Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Pflugers Arch ; 476(1): 101-110, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37770586

RESUMEN

Acid-sensing ion channels (ASICs) in dorsal root ganglion (DRG) neurons play an important role in inflammatory pain. The objective of this study is to observe the regulatory role of ASICs in monosodium urate (MSU) crystal-induced gout pain and explore the basis for ASICs in DRG neurons as a target for gout pain treatment. The gout arthritis model was induced by injecting MSU crystals into the ankle joint of mice. The circumference of the ankle joint was used to evaluate the degree of swelling; the von Frey filaments were used to determine the withdrawal threshold of the paw. ASIC currents and action potentials (APs) were recorded by patch clamp technique in DRG neurons. The results displayed that injecting MSU crystals caused ankle edema and mechanical hyperalgesia of the paw, which was relieved after amiloride treatment. The ASIC currents in DRG neurons were increased to a peak on the second day after injecting MSU crystals, which were decreased after amiloride treatment. MSU treatment increased the current density of ASICs in different diameter DRG cells. MSU treatment does not change the characteristics of AP. The results suggest that ASICs in DRG neurons participate in MSU crystal-induced gout pain.


Asunto(s)
Gota , Ácido Úrico , Ratones , Animales , Ácido Úrico/farmacología , Canales Iónicos Sensibles al Ácido , Amilorida , Gota/inducido químicamente , Dolor
2.
Circ Res ; 130(9): 1306-1317, 2022 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-35317607

RESUMEN

BACKGROUND: Transcriptional remodeling is known to contribute to heart failure (HF). Targeting stress-dependent gene expression mechanisms may represent a clinically relevant gene therapy option. We recently uncovered a salutary mechanism in the heart whereby JP2 (junctophilin-2), an essential component of the excitation-contraction coupling apparatus, is site-specifically cleaved and releases an N-terminal fragment (JP2NT [N-terminal fragment of JP2]) that translocates into the nucleus and functions as a transcriptional repressor of HF-related genes. This study aims to determine whether JP2NT can be leveraged by gene therapy techniques for attenuating HF progression in a preclinical pressure overload model. METHODS: We intraventricularly injected adeno-associated virus (AAV) (2/9) vectors expressing eGFP (enhanced green fluorescent protein), JP2NT, or DNA-binding deficient JP2NT (JP2NTΔbNLS/ARR) into neonatal mice and induced cardiac stress by transaortic constriction (TAC) 9 weeks later. We also treated mice with established moderate HF from TAC stress with either AAV-JP2NT or AAV-eGFP. RNA-sequencing analysis was used to reveal changes in hypertrophic and HF-related gene transcription by JP2NT gene therapy after TAC. Echocardiography, confocal imaging, and histology were performed to evaluate heart function and pathological myocardial remodeling following stress. RESULTS: Mice preinjected with AAV-JP2NT exhibited ameliorated cardiac remodeling following TAC. The JP2NT DNA-binding domain is required for cardioprotection as its deletion within the AAV-JP2NT vector prevented improvement in TAC-induced cardiac dysfunction. Functional and histological data suggest that JP2NT gene therapy after the onset of cardiac dysfunction is effective at slowing the progression of HF. RNA-sequencing analysis further revealed a broad reversal of hypertrophic and HF-related gene transcription by JP2NT overexpression after TAC. CONCLUSIONS: Our prevention- and intervention-based approaches here demonstrated that AAV-mediated delivery of JP2NT into the myocardium can attenuate stress-induced transcriptional remodeling and the development of HF when administered either before or after cardiac stress initiation. Our data indicate that JP2NT gene therapy holds great potential as a novel therapeutic for treating hypertrophy and HF.


Asunto(s)
Insuficiencia Cardíaca , Animales , ADN , Dependovirus , Modelos Animales de Enfermedad , Terapia Genética , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/terapia , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , ARN , Remodelación Ventricular
3.
Inflammopharmacology ; 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38829504

RESUMEN

OBJECTIVE: To investigate whether honokiol (HNK) acted as an analgesic in connection with inhibiting the voltage-gated proton channel (Hv1). METHODS: The model of gouty arthritis was induced by injecting monosodium urate (MSU) crystals into the hind ankle joint of mice. HNK was given by intragastric administration. Ankle swelling degree and mechanical allodynia were evaluated using ankle joint circumference measurement and von Frey filaments, respectively. Hv1 current, tail current, and action potential in dorsal root ganglion (DRG) neurons were recorded with patch-clamp techniques. RESULTS: HNK (10, 20, 40 mg/kg) alleviated inflammatory response and mechanical allodynia in a dose-dependent manner. In normal DRG neurons, 50 µM Zn2+ or 2-GBI significantly inhibited the Hv1 current and the current density of Hv1 increased with increasing pH gradient. The amplitude of Hv1 current significantly increased on the 3rd after MSU treatment, and HNK dose-dependently reversed the upregulation of Hv1 current. Compared with MSU group, 40 mg/kg HNK shifted the activation curve to the direction of more positive voltage and increased reversal potential to the normal level. In addition, 40 mg/kg HNK reversed the down-regulation of tail current deactivation time constant (τtail) but did not alter the neuronal excitability of DRG neurons in gouty mice. CONCLUSION: HNK may be a potential analgesic by inhibiting Hv1 current.

4.
Inflamm Res ; 71(4): 485-495, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35298670

RESUMEN

OBJECTIVE: The transient receptor potential vanilloid subtype 1 (TRPV1) channel is considered to play an important regulatory role in the process of pain. The purpose of this study is to observe the change characteristics of TRPV1 channel in MSU-induced gouty arthritis and to find a new target for clinical treatment of gout pain. METHODS: Acute gouty arthritis was induced by injection of monosodium urate (MSU) crystals into the ankle joint of mice. The swelling degree was evaluated by measuring the circumference of the ankle joint. Mechanical hyperalgesia was conducted using the electronic von Frey. Calcium fluorescence and TRPV1 current were recorded by applying laser scanning confocal microscope and patch clamp in dorsal root ganglion (DRG) neurons, respectively. RESULTS: MSU treatment resulted in significant inflammatory response and mechanical hyperalgesia. The peak swelling degree appeared at 12 h, and the minimum pain threshold appeared at 8 h after MSU treatment. The fluorescence intensity of capsaicin-induced calcium response and TRPV1 current were increased in DRG cells from MSU-treated mice. The number of cells that increased calcium response after MSU treatment was mainly distributed in small-diameter DRG cells. However, the action potential was not significantly changed in small-diameter DRG cells after MSU treatment. CONCLUSIONS: These findings identified an important role of TRPV1 in mediating mechanical hyperalgesia in MSU-induced gouty arthritis and further suggest that TRPV1 can be regarded as a potential new target for the clinical treatment of gouty arthritis.


Asunto(s)
Artritis Gotosa , Canales de Potencial de Receptor Transitorio , Animales , Artritis Gotosa/inducido químicamente , Artritis Gotosa/tratamiento farmacológico , Calcio , Edema , Hiperalgesia/inducido químicamente , Ratones , Dolor , Canales Catiónicos TRPV , Canales de Potencial de Receptor Transitorio/uso terapéutico , Ácido Úrico
5.
Clin Exp Pharmacol Physiol ; 48(3): 347-354, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33064853

RESUMEN

The voltage-gated sodium channel (VGSC) currents in dorsal root ganglion (DRG) neurons contain mainly TTX-sensitive (TTX-S) and TTX-resistant (TTX-R) Na+ currents. Magnolol (Mag), a hydroxylated biphenyl compound isolated from the bark of Magnolia officinalis, has been well documented to exhibit analgesic effects, but its mechanism is not yet fully understood. The aim of the present study was to investigate whether the antinociceptive effects of Mag is through inhibition of Na+ currents. Na+ currents in freshly isolated mouse DRG neurons were recorded with the whole cell patch clamp technique. Results showed that Mag inhibited TTX-S and TTX-R Na+ currents in a concentration-dependent manner. The IC50 values for block of TTX-S and TTX-R Na+ currents were 9.4 and 7.0 µmol/L, respectively. Therefore, TTX-R Na+ current was more susceptible to Mag than TTX-S Na+ current. For TTX-S Na+ channel, 10 µmol/L Mag shifted the steady state inactivation curve toward more negative by 9.8 mV, without affecting the activation curve. For TTX-R Na+ channel, 7 µmol/L Mag shifted the steady state activation and inactivation curves toward more positive and negative potentials by 6.5 and 11.7 mV, respectively. In addition, Mag significantly postponed recovery of TTX-S and TTX-R Na+ currents from inactivation, and produced frequency dependent blocks of both subtypes of Na+ currents. These results suggest that the inhibitory effects of Mag on Na+ channels may contribute to its analgesic effect.


Asunto(s)
Compuestos de Bifenilo , Ganglios Espinales , Lignanos , Sodio , Técnicas de Placa-Clamp , Tetrodotoxina
6.
Inflammopharmacology ; 29(3): 869-877, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34021831

RESUMEN

Voltage-gated sodium channels are currently recognized as one of the targets of analgesics. Magnolol (Mag), an active component isolated from Magnolia officinalis, has been reported to exhibit analgesic effects. The objective of this study was to investigate whether the analgesic effect of Mag was associated with blocking Na+ channels. Inflammatory pain was induced by the injection of carrageenan into the hind paw of mice. Mag was administered orally. Mechanical hyperanalgesia was evaluated by using von Frey filaments. Na+ currents and neuronal excitability in acutely isolated mouse dorsal root ganglion (DRG) neurons were recorded with the whole-cell patch clamp technique. Results showed that Mag (10 ~ 40 mg/kg) dose-dependently inhibited the paw edema and reduced mechanical pain in the inflammatory animal model. Injection of carrageenan significantly increased the amplitudes of TTX-sensitive and TTX-resistant Na+ currents. Compared with the carrageenan group, Mag inhibited the upregulation of two types of Na+ currents induced by carrageenan in a dose-dependent manner. Mag 40 mg/kg shifted the inactivation curves of two types of Na+ currents to hyperpolarization and returned to normal animal level without changing their activation curves. Mag 40 mg/kg significantly reduced the percentage of cells firing multiple spikes and inhibited the neuronal hyperexcitability induced by carrageenan. Our data suggest that the analgesic effect of Mag may be associated with a decreased neuronal excitability by blocking Na+ current.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Compuestos de Bifenilo/uso terapéutico , Ganglios Espinales/efectos de los fármacos , Lignanos/uso terapéutico , Neuronas/efectos de los fármacos , Dolor/tratamiento farmacológico , Bloqueadores de los Canales de Sodio/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/farmacología , Compuestos de Bifenilo/farmacología , Carragenina/toxicidad , Células Cultivadas , Relación Dosis-Respuesta a Droga , Edema/inducido químicamente , Edema/tratamiento farmacológico , Edema/metabolismo , Ganglios Espinales/fisiología , Lignanos/farmacología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Neuronas/fisiología , Dolor/fisiopatología , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/fisiología
7.
Inflamm Res ; 69(6): 589-598, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32306120

RESUMEN

OBJECTIVE: To evaluate the role of K+ channels in pain following gouty arthritis. METHODS: The model of acute gouty arthritis was induced by monosodium urate (MSU) in mice. The swelling degree was determined by measuring the circumference of the ankle joint. Mechanical hyperalgesia was detected by von Frey filaments. Two types of K+ currents, A-type currents (IA) and delayed rectifier currents (IK), were recorded in dorsal root ganglion (DRG) neurons using patch-clamp techniques. RESULTS: The swelling degree reached its maximum at 10 h and the minimum pain threshold was maintained between 8 and 48 h after MSU treatment in mice. The amplitudes of IA and IK in DRG neurons were moderately increased on day 1 after MSU treatment, and then, they were gradually decreased with times and reached their minimums on day 4 (for IA) or 5 (for IK). Compared with control group, the activation curve of IA was significantly shifted to more positive potential and the recovery time of IA from inactivation was markedly prolonged, but inactivation and frequency dependence of IA appeared unaffected in MSU-treated group. Additionally, no change was observed in the activation curve of IK after MSU treatment. The excitability was significantly higher in the MSU group than in the control group. CONCLUSIONS: MSU-induced gout pain may be related to the hyperexcitability of DRG neurons elicited by decreasing K+ currents.


Asunto(s)
Artritis Gotosa/fisiopatología , Dolor/fisiopatología , Canales de Potasio con Entrada de Voltaje/fisiología , Animales , Artritis Gotosa/inducido químicamente , Ganglios Espinales/fisiología , Masculino , Ratones Endogámicos ICR , Neuronas/fisiología , Dolor/inducido químicamente , Ácido Úrico
8.
Proc Natl Acad Sci U S A ; 114(21): E4288-E4295, 2017 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-28490495

RESUMEN

Alternative splicing changes the CaV1.2 calcium channel electrophysiological property, but the in vivo significance of such altered channel function is lacking. Structure-function studies of heterologously expressed CaV1.2 channels could not recapitulate channel function in the native milieu of the cardiomyocyte. To address this gap in knowledge, we investigated the role of alternative exon 33 of the CaV1.2 calcium channel in heart function. Exclusion of exon 33 in CaV1.2 channels has been reported to shift the activation potential -10.4 mV to the hyperpolarized direction, and increased expression of CaV1.2Δ33 channels was observed in rat myocardial infarcted hearts. However, how a change in CaV1.2 channel electrophysiological property, due to alternative splicing, might affect cardiac function in vivo is unknown. To address these questions, we generated mCacna1c exon 33-/--null mice. These mice contained CaV1.2Δ33 channels with a gain-of-function that included conduction of larger currents that reflects a shift in voltage dependence and a modest increase in single-channel open probability. This altered channel property underscored the development of ventricular arrhythmia, which is reflected in significantly more deaths of exon 33-/- mice from ß-adrenergic stimulation. In vivo telemetric recordings also confirmed increased frequencies in premature ventricular contractions, tachycardia, and lengthened QT interval. Taken together, the significant decrease or absence of exon 33-containing CaV1.2 channels is potentially proarrhythmic in the heart. Of clinical relevance, human ischemic and dilated cardiomyopathy hearts showed increased inclusion of exon 33. However, the possible role that inclusion of exon 33 in CaV1.2 channels may play in the pathogenesis of human heart failure remains unclear.


Asunto(s)
Potenciales de Acción/genética , Canales de Calcio Tipo L/genética , Síndrome de QT Prolongado/genética , Taquicardia/genética , Complejos Prematuros Ventriculares/genética , Potenciales de Acción/fisiología , Empalme Alternativo/genética , Animales , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/metabolismo , Células Cultivadas , Colforsina/farmacología , Fenómenos Electrofisiológicos/genética , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Isoproterenol/farmacología , Síndrome de QT Prolongado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Nifedipino/farmacología , Ratas , Eliminación de Secuencia/genética , Taquicardia/patología , Complejos Prematuros Ventriculares/patología
9.
J Neuroinflammation ; 16(1): 83, 2019 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-30975172

RESUMEN

BACKGROUND: Neuropathic pain is a serious clinical problem that needs to be solved urgently. ASK1 is an upstream protein of p38 and JNK which plays important roles in neuroinflammation during the induction and maintenance of chronic pain. Therefore, inhibition of ASK1 may be a novel therapeutic approach for neuropathic pain. Here, we aim to investigate the effects of paeoniflorin on ASK1 and neuropathic pain. METHODS: The mechanical and thermal thresholds of rats were measured using the Von Frey test. Cell signaling was assayed using western blotting and immunohistochemistry. RESULTS: Chronic constrictive injury (CCI) surgery successfully decreased the mechanical and thermal thresholds of rats and decreased the phosphorylation of ASK1 in the rat spinal cord. ASK1 inhibitor NQDI1 attenuated neuropathic pain and decreased the expression of p-p38 and p-JNK. Paeoniflorin mimicked ASK1 inhibitor NQDI1 and inhibited ASK1 phosphorylation. Paeoniflorin decreased the expression of p-p38 and p-JNK, delayed the progress of neuropathic pain, and attenuated neuropathic pain. Paeoniflorin reduced the response of astrocytes and microglia to injury, decreased the expression of IL-1ß and TNF-α, and downregulated the expression of CGRP induced by CCI. CONCLUSIONS: Paeoniflorin is an effective drug for the treatment of neuropathic pain in rats via inhibiting the phosphorylation of ASK1, suggesting it may be effective in patients with neuropathic pain.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Apoptosis/efectos de los fármacos , Encefalitis/tratamiento farmacológico , Glucósidos/uso terapéutico , MAP Quinasa Quinasa Quinasa 5/metabolismo , Monoterpenos/uso terapéutico , Neuropatía Ciática/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Encefalitis/complicaciones , Hidroxiquinolinas/uso terapéutico , Hiperalgesia/fisiopatología , Interleucina-1beta/metabolismo , Masculino , Umbral del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Neuropatía Ciática/complicaciones
10.
J Membr Biol ; 251(4): 573-579, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29594433

RESUMEN

Voltage-gated K+ (KV) currents play a crucial role in regulating pain by controlling neuronal excitability, and are divided into transient A-type currents (IA) and delayed rectifier currents (IK). The dorsal root ganglion (DRG) neurons are heterogeneous and the subtypes of KV currents display different levels in distinct cell sizes. To observe correlations of the subtypes of KV currents with DRG cell sizes, KV currents were recorded by whole-cell patch clamp in freshly isolated mouse DRG neurons. Results showed that IA occupied a high proportion in KV currents in medium- and large-diameter DRG neurons, whereas IK possessed a larger proportion of KV currents in small-diameter DRG neurons. A lower correlation was found between the proportion of IA or IK in KV currents and cell sizes. These data suggest that IA channels are mainly expressed in medium and large cells and IK channels are predominantly expressed in small cells.


Asunto(s)
Ganglios Espinales/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Animales , Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología , Ratones , Técnicas de Placa-Clamp
11.
Cytokine ; 108: 151-159, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29609137

RESUMEN

Therapeutic resistance leading to tumor relapse is a major challenge in breast cancer (BCa) treatment. Numerous factors involved in multiple mechanisms promote the development of tumor chemo/radio-resistance. Cytokines/chemokines are important inflammatory factors and highly related to tumorigenesis, metastasis and tumors responses to treatment. A large number of studies have demonstrated that the network of cytokines activates multiple cell signaling pathways to promote tumor cell survival, proliferation, invasion, and migration. Particularly in BCa, cytokines-enhanced the epithelial-mesenchymal transition (EMT) process plays a pivotal role in the progression of metastatic phenotypes and resistance to the traditional chemo/radio-therapy. Virtually, therapeutic resistance is not entirely determined by tumor cell intrinsic characteristics but also dependent upon synchronized effects by numerous of local microenvironmental factors. Emerging evidence highlighted that exosomes secreted from various types of cells promote intercellular communication by transferring bioactive molecules including miRNAs and cytokines, suggesting that exosomes are essential for sustentation of tumor progression and therapeutic resistance within the tumor microenvironment. In this review, we discuss the mechanisms by which cytokines promote therapeutic resistance of BCa and suggest a potential approach for improving BCa therapeutics by inhibition of exosome function.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Citocinas/inmunología , Resistencia a Antineoplásicos , Exosomas/inmunología , Microambiente Tumoral/inmunología , Animales , Neoplasias de la Mama/inmunología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Femenino , Humanos , Ratones , MicroARNs , Recurrencia Local de Neoplasia/inmunología , Transducción de Señal
12.
Brain Behav Immun ; 68: 111-122, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29017971

RESUMEN

Postoperative pain is a common form of acute pain that, if not managed effectively, can become chronic pain. Evidence has shown that glia, especially microglia, mediate neuroinflammation, which plays a vital role in pain sensitization. Moreover, toll-like receptor 4 (TLR4), the tumor necrosis factor receptor (TNF-R), the interleukin-1 receptor (IL-1R), and the interleukin-6 receptor (IL-6R) have been considered key components in central pain sensitization and neuroinflammation. Therefore, we hypothesized that activation of the body's endogenous "immune brakes" will inhibit these receptors and achieve inflammation tolerance as well as relieve postoperative pain. After searching for potential candidates to serve as this immune brake, we identified and focused on the suppressor of cytokine signaling 3 (SOCS3) gene. To regulate SOCS3 expression, we used paeoniflorin to induce heat shock protein 70 (HSP70)/TLR4 signaling. We found that paeoniflorin significantly induced SOCS3 expression both in vitro and in vivo and promoted the efflux of HSP70 from the cytoplasm to the extracellular environment. Furthermore, paeoniflorin markedly attenuated incision-induced mechanical allodynia, and this effect was abolished by small interfering RNAs targeting SOCS3. These findings demonstrated an effective and safe strategy to alleviate postoperative pain.


Asunto(s)
Dolor Postoperatorio/inmunología , Dolor Postoperatorio/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/fisiología , Animales , Citocinas/metabolismo , Tolerancia a Medicamentos/fisiología , Glucósidos/farmacología , Proteínas HSP70 de Choque Térmico/metabolismo , Hiperalgesia/metabolismo , Inflamación/metabolismo , Ratones , Microglía/fisiología , Monoterpenos/farmacología , Neuralgia/metabolismo , Neuroglía/fisiología , Neuroinmunomodulación/fisiología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Receptor Toll-Like 4/metabolismo
13.
Neurochem Res ; 43(2): 450-457, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29177805

RESUMEN

Voltage-gated potassium (KV) currents, subdivided into rapidly inactivating A-type currents (I A) and slowly inactivating delayed rectifier currents (I K), play a fundamental role in modulating pain by controlling neuronal excitability. The effects of Honokiol (Hon), a natural biphenolic compound derived from Magnolia officinalis, on KV currents were investigated in freshly isolated mouse dorsal root ganglion neurons using the whole-cell patch clamp technique. Results showed that Hon inhibited I A and I K in concentration-dependent manner. The IC50 values for block of I A and I K were 30.5 and 25.7 µM, respectively. Hon (30 µM) shifted the steady-state activation curves of I A and I K to positive potentials by 17.6 and 16.7 mV, whereas inactivation and recovery from the inactivated state of I A were unaffected. These results suggest that Hon preferentially interacts with the active states of the I A and I K channels, and has no effect on the resting state and inactivated state of the I A channel. Blockade on K+ channels by Hon may contribute to its antinociceptive effect, especially anti-inflammatory pain.


Asunto(s)
Compuestos de Bifenilo/farmacología , Ganglios Espinales/citología , Activación del Canal Iónico/efectos de los fármacos , Lignanos/farmacología , Neuronas/efectos de los fármacos , Canales de Potasio/efectos de los fármacos , Animales , Compuestos de Bifenilo/química , Células Cultivadas , Ganglios Espinales/efectos de los fármacos , Lignanos/química , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Neuronas/metabolismo , Técnicas de Placa-Clamp/métodos , Potasio/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo
14.
Biochem Biophys Res Commun ; 490(4): 1189-1196, 2017 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-28669730

RESUMEN

By analyzing The Cancer Genome Atlas (TCGA) datasets, we discovered that the zinc finger protein 692 (ZNF692) were over-expressed in Lung adenocarcinoma (LUAD) tissues compared to adjacent non-tumor tissues (P < 0.0001). In this study, we investigated the function of ZNF692 in the progression of LUAD. We found that ZNF692 knockdown inhibited LUAD cells proliferation, migration, and invasion both in vitro and in vivo. And LUAD cell apoptosis was induced following the down-regulation of ZNF692. Our results show that ZNF692 is over-expressed in LUAD tissues compared to adjacent normal tissues, and hyper-expression of ZNF692 in LUAD is an independent risk factor for worse overall survival in LUAD patients (HR: 8.800, 95%CI: 1.082-71.560, P = 0.042) by Tissue Microarray stain assay (TMA). GO analysis indicated that most genes were enriched in metabolic process which were associated highly with ZNF692 levels. Collectively, our results suggested that ZNF692 may serve as a potential oncogene and biomarker in LUAD by influencing cell metabolism.


Asunto(s)
Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Movimiento Celular , Proteínas de Unión al ADN/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Factores de Transcripción/metabolismo , Adenocarcinoma del Pulmón , Proliferación Celular , Humanos , Células Tumorales Cultivadas
15.
J Neuroinflammation ; 14(1): 211, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-29096659

RESUMEN

BACKGROUND: Morphine tolerance is a clinical challenge, and its pathogenesis is closely related to the neuroinflammation mediated by Toll-like receptor 4 (TLR4). In Chinese pain clinic, lidocaine is combined with morphine to treat chronic pain. We found that lidocaine sufficiently inhibited neuroinflammation induced by morphine and improved analgesic tolerance on the basis of non-affecting pain threshold. METHODS: CD-1 mice were utilized for tail-flick test to evaluate morphine tolerance. The microglial cell line BV-2 was utilized to investigate the mechanism of lidocaine. Neuroinflammation-related cytokines were measured by western blotting and real-time PCR. The level of suppressor of cytokine signaling 3 (SOCS3) and adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK)-related signaling pathway was evaluated by western blotting, real-time PCR, enzyme-linked immunosorbent assay (ELISA), and immunofluorescence staining. RESULTS: Lidocaine potentiated an anti-nociceptive effect of morphine and attenuated the chronic analgesic tolerance. Lidocaine suppressed morphine-induced activation of microglia and downregulated inflammatory cytokines, interleukin-1ß (IL-1ß), and tumor necrosis factor-alpha (TNF-α) via upregulating SOCS3 by activating AMPK. Lidocaine enhanced AMPK phosphorylation in a calcium-dependent protein kinase kinase ß (CaMKKß)-dependent manner. Furthermore, lidocaine decreased the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and inhibited the nuclear factor-κB (NF-κB) in accordance with the inhibitory effects to TLR4. CONCLUSIONS: Lidocaine as a prevalent local anesthetic suppresses morphine tolerance efficiently. AMPK-dependent upregulation of SOCS3 by lidocaine plays a crucial role in the improvement of analgesic tolerance.


Asunto(s)
Mediadores de Inflamación/antagonistas & inhibidores , Lidocaína/administración & dosificación , Morfina/administración & dosificación , Proteínas Quinasas/biosíntesis , Médula Espinal/efectos de los fármacos , Proteína 3 Supresora de la Señalización de Citocinas/biosíntesis , Quinasas de la Proteína-Quinasa Activada por el AMP , Analgésicos Opioides/administración & dosificación , Anestésicos Locales/administración & dosificación , Animales , Línea Celular , Quimioterapia Combinada , Tolerancia a Medicamentos/fisiología , Inflamación/metabolismo , Inflamación/patología , Inflamación/prevención & control , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Médula Espinal/metabolismo , Médula Espinal/patología
16.
Anesthesiology ; 123(5): 1170-85, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26378398

RESUMEN

BACKGROUND: Activation of adenosine monophosphate-activated kinase (AMPK) has been associated with the inhibition of inflammatory nociception and the attenuation of morphine antinociceptive tolerance. In this study, the authors investigated the impact of AMPK activation through resveratrol treatment on bone cancer pain. METHODS: The nociception was assessed by measuring the incidence of foot withdrawal in response to mechanical indentation in rats (n = 8). Cytokine expression was measured using quantitative polymerase chain reaction (n = 8). Cell signalings were assayed by western blot (n = 4) and immunohistochemistry (n = 5). The microglial cell line BV-2, primary astrocytes, and neuron-like SH-SY5Y cells were cultured to investigate the in vitro effects. RESULTS: Resveratrol and 5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide, the AMPK activators, significantly attenuated bone cancer pain in rats with tumor cell implantation (TCI; threshold of mechanical withdrawal, resveratrol vs. vehicle: 10.1 ± 0.56 vs. 4.1 ± 0.37; 5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide vs. vehicle: 8.2 ± 0.17 vs. 4.1 ± 0.37, mean ± SEM); these effects were reversed by the AMPK inhibitor compound C (compound C vs. resveratrol: 6.2 ± 1.35 vs. 10.1 ± 0.56, mean ± SEM). Resveratrol has an AMPK-dependent inhibitory effect on TCI-evoked astrocyte and microglial activation. The antinociceptive effects of resveratrol were partially mediated by the reduced phosphorylation of mitogen-activated protein kinases and decreased production of proinflammatory cytokines in an AMPK-dependent manner. Furthermore, resveratrol potently inhibited inflammatory factors-mediated protein kinase B/mammalian target of rapamycin signaling in neurons. Acute pain evoked by proinflammatory cytokines in the spinal cord was significantly attenuated by resveratrol. CONCLUSIONS: AMPK activation in the spinal glia by resveratrol may have utility in the treatment of TCI-induced neuroinflammation, and our results further implicate AMPK as a novel target for the attenuation of bone cancer pain.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Neoplasias Óseas/enzimología , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Dolor/enzimología , Inhibidores de Proteínas Quinasas/uso terapéutico , Estilbenos/uso terapéutico , Animales , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología , Línea Celular Tumoral , Femenino , Humanos , Inflamación/enzimología , Inflamación/prevención & control , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Dolor/patología , Dolor/prevención & control , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Sprague-Dawley , Resveratrol , Estilbenos/farmacología
17.
Cytotherapy ; 15(11): 1395-405, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23992829

RESUMEN

BACKGROUND: Hydrogen sulfide (H2S) is an endogenously generated gaseous transmitter known for its cytoprotective effect mediated by the PI3K-Akt signaling pathway. Human induced pluripotent stem cell (hiPSC)-derived mesenchymal stromal cells (MSCs), or hiPSC-MSCs, represent an alternative source of MSCs for autologous cell therapy. The big-conductance Ca(2+)-activated outward K(+) currents (BKCa), known to mediate cell proliferation, have been detected in >80% of hiPSC-MSCs. The present study aimed to explore the effect of H2S on survival and proliferation of hiPSC-MSCs and investigate the mediatory role of BKCa. METHODS: Effects of H2S on proliferation and survival of hiPSC-MSCs were measured by 5-bromo-2-deoxyuridine incorporation, population doubling and cell cycle assays, and by 3-(4,5)-dimethylthiahiazo(-z-y1)-3,5-di-phenytetrazoliumromide assay and 4'-6-diamidino-2-phenylindole staining, respectively. BKCa was recorded by means of the whole-cell patch-clamp technique. The expressions of KCa 1.1 (encoding BKCa) and apoptosis-related genes were measured by reverse transcriptase-polymerase chain reaction. The phosphorylation of Akt was assessed by Western blot analysis. RESULTS: Exogenously administered NaHS (an H2S donor, 50-300 µmol/L) significantly promoted proliferation of hiPSC-MSCs. NaHS prevented the hypoxia-induced apoptosis and suppressed BKCa currents without altering the expression levels of α- and ß-KCa 1.1. In addition, NaHS increased the phosphorylation of Akt and decreased the expression of Caspase 8 and Bax in hiPSC-MSCs. Paxilline (1 µmol/L), a BKCa blocker, showed similar effects on promoting cell proliferation and phosphorylation of Akt and suppression of apoptotic genes in hiPSC-MSCs. CONCLUSIONS: Our data confirmed that H2S arguments the proliferation and survival of hiPSC-MSCs through activation of the PI3K-Akt pathway and that such effects could be mediated through inhibition of BKCa.


Asunto(s)
Sulfuro de Hidrógeno/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Células Madre Mesenquimatosas/efectos de los fármacos , Apoptosis/efectos de los fármacos , Caspasa 8/biosíntesis , Hipoxia de la Célula/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Indoles/farmacología , Canales de Potasio de Gran Conductancia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Técnicas de Placa-Clamp , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína X Asociada a bcl-2/biosíntesis
18.
Neurochem Res ; 36(6): 947-54, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21340731

RESUMEN

Scutellarin (Scu), the main bioactive component of Erilgeron breviscapus, protects the brain against ischemic damages. To explore the therapeutic mechanism of Scu, we investigated the impact of Scu on sodium current (I(Na)) of freshly isolated mouse hippocampal CA1 neurons using the whole-cell patch clamp technique. Results showed that Scu inhibited I(Na) in concentration- and holding potential-dependent manners. At 50 µM, Scu markedly shifted the steady state inactivation curve of I (Na) towards a more negative potential, slowed down the recovery of I(Na) from inactivation state, and elicited a frequency-dependent block of I (Na). The shape of the current-voltage (I-V) curve and the steady state activation curve of I(Na) were unaffected by Scu treatment. These findings suggest that Scu is capable of inhibiting I(Na) in neurons through predominantly affecting the inactivated state of I(Na). Inhibition of Na(+) channels provides a novel pharmacological basis for the anti-ischemic application of Scu.


Asunto(s)
Apigenina/farmacología , Glucuronatos/farmacología , Hipocampo/efectos de los fármacos , Neuronas/efectos de los fármacos , Canales de Sodio/efectos de los fármacos , Animales , Hipocampo/citología , Ratones , Neuronas/citología , Técnicas de Placa-Clamp
19.
Inflammation ; 44(4): 1405-1415, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33515125

RESUMEN

The aim of the present study was to observe the changes of TTX-R, Nav1.8, and Nav1.9 Na+ currents in MSU-induced gouty arthritis mice, and to explore the possibility of Nav1.8 and Nav1.9 channels as potential targets for gout pain treatment. Acute gouty arthritis was induced by monosodium urate (MSU) in mice. Swelling degree was evaluated by measuring the circumference of the ankle joint. Mechanical allodynia was assessed by applying the electronic von Frey. Na+ currents were recorded by patch-clamp techniques in acute isolated dorsal root ganglion (DRG) neurons. MSU treatment significantly increased the swelling degree of ankle joint and decreased the mechanical pain threshold. The amplitude of TTX-R Na+ current was significantly increased and reached its peak on the 4th day after injection of MSU. For TTX-R Na+ channel subunits, Nav1.8 current density was significantly increased, but Nav1.9 current density was markedly decreased after MSU treatment. MSU treatment shifted the steady-state activation curves of TTX-R Na+ channel, Nav1.8 and Nav1.9 channels, and the inactivation curves of TTX-R Na+ channel and Nav1.8 channels to the depolarizing direction, but did not affect the inactivation curve of Nav1.9 channel. Compared with the normal group, the recovery of Nav1.8 channel was faster, while that of Nav1.9 channel was slower. The recovery of TTX-R Na+ channel remained unchanged after MSU treatment. Additionally, MSU treatment increased DRG neurons excitability by reducing action potential threshold. Nav1.8 channel, not Nav1.9 channel, may be involved in MSU-induced gout pain by increasing nerve excitability.


Asunto(s)
Artritis Gotosa/inducido químicamente , Artritis Gotosa/metabolismo , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Canal de Sodio Activado por Voltaje NAV1.9/metabolismo , Ácido Úrico/toxicidad , Animales , Artritis Gotosa/patología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Hiperalgesia/patología , Masculino , Ratones , Ratones Endogámicos ICR , Bloqueadores de los Canales de Sodio/farmacología
20.
Mol Oncol ; 15(5): 1528-1542, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33508878

RESUMEN

Chemotherapeutic resistance is a major obstacle in the control of advanced breast cancer (BCa). We have previously shown that small extracellular vesicles (sEVs) can transmit adriamycin resistance between BCa cells. Here, we describe that sEV-mediated TGF-ß1 intercellular transfer is involved in the drug-resistant transmission. sEVs were isolated and characterized from both sensitive and resistant cells. sEVs derived from the resistant cells were incubated with the sensitive cells and resulted in transmitting the resistant phenotype to the recipient cells. Cytokine antibody microarray revealed that most metastasis-associated cytokines present at the high levels in sEVs from the resistant cells compared with their levels in sEVs from the sensitive cells, particularly TGF-ß1 is enriched in sEVs from the resistant cells. The sEV-mediated TGF-ß1 intercellular transfer led to increasing Smad2 phosphorylation and improving cell survival by suppressing apoptosis and enhancing cell mobility. Furthermore, sEV-mediated drug-resistant transmission by delivering TGF-ß1 was validated using a zebrafish xenograft tumor model. These results elaborated that sEV-mediated TGF-ß1 intercellular transfer contributes to adriamycin resistance in BCa.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos , Vesículas Extracelulares/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Antibióticos Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos/genética , Embrión no Mamífero , Vesículas Extracelulares/fisiología , Femenino , Humanos , Células MCF-7 , Comunicación Paracrina/genética , Factor de Crecimiento Transformador beta1/genética , Microambiente Tumoral/genética , Pez Cebra/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA