivity. The reducing endogenous miRNA-547-5p by the i.t. delivering antagomir-miR-547-5p into naive rats caused significant changes in IL-33 and ST2 expressions in both the DRG and SDH, and pain sensitivity, which were similar to those induced by the CCI. Since increasing IL-33 by the i.t. infusion of recombinant IL-33 produced no change in the expression of miR-547-5p, and the CCI still reduced miR-547-5p expression in rats with the IL-33 knockdown, we conclude that the reduction of miR-547-5p can be an upstream event leading to the enhancement of IL-33/ST2 signaling induced by the CCI. The intravenous application of bone marrow stromal cells (BMSCs) reduced the depression of miR-547-5p in both the DRG and SDH, and pain hypersensitivity produced by the CCI or antagomir-miR547-5p application. However, the BMSC effect was significantly occluded by the pretreatment with miR-547-5p agomir or the IL-33 knockdown, demonstrating a novel mechanism underlying the BMSC therapy.


Assuntos
Interleucina-33/metabolismo , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Neuralgia/genética , Neuralgia/terapia , Receptores de Interleucina-1/metabolismo , Transdução de Sinais , Regiões 3' não Traduzidas/genética , Animais , Antagomirs/metabolismo , Sequência de Bases , Constrição Patológica , Gânglios Espinais/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Interleucina-33/genética , Masculino , MicroRNAs/genética , Neuralgia/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Receptores de Interleucina-1/genética , Corno Dorsal da Medula Espinal/metabolismo , Regulação para Cima/genética
7.
Mol Pain ; 16: 1744806920930858, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32484026

RESUMO

AIMS: The arcuate nucleus is a vital brain region for coursing of pain command. G protein-coupled kinase 6 (GRK6) accommodates signaling through G protein-coupled receptors. Studies have demonstrated that GRK6 is involved in inflammatory pain and neuropathic pain. The present study was designed to explore the role and the underlying mechanism of GRK6 in arcuate nucleus of chronic visceral pain. METHODS: Chronic visceral pain of rats was induced by neonatal maternal deprivation and evaluated by monitoring the threshold of colorectal distension. Western blotting, immunofluorescence, real-time quantitative polymerase chain reaction techniques, and Nissl staining were employed to determine the expression and mutual effect of GRK6 with nuclear factor κB (NF-κB). RESULTS: Expression of GRK6 in arcuate nucleus was significantly reduced in neonatal maternal deprivation rats when compared with control rats. GRK6 was mainly expressed in arcuate nucleus neurons, but not in astrocytes, and a little in microglial cells. Neonatal maternal deprivation reduced the percentage of GRK6-positive neurons of arcuate nucleus. Overexpression of GRK6 by Lentiviral injection into arcuate nucleus reversed chronic visceral pain in neonatal maternal deprivation rats. Furthermore, the expression of NF-κB in arcuate nucleus was markedly upregulated in neonatal maternal deprivation rats. NF-κB selective inhibitor pyrrolidine dithiocarbamate suppressed chronic visceral pain in neonatal maternal deprivation rats. GRK6 and NF-κB were expressed in the arcuate nucleus neurons. Importantly, overexpression of GRK6 reversed NF-κB expression at the protein level. In contrast, injection of pyrrolidine dithiocarbamate once daily for seven consecutive days did not alter GRK6 expression in arcuate nucleus of neonatal maternal deprivation rats. CONCLUSIONS: Present data suggest that GRK6 might be a pivotal molecule participated in the central mechanisms of chronic visceral pain, which might be mediated by inhibiting NF-κB signal pathway. Overexpression of GRK6 possibly represents a potential strategy for therapy of chronic visceral pain.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Dor Crônica/metabolismo , Regulação para Baixo , Quinases de Receptores Acoplados a Proteína G/genética , Privação Materna , NF-kappa B/metabolismo , Regulação para Cima/genética , Dor Visceral/metabolismo , Animais , Animais Recém-Nascidos , Dor Crônica/complicações , Regulação para Baixo/efeitos dos fármacos , Quinases de Receptores Acoplados a Proteína G/metabolismo , Masculino , NF-kappa B/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Pirrolidinas/farmacologia , Ratos Sprague-Dawley , Tiocarbamatos/farmacologia , Regulação para Cima/efeitos dos fármacos , Dor Visceral/complicações
8.
J Neurosci Res ; 98(2): 384-403, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31407399

RESUMO

The cAMP-dependent protein kinase A family (PKAs), protein kinase C family (PKCs), and Src family kinases (SFKs) are found to play important roles in pain hypersensitivity. However, more detailed investigations are still needed in order to understand the mechanisms underlying the actions of PKAs, PKCs, and SFKs. Neurons in the hypothalamic arcuate nucleus (ARC) are found to be involved in the regulation of pain hypersensitivity. Here we report that the action potential (AP) firing activity of ARC neurons in culture was up-regulated by application of the adenylate cyclase activator forskolin or the PKC activator PMA, and that the forskolin or PMA application-induced up-regulation of AP firing activity could be blocked by pre-application of the SFK inhibitor PP2. SFK activation also up-regulated the AP firing activity and this effect could be prevented by pre-application of the inhibitors of PKCs, but not of PKAs. Furthermore, we identified that forskolin or PMA application caused increases in the phosphorylation not only in PKAs at T197 or PKCs at S660 and PKCα/ßII at T638/641, but also in SFKs at Y416. The forskolin or PMA application-induced increase in the phosphorylation of PKAs or PKCs was not affected by pre-treatment with PP2. The regulations of the SFK and AP firing activities by PKCs were independent upon the translocation of either PKCα or PKCßII. Thus, it is demonstrated that PKAs may act as an upstream factor(s) to enhance SFKs while PKCs and SFKs interact reciprocally, and thereby up-regulate the AP firing activity in hypothalamic ARC neurons.


Assuntos
Potenciais de Ação/fisiologia , Núcleo Arqueado do Hipotálamo/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Neurônios/metabolismo , Proteína Quinase C/metabolismo , Quinases da Família src/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Células Cultivadas , Colforsina/farmacologia , Feminino , Masculino , Neurônios/efeitos dos fármacos , Oligopeptídeos/farmacologia , Técnicas de Patch-Clamp , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Vasodilatadores/farmacologia
9.
Cell Commun Signal ; 17(1): 68, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31215470

RESUMO

BACKGROUND: Cholecystokinin (CCK) is implicated in the regulation of nociceptive sensitivity of primary afferent neurons. Nevertheless, the underlying cellular and molecular mechanisms remain unknown. METHODS: Using patch clamp recording, western blot analysis, immunofluorescent labelling, enzyme-linked immunosorbent assays, adenovirus-mediated shRNA knockdown and animal behaviour tests, we studied the effects of CCK-8 on the sensory neuronal excitability and peripheral pain sensitivity mediated by A-type K+ channels. RESULTS: CCK-8 reversibly and concentration-dependently decreased A-type K+ channel (IA) in small-sized dorsal root ganglion (DRG) neurons through the activation of CCK type B receptor (CCK-BR), while the sustained delayed rectifier K+ current was unaffected. The intracellular subunit of CCK-BR coimmunoprecipitated with Gαo. Blocking G-protein signaling with pertussis toxin or by the intracellular application of anti-Gß antibody reversed the inhibitory effects of CCK-8. Antagonism of phosphatidylinositol 3-kinase (PI3K) but not of its common downstream target Akts abolished the CCK-BR-mediated IA response. CCK-8 application significantly activated JNK mitogen-activated protein kinase. Antagonism of either JNK or c-Src prevented the CCK-BR-mediated IA decrease, whereas c-Src inhibition attenuated the CCK-8-induced p-JNK activation. Application of CCK-8 enhanced the action potential firing rate of DRG neurons and elicited mechanical and thermal pain hypersensitivity in mice. These effects were mediated by CCK-BR and were occluded by IA blockade. CONCLUSION: Our findings indicate that CCK-8 attenuated IA through CCK-BR that is coupled to the Gßγ-dependent PI3K and c-Src-mediated JNK pathways, thereby enhancing the sensory neuronal excitability in DRG neurons and peripheral pain sensitivity in mice.


Assuntos
Proteína Tirosina Quinase CSK/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fosfatidilinositol 3-Quinase/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Receptor de Colecistocinina B/metabolismo , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/efeitos dos fármacos , Animais , Gânglios Espinais/citologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Nociceptividade/efeitos dos fármacos , Dor/patologia , Dor/fisiopatologia , Sincalida/farmacologia
10.
J Headache Pain ; 20(1): 87, 2019 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-31375062

RESUMO

BACKGROUND: Migraine is a debilitating neurological disorder involving abnormal trigeminovascular activation and sensitization. However, the underlying cellular and molecular mechanisms remain unclear. METHODS: A rat model of conscious migraine was established through the electrical stimulation (ES) of the dural mater surrounding the superior sagittal sinus. Using patch clamp recording, immunofluorescent labelling, enzyme-linked immunosorbent assays and western blot analysis, we studied the effects of ES on sensory neuronal excitability and elucidated the underlying mechanisms mediated by voltage-gated ion channels. RESULTS: The calcitonin gene-related peptide (CGRP) level in the jugular vein blood and the number of CGRP-positive neurons in the trigeminal ganglia (TGs) were significantly increased in rats with ES-induced migraine. The application of ES increased actional potential firing in both small-sized IB4-negative (IB4-) and IB4+ TG neurons. No significant changes in voltage-gated Na+ currents were observed in the ES-treated groups. ES robustly suppressed the transient outward K+ current (IA) in both types of TG neurons, while the delayed rectifier K+ current remained unchanged. Immunoblot analysis revealed that the protein expression of Kv4.3 was significantly decreased in the ES-treated groups, while Kv1.4 remained unaffected. Interestingly, ES increased the P/Q-type and T-type Ca2+ currents in small-sized IB4- TG neurons, while there were no significant changes in the IB4+ subpopulation of neurons. CONCLUSION: These results suggest that ES decreases the IA in small-sized TG neurons and increases P/Q- and T-type Ca2+ currents in the IB4- subpopulation of TG neurons, which might contribute to neuronal hyperexcitability in a rat model of ES-induced migraine.


Assuntos
Estimulação Elétrica/métodos , Seio Sagital Superior/metabolismo , Gânglio Trigeminal/metabolismo , Potenciais de Ação , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Masculino , Neurônios Aferentes/fisiologia , Ratos , Ratos Sprague-Dawley , Seio Sagital Superior/citologia , Gânglio Trigeminal/citologia
11.
J Pineal Res ; 64(4): e12476, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29437250

RESUMO

Recent studies implicate melatonin in the antinociceptive activity of sensory neurons. However, the underlying mechanisms are still largely unknown. Here, we identify a critical role of melatonin in functionally regulating Cav3.2 T-type Ca2+ channels (T-type channel) in trigeminal ganglion (TG) neurons. Melatonin inhibited T-type channels in small TG neurons via the melatonin receptor 2 (MT2 receptor) and a pertussis toxin-sensitive G-protein pathway. Immunoprecipitation analyses revealed that the intracellular subunit of the MT2 receptor coprecipitated with Gαo . Both shRNA-mediated knockdown of Gαo and intracellular application of QEHA peptide abolished the inhibitory effects of melatonin. Protein kinase C (PKC) antagonists abolished the melatonin-induced T-type channel response, whereas inhibition of conventional PKC isoforms elicited no effect. Furthermore, application of melatonin increased membrane abundance of PKC-eta (PKCη ) while antagonism of PKCη or shRNA targeting PKCη prevented the melatonin-mediated effects. In a heterologous expression system, activation of MT2 receptor strongly inhibited Cav3.2 T-type channel currents but had no effect on Cav3.1 and Cav3.3 current amplitudes. The selective Cav3.2 response was PKCη dependent and was accompanied by a negative shift in the steady-state inactivation curve. Furthermore, melatonin decreased the action potential firing rate of small TG neurons and attenuated the mechanical hypersensitivity in a mouse model of complete Freund's adjuvant-induced inflammatory pain. These actions were inhibited by T-type channel blockade. Together, our results demonstrated that melatonin inhibits Cav3.2 T-type channel activity through the MT2 receptor coupled to novel Gßγ -mediated PKCη signaling, subsequently decreasing the membrane excitability of TG neurons and pain hypersensitivity in mice.


Assuntos
Canais de Cálcio Tipo T/efeitos dos fármacos , Melatonina/farmacologia , Proteína Quinase C/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Animais , Canais de Cálcio Tipo T/metabolismo , Hiperalgesia/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Camundongos Endogâmicos ICR , Receptor MT2 de Melatonina/metabolismo , Células Receptoras Sensoriais/metabolismo , Transdução de Sinais/efeitos dos fármacos , Gânglio Trigeminal/efeitos dos fármacos , Gânglio Trigeminal/metabolismo
12.
Carcinogenesis ; 38(2): 168-176, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28034876

RESUMO

Polymorphisms in pre-miRNAs may affect its expression, then have effect on its target mRNAs and be associated with cancer susceptibility. In this study, we evaluated the association of an indel polymorphism rs57408770 in pre-miR-3131 with hepatocellular carcinoma (HCC) susceptibility in a Chinese population. The contribution of rs57408770 to HCC risk was investigated in two independent case-control sets (1051 HCC and 1058 controls). Logistic regression analysis showed that the insertion allele of rs57408770 was significantly associated with an increased risk for HCC occurrence in both case-control studies. Moreover, the results of genotype-phenotype correlation analysis from both in vivo and in vitro experiments showed that the insertion allele was significantly correlated with higher expression of mature miR-3131 comparing with the deletion allele. The RNA-Binding Protein Immunoprecipitation assay results indicated that rs57408770 could affect the expression level of mature miR-3131 probably through disturbing the binding of splicing factor SRp20 with pre-miR-3131. Furthermore, overexpression of miR-3131 displayed a proliferation promoting and anti-apoptosis effect on HCC cell lines, suggesting that miR-3131 may act as a proto-oncogene in HCC. Finally, human genome-wide gene expression profile assay was used to screen the targets of miR-3131. The overexpressed miR-3131 could lead to a significant decrease of DTHD1 and XAF1 mRNA level. Taken together, our findings provided evidence that rs57408770 may play a functional role in the carcinogenesis of HCC by affecting SRp20 binding with pre-miR-3131 and affecting the expression of mature miR-3131, subsequently affecting the expression of DTHD1 and XAF1, thus confers risk for HCC.


Assuntos
Carcinoma Hepatocelular/genética , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neoplasias Hepáticas/genética , MicroRNAs/genética , Proteínas de Neoplasias/genética , Proteínas Adaptadoras de Transdução de Sinal , Idoso , Proteínas Reguladoras de Apoptose , Carcinoma Hepatocelular/patologia , China , Feminino , Regulação Neoplásica da Expressão Gênica , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Mutação INDEL/genética , Neoplasias Hepáticas/patologia , Masculino , MicroRNAs/biossíntese , Pessoa de Meia-Idade , Proto-Oncogene Mas , Fatores de Risco
13.
Opt Express ; 25(16): 19479-19486, 2017 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-29041141

RESUMO

We propose and experimentally demonstrate an ultra-compact silicon photonic crystal nanobeam (PCN) cavity with an energy-efficient graphene micro-heater. Owing to the PCN cavity with an ultra-small optical mode volume of 0.145 µm3, the light-matter interaction is greatly enhanced and the thermo-optic (TO) tuning efficiency is increased. The TO tuning efficiency is measured to be as high as 1.5 nm/mW, which can be further increased to 3.75 nm/mW based on numerical simulations with an optimized structure. The time constants with a rise time constant of τrise = 1.11 µs and a fall time constant of τfall = 1.47 µs are obtained in the experiment.

14.
J Biol Chem ; 290(13): 8644-55, 2015 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-25678708

RESUMO

Recent studies have demonstrated that urotensin-II (U-II) plays important roles in cardiovascular actions including cardiac positive inotropic effects and increasing cardiac output. However, the mechanisms underlying these effects of U-II in cardiomyocytes still remain unknown. We show by electrophysiological studies that U-II dose-dependently potentiates L-type Ca(2+) currents (ICa,L) in adult rat ventricular myocytes. This effect was U-II receptor (U-IIR)-dependent and was associated with a depolarizing shift in the voltage dependence of inactivation. Intracellular application of guanosine-5'-O-(2-thiodiphosphate) and pertussis toxin pretreatment both abolished the stimulatory effects of U-II. Dialysis of cells with the QEHA peptide, but not scrambled peptide SKEE, blocked the U-II-induced response. The phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin as well as the class I PI3K antagonist CH132799 blocked the U-II-induced ICa,L response. Protein kinase C antagonists calphostin C and chelerythrine chloride as well as dialysis of cells with 1,2bis(2aminophenoxy)ethaneN,N,N',N'-tetraacetic acid abolished the U-II-induced responses, whereas PKCα inhibition or PKA blockade had no effect. Exposure of ventricular myocytes to U-II markedly increased membrane PKCß1 expression, whereas inhibition of PKCß1 pharmacologically or by shRNA targeting abolished the U-II-induced ICa,L response. Functionally, we observed a significant increase in the amplitude of sarcomere shortening induced by U-II; blockade of U-IIR as well as PKCß inhibition abolished this effect, whereas Bay K8644 mimicked the U-II response. Taken together, our results indicate that U-II potentiates ICa,L through the ßγ subunits of Gi/o-protein and downstream activation of the class I PI3K-dependent PKCß1 isoform. This occurred via the activation of U-IIR and contributes to the positive inotropic effect on cardiomyocytes.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Proteína Quinase C beta/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Sinalização do Cálcio , Ventrículos do Coração/citologia , Isoenzimas/metabolismo , Masculino , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Fosfatidilinositol 3-Quinases , Subunidades Proteicas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Sarcômeros/fisiologia , Urotensinas/fisiologia
15.
Sheng Li Xue Bao ; 68(5): 575-584, 2016 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-27778022

RESUMO

Hydrogen sulfide (H2S) contributes to visceral hyperalgesia in primary sensory neurons, but its role in central nervous system remains largely unknown. This study was to investigate the roles and underlying mechanisms of H2S and its endogenous synthesis enzymes in the arcuate nucleus (ARC) in rat pancreatic hyperalgesia. Chronic pancreatitis (CP) was induced in male adult Sprague-Dawley rats by intra-pancreatic ductal injection of trinitrobenzene sulfonic acid (TNBS). Abdominal hyperalgesia was assessed by referred somatic behaviors to mechanical stimulation of rat abdomen. Western blot analysis was performed to detect protein expression in the ARC. CP markedly upregulated cystathionine ß-synthetase (CBS) expression but did not alter cystathionine-γ-lyase level in the ARC at 4 weeks after TNBS injection. Although the expression of total GluN2B was not altered, CP greatly enhanced the phosphorylation level of GluN2B in the ARC when compared with age- and sex-matched control rats. CP also significantly increased expression of protein kinase Cγ (PKCγ) in the ARC. Arcuate microinjection of O-(Carboxymethyl) hydroxylamine hemihydrochloride (AOAA, an inhibitor of CBS) significantly attenuated abdominal pain in CP rats in a dose-dependent manner and reversed the CP-induced upregulation of p-GluN2B and PKCγ in the ARC. Furthermore, the GluN2B inhibitor or specific PKC inhibitor chelerythrine significantly attenuated abdominal hyperalgesia in CP rats. The p-GluN2B expression was also suppressed by PKC inhibitor. Taken together, our results suggest that the upregulation of CBS in the ARC leads to an activation of GluN2B via PKCγ, which may play an important role in generation of pain hypersensitivity of CP.


Assuntos
Núcleo Arqueado do Hipotálamo , Pancreatite Crônica , Doença Aguda , Animais , Cistationina beta-Sintase , Hiperalgesia , Masculino , Dor , Fosforilação , Proteína Quinase C , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato , Regulação para Cima
16.
Mol Pain ; 11: 15, 2015 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-25885215

RESUMO

Lumbar disc herniation (LDH) is a major cause of discogenic low back pain and sciatica, but the underlying mechanisms remain largely unknown. Hydrogen sulfide (H2S) is becoming recognized for its involvement in a wide variety of processes including inflammation and nociception. The present study was designed to investigate the roles of the H2S signaling pathway in the regulation of expression and function of purinergic receptors (P2XRs) in dorsal root ganglion (DRG) neurons from rats with LDH. LDH was induced by implantation of autologous nucleus pulposus (NP), harvested from rat tail, in lumbar 5 and 6 spinal nerve roots. Implantation of autologous NP induced persistent pain hypersensitivity, which was partially reversed by an intrathecal injection of A317491, a potent inhibitor of P2X3Rs and P2X2/3Rs. The NP induced persistent pain hypersensitivity was associated with the increased expression of P2X3Rs, but not P2X1Rs and P2X2Rs, receptors in L5-6 DRGs. NP implantation also produced a 2-fold increase in ATP-induced intracellular calcium signals in DRG neurons when compared to those of controls (P < 0.05). Interestingly, NP implantation significantly enhanced expression of the endogenous hydrogen sulfide producing enzyme, cystathionine-ß-synthetase (CBS). Systematic administration of O-(Carboxymethyl) hydroxylamine hemihydrochloride (AOAA), an inhibitor of CBS, suppressed the upregulation of P2X3R expression and the potentiation of ATP-induced intracellular calcium signals in DRG neurons (P < 0.05). Intrathecal injection of AOAA markedly attenuated NP induced- persistent pain hypersensitivity. Our results suggest that sensitization of P2X3Rs, which is likely mediated by CBS-H2S signaling in primary sensory neurons, contributes to discogenic pain. Targeting CBS/H2S-P2X3R signaling may represent a potential treatment for neuropathic pain caused by LDH.


Assuntos
Cistationina beta-Sintase/metabolismo , Hipersensibilidade/metabolismo , Deslocamento do Disco Intervertebral/metabolismo , Neuralgia/metabolismo , Receptores Purinérgicos P2X3/metabolismo , Animais , Modelos Animais de Doenças , Gânglios Espinais/metabolismo , Hiperalgesia/fisiopatologia , Masculino , Ratos Sprague-Dawley , Raízes Nervosas Espinhais/metabolismo
17.
Am J Physiol Gastrointest Liver Physiol ; 308(8): G710-9, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25634810

RESUMO

The mechanism of pain in chronic pancreatitis (CP) is poorly understood. The aim of this study was designed to investigate roles of norepinephrine (NE) and P2X receptor (P2XR) signaling pathway in the pathogenesis of hyperalgesia in a rat model of CP. CP was induced in male adult rats by intraductal injection of trinitrobenzene sulfonic acid (TNBS). Mechanical hyperalgesia was assessed by referred somatic behaviors to mechanical stimulation of rat abdomen. P2XR-mediated responses of pancreatic dorsal root ganglion (DRG) neurons were measured utilizing calcium imaging and whole cell patch-clamp-recording techniques. Western blot analysis and immunofluorescence were performed to examine protein expression. TNBS injection produced a significant upregulation of P2X3R expression and an increase in ATP-evoked responses of pancreatic DRG neurons. The sensitization of P2X3Rs was reversed by administration of ß-adrenergic receptor antagonist propranolol. Incubation of DRG neurons with NE significantly enhanced ATP-induced intracellular calcium signals, which were abolished by propranolol, and partially blocked by protein kinase A inhibitor H-89. Interestingly, TNBS injection led to a significant elevation of NE concentration in DRGs and the pancreas, an upregulation of ß2-adrenergic receptor expression in DRGs, and amplification of the NE-induced potentiation of ATP responses. Importantly, pancreatic hyperalgesia was markedly attenuated by administration of purinergic receptor antagonist suramin or A317491 or ß2-adrenergic receptor antagonist butoxamine. Sensitization of P2X3Rs, which was likely mediated by adrenergic signaling in primary sensory neurons, contributes to pancreatic pain, thus identifying a potential target for treating pancreatic pain caused by inflammation.


Assuntos
Sinalização do Cálcio , Gânglios Espinais/metabolismo , Hiperalgesia/metabolismo , Norepinefrina/metabolismo , Limiar da Dor , Pâncreas/inervação , Pancreatite Crônica/metabolismo , Receptores Purinérgicos P2X3/metabolismo , Células Receptoras Sensoriais/metabolismo , Trifosfato de Adenosina/metabolismo , Antagonistas de Receptores Adrenérgicos beta 2/farmacologia , Animais , Comportamento Animal , Sinalização do Cálcio/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Modelos Animais de Doenças , Gânglios Espinais/efeitos dos fármacos , Hiperalgesia/induzido quimicamente , Hiperalgesia/fisiopatologia , Hiperalgesia/prevenção & controle , Masculino , Potenciais da Membrana , Atividade Motora , Limiar da Dor/efeitos dos fármacos , Pancreatite Crônica/induzido quimicamente , Pancreatite Crônica/fisiopatologia , Inibidores de Proteínas Quinases/farmacologia , Antagonistas do Receptor Purinérgico P2X/farmacologia , Ratos Sprague-Dawley , Receptor Cross-Talk , Receptores Adrenérgicos beta 2/efeitos dos fármacos , Receptores Adrenérgicos beta 2/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Fatores de Tempo , Ácido Trinitrobenzenossulfônico
18.
Tumour Biol ; 36(7): 5467-72, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25687183

RESUMO

It has been reported that miR-21 is upregulated in hepatocellular carcinoma (HCC), and overexpressed miR-21 plays a key role in promoting cell cycle progression, reducing cell death and favoring angiogenesis and invasion. Overexpression of hepatocellular carcinoma, downregulated 1 (HEPN1) exhibits an antiproliferative effect on HepG2 cells, suggesting that silencing of HEPN1 may contribute to carcinogenesis of hepatocytes. In silico analysis revealed that HEPN1 may be a potential target of miR-21. Using quantitative reverse transcription PCR and Western blot, we found that HEPN1 was strikingly downregulated in both mRNA (fold change was 33.5, P < 0.0001) and protein levels in human HCC tumor tissues, in comparison with the adjacent non-tumor tissues. More importantly, the expression level of HEPN1 was inversely correlated with the expression of miR-21 in HCC (R (2) = 0.442, P < 0.0001). The combination between the 3' untranslated region (UTR) of HEPN1 with miR-21 was experimentally verified by a miRNA luciferase reporter approach. The suppressed cell proliferation upon stimulation of miR-21 inhibitor could be partially abolished by knocking down HEPN1, so inhibition of miR-21 expression in HCC cells profoundly suppressed cell proliferation partially by upregulating HEPN1 expression. Taken together, the current study suggested an underlying mechanism that miR-21 directly target HEPN1 and inhibit its expression during the carcinogenesis of HCC. HEPN1 may thus be a candidate as a therapeutic target for patients with HCC.


Assuntos
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , MicroRNAs/genética , Proteínas/metabolismo , Apoptose/genética , Carcinoma Hepatocelular/patologia , Ciclo Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Humanos , Neoplasias Hepáticas/patologia , MicroRNAs/metabolismo , Proteínas/genética , RNA Mensageiro/biossíntese
19.
J Pineal Res ; 58(3): 321-34, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25707622

RESUMO

Although melatonin receptors are widely expressed in the mammalian central nervous system and peripheral tissues, there are limited data regarding the functions of melatonin in cerebellar Purkinje cells. Here, we identified a novel functional role of melatonin in modulating P-type Ca(2+) channels and action-potential firing in rat Purkinje neurons. Melatonin at 0.1 µm reversibly decreased peak currents (I(Ba)) by 32.9%. This effect was melatonin receptor 1 (MT(R1)) dependent and was associated with a hyperpolarizing shift in the voltage dependence of inactivation. Pertussis toxin pretreatment, intracellular application of QEHA peptide, and a selective antibody raised against the Gß subunit prevented the inhibitory effects of melatonin. Pretreatment with phosphatidylinositol 3-kinase (PI3K) inhibitors abolished the melatonin-induced decrease in I(Ba). Surprisingly, melatonin responses were not regulated by Akt, a common downstream target of PI3K. Melatonin treatment significantly increased protein kinase C (PKC) activity 2.1-fold. Antagonists of PKC, but not of protein kinase A, abolished the melatonin-induced decrease in I(Ba). Melatonin application increased the membrane abundance of PKCδ, and PKCδ inhibition (either pharmacologically or genetically) abolished the melatonin-induced IBa response. Functionally, melatonin increased spontaneous action-potential firing by 53.0%; knockdown of MT(R1) and blockade of P-type channels abolished this effect. Thus, our results suggest that melatonin inhibits P-type channels through MT(R1) activation, which is coupled sequentially to the ßγ subunits of G(i/o)-protein and to downstream PI3K-dependent PKCδ signaling. This likely contributes to its physiological functions, including spontaneous firing of cerebellar Purkinje neurons.


Assuntos
Canais de Cálcio Tipo P/efeitos dos fármacos , Melatonina/farmacologia , Fosfatidilinositol 3-Quinase/metabolismo , Proteína Quinase C-delta/metabolismo , Células de Purkinje/efeitos dos fármacos , Animais , Canais de Cálcio Tipo P/metabolismo , Células de Purkinje/metabolismo , Ratos , Ratos Wistar
20.
J Neurosci ; 33(21): 9028-38, 2013 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-23699514

RESUMO

Patients with long-standing diabetes frequently demonstrate gastric hypersensitivity with an unknown mechanism. The present study was designed to investigate roles for nuclear factor-κB (NF-κB) and the endogenous H2S-producing enzyme cystathionine-ß-synthetase (CBS) signaling pathways by examining cbs gene methylation status in adult rats with diabetes. Intraperitoneal injection of streptozotocin (STZ) produced gastric hypersensitivity in female rats in response to gastric balloon distention. Treatment with the CBS inhibitor aminooxyacetic acid significantly attenuated STZ-induced gastric hypersensitivity in a dose-dependent fashion. Aminooxyacetic acid treatment also reversed hyperexcitability of gastric-specific dorsal root ganglion (DRG) neurons labeled by the dye DiI in diabetic rats. Conversely, the H2S donor NaHS enhanced neuronal excitability of gastric DRG neurons. Expression of CBS and p65 were markedly enhanced in gastric DRGs in diabetic rats. Blockade of NF-κB signaling using pyrrolidine dithiocarbamate reversed the upregulation of CBS expression. Interestingly, STZ treatment led to a significant demethylation of CpG islands in the cbs gene promoter region, as determined by methylation-specific PCR and bisulfite sequencing. STZ treatment also remarkably downregulated the expression of DNA methyltransferase 3a and 3b. More importantly, STZ treatment significantly enhanced the ability of cbs to bind DNA at the p65 consensus site, as shown by chromatin immunoprecipitation assays. Our findings suggest that upregulation of cbs expression is attributed to cbs promoter DNA demethylation and p65 activation and that the enhanced interaction of the cbs gene and p65 contributes to gastric hypersensitivity in diabetes. This finding may guide the development and evaluation of new treatment modalities for patients with diabetic gastric hypersensitivity.


Assuntos
Cistationina beta-Sintase/metabolismo , Diabetes Mellitus Experimental/complicações , Hipersensibilidade , NF-kappa B/metabolismo , Gastropatias/etiologia , Aminoácidos , Análise de Variância , Animais , Área Sob a Curva , Estudos de Casos e Controles , Imunoprecipitação da Cromatina , Ilhas de CpG/efeitos dos fármacos , Ilhas de CpG/fisiologia , Cistationina beta-Sintase/genética , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Metiltransferase 3A , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Eletromiografia , Inibidores Enzimáticos/farmacologia , Feminino , Gânglios Espinais/patologia , Hipersensibilidade/tratamento farmacológico , Hipersensibilidade/etiologia , Potenciais da Membrana/efeitos dos fármacos , Metilação/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Ácido Oxâmico/uso terapêutico , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/fisiologia , Gastropatias/tratamento farmacológico , Sulfitos/farmacologia , Regulação para Cima/efeitos dos fármacos , DNA Metiltransferase 3B
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa