Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 177
Filtrar
1.
Sci Rep ; 14(1): 5375, 2024 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-38438412

RESUMEN

Propyl gallate (PG) exhibits an anti-growth effect on various cell types. The present study investigated the impact of PG on the levels of reactive oxygen species (ROS) and glutathione (GSH) in primary human pulmonary fibroblast (HPF) cells. Moreover, the effects of N-acetyl cysteine (NAC, an antioxidant), L-buthionine sulfoximine (BSO, a GSH synthesis inhibitor), and small interfering RNA (siRNAs) against various antioxidant genes on ROS and GSH levels and cell death were examined in PG-treated HPF cells. PG (100-800 µM) increased the levels of total ROS and O2·- at early time points of 30-180 min and 24 h, whereas PG (800-1600 µM) increased GSH-depleted cell number at 24 h and reduced GSH levels at 30-180 min. PG downregulated the activity of superoxide dismutase (SOD) and upregulated the activity of catalase in HPF cells. Treatment with 800 µM PG increased the number of apoptotic cells and cells that lost mitochondrial membrane potential (MMP; ΔΨm). NAC treatment attenuated HPF cell death and MMP (ΔΨm) loss induced by PG, accompanied by a decrease in GSH depletion, whereas BSO exacerbated the cell death and MMP (ΔΨm) loss without altering ROS and GSH depletion levels. Furthermore, siRNA against SOD1, SOD2, or catalase attenuated cell death in PG-treated HPF cells, whereas siRNA against GSH peroxidase enhanced cell death. In conclusion, PG induced cell death in HPF cells by increasing ROS levels and depleting GSH. NAC was found to decrease HPF cell death induced by PG, while BSO enhanced cell death. The findings shed light on how manipulating the antioxidant system influence the cytotoxic effects of PG in HPF cells.


Asunto(s)
Chrysanthemum , Galato de Propilo , Humanos , Galato de Propilo/farmacología , Antioxidantes/farmacología , Especies Reactivas de Oxígeno , Catalasa , Muerte Celular , Fibroblastos , Glutatión , Butionina Sulfoximina/farmacología , ARN Interferente Pequeño/genética
2.
Ann Med ; 56(1): 2319853, 2024 12.
Artículo en Inglés | MEDLINE | ID: mdl-38373208

RESUMEN

Propyl gallate (PG) has been found to exert an inhibitory effect on the growth of different cell types, including lung cancer cells. However, little is known about the cytotoxicological effects of PG specifically on normal primary lung cells. The current study examined the cellular effects and cell death resulting from PG treatment in human pulmonary fibroblast (HPF) cells. DNA flow cytometry results demonstrated that PG (100-1,600 µM) had a significant impact on the cell cycle, leading to G1 phase arrest. Notably, 1,600 µM PG slightly increased the number of sub-G1 cells. Additionally, PG (400-1,600 µM) resulted in the initiation of cell death, a process that coincided with a loss of mitochondrial membrane potential (MMP; ΔΨm). This loss of MMP (ΔΨm) was evaluated using a FACS cytometer. In PG-treated HPF cells, inhibitors targeting pan-caspase, caspase-3, caspase-8, and caspase-9 showed no significant impact on the quantity of annexin V-positive and MMP (ΔΨm) loss cells. The administration of siRNA targeting Bax or caspase-3 demonstrated a significant attenuation of PG-induced cell death in HPF cells. However, the use of siRNAs targeting p53, Bcl-2, or caspase-8 did not exhibit any notable effect on cell death. Furthermore, none of the tested MAPK inhibitors, including MEK, c-Jun N-terminal kinase (JNK), and p38, showed any impact on PG-induced cell death or the loss of MMP (ΔΨm) in HPF cells. In conclusion, PG induces G1 phase arrest of the cell cycle and cell death in HPF cells through apoptosis and/or necrosis. The observed HPF cell death is mediated by the modulation of Bax and caspase-3. These findings offer insights into the cytotoxic and molecular effects of PG on normal HPF cells.


Asunto(s)
Glutatión , Galato de Propilo , Humanos , Galato de Propilo/metabolismo , Galato de Propilo/farmacología , Caspasa 8/metabolismo , Caspasa 8/farmacología , Proteína X Asociada a bcl-2/metabolismo , Proteína X Asociada a bcl-2/farmacología , Caspasa 3/metabolismo , Caspasa 3/farmacología , Glutatión/metabolismo , Glutatión/farmacología , Especies Reactivas de Oxígeno/metabolismo , Proliferación Celular , Muerte Celular , Apoptosis , Pulmón , Fibroblastos/metabolismo
3.
Molecules ; 28(18)2023 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-37764247

RESUMEN

Ebselen is a glutathione (GSH) peroxidase (GPx) mimic originally developed to reduce reactive oxygen species (ROS). However, little is known about its cytotoxicological effects on lung cells. Therefore, this study aimed to investigate the effects of Ebselen on the cell growth and cell death of A549 lung cancer cells, Calu-6 lung cancer cells, and primary normal human pulmonary fibroblast (HPF) cells in relation to redox status. The results showed that Ebselen inhibited the growth of A549, Calu-6, and HPF cells with IC50 values of approximately 12.5 µM, 10 µM, and 20 µM, respectively, at 24 h. After exposure to 15 µM Ebselen, the proportions of annexin V-positive cells were approximately 25%, 65%, and 10% in A549, Calu-6, and HPF cells, respectively. In addition, Ebselen induced arrest at the S phase of the cell cycle in A549 cells and induced G2/M phase arrest in Calu-6 cells. Treatment with Ebselen induced mitochondrial membrane potential (MMP; ΔΨm) loss in A549 and Calu-6 cells. Z-VAD, a pan-caspase inhibitor, did not decrease the number of annexin V-positive cells in Ebselen-treated A549 and Calu-6 cells. Intracellular ROS levels were not significantly changed in the Ebselen-treated cancer cells at 24 h, but GSH depletion was efficiently induced in these cells. Z-VAD did not affect ROS levels or GSH depletion in Ebselen-treated A549 or Ebselen-treated Calu-6 cells. In conclusion, Ebselen inhibited the growth of lung cancer and normal fibroblast cells and induced cell cycle arrest and cell death in lung cancer cells with GSH depletion.


Asunto(s)
Apoptosis , Neoplasias Pulmonares , Humanos , Especies Reactivas de Oxígeno/metabolismo , Anexina A5 , Línea Celular Tumoral , Muerte Celular , Glutatión/metabolismo , Puntos de Control del Ciclo Celular , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Pulmón/metabolismo , Proliferación Celular
4.
Molecules ; 27(21)2022 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-36364165

RESUMEN

Tempol (4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl) is a stable, cell-permeable redox-cycling nitroxide water-soluble superoxide dismutase (SOD) mimetic agent. However, little is known about its cytotoxic effects on lung-related cells. Thus, the present study investigated the effects of Tempol on cell growth and death as well as changes in reactive oxygen species (ROS) and glutathione (GSH) levels in Calu-6 and A549 lung cancer cells, normal lung WI-38 VA-13 cells, and primary pulmonary fibroblast cells. Results showed that Tempol (0.5~4 mM) dose-dependently inhibited the growth of lung cancer and normal cells with an IC50 of approximately 1~2 mM at 48 h. Tempol induced apoptosis in lung cells with loss of mitochondrial membrane potential (MMP; ∆Ψm) and activation of caspase-3. There was no significant difference in susceptibility to Tempol between lung cancer and normal cells. Z-VAD, a pan-caspase inhibitor, significantly decreased the number of annexin V-positive cells in Tempol-treated Calu-6, A549, and WI-38 VA-13 cells. A 2 mM concentration of Tempol increased ROS levels, including O2•- in A549 and WI-38 VA-13 cells after 48 h, and specifically increased O2•- levels in Calu-6 cells. In addition, Tempol increased the number of GSH-depleted cells in Calu-6, A549, and WI-38 VA-13 cells at 48 h. Z-VAD partially downregulated O2•- levels and GSH depletion in Tempol-treated these cells. In conclusion, treatment with Tempol inhibited the growth of both lung cancer and normal cells via apoptosis and/or necrosis, which was correlated with increased O2•- levels and GSH depletion.


Asunto(s)
Apoptosis , Neoplasias Pulmonares , Humanos , Especies Reactivas de Oxígeno/metabolismo , Glutatión/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Potencial de la Membrana Mitocondrial , Proliferación Celular
5.
Molecules ; 27(16)2022 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-36014444

RESUMEN

Auranofin, as a thioredoxin reductase (TrxR) inhibitor, has promising anti-cancer activity in several cancer types. However, little is known about the inhibitory effect of auranofin on lung cancer cell growth. We, therefore, investigated the antigrowth effects of auranofin in various lung cancer cells with respect to cell death, reactive oxygen species (ROS), and glutathione (GSH) levels. Treatment with 0~5 µM auranofin decreased cell proliferation and induced cell death in Calu-6, A549, SK-LU-1, NCI-H460, and NCI-H1299 lung cancer cells at 24 h. In addition, 0~5 µM auranofin increased ROS levels, including O2•-, and depleted GSH levels in these cells. N-acetyl cysteine (NAC) prevented growth inhibition and mitochondrial membrane potential (MMP, ∆Ψm) loss in 3 and 5 µM auranofin-treated Calu-6 and A549 cells at 24 h, respectively, and decreased ROS levels and GSH depletion in these cells. In contrast, L-buthionine sulfoximine (BSO) enhanced cell death, MMP (∆Ψm) loss, ROS levels, and GSH depletion in auranofin-treated Calu-6 and A549 cells. Treatment with 3 and 5 µM auranofin induced caspase-3 activation and poly (ADP ribose) polymerase (PARP) cleavage in Calu-6 and A549 cells, respectively. Both were prevented by NAC, but enhanced by BSO. Moreover, TrxR activity was reduced in auranofin-treated Calu-6 and A549 cells. That activity was decreased by BSO, but increased by NAC. In conclusion, these findings demonstrate that auranofin-induced cell death is closely related to oxidative stress resulted from increased ROS levels and GSH depletion in lung cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Auranofina , Neoplasias Pulmonares , Acetilcisteína/metabolismo , Acetilcisteína/farmacología , Apoptosis , Auranofina/farmacología , Butionina Sulfoximina/farmacología , Línea Celular Tumoral , Proliferación Celular , Glutatión/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Potencial de la Membrana Mitocondrial , Especies Reactivas de Oxígeno/metabolismo
6.
Molecules ; 27(14)2022 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-35889456

RESUMEN

Propyl gallate [3,4,5-trihydroxybenzoic acid propyl ester; PG] exhibits an anti-growth effect in various cells. In this study, the anti-apoptotic effects of various caspase inhibitors were evaluated in PG-treated Calu-6 and A549 lung cancer cells in relation to reactive oxygen species (ROS) and glutathione (GSH) levels. Treatment with 800 µM PG inhibited the proliferation and induced the cell death of both Calu-6 and A549 cells at 24 h. Each inhibitor of pan-caspase, caspase-3, caspase-8, and caspase-9 reduced the number of dead and sub-G1 cells in both PG-treated cells at 24 h. PG increased ROS levels, including O2∙-, in both lung cancer cell lines at 24 h. Generally, caspase inhibitors appeared to decrease ROS levels in PG-treated lung cancer cells at 24 h and somewhat reduced O2∙- levels. PG augmented the number of GSH-depleted Calu-6 and A549 cells at 24 h. Caspase inhibitors did not affect the level of GSH depletion in PG-treated A549 cells but differently and partially altered the depletion level in PG-treated Calu-6 cells. In conclusion, PG exhibits an anti-proliferative effect in Calu-6 and A549 lung cancer cells and induced their cell death. PG-induced lung cancer death was accompanied by increases in ROS levels and GSH depletion. Therefore, the anti-apoptotic effects of caspase inhibitors were, at least in part, related to changes in ROS and GSH levels.


Asunto(s)
Neoplasias Pulmonares , Galato de Propilo , Apoptosis , Inhibidores de Caspasas/farmacología , Proliferación Celular , Glutatión/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Potencial de la Membrana Mitocondrial , Galato de Propilo/farmacología , Especies Reactivas de Oxígeno/metabolismo
7.
J Appl Toxicol ; 42(3): 436-449, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34464457

RESUMEN

Propyl gallate (3,4,5-trihydroxybenzoic acid propyl ester, PG) has an anti-proliferative effect in various cells. In this study, Calu-6 and A549 lung cancer cells were used to examine the anti-proliferative effect of PG in relation to reactive oxygen species (ROS) and glutathione (GSH) levels. PG (100-1,600 µM) dose-dependently inhibited the proliferation of Calu-6 and A549 cells at 24 h, and PG at 800-1,600 µM strongly induced cell death in both cell lines. PG (800-1,600 µM) increased cellular metabolism in Calu-6 but not A549 cells at 4 h. PG either increased or decreased ROS levels, including O2 ˙- and ˙OH, depending on the incubation doses and times of 1 or 24 h. Even these effects differed between Calu-6 and A549 cell types. PG reduced the activity of superoxide dismutase (SOD) in Calu-6 cells, and it augmented the activity of catalase in A549 cells. PG dose-dependently increased the number of GSH depleted cells in both Calu-6 and A549 cells at 24 h. In addition, PG decreased GSH levels in both lung cancer cells at 1 h. Furthermore, diethyldithiocarbamate (DDC; an inhibitor of SOD) and 3-amino-1,2,4-triazole (AT; an inhibitor of catalase) differently affected cellular metabolism, ROS and GSH levels in PG-treated and PG-untreated Calu-6 and A549 cells at 1 h. In conclusion, PG dose-dependently decreased the proliferation of Calu-6 and A549 lung cancer cells, which was related to changes in ROS levels and the depletion of GSH.


Asunto(s)
Antioxidantes/farmacología , Glutatión/metabolismo , Galato de Propilo/farmacología , Especies Reactivas de Oxígeno/metabolismo , Células A549 , Humanos
8.
Sci Rep ; 11(1): 14869, 2021 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-34290305

RESUMEN

Tempol (4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl) is a potential redox agent in cells. The present study investigated changes in cellular reactive oxygen species (ROS) and glutathione (GSH) levels and in antioxidant enzymes, in Tempol-treated Calu-6 and A549 lung cancer cells, normal lung WI-38 VA-13 cells, and primary pulmonary fibroblasts. Results demonstrated that Tempol (0.5-4 mM) either increased or decreased general ROS levels in lung cancer and normal cells at 48 h and specifically increased O2•- levels in these cells. In addition, Tempol differentially altered the expression and activity of antioxidant enzymes such as superoxide dismutase, catalase, and thioredoxin reductase1 (TrxR1) in A549, Calu-6, and WI-38 VA-13 cells. In particular, Tempol treatment increased TrxR1 protein levels in these cells. Tempol at 1 mM inhibited the growth of lung cancer and normal cells by about 50% at 48 h but also significantly induced cell death, as evidenced by annexin V-positive cells. Furthermore, down-regulation of TrxR1 by siRNA had some effect on ROS levels as well as cell growth inhibition and death in Tempol-treated or -untreated lung cells. In addition, some doses of Tempol significantly increased the numbers of GSH-depleted cells in both cancer cells and normal cells at 48 h. In conclusion, Tempol differentially increased or decreased levels of ROS and various antioxidant enzymes in lung cancer and normal cells, and induced growth inhibition and death in all lung cells along with an increase in O2•- levels and GSH depletion.


Asunto(s)
Antioxidantes , Óxidos N-Cíclicos/farmacología , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Pulmón/enzimología , Pulmón/patología , Oxidación-Reducción/efectos de los fármacos , Células A549 , Catalasa/metabolismo , Células Cultivadas , Glutatión/metabolismo , Humanos , Pulmón/citología , Pulmón/metabolismo , Neoplasias Pulmonares/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Marcadores de Spin , Superóxido Dismutasa/metabolismo , Tiorredoxina Reductasa 1/metabolismo , Factores de Tiempo
9.
Toxicol In Vitro ; 74: 105176, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33865947

RESUMEN

Propyl gallate (PG) has an anti-growth effect in lung cancer cells. The present study investigated the effects of mitogen-activated protein kinase (MAPK; MEK, JNK, and p38) inhibitors on PG-treated Calu-6 and A549 lung cancer cells in relation to cell death as well as reactive oxygen species (ROS) and glutathione (GSH) levels. PG induced cell death in both Calu-6 and A549 lung cancer cells at 24 h, which was accompanied by loss of mitochondrial membrane potential (MMP; ΔΨm). All of the tested MAPK inhibitors increased cell death in both PG-treated lung cancer cell lines. In particular, MEK inhibitor strongly enhanced cell death and MMP (ΔΨm) loss in PG-treated Calu-6 cells and p38 inhibitor had the same effects in A549 cells as well. PG increased ROS levels and caused GSH depletion in both cell lines at 24 h. MAPK inhibitors increased O2•- levels and GSH depletion in PG-treated Calu-6 cells, and JNK and p38 inhibitors increased ROS levels and GSH depletion in PG-treated A549 cells. In conclusion, MAPK inhibitors increased cell death in PG-treated Calu-6 and A549 lung cancer cells. Enhanced cell death and GSH depletion in Calu-6 cells caused by the MEK inhibitor were related to increased O2•- levels, and the effects of the p38 inhibitor in A549 cells were correlated with increased general ROS levels.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Galato de Propilo/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Glutatión/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
10.
Oncol Rep ; 44(6): 2783-2791, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33125113

RESUMEN

Propyl gallate (3,4,5­trihydroxybenzoic acid propyl ester; PG) is a synthetic phenolic antioxidant which exerts many effects on tissue and cell functions. In the present study, Calu­6 and A549 lung cancer cells were used to examine the molecular mechanism of the anti­growth effects of PG in relation to apoptosis and cell cycle arrest. PG inhibited the growth of both lung cancer cell types in a dose­dependent manner with an IC50 of 800 µM at 24 h based on MTT assays. DNA flow cytometry showed that PG induced G1 phase arrest of the cell cycle in Calu­6 and A549 cells. In addition, PG induced apoptosis in both lung cancer cell types, as evidenced by sub­G1 cell population and Annexin V­stained cells. Western blot results demonstrated that PG decreased the Bcl­2 level which was accompanied by an increase in the cleaved form of poly(ADP­ribose) polymerase (PARP). PG also triggered loss of mitochondrial membrane potential (MMP; ∆Ψm) and decreased MMP (∆Ψm) levels in both lung cancer cell types, as assessed by FACS analysis. Furthermore, PG upregulated the activities of caspase­3 and caspase­8 in Calu­6 cells. In conclusion, PG treatment inhibited the growth of lung cancer cells, especially Calu­6 cells via caspase­dependent apoptosis as well as G1 phase arrest of the cell cycle.


Asunto(s)
Antioxidantes/farmacología , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Galato de Propilo/farmacología , Células A549 , Antioxidantes/uso terapéutico , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Neoplasias Pulmonares/patología , Galato de Propilo/uso terapéutico , Regulación hacia Arriba/efectos de los fármacos
11.
Oncol Rep ; 44(6): 2715-2724, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33125149

RESUMEN

Auranofin, an inhibitor of thioredoxin reductase (TrxR), inhibits the growth of a variety of cancer cells. In the present study, various lung cancer cells were used to investigate the molecular basis of anti­cancer effects of auranofin, including cell death via apoptosis or necrosis and cell cycle arrest. Generally, auranofin inhibited the growth of the tested lung cancer cell lines in a dose­dependent manner with an IC50 of 3­4 µM at 24 h. This agent significantly decreased the activity of TrxR in Calu­6 and A549 lung cancer cells. In addition, auranofin (3­5 µM) triggered necrosis in lung cancer cells measured by the release of lactate dehydrogenase (LDH) into culture media. Auranofin increased the percentages of sub­G1 cells in Calu­6 and A549 cells. DNA flow cytometry showed that auranofin induced G2/M phase arrest of Calu­6 cells. This agent also efficiently induced apoptosis, accompanied by loss of mitochondrial membrane potential (MMP; ∆Ψm), increases in cleavage forms of caspase­3 and poly (ADP­ribose) polymerase (PARP), and a high ratio of BAX to Bcl­2 proteins. Furthermore, various caspase inhibitors reduced apoptosis and MMP (∆Ψm) loss in auranofin­treated Calu­6 cells. In particular, the pan­caspase inhibitor, benzyloxycarbonyl­Val­Ala­Asp­fluoromethylketone (Z­VAD), decreased cleavage forms of caspase­3, ­8, and ­9 in these cells. In conclusion, auranofin inhibited the proliferation of lung cancer cells, especially Calu­6 cells, via cell cycle arrest and cell death due to necrosis or caspase­dependent apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Auranofina/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Necroptosis/efectos de los fármacos , Auranofina/uso terapéutico , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Inhibidores de Caspasas/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Neoplasias Pulmonares/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Oligopéptidos/farmacología
12.
Int J Mol Sci ; 21(7)2020 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-32290325

RESUMEN

Arsenic trioxide (ATO; As2O3) has anti-cancer effects in various solid tumors as well as hematological malignancy. Valproic acid (VPA), which is known to be a histone deacetylase inhibitor, has also anti-cancer properties in several cancer cells including lung cancer cells. Combined treatment of ATO and VPA (ATO/VPA) could synergistically enhance anti-cancer effects and reduce ATO toxicity ATO. In this study, the combined anti-cancer effects of ATO and VPA (ATO/VPA) was investigated in NCI-H460 and NCI-H1299 lung cancer cells in vitro and in vivo. A combination of 3 µM ATO and 3 mM VPA (ATO/VPA) strongly inhibited the growths of both lung cancer cell types. DNA flow cytometry indicated that ATO/VPA significantly induced G2/M-phase arrest in both cell lines. In addition, ATO/VPA strongly increased the percentages of sub-G1 cells and annexin V-FITC positive cells in both cells. However, lactate dehydrogenase (LDH) release from cells was not increased in ATO/VPA-treated cells. In addition, ATO/VPA increased apoptosis in both cell types, accompanied by loss of mitochondrial membrane potential (MMP, ∆Ψm), activation of caspases, and cleavage of anti-poly ADP ribose polymerase-1. Moreover, a pan-caspase inhibitor, Z-VAD, significantly reduced apoptotic cell death induced by ATO/VPA. In the xenograft model, ATO/VPA synergistically inhibited growth of NCI-H460-derived xenograft tumors. In conclusion, the combination of ATO/VPA effectively inhibited the growth of lung cancer cells through G2/M-phase arrest and apoptotic cell death, and had a synergistic antitumor effect in vivo.


Asunto(s)
Apoptosis/efectos de los fármacos , Trióxido de Arsénico/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Ácido Valproico/farmacología , Animales , Antineoplásicos/farmacología , Biomarcadores , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Humanos , Neoplasias Pulmonares , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Oncol Rep ; 43(1): 358-367, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31789420

RESUMEN

The thioredoxin (Trx) system is an important enzymatic complex involved in cellular redox homeostasis. Arsenic trioxide (ATO; As2O3) is known to trigger cell death in vascular smooth muscle cells (VSMCs) via oxidative stress. In the present study, the effects of changes in thioredoxin 1 (Trx1) and Trx reductase1 (TrxR1) on cell growth, death, reactive oxygen species (ROS), and glutathione (GSH) levels were evaluated in ATO­treated human pulmonary artery smooth muscle cells (HPASMCs). ATO inhibited growth and induced cell death in the HPASMCs at 24 h. Overexpression of Trx1 and TrxR1 using adenoviruses attenuated cell growth inhibition caused by ATO and partially prevented cell death. ATO increased ROS levels including the mitochondrial superoxide anion (O2•-) at 5 min. Administration of adTrx1 or adTrxR1 reduced the increased mitochondrial O2•- level in these cells. HPASMCs treated with Trx1 or TrxR1 siRNA showed increases in ROS levels with or without treatment of ATO at 5 min. Although ATO transiently increased GSH levels at 5 min, Trx1 and TrxR1 siRNAs reduced the increased GSH levels in these cells. In addition, PX­12 (a Trx1 inhibitor) and auranofin (a TrxR1 inhibitor) diminished the cellular metabolism in HPASMCs at 4 h, accompanied by an increase in ROS level and a decrease in GSH level. In conclusion, upregulation of Trx1 and TrxR1 somewhat decreased cell growth inhibition and death in ATO­treated HPASMCs, which was accompanied by reduced oxidative stress.


Asunto(s)
Trióxido de Arsénico/farmacología , Músculo Liso Vascular/citología , Tiorredoxina Reductasa 1/genética , Tiorredoxinas/genética , Auranofina/farmacología , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dependovirus/genética , Glutatión/metabolismo , Humanos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Estrés Oxidativo/efectos de los fármacos , Arteria Pulmonar , Especies Reactivas de Oxígeno/metabolismo , Tiorredoxina Reductasa 1/metabolismo , Tiorredoxinas/metabolismo , Regulación hacia Arriba/efectos de los fármacos
14.
Toxicol In Vitro ; 61: 104590, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31279089

RESUMEN

The thioredoxin (Trx) system controls cellular redox in vascular smooth muscle cells. The present study investigated the roles of Trx1 and Trx reductase1 (TrxR1) proteins in regulation of cell growth, death, reactive oxygen species (ROS) and glutathione (GSH) levels in hydrogen peroxide (H2O2)-treated human pulmonary artery smooth muscle (HPASM) cells. H2O2 induced growth inhibition and cell death in HPASM cells over 24 h. Overexpression of Trx1 and TrxR1 using adenoviruses significantly weakened cell growth inhibition and cell death caused by H2O2. Increases in ROS levels including mitochondrial superoxide anion (O2•-) were observed as early as 5-30 min after H2O2 addition. Administration of adTrxR1 attenuated H2O2-induced increases in ROS levels at 30-180 min. adTrx1 and adTrxR1 significantly reduced the increases in O2•- level in H2O2-treated HPASM cells at 24 h. Furthermore, HPASM cells transfected with Trx1 or TrxR1 siRNA showed increases in ROS levels with or without H2O2 at 5 min. While H2O2 transiently decreased GSH level at 5 min, Trx1 and TrxR1 siRNA intensified the decrease in GSH level. In conclusion, upregulation of Trx1 and TrxR1 significantly attenuated cell growth inhibition and death in H2O2-treated HPASM cells. As a whole, Trx-related adenoviruses diminished H2O2-induced ROS level in HPASM cells whereas Trx-related siRNAs increased ROS levels and decreased GSH level in these cells.


Asunto(s)
Peróxido de Hidrógeno/toxicidad , Miocitos del Músculo Liso/efectos de los fármacos , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Tiorredoxinas/metabolismo , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Glutatión/metabolismo , Humanos , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/citología , ARN Interferente Pequeño/administración & dosificación , Especies Reactivas de Oxígeno/metabolismo , Reductasa de Tiorredoxina-Disulfuro/genética , Tiorredoxinas/genética , Transfección , Regulación hacia Arriba
15.
BMC Public Health ; 19(1): 267, 2019 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-30841877

RESUMEN

BACKGROUND: Cigarette smoking is a major health risk, particularly in male South Koreans. Smoking cessation can benefit health; however, the process of quitting smoking is difficult to some smokers and shows its relationship to their stress level. The hypothesis of this study is that who has failed attempts to stop smoking induce more stress than habitual smoking. METHODS: To test this, the analysis on the association between smoking cessation attempts and stress levels in smokers was performed. The Korean Community Health Survey (2011-2016) data with the total of 488,417 participants' data were used for this study. Survey data were analyzed using the chi-square test and logistic regression. As the dependent variable, self-reported level of stress was selected. RESULTS: Of the subject population, 78.3% (63.3% males, 81.4% females) felt stressed. Among participants who successfully stopped smoking, 73.0% (72.6% males, 78.1% females) reported feeling stressed. In contrast, of those who failed to stop smoking, 83.3% (83.6% males, 86.3% females) reported high stress levels. Among those who did not attempt smoking cessation, 81.1% (81.2% males, 80.3% females) responded that they experienced stress. Those who failed to stop smoking had higher odds of stress than those who did not attempt smoking cessation [odds ratio (OR) 1.11, 95% confidence interval (CI) 1.09-1.14, p < 0.001]. Those who successfully stopped smoking had lower odds of stress than those who did not attempt smoking cessation (OR 0.87, 95% CI 0.86-0.89, p < 0.001). CONCLUSION: The study found an association between unsuccessful smoking cessation and stress level. As the result, people who failed smoking cessation showed higher stress. These data should be considered in health policy recommendations for smokers.


Asunto(s)
Cese del Hábito de Fumar/psicología , Estrés Psicológico/epidemiología , Adulto , Fumar Cigarrillos/psicología , Femenino , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Oportunidad Relativa , República de Corea/epidemiología , Adulto Joven
16.
Comput Struct Biotechnol J ; 17: 186-194, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30809323

RESUMEN

The emergence and reemergence of coronavirus epidemics sparked renewed concerns from global epidemiology researchers and public health administrators. Mathematical models that represented how contact tracing and follow-up may control Severe Acute Respiratory Syndrome (SARS) and Middle East Respiratory Syndrome (MERS) transmissions were developed for evaluating different infection control interventions, estimating likely number of infections as well as facilitating understanding of their likely epidemiology. We reviewed mathematical models for contact tracing and follow-up control measures of SARS and MERS transmission. Model characteristics, epidemiological parameters and intervention parameters used in the mathematical models from seven studies were summarized. A major concern identified in future epidemics is whether public health administrators can collect all the required data for building epidemiological models in a short period of time during the early phase of an outbreak. Also, currently available models do not explicitly model constrained resources. We urge for closed-loop communication between public health administrators and modelling researchers to come up with guidelines to delineate the collection of the required data in the midst of an outbreak and the inclusion of additional logistical details in future similar models.

17.
J Clin Med ; 8(1)2019 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-30621064

RESUMEN

Imperforate hymen (IH) is an uncommon congenital anomaly of the female genital tract, with the hymen completely obstructing the vaginal opening. Despite the simple diagnosis and treatment of IH, missed or delayed diagnosis is often a clinical problem owing to its low incidence, nonspecific symptoms, or insufficient physical examination. The aim of this study is to identify the characteristics, clinical presentations, treatment modalities, and outcomes of imperforate hymen patients. In this study, a literature search of PubMed, Scopus and Medline databases was performed for sources published up to 3 July 2018 for English-language studies with the term "imperforate hymen". The literature review identified 251 citations and 155 articles (143 case reports, 12 case series) containing 253 patients who were finally included (two papers were not written in English). Among 236 postnatal patients, the mean age of the patients was 10.7 ± 4.7 years. Abdominal pain (54.2%), urinary retention (20.3%), abnormal menstruation (14.0%), dysuria (9.7%), increased urinary frequency (5.1%), severe presentation of renal failure (n = 5, 2.1%), and urinary tract infection (n = 1, 0.4%) were presented. Most patients diagnosed with the condition underwent surgical treatment (83.5%), most of whom were treated via a hymenotomy (35.2%) and hymenectomy (36.4%), and the use of prophylactic antibiotics were only used in 7 patients. There were no differences in outcomes between two surgical methods. In addition, 141 (59.7%) patients showed improvement and 5 deceased patients were not related to IH or the operation itself; Complications, such as vaginal adhesion, were only noted in 6.6% of patients. In addition, among 17 cases of newborns with a diagnosis of IH before birth, hymenectomy (n = 5, 29.4%) and hymenotomy (n = 9, 52.9%) were the main treatment modalities and showed improved prognosis in 52.9% of newborns. Because IH diagnosis is easy and postsurgical prognosis is good, clinicians should carefully examine every female patient at birth. IH should be considered regarding adolescent girls with abdominal pain, lower back pain, or urinary retention, and perform appropriate physical examinations of the genital introitus. In addition, accurate diagnosis as IH, not misdiagnosing as vaginal septum or agenesis, is important to prevent severe complications such as stricture and ascending infection.

18.
Oncol Rep ; 40(3): 1787-1794, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29956787

RESUMEN

Hydrogen peroxide (H2O2) is frequently applied to cultured cells to induce oxidative stress. The present study investigated the molecular and cellular effects of exogenous H2O2 on Calu­6 and A549 lung cancer cells. Based on MTT assays, H2O2 inhibited the growth of Calu­6 and A549 cells with IC50 values of ~50 and 100 µM at 24 h, respectively. Cells treated with H2O2 demonstrated a considerable G1­phase arrest of the cell cycle. H2O2 dose­dependently augmented the numbers of dead (trypan blue­positive) and Annexin V­FITC­stained cells in these cells, which was accompanied by the reduction of Bcl­2 and pro­caspase­3 levels, as well as the upregulation of caspase­3 and ­8 activities. In addition, H2O2 triggered the failure of mitochondrial membrane potential (MMP; ΔΨm). However, relatively higher doses of H2O2 did not raise the percentages of sub­G1 cells in these cell lines. All the tested caspase inhibitors (Z­VAD for pan­caspases, Z­DEVD for caspase­3, Z­IETD for caspase­8 and Z­LEHD for caspase­9) decreased the percentages of sub­G1 and Annexin V­FITC­stained cells in the H2O2­treated Calu­6 and A549 cells. However, caspase inhibitors did not significantly prevent the loss of MMP (ΔΨm) in H2O2­treated lung cancer cells. In conclusion, H2O2 inhibited the growth of Calu­6 and A549 lung cancer cells through cell death and G1­phase arrest. H2O2­induced cell death resulted from necrosis, as well as caspase­dependent apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Neoplasias Pulmonares/patología , Oxidantes/farmacología , Caspasas/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Células Tumorales Cultivadas
19.
Environ Toxicol ; 2018 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-29708299

RESUMEN

Arsenic trioxide (ATO; As2 O3 ) induces cell death in various cells via oxidative stress. Expose to chronic arsenic is involved in the development of vascular diseases. However, little is known about the cytotoxic effects of ATO on human normal vascular smooth muscle cells (VSMCs). Thus, in this study, we investigated the effects of ATO on cell growth and death in human pulmonary artery smooth muscle (HPASM) cells in relation to reactive oxygen species (ROS) and glutathione (GSH) levels. ATO treatment decreased the growth of HPASM cells with an IC50 of ∼30-50 µM at 24 h, and ATO induced HPASM cell death via apoptosis or necrosis dependent on the doses of it at this time. Treatment with 50 µM ATO did not increase ROS levels at the early time points, but it significantly increased mitochondrial O2•- levels at 24 h. ATO also induced GSH depletion in HPASM cells. N-acetyl cysteine (NAC; a well-known antioxidant) did not significantly affect apoptotic cell death, ROS levels, or GSH depletion in ATO-treated HPASM cells. However, l-buthionine sulfoximine (BSO; an inhibitor of GSH synthesis) intensified mitochondrial O2•- levels in ATO-treated HPASM cells, and significantly increased cell death and GSH depletion in these cells as well. In summary, we provided the first evidence that ATO inhibited the growth of HPASM cells, and induced apoptotic and/or necrotic cell death in these cells, accompanied by increases in mitochondrial O2•- level and GSH depletion.

20.
Mol Cell Biochem ; 441(1-2): 125-134, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28887781

RESUMEN

Exogenous hydrogen peroxide (H2O2) induces oxidative stress and apoptosis in cancer cells. This study evaluated the antiapoptotic effects of pan-caspase and caspase-3, -8, or -9 inhibitors on H2O2-treated Calu-6 and A549 lung cancer cells in relation to reactive oxygen species (ROS) and glutathione (GSH). Treatment with 50-500 µM H2O2 inhibited the growth of Calu-6 and A549 cells at 24 h and induced apoptosis in these cells. All the tested caspase inhibitors significantly prevented cell death in H2O2-treated lung cancer cells. H2O2 increased intracellular ROS levels, including that of O 2·- , at 1 and 24 h. It also increased the activity of catalase but decreased the activity of SOD. In addition, H2O2 triggered GSH deletion in Calu-6 and A549 cells at 24 h. It reduced GSH levels in Calu-6 cells at 1 h but increased them at 24 h. Caspase inhibitors decreased O 2·- levels in H2O2-treated Calu-6 cells at 1 h and these inhibitors decreased ROS levels, including that of O 2·- , in H2O2-treated A549 cells at 24 h. Caspase inhibitors partially attenuated GSH depletion in H2O2-treated A549 cells and increased GSH levels in these cells at 24 h. However, the inhibitors did not affect GSH deletion and levels in Calu-6 cells at 24 h. In conclusion, H2O2 induced caspase-dependent apoptosis in Calu-6 and A549 cells, which was accompanied by increases in ROS and GSH depletion. The antiapoptotic effects of caspase inhibitors were somewhat related to the suppression of H2O2-induced oxidative stress and GSH depletion.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores de Caspasas/farmacología , Caspasas/metabolismo , Peróxido de Hidrógeno/farmacología , Neoplasias Pulmonares/enzimología , Proteínas de Neoplasias/antagonistas & inhibidores , Estrés Oxidativo/efectos de los fármacos , Células A549 , Humanos , Neoplasias Pulmonares/patología , Proteínas de Neoplasias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...