Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
J Virol ; 95(20): e0023121, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34379499

RESUMO

The NS1 protein of the influenza A virus plays a critical role in regulating several biological processes in cells, including the type I interferon (IFN) response. We previously profiled the cellular factors that interact with the NS1 protein of influenza A virus and found that the NS1 protein interacts with proteins involved in RNA splicing/processing, cell cycle regulation, and protein targeting processes, including 14-3-3ε. Since 14-3-3ε plays an important role in retinoic acid-inducible gene I (RIG-I) translocation to mitochondrial antiviral-signaling protein (MAVS) to activate type I IFN expression, the interaction of the NS1 and 14-3-3ε proteins may prevent the RIG-I-mediated IFN response. In this study, we confirmed that the 14-3-3ε protein interacts with the N-terminal domain of the NS1 protein and that the NS1 protein inhibits RIG-I-mediated IFN-ß promoter activation in 14-3-3ε-overexpressing cells. In addition, our results showed that knocking down 14-3-3ε can reduce IFN-ß expression elicited by influenza A virus and enhance viral replication. Furthermore, we found that threonine in the 49th amino acid position of the NS1 protein plays a role in the interaction with 14-3-3ε. Influenza A virus expressing C terminus-truncated NS1 with a T49A mutation dramatically increases IFN-ß mRNA in infected cells and causes slower replication than that of virus without the T-to-A mutation. Collectively, this study demonstrates that 14-3-3ε is involved in influenza A virus-initiated IFN-ß expression and that the interaction of the NS1 protein and 14-3-3ε may be one of the mechanisms for inhibiting type I IFN activation during influenza A virus infection. IMPORTANCE Influenza A virus is an important human pathogen causing severe respiratory disease. The virus has evolved several strategies to dysregulate the innate immune response and facilitate its replication. We demonstrate that the NS1 protein of influenza A virus interacts with the cellular chaperone protein 14-3-3ε, which plays a critical role in retinoic acid-inducible gene I (RIG-I) translocation that induces type I interferon (IFN) expression, and that NS1 protein prevents RIG-I translocation to the mitochondrial membrane. The interaction site for 14-3-3ε is the RNA-binding domain (RBD) of the NS1 protein. Therefore, this research elucidates a novel mechanism by which the NS1 RBD mediates IFN-ß suppression to facilitate influenza A viral replication. Additionally, the findings reveal the antiviral role of 14-3-3ε during influenza A virus infection.


Assuntos
Proteínas 14-3-3/imunologia , Influenza Humana/imunologia , Interferon beta/metabolismo , Proteínas 14-3-3/metabolismo , Linhagem Celular Tumoral , Proteína DEAD-box 58/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata/imunologia , Vírus da Influenza A/metabolismo , Influenza Humana/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/metabolismo , Interferon beta/fisiologia , Regiões Promotoras Genéticas/genética , Processamento de Proteína Pós-Traducional , RNA Viral/genética , Receptores Imunológicos/metabolismo , Proteínas não Estruturais Virais/imunologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/genética
2.
Cell Mol Neurobiol ; 42(7): 2205-2217, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33954807

RESUMO

Epidemiologic studies have indicated that chronic hypertension may facilitate the progression of abnormal behavior, such as emotional irritability, hyperactivity, and attention impairment. However, the mechanism of how chronic hypertension affects the brain and neuronal function remains unclear. In this study, 58-week-old male spontaneously hypertensive rats (SHR) and age-matched Wistar-Kyoto (WKY) control rats were used. Their locomotor activity and neuronal function were assessed by the open field test, novel object, and Y maze recognition test. Moreover brain tissues were analyzed. We found that the aged SHR exhibited significant locomotor hyperactivity when compared to the WKY rats. However, there was no significant difference in novel object and novel arm recognition between aged SHR and the WKY rats. In the analysis of synaptic membrane protein, the expression of glutamatergic receptors, such as the N-methyl-D-aspartate (NMDA) receptor receptors subunits 2B (GluN2B) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor 1 (GluA1) in the hippocampus of SHR were significantly higher than those of WKY rats. In addition, in the synaptic membrane of SHR's hippocampus and medial prefrontal cortex (mPFC), a down-regulation of astrocytes was found, though the excitatory amino acid transporter 2 (EAAT2) remained constant. Moreover, a down-regulation of microglia in the hippocampus and mPFC was seen in the SHR brain. Long-term exposure to high blood pressure causes upregulation of glutamate receptors. The upregulation of glutamatergic receptors in hippocampus may contribute to the hyper-locomotor activity of aged rodents and may as a therapeutic target in hypertension-induced irritability and hyperactivity.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade , Hipertensão , Animais , Ácido Glutâmico , Hipocampo , Masculino , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptores de AMPA , Receptores de N-Metil-D-Aspartato , Regulação para Cima
3.
Int J Mol Sci ; 21(4)2020 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-32070065

RESUMO

Croton is an extensive flowering plant genus in the spurge family, Euphorbiaceae. Three croton compounds with the common ent-kaurane skeleton have been purified from Croton tonkinensis. METHODS: We examined any modifications of croton components (i.e., croton-01 [ent-18-acetoxy-7α-hydroxykaur-16-en-15-one], croton-02 [ent-7α,14ß-dihydroxykaur-16-en-15-one] and croton-03 [ent-1ß-acetoxy-7α,14ß-dihydroxykaur-16-en-15-one] on either hyperpolarization-activated cation current (Ih) or erg-mediated K+ current identified in pituitary tumor (GH3) cells and in rat insulin-secreting (INS-1) cells via patch-clamp methods. RESULTS: Addition of croton-01, croton-02, or croton-03 effectively and differentially depressed Ih amplitude. Croton-03 (3 µM) shifted the activation curve of Ih to a more negative potential by approximately 11 mV. The voltage-dependent hysteresis of Ih was also diminished by croton-03 administration. Croton-03-induced depression of Ih could not be attenuated by SQ-22536 (10 µM), an inhibitor of adenylate cyclase, but indeed reversed by oxaliplatin (10 µM). The Ih in INS-1 cells was also depressed effectively by croton-03. CONCLUSION: Our study highlights the evidence that these ent-kaurane diterpenoids might conceivably perturb these ionic currents through which they have high influence on the functional activities of endocrine or neuroendocrine cells.


Assuntos
Croton/química , Diterpenos do Tipo Caurano/farmacologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/antagonistas & inibidores , Neoplasias Hipofisárias/tratamento farmacológico , Adenilil Ciclases/genética , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Diterpenos do Tipo Caurano/química , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/química , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Estrutura Molecular , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/patologia , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Folhas de Planta/química , Ratos
4.
Mol Pain ; 15: 1744806919855204, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31161874

RESUMO

Traumatic rib fracture can cause severe pain and is usually associated with the depression of respiratory drive followed by severe respiratory complications. It is critical for patients with rib fracture to receive adequate analgesia. However, strong opioids and other analgesics often produces side effects and may even cause respiratory suppression. Meanwhile, rib fixation now has become a popular method for treating rib fracture patients. However, the actual molecular mechanism leading to its effectiveness as an analgesia has not been fully investigated, and the best analgesic method for its use in rib fracture patients has not yet been determined. We developed a new animal model for rib fracture and evaluated changes in pain severity after rib fixation. Our data indicated significantly better analgesic behavior if a soft string rib fixation is performed, which is associated with cytokine (interleukine-6 and interleukine-10) decreases in the spinal cord and co-localization with glia cells. Our results provided a treatment suggestion for rib fracture patients and the possible molecular mechanism for the analgesic effects. Further molecular mechanisms and the best therapeutic methods are still needed for this severe painful condition.


Assuntos
Analgésicos/farmacologia , Citocinas/metabolismo , Fixação de Fratura , Fraturas das Costelas/cirurgia , Costelas/cirurgia , Medula Espinal/patologia , Animais , Astrócitos/metabolismo , Densidade Óssea , Densitometria , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Neuroglia/metabolismo , Osteogênese , Dor/patologia , Ratos Sprague-Dawley , Fraturas das Costelas/diagnóstico por imagem , Fraturas das Costelas/patologia , Costelas/diagnóstico por imagem , Costelas/patologia , Microtomografia por Raio-X , Raios X
5.
J Biomed Sci ; 26(1): 74, 2019 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-31627754

RESUMO

BACKGROUND: Complex regional pain syndrome (CRPS) is related to microcirculation impairment caused by tissue hypoxia and peripheral cytokine overproduction in the affected human limb and chronic post-ischemic pain (CPIP) is considered as an animal model for this intractable disease. Previous studies suggest that the pathogenesis of CPIP involves the hypoxia inducible factor-1α (HIF-1α) and an exaggerated regional inflammatory and free radical response. The inhibition of HIF-1α is known to relieve CPIP. So, propofol, as a free radical scavenger, is very likely to be beneficial in terms of relieving CPIP. METHODS: We set up a CPIP model using the hindpaw of mice. We administered propofol (10 mg/kg) just after the reperfusion period (early stage) and also on the second day (late stage), as treatment. The analysis evaluated the expression of HIF-1α, free radicals, and inflammasome. RESULTS: Propofol administration produced obvious analgesia in both mechanical and thermal evaluation in the early stage of CPIP (2 h after reperfusion). Only a mild analgesic effect was found in the late stage (48 h later after reperfusion). In the early stage, the expression of HIF-1α and the inflammasome marker (NALP1) along with caspase-1 were suppressed by propofol. The free radical level also decreased in the propofol group. But those molecular changes were not founded in the late stage of CPIP. CONCLUSION: Our data demonstrated that propofol produces mice analgesia in the early stage of CPIP and this effect is associated with inhibition of free radical, hypoxia inducible factor and inflammasome.


Assuntos
Analgesia , Síndromes da Dor Regional Complexa/tratamento farmacológico , Hipnóticos e Sedativos/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Inflamassomos/genética , Propofol/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Administração Intravenosa , Anestésicos Intravenosos/farmacologia , Animais , Sequestradores de Radicais Livres/farmacologia , Regulação da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamassomos/metabolismo , Masculino , Camundongos
6.
Drug Dev Res ; 80(6): 846-856, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31301190

RESUMO

Nalbuphine (NAL) is recognized as a mixer with the κ-opioid receptor agonist and the µ-opioid receptor antagonist. However, whether this drug causes any modifications in neuronal ionic currents is unclear. The effects of NAL on ionic currents in mHippoE-14 hippocampal neurons were investigated. In the whole-cell current recordings, NAL suppressed the peak amplitude of voltage-gated Na+ current (INa ) with an IC50 value of 1.9 µM. It shifted the steady-state inactivation curve of peak INa to the hyperpolarized potential, suggesting that there is the voltage dependence of NAL-mediated inhibition of peak INa . In continued presence of NAL, subsequent application of either dynorphin A1-13 (1 µM) or naloxone (30 µM) failed to modify its suppression of peak INa . Tefluthrin (Tef; 10 µM), a pyrethroid known to activate INa , increased peak INa with slowed current inactivation; however, further application of NAL suppressed Tef-mediated suppression of peak INa followed by an additional slowing of current inactivation. In addition, NAL suppressed the amplitude of M-type K+ current [IK(M) ] with an IC50 value of 5.7 µM, while it slightly suppressed erg-mediated and delayed-rectifier K+ currents. In the inside-out current recordings, NAL failed to modify the activity of large-conductance Ca2+ -activated K+ channels. In differentiated NG108-15 neuronal cells, NAL also suppressed the peak INa , and subsequent addition of Tef reversed NAL-induced suppression of INa . Our study highlights the evidence that in addition to modulate opioid receptors, NAL has the propensity to interfere with ionic currents including INa and IK(M) , thereby influencing the functional activities of central neurons.


Assuntos
Analgésicos Opioides/farmacologia , Canais de Potássio de Retificação Tardia/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Nalbufina/farmacologia , Neurônios/efeitos dos fármacos , Receptores Opioides kappa/agonistas , Receptores Opioides mu/antagonistas & inibidores , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Animais , Linhagem Celular , Canais de Potássio de Retificação Tardia/fisiologia , Canais de Potássio Éter-A-Go-Go/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Camundongos , Neurônios/fisiologia
7.
Anesth Analg ; 126(2): 661-669, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28858902

RESUMO

BACKGROUND: Huntington disease (HD) affects the nervous system and leads to mental and motor dysfunction. Previous studies have shown that HD is caused by the exon 1 region of the huntingtin (HTT) gene having expanded CAG trinucleotide repeats. However, few studies have focused on the relationship between HD and pain. The purpose of this study is to investigate the relationship between HD and pain response. METHODS: We used clinical similar transgenic HD mice carrying a mutant HTT exon 1 containing 84 CAG trinucleotide repeats to evaluate the relationship between HD and pain. Inflammatory pain models were induced by either formalin or complete Freund adjuvant injection over the hind paw. Spinal cord, dorsal root ganglion, and paw skin tissues were harvested at the end of the behavioral inflammatory pain studies. Immunofluorescence assay, Western blotting, and enzyme-linked immunosorbent assay were used to identify changes in cells and cytokines. RESULTS: Our data demonstrate that preonset HD mice exhibited less pain behavior than wild-type (WT) mice in both young (n = 11 [WT], 13 [HD]) and aged (n = 8 [WT], 9 [HD]) mice. Western blotting and immunohistological examination of lumbar spinal cord tissue and dorsal root ganglion indicate less activation of glial cells and astrocytes in young HD mice (n = 6-7) compared to that in WT mice (n = 6-7). The production levels of tumor necrosis factor-α, interleukin-1ß, and substance P were also lower in young HD mice (n = 6-7). CONCLUSIONS: Our data demonstrate less pain behavior and pain-related cytokine response at the spinal cord level for HD mice compared to those for WT mice. Further studies are needed for determining the mechanism as to how mutant HTT leads to altered pain behavior and pain-related cytokine response.


Assuntos
Modelos Animais de Doenças , Proteína Huntingtina/genética , Doença de Huntington/genética , Medição da Dor/métodos , Dor/genética , Animais , Feminino , Doença de Huntington/patologia , Masculino , Camundongos , Camundongos Transgênicos , Dor/patologia
8.
J Nanobiotechnology ; 16(1): 49, 2018 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-29769077

RESUMO

BACKGROUND: Nanoparticles have become one of the most promising among the potential materials used for biomedical applications. However, few researchers have focused on their effects on analgesia. Despite the fact that various nanoparticles have been evaluated for drug delivery and MRI imaging contrast enhancement in clinical settings, no reports have investigated the in vivo synergy of ketorolac iron-oxide nanoparticle conjugates to improve the analgesic effect. METHODS: Ketorolac conjugated magnetic iron oxide nanoparticles (Keto-SPIO) were synthesized via two-stage additions of protective agents and chemical co-precipitation. ICR mice were used to develop inflammatory pain models induced by Complete Freund's adjuvant (CFA) injection in the hind paw. Different magnet field strengths and polarities were applied to the spinal cord after injecting Keto-SPIO into the theca space. Analgesia behavior was evaluated with the up-down method via von Frey microfilament measurement. Spinal cord tissues were harvested at the end analgesia time point upon induction of the inflammatory pain. The presence of the two cyclooxygenases (COX) in the spinal cord was examined via Western blotting to quantify the changes after intra-thecal Keto-SPIO administration. RESULTS: Intrathecal Keto-SPIO administration demonstrated a magnetic field-dependent analgesia effect in CFA pain model with a significant reduction in COX expression. CONCLUSIONS: Our results indicated that intrathecal administration of the Keto-SPIO combined magnet field modulated delivery significantly promoted an analgesia effect with suppression of COX in the mice inflammatory pain model.


Assuntos
Anti-Inflamatórios não Esteroides/farmacocinética , Cetorolaco/farmacocinética , Nanopartículas de Magnetita/química , Nanoconjugados/química , Manejo da Dor/métodos , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Inflamação/tratamento farmacológico , Injeções Espinhais , Cetorolaco/administração & dosagem , Cetorolaco/farmacologia , Cetorolaco/uso terapêutico , Campos Magnéticos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Dor/fisiopatologia , Tamanho da Partícula , Prostaglandina-Endoperóxido Sintases/metabolismo
9.
Nanomedicine ; 13(6): 1975-1981, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28539274

RESUMO

Few studies have investigated the effects of iron oxide nanoparticles (NPs) on analgesia. We developed inflammatory pain models via complete Freund's adjuvant injection over the hind paw in CD1 mice. Various doses of magnetite (Fe3O4) NPs were injected into the paw. Analgesia behavior was checked with von Frey microfilament and thermal irradiation measurements. Paw skin tissues were harvested at the maximal analgesia time point. The presence of activated white cells (CD68, myeloperoxidase) and free radical (reactive oxygen species, ROS) production was also checked. Western blotting was used to identify the changes of ROS production enzymes. Fe3O4 NPs demonstrated a dose-related analgesia effect with significant reduction in inflammatory cells, pro-inflammatory markers, and ROS production in the lesion paw. ROS production enzyme expression also declined. The results indicate that local Fe3O4 NP administration induced significant analgesia via attenuation of inflammatory cell infiltration and pro-inflammatory signaling as well as scavenging of microenvironment free radicals in a mouse inflammatory pain model.


Assuntos
Analgesia/métodos , Modelos Animais de Doenças , Compostos Férricos/uso terapêutico , Inflamação/tratamento farmacológico , Nanopartículas/uso terapêutico , Dor/tratamento farmacológico , Adjuvantes Imunológicos/toxicidade , Animais , Adjuvante de Freund , Inflamação/induzido quimicamente , Inflamação/patologia , Masculino , Camundongos , Dor/induzido quimicamente , Dor/patologia
10.
J Virol ; 89(20): 10273-85, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26223635

RESUMO

UNLABELLED: The NS1 protein of influenza virus has multiple functions and is a determinant of virulence. Influenza viruses with NS1 deletions (DelNS1 influenza viruses) are a useful tool for studying virus replication and can serve as effective live attenuated vaccines, but deletion of NS1 severely diminishes virus replication, hampering functional studies and vaccine production. We found that WSN-DelNS1 viruses passaged in cells consistently adapted to gain an A14U substitution in the 3' noncoding region of the M segment of viral RNA (vRNA) which restored replicative ability. DelNS1-M-A14U viruses cannot inhibit interferon expression in virus infected-cells, providing an essential model for studying virus replication in the absence of the NS1 protein. Characterization of DelNS1-M-A14U virus showed that the lack of NS1 has no apparent effect on expression of other viral proteins, with the exception of M mRNAs. Expression of the M transcripts, M1, M2, mRNA3, and mRNA4, is regulated by alternative splicing. The A14U substitution changes the splicing donor site consensus sequence of mRNA3, altering expression of M transcripts, with M2 expression significantly increased and mRNA3 markedly suppressed in DelNS1-M-A14U, but not DelNS1-M-WT, virus-infected cells. Further analysis revealed that the A14U substitution also affects promoter function during replication of the viral genome. The M-A14U mutation increases M vRNA synthesis in DelNS1 virus infection and enhances alternative splicing of M2 mRNA in the absence of other viral proteins. The findings demonstrate that NS1 is directly involved in influenza virus replication through modulation of alternative splicing of M transcripts and provide strategic information important to construction of vaccine strains with NS1 deletions. IMPORTANCE: Nonstructural protein (NS1) of influenza virus has multiple functions. Besides its role in antagonizing host antiviral activity, NS1 is also believed to be involved in regulating virus replication, but mechanistic details are not clear. The NS1 protein is a virulence determinant which inhibits both innate and adaptive immunity and live attenuated viruses with NS1 deletions show promise as effective vaccines. However, deletion of NS1 causes severe attenuation of virus replication during infection, impeding functional studies and vaccine development. We characterized a replication-competent DelNS1 virus which carries an A14U substitution in the 3' noncoding region of the vRNA M segment. We found that M-A14U mutation supports virus replication through modulation of alternative splicing of mRNAs transcribed from the M segment. Our findings give insight into the role of NS1 in influenza virus replication and provide an approach for constructing replication-competent strains with NS1 deletions for use in functional and vaccine studies.


Assuntos
Processamento Alternativo , Genoma Viral , Vírus da Influenza A/genética , RNA Viral/genética , Proteínas não Estruturais Virais/genética , Replicação Viral/genética , Regiões 3' não Traduzidas , Animais , Sequência de Bases , Chlorocebus aethiops , Cães , Células HEK293 , Humanos , Vírus da Influenza A/metabolismo , Células Madin Darby de Rim Canino , Dados de Sequência Molecular , Mutação Puntual , Sítios de Splice de RNA , RNA Viral/metabolismo , Deleção de Sequência , Células Vero , Proteínas não Estruturais Virais/deficiência
11.
PLoS Pathog ; 10(6): e1004199, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24968230

RESUMO

The primary role of cytoplasmic viral RNA-dependent RNA polymerase (RdRp) is viral genome replication in the cellular cytoplasm. However, picornaviral RdRp denoted 3D polymerase (3D(pol)) also enters the host nucleus, where its function remains unclear. In this study, we describe a novel mechanism of viral attack in which 3D(pol) enters the nucleus through the nuclear localization signal (NLS) and targets the pre-mRNA processing factor 8 (Prp8) to block pre-mRNA splicing and mRNA synthesis. The fingers domain of 3D(pol) associates with the C-terminal region of Prp8, which contains the Jab1/MPN domain, and interferes in the second catalytic step, resulting in the accumulation of the lariat form of the splicing intermediate. Endogenous pre-mRNAs trapped by the Prp8-3D(pol) complex in enterovirus-infected cells were identified and classed into groups associated with cell growth, proliferation, and differentiation. Our results suggest that picornaviral RdRp disrupts pre-mRNA splicing processes, that differs from viral protease shutting off cellular transcription and translation which contributes to the pathogenesis of viral infection.


Assuntos
Núcleo Celular/metabolismo , Citoplasma/enzimologia , Enterovirus Humano A/metabolismo , Splicing de RNA , Proteínas de Ligação a RNA/antagonistas & inibidores , DNA Polimerase Dirigida por RNA/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular , Núcleo Celular/virologia , Citoplasma/metabolismo , Citoplasma/virologia , Enterovirus/metabolismo , Enterovirus/patogenicidade , Enterovirus Humano A/patogenicidade , Humanos , Sinais de Localização Nuclear , Poliovirus/metabolismo , Poliovirus/patogenicidade , Biossíntese de Proteínas , Domínios e Motivos de Interação entre Proteínas , Precursores de RNA/antagonistas & inibidores , Precursores de RNA/metabolismo , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , DNA Polimerase Dirigida por RNA/química , DNA Polimerase Dirigida por RNA/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Rhinovirus/metabolismo , Rhinovirus/patogenicidade , Especificidade da Espécie , Transcrição Gênica , Carga Viral , Proteínas Virais/química , Proteínas Virais/genética , Virulência
12.
Clin Exp Pharmacol Physiol ; 43(3): 355-9, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26711019

RESUMO

Complex regional pain syndrome (CRPS) is related to microcirculation impairment associated with tissue hypoxia and peripheral cytokine overproduction in the affected limb. Previous studies suggest that the pathogenesis involves hypoxia inducible factor-1α (HIF-1α) and exaggerated regional inflammatory response. 1-methylpropyl 2-imidazolyl disulfide (PX-12) acts as the thioredoxin-1 (Trx-1) inhibitor and decreases the level of HIF-1α, and can rapidly be metabolized for Trx-1 redox inactivation. This study hypothesized that PX-12 can decrease the cytokine production for nociceptive sensitization in the hypoxia-induced pain model. CD1 mice weighing around 30 g were used. The animal CRPS model was developed via the chronic post-ischaemic pain (CPIP) model. The model was induced by using O-rings on the ankles of the mice hind limbs to produce 3-h ischaemia-reperfusion injury on the paw. PX-12 (25 mg/kg, 5 mg/kg) was given through tail vein injection immediately after ischaemia. Animal behaviour was tested using the von Frey method for 7 days. Local paw skin tissue was harvest from three groups (control, 5 mg/kg, 25 mg/kg) 2 h after injection of PX-12. The protein expression of interleukin-1ß (IL-1ß) and HIF-1α was analysed with the Western blotting method. Mice significantly present an anti-allodynia effect in a dose-related manner after the PX-12 administration. Furthermore, PX-12 not only decreased the expression of HIF-1α but also decreased the expression of IL-1ß over the injured palm. This study, therefore, shows the first evidence of the anti-allodynia effect of PX-12 in a CPIP animal model for pain behaviour. The study concluded that inhibition of HIF-1α may produce an analgesic effect and the associated suppression of inflammatory cytokine IL-1ß in a CPIP model.


Assuntos
Síndromes da Dor Regional Complexa/complicações , Citocinas/metabolismo , Dissulfetos/farmacologia , Hiperalgesia/complicações , Hiperalgesia/tratamento farmacológico , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Imidazóis/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Dissulfetos/uso terapêutico , Hiperalgesia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imidazóis/uso terapêutico , Inflamação/metabolismo , Masculino , Camundongos
13.
Biomedicines ; 11(10)2023 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-37893024

RESUMO

Cannabidiol (CBD) is a naturally occurring compound found in the Cannabis plant that is known for its potential therapeutic effects. However, its impact on membrane ionic currents remains a topic of debate. This study aimed to investigate how CBD modifies various types of ionic currents in pituitary GH3 cells. Results showed that exposure to CBD led to a concentration-dependent decrease in M-type K+ currents (IK(M)), with an IC50 of 3.6 µM, and caused the quasi-steady-state activation curve of the current to shift to a more depolarized potential with no changes in the curve's steepness. The CBD-mediated block of IK(M) was not reversed by naloxone, suggesting that it was not mediated by opioid receptors. The IK(M) elicited by pulse-train stimulation was also decreased upon exposure to CBD. The magnitude of erg-mediated K+ currents was slightly reduced by adding CBD (10 µM), while the density of voltage-gated Na+ currents elicited by a short depolarizing pulse was not affected by it. Additionally, CBD decreased the magnitude of hyperpolarization-activated cation currents (Ih) with an IC50 of 3.3 µM, and the decrease was reversed by oxaliplatin. The quasi-steady-state activation curve of Ih was shifted in the leftward direction with no changes in the slope factor of the curve. CBD also diminished the strength of voltage-dependent hysteresis on Ih elicited by upright isosceles-triangular ramp voltage. Collectively, these findings suggest that CBD's modification of ionic currents presented herein is independent of cannabinoid or opioid receptors and may exert a significant impact on the functional activities of excitable cells occurring in vitro or in vivo.

14.
Minerva Anestesiol ; 89(5): 434-444, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36762984

RESUMO

INTRODUCTION: Association between sugammadex and risk of postoperative nausea and vomiting remains unclear. EVIDENCE ACQUISITION: We performed meta-analysis of randomized controlled trials with trial sequential analysis to compare sugammadex with neostigmine in adults receiving elective surgery under general anesthesia with postoperative extubation. Databases of MEDLINE, Embase, and Cochrane Central Register of Controlled Trials were searched from inception to April 15, 2022. Primary outcome was risk of postoperative nausea and vomiting after patients received sugammadex or neostigmine. Secondary outcomes were incidences of sugammadex-related complications. EVIDENCE SYNTHESIS: Meta-analysis of 40 trials with 5455 patients showed an overall lower risk of postoperative nausea and vomiting in the sugammadex group than in the neostigmine group (risk ratio: 0.85, 95% CI [0.76-0.94], heterogeneity I2=4%, P=0.002). Subgroup analyses demonstrated a lower risk of postoperative nausea and vomiting associated with sugammadex than with neostigmine: 1) in the postanesthesia care unit (risk ratio: 0.77, 95% CI [0.66-0.90], I2=8%, P=0.001) but not in wards; 2) under volatile anesthetics but not total intravenous anesthesia; 3) regardless of the administration of prophylactic antiemetics; and 4) when sugammadex was administered at 2 mg/kg but not 4 mg/kg. No major complications such as cardiac arrest or refractory bradycardia were noted and every patient achieved adequate neuromuscular recovery before extubation in all of the included trials. The overall quality of evidence was moderate. CONCLUSIONS: Sugammadex was associated with a lower risk of postoperative nausea and vomiting compared with neostigmine immediately after surgery, especially for patients receiving volatile anesthetics regardless of the use of prophylactic antiemetics.


Assuntos
Antieméticos , Bloqueio Neuromuscular , Adulto , Humanos , Sugammadex , Neostigmina/uso terapêutico , Náusea e Vômito Pós-Operatórios/epidemiologia , Náusea e Vômito Pós-Operatórios/prevenção & controle , Bloqueio Neuromuscular/efeitos adversos , Ensaios Clínicos Controlados Aleatórios como Assunto , Período de Recuperação da Anestesia
15.
Microbes Infect ; 25(1-2): 105044, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36096357

RESUMO

The World Health Organization has highlighted the importance of an international standard (IS) for severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) neutralizing antibody titer detection to calibrate diagnostic techniques. We applied an IS to calibrate neutralizing antibody titers (NTs) (international units/mL) in response to coronavirus disease 2019 (COVID-19) vaccination. Moreover, the association between different factors and neutralizing antibodies was analyzed. A total of 1667 serum samples were collected from participants receiving different COVID-19 vaccines. Antibody titers were determined by a microneutralization assay using live viruses in a biosafety level 3 (BSL-3) laboratory and a commercial serological MeDiPro kit. The titer determined using the MeDiPro kit was highly correlated with the NT determined using live viruses and calibrated using IS. Fever and antipyretic analgesic treatment were related to neutralizing antibody responses in ChAdOx1-S and BNT162b2 vaccinations. Individuals with diabetes showed a low NT elicited by MVC-COV1901. Individuals with hypertension receiving the BNT162b2 vaccine had lower NTs than those without hypertension. Our study provided the international unit (IU) values of NTs in vaccinated individuals for the development of vaccines and implementation of non-inferiority trials. Correlation of the influencing factors with NTs can provide an indicator for selecting COVID-19 vaccines based on personal attributes.


Assuntos
COVID-19 , Hipertensão , Humanos , Vacinas contra COVID-19 , Vacina BNT162 , COVID-19/prevenção & controle , SARS-CoV-2 , Anticorpos Neutralizantes , Vacinação , Anticorpos Antivirais
16.
Mol Pain ; 8: 18, 2012 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-22439811

RESUMO

BACKGROUND: Activation of spinal cord glial cells such as microglia and astrocytes has been shown to regulate chronic opioid-induced antinociceptive tolerance and hyperalgesia, due to spinal up-regulation of the proinflammatory cytokines such as interleukin-1 beta (IL-1ß). Matrix metalloprotease-9 (MMP-9) has been implicated in IL-1ß activation in neuropathic pain. However, it is unclear whether acute opioid treatment can activate glial cells in the peripheral nervous system. We examined acute morphine-induced activation of satellite glial cells (SGCs) and up-regulation of IL-1ß in dorsal root ganglia (DRGs), and further investigated the involvement of MMP-9 in these opioid-induced peripheral changes. RESULTS: Subcutaneous morphine injection (10 mg/kg) induced robust peripheral glial responses, as evidenced by increased GFAP expression in DRGs but not in spinal cords. The acute morphine-induced GFAP expression is transient, peaking at 2 h and declining after 3 h. Acute morphine treatment also increased IL-1ß immunoreactivity in SGCs and IL-1ß activation in DRGs. MMP-9 and GFAP are expressed in DRG neurons and SGCs, respectively. Confocal analysis revealed a close proximity of MMP-9 and GFAP immunostaining. Importantly, morphine-induced DRG up-regulation of GFAP expression and IL-1ß activation was abolished after Mmp9 deletion or naloxone pre-treatment. Finally, intrathecal injections of IL-1ß-selective siRNA not only reduced DRG IL-1ß expression but also prolonged acute morphine-induced analgesia. CONCLUSIONS: Acute morphine induces opioid receptors- and MMP-9-dependent up-regulation of GFAP expression and IL-1ß activation in SGCs of DRGs. MMP-9 could mask and shorten morphine analgesia via peripheral neuron-glial interactions. Targeting peripheral glial activation might prolong acute opioid analgesia.


Assuntos
Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Interleucina-1beta/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Morfina/farmacologia , Neuroglia/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Western Blotting , Imuno-Histoquímica , Masculino , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Knockout , Neuroglia/efeitos dos fármacos , Reação em Cadeia da Polimerase em Tempo Real
17.
Mol Pain ; 8: 19, 2012 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-22444868

RESUMO

BACKGROUND: Despite decades of intense research efforts, actions of acute opioids are not fully understood. Increasing evidence suggests that in addition to well-documented antinociceptive effects opioids also produce paradoxical hyperalgesic and excitatory effects on neurons. However, most studies focus on the pronociceptive actions of chronic opioid exposure. Matrix metalloproteinase 9 (MMP-9) plays an important role in neuroinflammation and neuropathic pain development. We examined MMP-9 expression and localization in dorsal root ganglia (DRGs) after acute morphine treatment and, furthermore, the role of MMP-9 in modulating acute morphine-induced analgesia and hyperalgesia in mice. RESULTS: Subcutaneous morphine induced a marked up-regulation of MMP-9 protein in DRGs but not spinal cords. Morphine also increased MMP-9 activity and mRNA expression in DRGs. MMP-9 up-regulation peaked at 2 h but returned to the baseline after 24 h. In DRG tissue sections, MMP-9 is expressed in small and medium-sized neurons that co-express mu opioid receptors (MOR). In DRG cultures, MOR agonists morphine, DAMGO, and remifentanil each increased MMP-9 expression in neurons, whereas the opioid receptor antagonist naloxone and the MOR-selective antagonist D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH2 (CTAP) suppressed morphine-induced MMP-9 expression. Notably, subcutaneous morphine-induced analgesia was enhanced and prolonged in Mmp9 knockout mice and also potentiated in wild-type mice receiving intrathecal injection of MMP-9 inhibitors. Consistently, intrathecal injection of specific siRNA targeting MMP-9 reduced MMP-9 expression in DRGs and enhanced and prolonged morphine analgesia. Subcutaneous morphine also produced heat hyperalgesia at 24 h, but this opioid-induced hyperalgesia was not enhanced after MMP-9 deletion or inhibition. CONCLUSIONS: Transient MMP-9 up-regulation in DRG neurons can mask opioid analgesia, without modulating opioid-induced hyperalgesia. Distinct molecular mechanisms (MMP-9 dependent and independent) control acute opioid-induced pronociceptive actions (anti-analgesia in the first several hours and hyperalgesia after 24 h). Targeting MMP-9 may improve acute opioid analgesia.


Assuntos
Analgésicos Opioides/farmacologia , Metaloproteinase 9 da Matriz/metabolismo , Morfina/farmacologia , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/enzimologia , Animais , Western Blotting , Imuno-Histoquímica , Interleucina-1beta/metabolismo , Masculino , Camundongos , Reação em Cadeia da Polimerase em Tempo Real
18.
PLoS One ; 17(1): e0262074, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35061744

RESUMO

INTRODUCTION: Previous studies have demonstrated that cytokines, transforming growth factor (TGF-ß1), and brain-derived neurotrophic factor (BDNF) can impact the intensity of pain in rodents. However, the roles of cytokines, TGF-ß1 and BDNF in humans with chronic pain in osteoarthritis remains unclear, and no comparison between plasma and central cerebral spinal fluid (CSF) has been conducted. METHODS: Patients with osteoarthritis who were scheduled to receive spinal anesthesia were enrolled. The intensity of pain was evaluated with a visual analogue scale (VAS). In addition, patients with genitourinary system (GU) diseases and without obvious pain (VAS 0-1) were included as a comparison (control) group. The levels of TGF-ß1, BDNF, tumor necrosis factor-α (TNF-α), and interleukin (IL)-8 within the CSF and plasma were collected and evaluated before surgery. RESULTS: The plasma and CSF TGF-ß1 levels were significantly lower in the osteoarthritis patients with pain (VAS ≥ 3) than in the GU control patients. Downregulation of plasma BDNF was also found in osteoarthritis patients with pain. The Spearman correlation analysis showed that the VAS pain scores were significantly negatively correlated with the levels of TGF-ß1 in the CSF of patients with osteoarthritis. However, there was no significant correlations between the pain scores and the levels of BDNF, TNF-α, and IL-8 in either the CSF or plasma. CONCLUSIONS: TGF-ß1 but not BDNF, TNF-α, or IL-8 may be an important biological indicator in the CSF of osteoarthritis patients with chronic pain.


Assuntos
Biomarcadores/análise , Dor Crônica/patologia , Osteoartrite/patologia , Fator de Crescimento Transformador beta1/sangue , Idoso , Biomarcadores/sangue , Biomarcadores/líquido cefalorraquidiano , Fator Neurotrófico Derivado do Encéfalo/sangue , Fator Neurotrófico Derivado do Encéfalo/líquido cefalorraquidiano , Dor Crônica/complicações , Feminino , Humanos , Interleucina-8/sangue , Interleucina-8/líquido cefalorraquidiano , Masculino , Pessoa de Meia-Idade , Osteoartrite/complicações , Índice de Gravidade de Doença , Fator de Crescimento Transformador beta1/líquido cefalorraquidiano , Doenças Urogenitais/complicações , Doenças Urogenitais/patologia
19.
Pharmaceutics ; 14(2)2022 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-35214098

RESUMO

Our previous studies have revealed the ultrasmall superparamagnetic iron oxide in the amine group USPIO-101 has an analgesic effect on inflammatory pain. Here, we further investigated its effect on the spinal cord and brain via electrophysiological and molecular methods. We used a mouse inflammatory pain model, induced by complete Freund's adjuvant (CFA), and measured pain thresholds via von Frey methods. We also investigated the effects of USPIO-101 via an extracellular electrophysiological recording at the spinal dorsal horn synapses and hippocampal Schaffer collateral-CA1 synapses, respectively. The mRNA expression of pro-inflammatory cytokines was detected by quantitative real-time polymerase chain reaction (RT-qPCR). Our results showed intrathecal USPIO-101 produces similar analgesic behavior in mice with chronic inflammatory pain via intrathecal or intraplantar administration. The potentiated low-frequency stimulation-induced spinal cord long-term potentiation (LTP) at the spinal cord superficial dorsal horn synapses could decrease via USPIO-101 in mice with chronic inflammatory pain. However, the mRNA expression of cyclooxygenase-2 was enhanced with lipopolysaccharide (LPS) stimulation in microglial cells, and we also found USPIO-101 at 30 µg/mL could decrease the magnitude of hippocampal LTP. These findings revealed that intrathecal USPIO-101 presented an analgesia effect at the spinal cord level, but had neurotoxicity risk at higher doses.

20.
Cells ; 11(2)2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-35053411

RESUMO

Epidemiologic studies have indicated that dyslipidemia may facilitate the progression of neuronal degeneration. However, the effects of chronic dyslipidemia on brain function, especially in older individuals, remain unclear. In this study, middle-aged 37-week-old male Wistar-Kyoto rats were fed a normal diet (ND) or a 45% high-fat diet (HFD) for 30 weeks (i.e., until 67 weeks of age). To study the effects of chronic dyslipidemia on the brain, we analyzed spontaneous locomotor activity, cognitive function, and brain tissues in both groups of rats after 30 weeks. Compared with age-matched rats fed a ND, Wistar-Kyoto rats fed a HFD had dyslipidemia and showed decreased movement but normal recognition of a novel object. In our brain analyses, we observed a significant decrease in astrocytes and tyrosine hydroxylase-containing neurons in the substantia nigra and locus coeruleus of rats fed a HFD compared with rats fed a ND. However, hippocampal pyramidal neurons were not affected. Our findings indicate that the long-term consumption of a HFD may cause lipid metabolism overload in the brain and damage to glial cells. The decrease in astrocytes may lead to reduced protection of the brain and affect the survival of tyrosine hydroxylase-containing neurons but not pyramidal neurons of the hippocampus.


Assuntos
Envelhecimento/patologia , Encéfalo/patologia , Dieta Hiperlipídica , Comportamento Alimentar , Neuroglia/patologia , Neurônios/patologia , Tirosina 3-Mono-Oxigenase/metabolismo , Animais , Astrócitos/patologia , Cognição , Neurônios Dopaminérgicos/patologia , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/patologia , Locus Cerúleo/metabolismo , Microglia/patologia , Atividade Motora , Norepinefrina/metabolismo , Células Piramidais/patologia , Ratos Endogâmicos WKY , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA