Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Physiol Investig ; 67(3): 103-106, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38857206

RESUMO

A recent study investigated the correlation between telmisartan (TEL) exposure and Alzheimer's disease (AD) risk among African Americans (AAs) and European Americans. Their findings indicated that moderate-to-high TEL exposure was linked to a decreased incidence of AD among AAs. These results suggest a potential association between TEL and a reduced risk of AD specifically within the AA population. Here, we investigated the effects of TEL, either alone or in combination with ranolazine (Ran) or dapagliflozin (Dapa), on voltage-gated Na + currents ( INa ) in Neuro-2a cells. TEL, primarily used for treating hypertension and cardiovascular disorders, showed a stimulatory effect on INa , while Ran and Dapa reversed this stimulation. In Neuro-2a cells, we demonstrated that with exposure to TEL, the transient ( INa(T) ) and late ( INa(L) ) components of INa were differentially stimulated with effective EC 50 's of 16.9 and 3.1 µM, respectively. The research implies that TEL's impact on INa might be associated with enhanced neuronal excitability. This study highlights the complex interplay between TEL, Ran, and Dapa on INa and their potential implications for AD, emphasizing the need for further investigation to understand the mechanisms involved.


Assuntos
Acetanilidas , Compostos Benzidrílicos , Benzimidazóis , Benzoatos , Glucosídeos , Neuroblastoma , Piperazinas , Ranolazina , Telmisartan , Telmisartan/farmacologia , Telmisartan/uso terapêutico , Glucosídeos/farmacologia , Glucosídeos/uso terapêutico , Benzimidazóis/farmacologia , Benzimidazóis/uso terapêutico , Ranolazina/farmacologia , Ranolazina/uso terapêutico , Benzoatos/farmacologia , Benzoatos/uso terapêutico , Neuroblastoma/tratamento farmacológico , Neuroblastoma/patologia , Linhagem Celular Tumoral , Animais , Acetanilidas/farmacologia , Piperazinas/farmacologia , Piperazinas/uso terapêutico , Camundongos , Compostos Benzidrílicos/farmacologia , Compostos Benzidrílicos/uso terapêutico , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos
2.
Eur J Pharmacol ; 971: 176518, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38556119

RESUMO

XAV-939(XAV) is a chemical compound that inhibits the activity of tankyrase. However, the precise way in which XAV alters membrane ionic currents is not well understood. In this study,our goal was to examine the impact of XAV on the ionic currents in mouse MA-10 Leydig cells, specifically focusing on the magnitude, gating properties,and voltage-dependent hysteresis of erg-mediated K+currents(IK(erg)). In our whole-cell current recordings we observed that the addition of XAV inhibited the density of IK(erg) in a concentration-dependent manner with an IC50 of 3.1 µM. Furthermore we found that continued exposure to XAV, further addition of neither liraglutide nor insulin-like growth factor-1 counteracted XAV-mediated inhibition of IK(erg). Additionally the presence of XAV suppressed the mean current versus voltage relationship of IK(erg) across the entire voltage-clamp step analyzed. This compound shifted the steady-state activation curve of IK(erg) to a less negative potential by approximately 12 mV. The presence of XAV increased the time constant of deactivating IK(erg) in MA-10 cells. The voltage-dependent clockwise hysteresis of IK(erg) responding to prolonged upright isosceles-triangular ramp voltage became diminished by adding XAV; moreover subsequent addition of NS3623 effectively reversed XAV-induced decrease of hysteretic area of IK(erg). XAV also inhibited the proliferation of this cell line and the IC50 value of XAV-induced inhibition of cell proliferation was 2.8M. Overall the suppression of IK(erg) by XAV may serve as a significant ionic mechanism that contribute to the functional properties of MA-10 cells. However, it is important to note that this effect cannot be attributed solely to the inhibition of tankyrase.


Assuntos
Compostos Heterocíclicos com 3 Anéis , Neoplasias , Tanquirases , Camundongos , Masculino , Animais , Linhagem Celular
4.
Cancer Med ; 12(3): 3260-3275, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36000705

RESUMO

Arsenic compounds have been applied treating acute promyelocytic 1eukemia and solid tumors with brief mechanism investigations. In fact, we have demonstrated that sodium arsenite plus dimethylarsenic acid could activate apoptosis in MA-10 mouse Leydig tumor cells by inducing caspase pathways. However, detail underlying mechanisms how caspase cascade is regulated remains elusive. Therefore, the apoptotic mechanism of sodium arsenite plus dimethylarsenic acid were examined in MA-10 cells in this study. Our results reveal that Fas/FasL protein expressions were stimulated by sodium arsenite plus dimethylarsenic acid in MA-10 cells. In addition, reactive oxygen species (ROS) generation, cytochrome C release, Bid truncation, and Bax translocation were induced in MA-10 cells by arsenic compounds. Moreover, activation of p38, JNK and ERK1/2, MAPK pathways was stimulated while Akt phosphorylated levels and Akt expression were decreased by sodium arsenite plus dimethylarsenic in MA-10 cells. In conclusion, sodium arsenite and dimethylarsenic acid did activate MAPK pathway plus ROS generation, but suppress Akt pathway, to modulate caspase pathway and then induce MA-10 cell apoptosis.


Assuntos
Arsenitos , Neoplasias , Animais , Camundongos , Proteínas Proto-Oncogênicas c-akt , Espécies Reativas de Oxigênio/metabolismo , Apoptose , Arsenitos/farmacologia , Caspases
5.
Diagnostics (Basel) ; 12(12)2022 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-36553170

RESUMO

In thoracic surgery, the double lumen endotracheal tube (DLT) is used for differential ventilation of the lung. DLT allows lung collapse on the surgical side that requires access to the thoracic and mediastinal areas. DLT placement for a given patient depends on two settings: a tube of the correct size (or 'size') and to the correct insertion depth (or 'depth'). Incorrect DLT placements cause oxygen desaturation or carbon dioxide retention in the patient, with possible surgical failure. No guideline on these settings is currently available for anesthesiologists, except for the aid by bronchoscopy. In this study, we aimed to predict DLT 'depths' and 'sizes' applied earlier on a group of patients (n = 231) using a computer modeling approach. First, for these patients we retrospectively determined the correlation coefficient (r) of each of the 17 body parameters against 'depth' and 'size'. Those parameters having r > 0.5 and that could be easily obtained or measured were selected. They were, for both DLT settings: (a) sex, (b) height, (c) tracheal diameter (measured from X-ray), and (d) weight. For 'size', a fifth parameter, (e) chest circumference was added. Based on these four or five parameters, we modeled the clinical DLT settings using a Support Vector Machine (SVM). After excluding statistical outliers (±2 SD), 83.5% of the subjects were left for 'depth' in the modeling, and similarly 85.3% for 'size'. SVM predicted 'depths' matched with their clinical values at a r of 0.91, and for 'sizes', at an r of 0.82. The less satisfactory result on 'size' prediction was likely due to the small target choices (n = 4) and the uneven data distribution. Furthermore, SVM outperformed other common models, such as linear regression. In conclusion, this first model for predicting the two DLT key settings gave satisfactory results. Findings would help anesthesiologists in applying DLT procedures more confidently in an evidence-based way.

6.
Life (Basel) ; 11(12)2021 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-34947927

RESUMO

Despite improvements in cancer treatments resulting in higher survival rates, the proliferation and metastasis of tumors still raise new questions in cancer therapy. Therefore, new drugs and strategies are still needed. Midazolam (MDZ) is a common sedative drug acting through the γ-aminobutyric acid receptor in the central nervous system and also binds to the peripheral benzodiazepine receptor (PBR) in peripheral tissues. Previous studies have shown that MDZ inhibits cancer cell proliferation but increases cancer cell apoptosis through different mechanisms. In this study, we investigated the possible anticancer mechanisms of MDZ on different cancer cell types. MDZ inhibited transforming growth factor ß (TGF-ß)-induced cancer cell proliferation of both A549 and MCF-7 cells. MDZ also inhibited TGF-ß-induced cell migration, invasion, epithelial-mesenchymal-transition, and Smad phosphorylation in both cancer cell lines. Inhibition of PBR by PK11195 rescued the MDZ-inhibited cell proliferation, suggesting that MDZ worked through PBR to inhibit TGF-ß pathway. Furthermore, MDZ inhibited proliferation, migration, invasion and levels of mesenchymal proteins in MDA-MD-231 triple-negative breast cancer cells. Together, MDZ inhibits cancer cell proliferation both in epithelial and mesenchymal types and EMT, indicating an important role for MDZ as a candidate to treat lung and breast cancers.

7.
Acta Biochim Biophys Sin (Shanghai) ; 53(2): 238-248, 2021 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-33410473

RESUMO

Naltrexone is widely used for alleviating opioid-related side effects in cancer patients. However, the effects of naltrexone on cancer progression are controversial in the literature. The present study was carried out to investigate the effects of successive treatment with clinically relevant doses of naltrexone on the malignant biological behaviors of bladder cancer cells. The human bladder cancer T24 cells and mouse bladder cancer MB49 cells were treated with naltrexone. Cell proliferation, migration, and invasion abilities were analyzed. Morphological changes of the cells were confirmed by F-actin immunofluorescence staining. Epithelial-mesenchymal transition (EMT)-related markers and transcriptional factors, as well as activation of the phosphatidylinositol 3 kinase (PI3K)/AKT signaling pathway, were analyzed. Results showed that, compared with the control group, successive treatment with naltrexone significantly promoted the proliferation and decreased the apoptosis of bladder cancer cells, together with increase in cell migration and invasion ability. Continuous treatment with naltrexone also significantly reduced the expression of epithelial markers (E-cadherin and cytokeratin 19), increased the expression of mesenchymal markers (N-cadherin and vimentin) and EMT-inducing transcription factors (Snail and Slug), and further shifted the morphological phenotype of bladder cancer cells to a mesenchymal phenotype. The PI3K/AKT signaling pathway was activated by successive treatment with naltrexone. Notably, incubation with the specific PI3K inhibitor LY294002 together with naltrexone reversed the naltrexone-induced EMT progression. In conclusion, successive treatment with naltrexone may be favorable for the progression of bladder tumors by activating the PI3K/AKT signaling pathway and inducing EMT. Long-term exposure to naltrexone should be used cautiously in patients with bladder cancer.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Naltrexona/farmacologia , Neoplasias da Bexiga Urinária/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia
8.
Int J Nanomedicine ; 15: 6961-6973, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33061362

RESUMO

BACKGROUND: Although graphene quantum dots (GQDs) have received considerable research attention for their applications in various fields, the use of GQDs, such as nitrogen-doped GQDs (N-GQDs) and amino-functionalized N-GQDs (amino-N-GQDs), as photosensitizers to facilitate photodynamic therapy (PDT) has received limited research intention. To address this research gap, this study prepared novel amino-N-GQDs and investigated their properties. METHODS: The amino-N-GQDs subjected to two-photon excitation (TPE) exhibited remarkable bactericidal capability in PDT. The bonding compositions of nitrogen and the amino-functionalized group played a critical role in their antimicrobial effects. RESULTS: Compared with amino-group-free N-GQDs and amino-N-free GQDs, the amino-N-GQDs generated a higher amount of reactive oxygen species, demonstrating their superior efficacy for two-photon PDT. Additionally, the intrinsic luminescence properties and high photostability of the amino-N-GQDs demonstrate their suitability as an effective two-photon contrast agent for tracking bacteria during two-photon biomedical imaging. CONCLUSION: The amino-N-GQD and their remarkable properties may provide an efficient alternative approach for observing and easily eliminating malignant microbes in the future.


Assuntos
Antibacterianos/farmacologia , Meios de Contraste/química , Nitrogênio/farmacologia , Fotoquimioterapia/métodos , Pontos Quânticos/química , Antibacterianos/química , Bacillus subtilis/efeitos dos fármacos , Grafite/química , Luminescência , Nitrogênio/química , Fótons , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia
9.
Oncol Rep ; 44(6): 2701-2714, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33125114

RESUMO

Arsenic is a well­documented environmental toxicant that can induce neurotoxicity and peripheral vascular diseases. In fact, arsenic trioxide has been used to treat various cancer types. Oral cancer has been in the top ten common cancers for decades in Taiwan, and the incidence rate is continuously increasing. The majority of oral cancers are associated with excessive tobacco, alcohol consumption and betel chewing. To the best of our knowledge, no study has revealed the effect of arsenic compounds on oral cancers. Thus, the present study used OEC­M1 oral squamous carcinoma cells treated with sodium arsenite (NaAsO2) and dimethylarsenic acid (DMA) to determine whether both arsenic compounds could exert anticancer effects on oral cancer. The results demonstrated that NaAsO2 and DMA induced rounding up and membrane blebbing in OEC­M1 cells, which are morphological characteristics of apoptosis. Annexin V/PI double staining analysis further confirmed that both arsenic compounds induced apoptosis of OEC­M1 cells. In addition, NaAsO2 and DMA significantly decreased the survival rate and increased the percentage of OEC­M1 cells in the subG1 and G2/M phases (P<0.05). Furthermore, both arsenic compounds significantly activated the cleavage of caspase­8, ­9, ­3 and PARP, and the phosphorylation of JNK, ERK1/2 and p38 in OEC­M1 cells (P<0.05). Collectively, the findings of the present study indicated that NaAsO2 and DMA stimulate extrinsic and intrinsic apoptotic pathways through the activation of the MAPK pathways to induce apoptosis of OEC­M1 cells, suggesting that NaAsO2 and DMA may be used as novel anticancer drugs for oral cancers.


Assuntos
Apoptose/efeitos dos fármacos , Arsenitos/farmacologia , Carcinoma/tratamento farmacológico , Neoplasias Gengivais/tratamento farmacológico , Compostos de Sódio/farmacologia , Arsenitos/uso terapêutico , Carcinoma/patologia , Caspases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Neoplasias Gengivais/patologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Compostos de Sódio/uso terapêutico
10.
Drug Des Devel Ther ; 14: 2987-3000, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32801639

RESUMO

INTRODUCTION: Human gestational choriocarcinoma, a type of gestational trophoblastic disease, occurs after miscarriage, abortion, ectopic pregnancy, or molar pregnancy. Despite recent advances in the mechanism of anticancer drugs that induce human gestational choriocarcinoma apoptosis or block its growth, new therapeutic approaches are needed to be established. Cordycepin is an active anti-cancer component extracted from Cordyceps sinensis. It prevents cell proliferation both in vitro and in vivo. MATERIALS AND METHODS: Here, we examined cell growth by counting cell numbers, and performing a flow cytometry assay and EdU incorporation assay. Centrosome and cytoskeleton-related structures were observed by immunofluorescence assay. The DNA damage-related signaling was examined by Western blot assay. RESULTS: Here, we showed that cordycepin inhibited human gestational choriocarcinoma cell proliferation and induced cell death. In addition, treatment with cordycepin activated DNA-PK and ERK, thus inducing centrosome amplification and aberrant mitosis. These amplified centrosomes also disrupted microtubule arrays and actin networks, thus leading to defective cell adhesion. Furthermore, cordycepin induced autophagy for triggering cell death. CONCLUSION: Thus, our study demonstrates that cordycepin inhibits cell proliferation and disrupts the cytoskeleton by triggering centrosome amplification.


Assuntos
Antineoplásicos/farmacologia , Centrossomo/efeitos dos fármacos , Coriocarcinoma/tratamento farmacológico , Desoxiadenosinas/farmacologia , Doença Trofoblástica Gestacional/tratamento farmacológico , Homeostase/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Coriocarcinoma/patologia , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Doença Trofoblástica Gestacional/patologia , Humanos , Gravidez
11.
Eur J Pharmacol ; 879: 173141, 2020 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-32353360

RESUMO

Dapagliflozin (DAPA) or canagliflozin (CANA), Na+-dependent glucose co-transporter type 2 (SGLT2) inhibitors, were used for treatment of type II diabetes mellitus. Addition of DAPA or CANA suppressed M-type K+ current (IK(M)) in pituitary tumor (GH3) and pheochromocytoma PC12 cells. The IC50 value for DAPA- or CANA-mediated inhibition of IK(M) in GH3 cells was 0.11 or 0.42 µM, respectively. The presence of DAPA (0.1 µM) shifted the steady-state activation of IK(M) to less depolarized potential without changing the gating charge of the current. During high-frequency depolarizing pulses, IK(M) magnitude was reduced by DAPA; however, DAPA-induced block of IK(M) remained effective. The amplitude of neither erg-mediated K+ current nor hyperpolarization-activated cation current in GH3 cells was modified in the presence of 1 µM DAPA. Alternatively, addition of DAPA, CANA, phlorizin or chlorotoxin effectively suppressed α-methylglucoside-(αMG-) induced current (IαMG) in GH3 cells, albeit inability of tefluthrin (activator of INa) to suppress this current. DAPA shifted the charge-voltage relation of presteady-state IαMG in a rightward and downward direction with no change in the gating charge of the IαMG. Under current-clamp recordings, subsequent additions of DAPA, but still in the continued presence of αMG, increased the firing rate of spontaneous action potentials stimulated by αMG. Our results suggested that activity of SGLT was expressed functionally in GH3 and PC12 cells. Therefore, inhibitory actions of DAPA or CANA on the amplitude and gating of IK(M) might provide a yet unidentified mechanism through which the SGLT1 or SGLT2 activity were attenuated in unclamped cells occurring in vivo.


Assuntos
Neoplasias das Glândulas Suprarrenais/fisiopatologia , Compostos Benzidrílicos/farmacologia , Canagliflozina/farmacologia , Glucosídeos/farmacologia , Feocromocitoma/fisiopatologia , Neoplasias Hipofisárias/fisiopatologia , Canais de Potássio/fisiologia , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Neoplasias das Glândulas Suprarrenais/metabolismo , Animais , Linhagem Celular Tumoral , Metilglucosídeos/farmacologia , Feocromocitoma/metabolismo , Neoplasias Hipofisárias/metabolismo , Ratos
12.
Int J Mol Med ; 46(1): 439-448, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32319554

RESUMO

Propofol is an anesthetic agent moderating GABA receptors in the nervous system. A number of studies have demonstrated that propofol exerts a negative effect on neural stem cell development in the neonatal mouse hippocampus. However, to the best of our knowledge, there is no study available to date illustrating whether neonatal exposure to propofol affects Leydig stem/progenitor cell development for normal male reproductive development and functions, and the regulatory mechanism remains elusive. In the present study, TM3 cells, a mouse Leydig stem/progenitor cell line, was treated with propofol. The data illustrated that propofol significantly reduced TM3 cell viability. TM3 subG1 phase cell numbers were significantly increased by propofol assayed by flow cytometric analysis. Annexin V/PI double staining assay of the TM3 Leydig cells also demonstrated that propofol increased TM3 cell apoptosis. In addition, cleaved caspase­8, ­9 and ­3 and/or poly(ADP­ribose) polymerase (PARP) were significantly activated by propofol in the TM3 cells. Furthermore, the expression levels of phospho­JNK, phospho­ERK1/2 and phospho­p38 were activated by propofol in the TM3 cells, indicating that propofol induced apoptosis through the mitogen­activated protein kinase (MAPK) pathway. Additionally, propofol diminished the phosphorylation of Akt to increase the apoptosis of TM3 cells. On the whole, the findings of the present study demonstrate that propofol induces TM3 cell apoptosis by activating caspases and MAPK pathways, as well as by inhibiting the Akt pathway in TM3 cells. These findings illustrate that propofol affects the viability of Leydig stem/progenitor cells possibly related to the development of the male reproductive system.


Assuntos
Apoptose/efeitos dos fármacos , Caspases/metabolismo , Células Intersticiais do Testículo/efeitos dos fármacos , Propofol/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células-Tronco/efeitos dos fármacos , Animais , Caspase 8/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Intersticiais do Testículo/citologia , Células Intersticiais do Testículo/metabolismo , Masculino , Camundongos , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/citologia , Células-Tronco/metabolismo
13.
Int J Mol Sci ; 21(1)2020 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-31948124

RESUMO

Pterostilbene (PTER), a natural dimethylated analog of resveratrol, has been demonstrated to produce anti-neoplastic or neuroprotective actions. However, how and whether this compound can entail any perturbations on ionic currents in electrically excitable cells remains unknown. In whole-cell current recordings, addition of PTER decreased the amplitude of macroscopic Ih during long-lasting hyperpolarization in GH3 cells in a concentration-dependent manner, with an effective IC50 value of 0.84 µM. Its presence also shifted the activation curve of Ih along the voltage axis to a more hyperpolarized potential, by 11 mV. PTER at a concentration greater than 10 µM could also suppress l-type Ca2+ and transient outward K+ currents in GH3 cells. With the addition of PTER, IK(Ca) amplitude was increased, with an EC50 value of 2.23 µM. This increase in IK(Ca) amplitude was attenuated by further addition of verruculogen, but not by tolbutamide or TRAM-39. Neither atropine nor nicotine, in the continued presence of PTER, modified the PTER-stimulated IK(Ca). PTER (10 µM) slightly suppressed the amplitude of l-type Ca2+ current and transient outward K+ current. The presence of PTER (3 µM) was also effective at increasing the open-state probability of large-conductance Ca2+-activated K+ (BKCa) channels identified in hippocampal mHippoE-14 neurons; however, its inability to alter single-channel conductance was detected. Our study highlights evidence to show that PTER has the propensity to perturb ionic currents (e.g., Ih and IK(Ca)), thereby influencing the functional activities of neurons, and neuroendocrine or endocrine cells.


Assuntos
Potenciais da Membrana/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Estilbenos/farmacologia , Animais , Cálcio/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Polaridade Celular/efeitos dos fármacos , Polaridade Celular/fisiologia , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/fisiologia , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/fisiologia , Potenciais da Membrana/fisiologia , Camundongos , Neurônios/química , Neurônios/metabolismo , Neurônios/fisiologia , Técnicas de Patch-Clamp , Hipófise/efeitos dos fármacos , Hipófise/fisiologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/fisiologia , Ratos
14.
Fundam Clin Pharmacol ; 34(2): 213-221, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31618480

RESUMO

Quercetin is a flavonol polyphenol widely found in many vegetables, grains, and fruits. Quercetin has been shown to inhibit proliferation and invasion of various glioma cells and is regarded as a potential anticancer agent against glioma. However, whether and how this drug could affect brain blood vessels and endothelial cells (EC) are less understood. Further, there is hitherto no report on how quercetin affects brain EC Ca2+ homeostasis. In this report, we investigated the effects of quercetin on Ca2+ homeostasis in mouse brain bEnd.3 EC. We demonstrated that quercetin raised cytosolic Ca2+ level in a concentration-dependent manner. Quercetin-triggered Ca2+ signal composed of both internal Ca2+ release and extracellular Ca2+ influx. Quercetin caused Ca2+ release from the endoplasmic reticulum, and consistently, inhibition of inositol 1,4,5-trisphosphate receptor (IP3R) by xestospongin C (XeC) suppressed quercetin-triggered Ca2+ release. Quercetin also caused Ca2+ release from lysosomes, an observation in concordance with the inhibition of quercetin-triggered Ca2+ release by trans-Ned-19, a blocker of two-pore channels. As quercetin depleted intracellular Ca2+ storage, it suppressed ATP-induced Ca2+ release and thereby blunted ATP-triggered Ca2+ signaling. In addition, quercetin co-treatment significantly suppressed ATP-stimulated nitric oxide release. Our work therefore showed, for the first time, quercetin perturbed intracellular Ca2+ stores and strongly suppressed ATP-triggered response in bEnd.3 cells.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Células Endoteliais/efeitos dos fármacos , Quercetina/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Antioxidantes/administração & dosagem , Antioxidantes/farmacologia , Linhagem Celular , Relação Dose-Resposta a Droga , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Células Endoteliais/metabolismo , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Camundongos , Óxido Nítrico/metabolismo , Quercetina/administração & dosagem
15.
Oncol Rep ; 41(6): 3565-3574, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31002349

RESUMO

In the western world, there is an increasing trend of occurrence in testicular cancer. Treatment of malignant testicular cancer is primarily combined surgery with various chemical drugs. Propofol has been frequently used as an anesthetic and sedative induction agent, which could modulate different γ­aminobutyric acid receptors in the central nervous system. Studies demonstrated that propofol activates endoplasmic reticulum stress to induce apoptosis in lung cancer. However, it remains elusive whether propofol regulates caspase and/or mitogen­activated protein kinase (MAPK) pathways to induce apoptosis in Leydig tumor cells. In the present study, MA­10 mouse Leydig tumor cells were treated with propofol, and possible signal pathways associated with apoptosis were investigated. Results demonstrated that increasing dosage of propofol (300­600 µM) for 24 h significantly decreased cell viability in MA­10 cells (P<0.05). In flow cytometry analysis, the amount of sub­G1 phase cell numbers in MA­10 cells was significantly increased by propofol (P<0.05). Additionally, Annexin V/propidium iodide double staining further confirmed that propofol could induce MA­10 cell apoptosis. Furthermore, cleaved caspase­8, ­9 and ­3, and/or poly(ADP­ribose) polymerase were significantly activated following treatment of propofol in MA­10 cells. In addition, c­Jun N­terminal kinase, extracellular signal­regulated kinase 1/2, and p38 were significantly activated by propofol in MA­10 cells (P<0.05), indicating that propofol may induce apoptosis through the MAPK pathway. Additionally, propofol diminished the phosphorylation of Akt to activate apoptosis in MA­10 cells. In conclusion, propofol may induce MA­10 cell apoptosis by activating caspase and MAPK pathways, and inhibiting the Akt pathway in MA­10 cells, demonstrating that propofol may be a potential anticancer agent against Leydig cell cancer.


Assuntos
Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Propofol/farmacologia , Neoplasias Testiculares/tratamento farmacológico , Animais , Caspases/genética , Proliferação de Células , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Tumor de Células de Leydig , MAP Quinase Quinase 1/genética , Masculino , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais/efeitos dos fármacos , Neoplasias Testiculares/genética , Neoplasias Testiculares/patologia
16.
J Cell Mol Med ; 22(5): 2896-2907, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29516686

RESUMO

Midazolam, a benzodiazepine derivative, is widely used for sedation and surgery. However, previous studies have demonstrated that Midazolam is associated with increased risks of congenital malformations, such as dwarfism, when used during early pregnancy. Recent studies have also demonstrated that Midazolam suppresses osteogenesis of mesenchymal stem cells (MSCs). Given that hypertrophic chondrocytes can differentiate into osteoblast and osteocytes and contribute to endochondral bone formation, the effect of Midazolam on chondrogenesis remains unclear. In this study, we applied a human MSC line, the KP cell, to serve as an in vitro model to study the effect of Midazolam on chondrogenesis. We first successfully established an in vitro chondrogenic model in a micromass culture or a 2D high-density culture performed with TGF-ß-driven chondrogenic induction medium. Treatment of the Midazolam dose-dependently inhibited chondrogenesis, examined using Alcian blue-stained glycosaminoglycans and the expression of chondrogenic markers, such as SOX9 and type II collagen. Inhibition of Midazolam by peripheral benzodiazepine receptor (PBR) antagonist PK11195 or small interfering RNA rescued the inhibitory effects of Midazolam on chondrogenesis. In addition, Midazolam suppressed transforming growth factor-ß-induced Smad3 phosphorylation, and this inhibitory effect could be rescued using PBR antagonist PK11195. This study provides a possible explanation for Midazolam-induced congenital malformations of the musculoskeletal system through PBR.


Assuntos
Condrogênese/efeitos dos fármacos , Antagonistas de Receptores de GABA-A/farmacologia , Células-Tronco Mesenquimais/metabolismo , Midazolam/farmacologia , Receptores de GABA-A/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Humanos , Isoquinolinas/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo
17.
Biochem Pharmacol ; 151: 79-88, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29477572

RESUMO

GMQ (2-guanidine-4-methylquinazoline or N-(4-methyl-2-quinazolinyl)-guanidine hydrochloride), an agonist of acid-sensing ion channel type 3, has been increasingly used for in vivo studies of alternations in nociceptic behavior. In this study, we tried to investigate whether GMQ has any possible effect on other types of ion channels. Addition of GMQ to pituitary GH3 cells raised the amplitude of Ca2+-activated K+ currents (IK(Ca)), which was reversed by verruculogen or PF1022A, but not by TRAM-39. Under inside-out current recordings, addition of GMQ into bath enhanced the probability of large-conductance Ca2+-activated K+ (BKCa) channels with an EC50 value of 0.95 µM. The activation curve of BKCa channels during exposure to GMQ shifted to a lower depolarized potential, with no change in the gating charge of the curve; however, there was a reduction of free energy for channel activation in its presence. As cells were exposed to GMQ, the amplitude of ion currents were suppressed, including delayed rectifying K+ current, voltage-gated Na+ and L-type Ca2+ currents. In Rolf B1.T olfactory sensory neuron, addition of GMQ was able to induce inward current and to suppress peak INa. Taken together, findings from these results indicated that in addition to the activation of ASIC3 channels, this compound might directly produce additional actions on various types of ion channels. Caution should be taken in the interpretation of in vivo experimental results when GMQ or other structurally similar compounds are used as targets to characterize the potential functions of ASIC3 channels.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Guanidinas/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Neurônios Receptores Olfatórios/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Quinazolinas/farmacologia , Agonistas de Canais de Sódio/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Transporte de Íons , Neurônios Receptores Olfatórios/metabolismo , Técnicas de Patch-Clamp , Hipófise/metabolismo , Ratos
18.
Toxicol Lett ; 285: 104-112, 2018 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-29306026

RESUMO

Voltage-gated Na+ currents (INa), known to contain many components (e.g., transient, resurgent and persistent INa) with unique gating properties, are involved in the generation and propagation of action potentials in excitable cells. In this study, how tefluthrin (Tef), a synthetic pyrethoid, and telmisartan (TEL), blocker of angiotensin II receptors can perturb those components of INa was investigated. The presence of either Tef or TEL increased the values of the gating charges of INa involved in the activation (za) and inactivation (zi). Tef also increased the amplitude of resurgent INa (INa(R)) or persistent INa (INa(P)) in a pituitary cell line (GH3), while TEL produced minimal effects on them. Subsequent addition of either ranolazine (a blocker of late INa) or d-limonene (a monoterpene), could reverse the changes by TEL or Tef on za or zi. In SCN5A-expressing HEK293T cells, addition of Tef or TEL also increased the peak amplitude and the inactivation time constant of INa which was accompanied by the increased za value of INa. Taken together, the results indicated that Tef- or TEL-mediated changes in the gating kinetics of INa are linked to their actions on functional activity of neurons, neuroendocrine or endocrine cells.


Assuntos
Benzimidazóis/farmacologia , Benzoatos/farmacologia , Ciclopropanos/farmacologia , Hidrocarbonetos Fluorados/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Somatotrofos/efeitos dos fármacos , Canais de Sódio Disparados por Voltagem/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Cicloexenos/farmacologia , Células HEK293 , Humanos , Limoneno , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Ranolazina/farmacologia , Ratos , Somatotrofos/metabolismo , Telmisartan , Terpenos/farmacologia , Transfecção , Canais de Sódio Disparados por Voltagem/genética
19.
Cell Physiol Biochem ; 41(5): 2053-2066, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28456794

RESUMO

BACKGROUND: Artemisinin (ART) is an anti-malarial agent reported to influence endocrine function. METHODS: Effects of ART on ionic currents and action potentials (APs) in pituitary tumor (GH3) cells were evaluated by patch clamp techniques. RESULTS: ART inhibited the amplitude of delayed-rectifier K+ current (IK(DR)) in response to membrane depolarization and accelerated the process of current inactivation. It exerted an inhibitory effect on IK(DR) with an IC50 value of 11.2 µM and enhanced IK(DR) inactivation with a KD value of 14.7 µM. The steady-state inactivation curve of IK(DR) was shifted to hyperpolarization by 10 mV. Pretreatment of chlorotoxin (1 µM) or iloprost (100 nM) did not alter the magnitude of ART-induced inhibition of IK(DR) in GH3 cells. ART also decreased the peak amplitude of voltage-gated Na+ current (INa) with a concentration-dependent slowing in inactivation rate. Application of KMUP-1, an inhibitor of late INa, was effective at reversing ART-induced prolongation in inactivation time constant of INa. Under current-clamp recordings, ART alone reduced the amplitude of APs and prolonged the duration of APs. CONCLUSION: Under ART exposure, the inhibitory actions on both IK(DR) and INa could be a potential mechanisms through which this drug influences membrane excitability of endocrine or neuroendocrine cells appearing in vivo.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Artemisininas/farmacologia , Canais de Potássio de Retificação Tardia/antagonistas & inibidores , Lactonas/farmacologia , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias Hipofisárias/tratamento farmacológico , Prolactinoma/tratamento farmacológico , Animais , Canais de Potássio de Retificação Tardia/metabolismo , Relação Dose-Resposta a Droga , Proteínas de Neoplasias/metabolismo , Neoplasias Hipofisárias/metabolismo , Neoplasias Hipofisárias/patologia , Prolactinoma/metabolismo , Prolactinoma/patologia , Ratos
20.
Onco Targets Ther ; 9: 2519-33, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27175086

RESUMO

PURPOSE: Midazolam is widely used as a sedative and anesthetic induction agent by modulating the different GABA receptors in the central nervous system. Studies have also shown that midazolam has an anticancer effect on various tumors. In a previous study, we found that midazolam could induce MA-10 mouse Leydig tumor cell apoptosis by activating caspase cascade. However, the detailed mechanism related to the upstream and downstream pathways of the caspase cascade, such as endoplasmic reticulum (ER) stress, autophagy, and p53 pathways plus cell cycle regulation in MA-10 mouse Leydig tumor cells, remains elusive. METHODS: Flow cytometry assay and Western blot analyses were exploited. RESULTS: Midazolam significantly decreased cell viability but increased sub-G1 phase cell numbers in MA-10 cells (P<0.05). Annexin V/propidium iodide double staining further confirmed that midazolam induced apoptosis. In addition, expressions of Fas and Fas ligand could be detected in MA-10 cells with midazolam treatments, and Bax translocation and cytochrome c release were also involved in midazolam-induced MA-10 cell apoptosis. Moreover, the staining and expression of LC3-II proteins could be observed with midazolam treatment, implying midazolam could induce autophagy to control MA-10 cell apoptosis. Furthermore, the expressions of p-EIF2α, ATF4, ATF3, and CHOP could be induced by midazolam, indicating that midazolam could stimulate apoptosis through ER stress in MA-10 cells. Additionally, the expressions of cyclin A, cyclin B, and CDK1 could be inhibited by midazolam, and the phosphorylation of p53, P27, and P21 could be adjusted by midazolam, suggesting that midazolam could manage cell cycle through the regulation of p53 pathway to induce apoptosis in MA-10 cells. CONCLUSION: Midazolam could induce cell apoptosis through the activation of ER stress and the regulation of cell cycle through p53 pathway with the involvement of autophagy in MA-10 mouse Leydig tumor cells.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA