Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Cell Physiol ; 234(3): 2756-2764, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30145789

RESUMO

The upregulation of nociceptive ion channels expressed in dorsal root ganglia (DRG) contributes to the development and retaining of diabetic pain symptoms. The flavonoid quercetin (3,3',4',5,7-pentahydroxyflavone) is a component extracted from various fruits and vegetables and exerts anti-inflammatory, analgesic, anticarcinogenic, antiulcer, and antihypertensive effects. However, the exact mechanism underlying quercetin's analgesic action remains poorly understood. The aim of this study was to investigate the effects of quercetin on diabetic neuropathic pain related to the P2X4 receptor in the DRG of type 2 diabetic rat model. Our data showed that both mechanical withdrawal threshold and thermal withdrawal latency in diabetic rats treated with quercetin were higher compared with those in untreated diabetic rats. The expression levels of P2X4 messenger RNA and protein in the DRG of diabetic rats were increased compared with the control rats, while quercetin treatment significantly inhibited such enhanced P2X4 expression in diabetic rats. The satellite glial cells (SGCs) enwrap the neuronal soma in the DRG. Quercetin treatment also lowered the elevated coexpression of P2X4 and glial fibrillary acidic protein (a marker of SGCs) and decreased the upregulation of phosphorylated p38 mitogen-activated protein kinase (p38MAPK) in the DRG of diabetic rats. Quercetin significantly reduced the P2X4 agonist adenosine triphosphate-activated currents in HEK293 cells transfected with P2X4 receptors. Thus, our data demonstrate that quercetin may decrease the upregulation of the P2X4 receptor in DRG SGCs, and consequently inhibit P2X4 receptor-mediated p38MAPK activation to relieve the mechanical and thermal hyperalgesia in diabetic rats.


Assuntos
Neuropatias Diabéticas/tratamento farmacológico , Gânglios Espinais/efeitos dos fármacos , Quercetina/farmacologia , Receptores Purinérgicos P2X4/efeitos dos fármacos , Animais , Diabetes Mellitus Experimental/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Neuralgia/tratamento farmacológico , Neuroglia/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos Sprague-Dawley , Receptores Purinérgicos P2X4/metabolismo
2.
Int J Neurosci ; 129(8): 784-793, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30621504

RESUMO

Aim: This study investigated whether the neuronal P2X3 receptor in rat dorsal root ganglia (DRG) mediated the effects of hesperidin on neuropathic pain. Materials and methods: The chronic constriction injury (CCI) model was used as a model of neuropathic pain. The mechanical withdrawal threshold (MWT) and thermal withdrawal latency (TWL) were measured. The mRNA and protein expression levels were assayed by real-time RT-PCR and Western blotting. Results: The results showed that mechanical and thermal hyperalgesia in the CCI rats were increased as compared to those in the sham group. The expression levels of P2X3 mRNA and protein in CCI rats were higher than those in the sham group. Dual-labelling immunofluorescence showed that the elevated P2X3 receptor was co-expressed with the neuronal marker NeuN in the DRG of CCI rats. Hesperidin treatment decreased both the mechanical and thermal hyperalgesia, and upregulated P2X3 expression in the CCI rats. Hesperidin treatment also reduced the ERK1/2 phosphorylation in the DRG of CCI rats. Moreover, hesperidin inhibited the P2X3 agonist ATP-induced currents in HEK293 cells transfected with the P2X3 plasmid. Therefore, hesperidin treatment could reverse the elevated expression of neuronal P2X3 receptor and reduce the activation of ERK1/2 in the DRG of CCI rats. Conclusions: Our findings suggested that hesperidin inhibited the nociceptive transmission mediated by the P2X3 receptor in neurons of DRG, and thus, relieved the mechanical and thermal hyperalgesia in CCI rats.


Assuntos
Gânglios Espinais , Hesperidina/farmacologia , Hiperalgesia , Neuralgia , Nociceptividade/efeitos dos fármacos , Receptores Purinérgicos P2X3 , Animais , Modelos Animais de Doenças , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Células HEK293 , Humanos , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Masculino , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2X3/efeitos dos fármacos , Receptores Purinérgicos P2X3/metabolismo
3.
J Cell Physiol ; 233(4): 3375-3383, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28949406

RESUMO

Superior cervical ganglia (SCG) innervate the myocardium and participate in sympathoexcitatory transmission. P2Y12 receptor is expressed in satellite glial cells (SGCs). This study seeks to clarify whether the P2Y12 receptor is involved in the sympathoexcitation reflex after myocardial ischemia (MI). MI model was induced by occlusion of the left coronary artery. P2Y12 were assayed by real time PCR and Western blotting. Our results showed that expression levels of P2Y12 mRNA and protein were significantly higher in the MI group than in the sham group. Administration of P2Y12 short hairpin RNA (shRNA) caused downregulation of the P2Y12 receptor in the SCG. In MI rats plus P2Y12 shRNA treatment group, the abnormal changes in diastolic blood pressure (DBP), systolic blood pressure (SBP), heart rate (HR), electrocardiograms (ECGs), and cardiac tissue structures were alleviated. When the treatment of P2Y12 shRNA in MI rats, upregulated co-expression values of P2Y12 and glial fibrillary acidic protein (GFAP), the upregulation of tumor necrosis factor α (TNF-α) and phosphorylated P38 mitogen activated protein kinase (p-P38 MAPK) in the SCG were decreased. Downregulation of the P2Y12 receptor in the SCG after MI may improve cardiac function by alleviating the sympathoexcitatory reflex.


Assuntos
Isquemia Miocárdica/metabolismo , Miocárdio/metabolismo , Receptores Purinérgicos P2Y12/metabolismo , Reflexo/fisiologia , Animais , Pressão Sanguínea/fisiologia , Regulação para Baixo/fisiologia , Coração/fisiologia , Frequência Cardíaca/fisiologia , Isquemia Miocárdica/patologia , Ratos Sprague-Dawley
4.
J Cell Physiol ; 233(12): 9620-9628, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29943819

RESUMO

Diabetic neuropathic pain is a common complication of type 2 diabetes mellitus (DM). Activation of satellite glial cells (SGCs) in the dorsal root ganglia (DRG) plays a crucial role in neuropathic pain through the release of proinflammatory cytokines. The P2Y12 receptor is expressed in SGCs of the DRG. In this study, our aim was to investigate the role of the P2Y12 receptor on the pathological changes in diabetic neuropathic pain. The present study showed that diabetic neuropathic pain increased mechanical and thermal hyperalgesia in type 2 DM model rats. The results showed that the expression levels of P2Y12 messenger RNA (mRNA) and protein in DRG SGCs were increased in DM model rats compared with control rats. Glial fibrillary acidic protein (GFAP) and interleukin-1ß (IL-1ß) expression levels in the DRG were increased in DM rats. Upregulation of GFAP is a marker of SGC activation. Targeting the P2Y12 receptor by short hairpin RNA (shRNA) decreased the upregulated expression of P2Y12 mRNA and protein, coexpression of P2Y12 and GFAP, the expression of GFAP, IL-1ß, and tumor necrosis factor-receptor 1 in the DRG of DM rats, and relieved mechanical and thermal hyperalgesia in DM rats. After treatment with the P2Y12 receptor shRNA, the enhancing integrated OPTICAL density (IOD) ratios of p-P38 MAPK to P38 mitogen activated protein kinase (MAPK) in the DM rats treated with P2Y12 shRNA were significantly lower than that in the untreated DM rats. Therefore, P2Y12 shRNA treatment decreased SGC activation to relieve mechanical and thermal hyperalgesia in DM rats.


Assuntos
Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 2/terapia , Neuropatias Diabéticas/terapia , Neuralgia/terapia , Neuroglia/metabolismo , RNA Interferente Pequeno/metabolismo , Receptores Purinérgicos P2Y12/metabolismo , Animais , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/patologia , Neuropatias Diabéticas/complicações , Neuropatias Diabéticas/patologia , Ativação Enzimática , Gânglios Espinais/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Interleucina-1beta/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Neuralgia/complicações , Neuralgia/patologia , Ratos Sprague-Dawley , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Regulação para Cima/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
Purinergic Signal ; 14(1): 47-58, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29159762

RESUMO

The direct neurotoxicity of HIV and neurotoxicity of combination antiretroviral therapy medications both contribute to the development of neuropathic pain. Activation of satellite glial cells (SGCs) in the dorsal root ganglia (DRG) plays a crucial role in mechanical and thermal hyperalgesia. The P2Y12 receptor expressed in SGCs of the DRG is involved in pain transmission. In this study, we explored the role of the P2Y12 receptor in neuropathic pain induced by HIV envelope glycoprotein 120 (gp120) combined with ddC (2',3'-dideoxycytidine). A rat model of gp120+ddC-induced neuropathic pain was used. Peripheral nerve exposure to HIV-gp120+ddC increased mechanical and thermal hyperalgesia in gp120+ddC-treated model rats. The gp120+ddC treatment increased expression of P2Y12 receptor mRNA and protein in DRG SGCs. In primary cultured DRG SGCs treated with gp120+ddC, the levels of [Ca2+]i activated by the P2Y12 receptor agonist 2-(Methylthio) adenosine 5'-diphosphate trisodium salt (2-MeSADP) were significantly increased. P2Y12 receptor shRNA treatment inhibited 2-MeSADP-induced [Ca2+]i in primary cultured DRG SGCs treated with gp120+ddC. Intrathecal treatment with a shRNA against P2Y12 receptor in DRG SGCs reduced the release of pro-inflammatory cytokines, decreased phosphorylation of p38 MAPK in the DRG of gp120+ddC-treated rats. Thus, downregulating the P2Y12 receptor relieved mechanical and thermal hyperalgesia in gp120+ddC-treated rats.


Assuntos
Proteína gp120 do Envelope de HIV , Neuralgia/metabolismo , Neuroglia/metabolismo , Receptores Purinérgicos P2/metabolismo , Zalcitabina/toxicidade , Animais , Fármacos Anti-HIV/toxicidade , Gânglios Espinais/metabolismo , Infecções por HIV/complicações , Hiperalgesia/metabolismo , Hiperalgesia/virologia , Masculino , Neuralgia/etiologia , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2Y12 , Regulação para Cima
6.
Mol Pain ; 13: 1744806917707667, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28554250

RESUMO

Background Chronic pain is a common symptom in human immunodeficiency virus (HIV)-1 infection/acquired immunodeficiency syndrome patients. The literature shows that the HIV envelope glycoprotein 120 (gp120) can directly cause hyperalgesia by stimulating primary sensory afferent nerves. The P2X7 receptor in the dorsal root ganglia (DRG) is closely related to neuropathic and inflammatory pain. In this study, we aimed to explore the effect of resveratrol (RES) on gp120-induced neuropathic pain that is mediated by the P2X7 receptor in the rat DRG. Results Mechanical hyperalgesia in rats treated with gp120 was increased compared with that in the sham group. The P2X7 expression levels in rats treated with gp120 were higher than those in the sham group. Co-localization of the P2X7 receptor and glial fibrillary acidic protein (GFAP, a marker of satellite glial cells [SGCs]) in the DRG SGCs of the gp120 group exhibited more intense staining than that of the sham group. RES decreased the mechanical hyperalgesia and P2X7 expression levels in gp120 treatment rats. Co-localization of the P2X7 receptor and GFAP in the gp120+ RES group was significantly decreased compared to the gp120 group. RES decreased the IL-1ß and TNF-α receptor (R) expression levels and ERK1/2 phosphorylation levels as well as increased IL-10 expression in the DRG of gp120-treated rats. Whole cell clamping demonstrated that RES significantly inhibited adenosine triphosphate-activated currents in HEK293 cells that were transfected with the P2X7 plasmid. Conclusions RES relieved mechanical hyperalgesia in gp120-treated rats by inhibiting the P2X7 receptor.


Assuntos
Proteína gp120 do Envelope de HIV/farmacologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Neuralgia/induzido quimicamente , Neuralgia/tratamento farmacológico , Receptores Purinérgicos P2X7/metabolismo , Estilbenos/uso terapêutico , Animais , Western Blotting , Eletrofisiologia , Células HEK293 , Humanos , Interleucina-10 , Masculino , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Resveratrol
7.
Purinergic Signal ; 13(4): 559-568, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28840511

RESUMO

Diabetic peripheral neuropathy (DPN) is the most common complication of diabetes mellitus (DM). More than 90% of all cases of DM belong to type 2 diabetes mellitus (T2DM). Emodin is the main active component of Radix et rhizoma rhei and has anti-bacterial, anti-viral, anti-ulcerogenic, anti-inflammatory, and anti-cancer effects. Nanoparticle encapsulation of drugs is beneficial for drug targeting and bioavailability as well as for lowering drug toxicity side effects. The aim of this study was to investigate the effects of nanoparticle-encapsulated emodin (nano emodin) on diabetic neuropathic pain (DNP) mediated by the Purin 2X3 (P2X3) receptor in the dorsal root ganglia (DRG). Mechanical withdrawal threshold (MWT) and thermal withdrawal latency (TWL) values in T2DM rats were lower than those of control rats. MWT and TWL in T2DM rats treated with nano emodin were higher compared with those in T2DM rats. Expression levels of P2X3 protein and messenger RNA (mRNA) in the DRG of T2DM rats were higher than those of controls, while levels in T2DM rats treated with nano emodin were significantly lower than those of the T2DM rats. Phosphorylation and activation of ERK1/2 in the T2DM DRG were decreased by nano emodin treatment. Nano emodin significantly inhibited currents activated by the P2X3 agonist α,ß-meATP in HEK293 cells transfected with the P2X3 receptor. Therefore, nano emodin treatment may relieve DNP by decreasing excitatory transmission mediated by the DRG P2X3 receptor in T2DM rats.


Assuntos
Neuropatias Diabéticas/metabolismo , Emodina/administração & dosagem , Gânglios Espinais/efeitos dos fármacos , Nanoconjugados , Receptores Purinérgicos P2X3/metabolismo , Animais , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Gânglios Espinais/metabolismo , Células HEK293 , Humanos , Masculino , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
9.
Neuron ; 112(10): 1676-1693.e12, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38513667

RESUMO

Neuronal loss is the central issue in Alzheimer's disease (AD), yet no treatment developed so far can halt AD-associated neurodegeneration. Here, we developed a monoclonal antibody (mAb2A7) against 217 site-phosphorylated human tau (p-tau217) and observed that p-tau217 levels positively correlated with brain atrophy and cognitive impairment in AD patients. Intranasal administration efficiently delivered mAb2A7 into male PS19 tauopathic mouse brain with target engagement and reduced tau pathology/aggregation with little effect on total soluble tau. Further, mAb2A7 treatment blocked apoptosis-associated neuronal loss and brain atrophy, reversed cognitive deficits, and improved motor function in male tauopathic mice. Proteomic analysis revealed that mAb2A7 treatment reversed alterations mainly in proteins associated with synaptic functions observed in murine tauopathy and AD brain. An antibody (13G4) targeting total tau also attenuated tau-associated pathology and neurodegeneration but impaired the motor function of male tauopathic mice. These results implicate p-tau217 as a potential therapeutic target for AD-associated neurodegeneration.


Assuntos
Doença de Alzheimer , Anticorpos Monoclonais , Tauopatias , Proteínas tau , Idoso , Idoso de 80 Anos ou mais , Animais , Feminino , Humanos , Masculino , Camundongos , Doença de Alzheimer/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Doença de Alzheimer/tratamento farmacológico , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/administração & dosagem , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/efeitos dos fármacos , Modelos Animais de Doenças , Imunoterapia/métodos , Camundongos Transgênicos , Degeneração Neural/patologia , Degeneração Neural/tratamento farmacológico , Fosforilação , Proteínas tau/metabolismo , Tauopatias/tratamento farmacológico
10.
Mol Ther Methods Clin Dev ; 27: 246-258, 2022 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-36320413

RESUMO

Fragile X syndrome (FXS) is caused by the loss of the fragile X messenger ribonucleoprotein 1 (FMRP) encoded by the FMR1 gene. Gene therapy using adeno-associated virus (AAV) to restore FMRP expression is a promising therapeutic strategy. However, so far AAV gene therapy tests for FXS only utilized rodent FMRPs driven by promoters other than the human FMR1 promoter. Restoration of human FMRP in appropriate cell types and at physiological levels, preferably driven by the human FMR1 promoter, would be more suitable for its clinical use. Herein, we generated two human FMR1 promoter subdomains that effectively drive gene expression. When AAVs expressing two different human FMRP isoforms under the control of a human FMR1 promoter subdomain were administered into bilateral ventricles of neonatal Fmr1 -/y and wild-type (WT) mice, both human FMRP isoforms were expressed throughout the brain in a pattern reminiscent to that of mouse FMRP. Importantly, human FMRP expression attenuated social behavior deficits and stereotyped and repetitive behavior, and reversed dysmorphological dendritic spines in Fmr1 -/y mice, without affecting WT mouse behaviors. Our results demonstrate that human FMR1 promoter can effectively drive human FMRP expression in the brain to attenuate Fmr1 -/y mouse deficits, strengthening the notion of using AAV gene therapy for FXS treatment.

11.
Signal Transduct Target Ther ; 7(1): 261, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35915083

RESUMO

Apolipoprotein E (APOE) plays a pivotal role in lipid including cholesterol metabolism. The APOE ε4 (APOE4) allele is a major genetic risk factor for Alzheimer's and cardiovascular diseases. Although APOE has recently been associated with increased susceptibility to infections of several viruses, whether and how APOE and its isoforms affect SARS-CoV-2 infection remains unclear. Here, we show that serum concentrations of APOE correlate inversely with levels of cytokine/chemokine in 73 COVID-19 patients. Utilizing multiple protein interaction assays, we demonstrate that APOE3 and APOE4 interact with the SARS-CoV-2 receptor ACE2; and APOE/ACE2 interactions require zinc metallopeptidase domain of ACE2, a key docking site for SARS-CoV-2 Spike protein. In addition, immuno-imaging assays using confocal, super-resolution, and transmission electron microscopies reveal that both APOE3 and APOE4 reduce ACE2/Spike-mediated viral entry into cells. Interestingly, while having a comparable binding affinity to ACE2, APOE4 inhibits viral entry to a lesser extent compared to APOE3, which is likely due to APOE4's more compact structure and smaller spatial obstacle to compete against Spike binding to ACE2. Furthermore, APOE ε4 carriers clinically correlate with increased SARS-CoV-2 infection and elevated serum inflammatory factors in 142 COVID-19 patients assessed. Our study suggests a regulatory mechanism underlying SARS-CoV-2 infection through APOE interactions with ACE2, which may explain in part increased COVID-19 infection and disease severity in APOE ε4 carriers.


Assuntos
COVID-19 , SARS-CoV-2 , Enzima de Conversão de Angiotensina 2/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Sítios de Ligação , COVID-19/genética , Humanos , Inflamação/genética , Ligação Proteica , Glicoproteína da Espícula de Coronavírus
12.
Natl Sci Rev ; 8(7): nwab024, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34691693

RESUMO

Loss-of-function mutations in sorting nexin 14 (SNX14) cause autosomal recessive spinocerebellar ataxia 20, which is a form of early-onset cerebellar ataxia that lacks molecular mechanisms and mouse models. We generated Snx14-deficient mouse models and observed severe motor deficits and cell-autonomous Purkinje cell degeneration. SNX14 deficiency disrupted microtubule organization and mitochondrial transport in axons by destabilizing the microtubule-severing enzyme spastin, which is implicated in dominant hereditary spastic paraplegia with cerebellar ataxia, and compromised axonal integrity and mitochondrial function. Axonal transport disruption and mitochondrial dysfunction further led to degeneration of high-energy-demanding Purkinje cells, which resulted in the pathogenesis of cerebellar ataxia. The antiepileptic drug valproate ameliorated motor deficits and cerebellar degeneration in Snx14-deficient mice via the restoration of mitochondrial transport and function in Purkinje cells. Our study revealed an unprecedented role for SNX14-dependent axonal transport in cerebellar ataxia, demonstrated the convergence of SNX14 and spastin in mitochondrial dysfunction, and suggested valproate as a potential therapeutic agent.

13.
Brain Res Bull ; 149: 216-221, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31051227

RESUMO

The satellite glial cells (SGCs) of the dorsal root ganglia (DRG) expressed P2X4 receptor. In this study, we investigated the abnormal sympathetic activity after myocardial ischemia (MI) involving P2X4 receptor in the cervical DRG SGC. The results showed that MI injury upregulated the P2X4 receptor mRNA and protein in DRG, and the upregulated P2X4 receptor was co-localized with glial fibrillary acidic protein (GFAP) in DRG SGCs. P2X4 short hairpin RNA (shRNA) treatment decreased the expression of P2X4 receptor, counteracted the upregulation of GFAP and IL-1ß and inhibited P38MAPK phosphorylation in DRG of MI rats. These results indicate that application of P2X4 shRNA may reduce P2X4-mediated nociceptive signal via inhibiting DRG afferents to alleviate the abnormal sympathetic activity induced by MI.


Assuntos
Gânglios Espinais/metabolismo , Gânglios Espinais/fisiopatologia , Isquemia Miocárdica/fisiopatologia , Neuroglia/metabolismo , Receptores Purinérgicos P2X4/metabolismo , Animais , Diabetes Mellitus Experimental/metabolismo , Neuropatias Diabéticas/metabolismo , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Isquemia Miocárdica/metabolismo , Neuralgia/metabolismo , Antagonistas do Receptor Purinérgico P2X/farmacologia , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Células Satélites Perineuronais/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
ACS Chem Neurosci ; 10(3): 1318-1325, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30475578

RESUMO

Activation of satellite glial cells (SGCs) in the dorsal root ganglia (DRG) is involved in mechanical and thermal hyperalgesia. The upregulated P2Y12 receptor expressed in SGCs of the DRG participates in the nociceptive transmission of neuropathic pain. Guanfu base A (GFA) has been reported to exhibit antiarrhythmic and anti-inflammatory effects. In this study, we explored the effects of GFA on P2Y12 receptor-mediated mechanical and thermal hyperalgesia in chronic constriction injury (CCI) rats. Sprague-Dawley rats were randomly divided into sham operation group (Sham), CCI operation group (CCI), CCI rats treated with guanfu base A group (CCI + GFA) and control rats treated with GFA group (Ctrl + GFA). Mechanical withdrawal threshold and thermal withdrawal latency were measured. P2Y12 expression in L4-L6 dorsal root ganglion (DRG) was detected by quantitative real-time PCR and Western blot. After CCI treatment, mechanical and thermal hyperalgesia and the expression values of P2Y12 receptor mRNA and protein in DRG were increased. Dual-labeling immunofluorescence showed that the coexpression of P2Y12 receptor and glial fibrillary acidic protein (GFAP) in the DRG of CCI rats was increased compared to sham rats. GFA relieved mechanical and thermal hyperalgesia in the CCI rats, decreased the expression of P2Y12 mRNA and protein and phosphorylation of p38 MAPK in the DRG, and increased the ADP-downregulated cAMP concentrations in HEK293 cells transfected with P2Y12 plasmid. After CCI rats were treated with GFA, the coexpression of P2Y12 receptor and GFAP in the DRG was significantly decreased compared to the untreated CCI group. Thus, downregulating the P2Y12 receptor relieved mechanical and thermal hyperalgesia in the CCI rats.


Assuntos
Analgésicos/farmacologia , Gânglios Espinais/efeitos dos fármacos , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Neuralgia/tratamento farmacológico , Receptores Purinérgicos P2/metabolismo , Animais , Constrição Patológica/tratamento farmacológico , Constrição Patológica/metabolismo , AMP Cíclico/metabolismo , Modelos Animais de Doenças , Gânglios Espinais/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Neuralgia/metabolismo , RNA Mensageiro/metabolismo , Distribuição Aleatória , Ratos Sprague-Dawley , Receptores Purinérgicos P2Y12/metabolismo , Nervo Isquiático , Fator de Necrose Tumoral alfa/metabolismo
15.
Neurochem Int ; 112: 259-266, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28827078

RESUMO

Human immunodeficiency virus (HIV) envelope glycoprotein (glycoprotein 120, gp120) can induce chronic neuropathic pain by directly stimulating primary sensory afferent neurons. Activation of satellite glial cells (SGCs) in dorsal root ganglia (DRG) plays an important role in the transmission of neuropathic pain. The P2Y12 receptor is expressed in SGCs of DRG. In this study, we investigated the role of the P2Y12 receptor in HIV gp120-induced neuropathic pain. The results showed that peripheral nerve exposure to HIV gp120 increased mechanical and thermal hyperalgesia in gp120-treated model rats. The gp120 treatment increased the expression of P2Y12 mRNA and protein in DRG SGCs. Treatment with P2Y12 short hairpin RNA (shRNA) in DRG SGCs decreased the upregulated expression of P2Y12 mRNA and protein in DRG SGCs as well as relieved mechanical and thermal hyperalgesia in gp120-treated rats. Reduction of P2Y12 receptor decreased co-expression of P2Y12 and glial fibrillary acidic protein (GFAP), expression of GFAP, interleukin (IL)-1ß, tumor necrosis factor (TNF)-receptor 1 (TNF-R1), and phosphorylation of Akt (p-Akt) proteins in DRG of gp120-treated rats. Upregulation of GFAP is a marker of SGC activation. Therefore, P2Y12 shRNA treatment decreased HIV gp120-induced mechanical and thermal hyperalgesia in gp120-treated rats.


Assuntos
Proteína gp120 do Envelope de HIV/toxicidade , Neuralgia/induzido quimicamente , Neuralgia/tratamento farmacológico , RNA Interferente Pequeno/administração & dosagem , Receptores Purinérgicos P2/administração & dosagem , Animais , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Masculino , Neuralgia/metabolismo , RNA Interferente Pequeno/biossíntese , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2/biossíntese , Receptores Purinérgicos P2Y12 , Resultado do Tratamento
16.
Oxid Med Cell Longev ; 2018: 5082817, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30210654

RESUMO

Insulin resistance and type 2 diabetes mellitus (T2DM) are highly prevalent around the world. Elevated concentrations of free fatty acids (FFAs) are closely related to insulin resistance and T2DM. P2X7 receptor is an ion channel gated by ATP, which is implicated in various scenarios including immune response, pain, and inflammation. In this study, we have explored whether P2X7 receptor is involved in pathological changes in human umbilical vein endothelial cells (HUVECs) induced by high FFA treatment, and the potential beneficial effects of evodiamine. Evodiamine could effectively suppress the enhanced expression of P2X7 receptor caused by high FFAs at both mRNA and protein levels. In addition, high FFA-induced cytotoxicity, the upregulated release of ATP, and production of reactive oxygen species (ROS) could be ameliorated by evodiamine in HUVECs. Evodiamine could also reverse the decreased NO formation and the increased adhesive events of immune cells at high FFAs. Moreover, evodiamine inhibited P2X7-dependent TNF-α expression and ERK 1/2 phosphorylation due to high FFAs. All these results indicated that evodiamine could correct the upregulated expression of P2X7 receptor induced under high FFA condition in HUVECs, and consequently suppressed oxidative stress and inflammatory responses.


Assuntos
Ácidos Graxos não Esterificados/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Extratos Vegetais/uso terapêutico , Quinazolinas/uso terapêutico , Receptores Purinérgicos P2X7/metabolismo , Células Endoteliais , Humanos , Extratos Vegetais/farmacologia , Quinazolinas/farmacologia , Receptores Purinérgicos P2X7/análise
17.
Front Pharmacol ; 9: 593, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29950989

RESUMO

Aim: In this study, we investigated whether andrographolide (Andro) can alleviate neuropathic pain induced by HIV gp120 plus ddC treatment and the mechanism of its action. Methods: The paw withdrawal threshold and the paw withdrawal latency were observed to assess pain behaviors in all groups of the rats, including control group, control combined with Andro treatment group, sham group, gp120 combined with ddC treatment group, gp120 plus ddC combined with A438079 treatment group, and gp120 plus ddC combined with Andro treatment by intrathecally injecting at a dose of 25 µg/20 µl group. The protein expression levels of the P2X7 receptor, tumor necrosis factor-α-receptor (TNFα-R), interleukin-1ß (IL-1ß), IL-10, phospho-extracellular regulated protein kinases (ERK) (p-ERK) in the L4-L6 dorsal root ganglia (DRG) were measured by western blotting. Real-time quantitative polymerase chain reaction was used to test the mRNA expression level of the P2X7 receptor. Double-labeling immunofluorescence was used to identify the co-localization of the P2X7 receptor with glial fibrillary acidic protein (GFAP) in DRG. Molecular docking was performed to identify whether the Andro interacted perfectly with the rat P2X7 (rP2X7) receptor. Results: Andro attenuated the mechanical and thermal hyperalgesia in gp120+ddC-treated rats and down-regulated the P2X7 receptor mRNA and protein expression in the L4-L6 DRGs of gp120+ddC-treated rats. Additionally, Andro simultaneously decreased the expression of TNFα-R and IL-1ß protein, increased the expression of IL-10 protein in L4-L6 DRGs, and inhibited the activation of ERK signaling pathways. Moreover, Andro decreased the co-expression of GFAP and the P2X7 receptor in the SGCs of L4-L6 DRG on 14th day after surgery. Conclusion: Andro decreased the hyperalgesia induced by gp120 plus ddC.

18.
Brain Res Bull ; 142: 289-296, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30118750

RESUMO

Diabetes mellitus (DM) is considered the primary cause of neuropathic pain. Osthole (7-methoxy-8[3-methylpent 2-enyl]coumarin) is a component extracted from Cnidium monnieri (L.) cusson plant seeds and has anti-inflammatory and anti-oxidative properties. The aim of the present study was to investigate the effects of osthole on diabetic neuropathic pain (DNP) involving the P2X4 receptor on satellite glial cells (SGCs) in the dorsal root ganglia (DRG) of type 2 diabetic rats. These data showed that the mechanical withdrawal threshold (MWT) and thermal withdrawal latency (TWL) in DM rats were lower than those in control rats. MWT and TWL in DM rats treated with osthole were higher compared with those in untreated DM rats. The expression levels of P2X4 mRNA and protein in the DRG of DM rats were higher compared with those in the control rats, while those in DM rats treated with osthole were significantly lower compared with those in the untreated DM rats. Osthole treatment decreased the co-expression levels of P2X4 and glial fibrillary acidic protein (GFAP) and reduced the up-regulated expression of interleukin-1 beta (IL-1ß), tumour necrosis factor-α (TNF-α), brain-derived neurotrophic factor (BDNF) and phosphorylated-p38MAPK and enhanced the down-regulation of IL-10 in DM rats. Thus, osthole treatment may act on the P2X4 receptor to alleviate the mechanical and thermal hyperalgesia in DM rats.


Assuntos
Analgésicos/farmacologia , Cumarínicos/farmacologia , Neuropatias Diabéticas/tratamento farmacológico , Gânglios Espinais/efeitos dos fármacos , Neuralgia/tratamento farmacológico , Receptores Purinérgicos P2X4/metabolismo , Animais , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Neuropatias Diabéticas/metabolismo , Neuropatias Diabéticas/patologia , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Hipoglicemiantes/farmacologia , Masculino , Neuralgia/metabolismo , Neuralgia/patologia , Limiar da Dor/efeitos dos fármacos , Limiar da Dor/fisiologia , Antagonistas do Receptor Purinérgico P2X/farmacologia , Distribuição Aleatória , Ratos Sprague-Dawley , Regulação para Cima/efeitos dos fármacos
19.
Front Physiol ; 9: 928, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30065662

RESUMO

Myocardial ischemia (MI) is one of the major causes of death in cardiac diseases. Purinergic signaling is involved in bidirectional neuronal-glial communication in the primary sensory ganglia. The sensory neuritis of cardiac afferent neurons in cervical dorsal root ganglion (cDRG) interacts with cardiac sympathetic efferent postganglionic neurons, forming feedback loops. The P2Y12 receptor is expressed in satellite glial cells (SGCs) of DRG. Baicalin is a major active ingredient extracted from natural herbal medicines, which has anti-inflammatory and strong anti-oxidation properties. In this study we investigated the effect of baicalin on P2Y12 receptor in the cervical DRG SGC-mediated sympathoexcitatory reflex, which is increased during MI. The results showed that the expression of P2Y12 receptor mRNA and protein in DRG, and the co-localization values of P2Y12 receptor and glial fibrillary acidic protein (GFAP) in cDRG SGCs were increased after MI. The activated SGCs increased IL-1ß protein expression and elevated Akt phosphorylation in cDRG. Baicalin treatment inhibited the upregulation of the P2Y12 receptor, GFAP protein and Akt phosphorylation in cDRG neurons/SGCs. The stellate ganglia (SG) affect cardiac sympathetic activity. Baicalin treatment also decreased the upregulation of the P2Y12 receptor, GFAP protein in the SG. The P2Y12 agonist, 2Me-SADP, increased [Ca2+]i in HEK293 cells transfected with the P2Y12 receptor plasmid and SGCs in cDRG. These results indicate that application of baicalin alleviates pathologic sympathetic activity induced by MI via inhibition of afferents in the cDRG.

20.
Neurochem Int ; 108: 27-33, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28192150

RESUMO

Neuropathic pain is a type of chronic pain caused by nervous system damage and dysfunction. The pathogenesis of chronic pain is complicated, and there are no effective therapies for neuropathic pain. Studies show that the P2X4 receptor expressed in the satellite glial cells (SGCs) of dorsal root ganglia (DRG) is related to neuropathic pain. Artemisinin is a monomeric component extracted from traditional Chinese medicine and has a variety of important pharmacological effects and potential applications. This study observed the effect of artemisinin on neuropathic pain and delineated its possible mechanism. The chronic constriction injury (CCI) rat model was used in this study. The results demonstrated that artemisinin relieved pain behaviors in the CCI rats, inhibited the expression of P2X4 receptor in the DRG, and decreased the ATP-activated currents in HEK293 cells transfected with P2X4 plasmid. Dual-labeling immunofluorescence showed that the coexpression of P2X4 receptor and glial fibrillary acidic protein (GFAP) in the DRG of CCI rats was increased compared to control rats. After CCI rats were treated with artemisinin, the coexpression of P2X4 receptor and GFAP in the DRG was significantly decreased compared to the CCI group. This finding suggested that artemisinin could inhibit the nociceptive transmission mediated by P2X4 receptor in the DRG SGCs and thus relieve pain behaviors in the CCI rats.


Assuntos
Artemisininas/uso terapêutico , Gânglios Espinais/metabolismo , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Medição da Dor/métodos , Receptores Purinérgicos P2X4/fisiologia , Animais , Artemisininas/farmacologia , Relação Dose-Resposta a Droga , Gânglios Espinais/efeitos dos fármacos , Células HEK293 , Humanos , Masculino , Medição da Dor/efeitos dos fármacos , Antagonistas do Receptor Purinérgico P2X/farmacologia , Antagonistas do Receptor Purinérgico P2X/uso terapêutico , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA