• Title/Summary/Keyword: N-Acetylcysteine

Search Result 133, Processing Time 0.024 seconds

Effects of N-acetylcysteine on biofilm formation by MBR sludge

  • Song, WonJung;Lade, Harshad;Yu, YoungJae;Kweon, JiHyang
    • Membrane and Water Treatment
    • /
    • v.9 no.3
    • /
    • pp.195-203
    • /
    • 2018
  • N-acetylcysteine (NAC) has been widely used as an initial mucolytic agent and is generally used as an antioxidant to help alleviate various inflammatory symptoms. NAC reduces bacterial extracellular polymeric substances (EPS) production, bacterial adhesion to the surface and strength of mature biofilm. The efficacy has been shown to inhibit proliferation of gram-positive and gram-negative bacteria. In membrane bioreactor (MBR) processes, which contain a variety of gram negative bacteria, biofilm formation has become a serious problem in stable operation. In this study, use of NAC as an inhibitor of biofilm contamination was investigated using the center for disease control (CDC) reactors with MBR sludge. Biomass reduction was confirmed with CLSM images of membrane surfaces by addition of NAC, which was more efficient as the concentration of NAC was increased to 1.5 mg/mL. NAC addition also showed decreases in EPS concentrations of the preformed biofilm, indicating that NAC was able to degrade EPS in the mature biofilm. NAC addition was also effective to inhibit biofilm formation by MBR sludge, which consisted of various microorganisms in consortia.

Suppression of Arsenic Trioxide-induced Apoptosis in HeLa Cells by N-Acetylcysteine

  • Han, Yong Hwan;Kim, Sung Zoo;Kim, Suhn Hee;Park, Woo Hyun
    • Molecules and Cells
    • /
    • v.26 no.1
    • /
    • pp.18-25
    • /
    • 2008
  • Arsenic trioxide (ATO) can affect many biological functions such as apoptosis and differentiation in various cells. We investigated the involvement of ROS and GSH in ATO-induced HeLa cell death using ROS scavengers, especially N-acetylcysteine (NAC). ATO increased intracellular ${O_2}^{{\cdot}-}$ levels and reduced intracellular GSH content. The ROS scavengers, Tempol, Tiron and Trimetazidine, did not significantly reduce levels of ROS or GSH depletion in ATO-treated HeLa cells. Nor did they reduce the apoptosis induced by ATO. In contrast, treatment with NAC reduced ROS levels and GSH depletion in the ATO-treated HeLa cells and prevented ATO-induced apoptosis. Treatment with exogenous SOD and catalase reduced the depletion of GSH content in ATO-treated cells. Catalase strongly protected the cells from ATO-induced apoptosis. In addition, treatment with SOD, catalase and NAC slightly inhibited the G1 phase accumulation induced by ATO. In conclusion, NAC protects HeLa cells from apoptosis induced by ATO by up-regulating intracellular GSH content and partially reducing the production of ${O_2}^{{\cdot}-}$.

Factors of Determining N-acetylcysteine Administration in Patients with Acute Acetaminophen Poisoning (급성 아세트아미노펜 중독에서 N-acetylcysteine 투여 결정 관련 인자)

  • Lee, Jeong Hwa;Choi, Sangchun;Yoon, Sang Kyu;Shin, Kyu Cheol
    • Journal of The Korean Society of Clinical Toxicology
    • /
    • v.18 no.2
    • /
    • pp.78-84
    • /
    • 2020
  • Purpose: In acute acetaminophen poisoning, the administration of N-acetylcysteine (NAC) can effectively treat the main complications, such as kidney injury and liver failure. In the current situation, measurements of the acetaminophen concentration are not checked in the usual medical facilities. Therefore, this study examined the factors of determining the administration of NAC in addition to the stated amount of intake. Methods: The medical records of patients who visited Ajou University Hospital emergency center with acetaminophen poisoning from January 2015 to December 2019 were reviewed retrospectively. One hundred and seventy-nine patients were initially included. Among these patients, 82 patients were finally selected according to the inclusion criteria in the study. The inclusion criteria were as follows: patients who were 15 years of age or older; those whose ingested dose, ingested time, and body weight were clearly identified; and patients whose acetaminophen sampling time was within 24 hours. Patients were divided into two groups: NAC administered vs. non-NAC administered. The following variables were compared in these two groups: ingested dose, ingested dose per body weight, hospital arrival time after ingestion, suicide attempt history, psychiatric disease history, classification of toxic/non-toxic groups, duration of hospitalization, and laboratory results. Results: Univariate analysis revealed the ingested dose per body weight, hospital arrival time after ingestion, suicide attempt history, and psychiatric disease history to be the determining factors in administering NAC. Logistic regression analysis confirmed that the ingested dose per body weight was the only significant factor leading to an NAC treatment decision. (Odds ratio=1.039, 95% Confidential interval=1.009-1.070, p=0.009) Conclusion: The ingested dose per body weight was the only determining factor for administering NAC in patients with acute acetaminophen poisoning. On the other hand, additional criteria or indicators for the NAC administration decision will be necessary considering the inaccuracy of the ingested dose per body weight and the efficiency of NAC administration.

Generation of Reactive Oxygen Species Contributes to the Development of Carbon Black Cytotoxicity to Vascular Cells

  • Lee, Jong-Gwan;Noh, Won-Jun;Kim, Hwa;Lee, Moo-Yeol
    • Toxicological Research
    • /
    • v.27 no.3
    • /
    • pp.161-166
    • /
    • 2011
  • Carbon black, a particulate form of pure elemental carbon, is an industrial chemical with the high potential of occupational exposure. Although the relationship between exposure to particulate matters (PM) and cardiovascular diseases is well established, the cardiovascular risk of carbon black has not been characterized clearly. In this study, the cytotoxicity of carbon black to vascular smooth muscle and endothelial cells were examined to investigate the potential vascular toxicity of carbon black. Carbon black with distinct particle size, N330 (primary size, 28~36 nm) and N990 (250~350 nm) were treated to A-10, rat aortic smooth muscle cells and human umbilical vein endothelial cell line, ECV304, and cell viability was assessed by lactate dehydrogenase (LDH) leakage assay. Treatment of carbon black N990 resulted in the significant reduction of viability in A-10 cells at 100 ${\mu}g$/ml, the highest concentration tested, while N330 failed to cause cell death. Cytotoxicity to ECV304 cells was induced only by N330 at higher concentration, 200 ${\mu}g$/ml, suggesting that ECV304 cells were relatively resistant to carbon black. Treatment of 100 ${\mu}g$/ml N990 led to the elevation of reactive oxygen species (ROS) detected by dichlorodihydrofluorescein (DCF) in A-10 cells. Pretreatment of antioxidants, N-acetylcysteine (NAC) and sulforaphane restored decreased viability of N990-treated A-10 cells, and N-acetylcysteine, but not sulforaphane, attenuated N990-induced ROS generation in A-10 cells. Taken together, present study shows that carbon black is cytotoxic to vascular cells, and the generation of reactive oxygen contributes to the development of cytotoxicity. ROS scavenging antioxidant could be a potential strategy to attenuate the toxicity induced by carbon black exposure.

Acetaminophen Poisoning (아세트아미노펜 중독)

  • Chung, Sung-Pil;Kim, Seung-Ho;Lee, Hahn-Shick
    • Journal of The Korean Society of Clinical Toxicology
    • /
    • v.6 no.1
    • /
    • pp.1-8
    • /
    • 2008
  • Acetaminophen (AAP) overdose can result in potentially serious hepatotoxicity. The ingested dose and time from ingestion to presentation are important prognostic factors. Toxic dose in adult is thought to be at least 10 g or 200 mg/kg. However, early management of acute overdose should be guided by the plasma AAP concentration. The antidote for AAP poisoning is N-acetylcysteine (NAC). It provides complete protection against hepatotoxicity if given within 8 h of acute overdose. If the concentration is above the possible toxicity line as predicted by the Rumack-Matthew nomogram, either the 72-hr oral or the 20-hr intravenous NAC regimen should be administered. NAC is also effective if started late in patients with established hepatic failure. This article summarizes the current consensus of clinical assessment and management for acute AAP overdose.

  • PDF

Effect of Depletion and Oxidation of Cellular GSH on Cytotoxicity of Mitomycin Small Cell Lung Cancer Cells

  • Lee, Chung-Soo
    • Biomolecules & Therapeutics
    • /
    • v.12 no.2
    • /
    • pp.92-100
    • /
    • 2004
  • Effect of the depletion or oxidation of GSH on mitomycin c (MMC)-induced mitochondrial damage and cell death was assessed in small cell lung cancer (SCLC) cells. MMC induced cell death and the decrease in the GSH contents in SCLC cells, which were inhibited by z-LEHD.fmk (a cell permeable inhibitor of caspase-9), z-DQMD.fmk (a cell permeable inhibitor of caspase-3) and thiol compound, N-acetylcysteine. MMC caused nuclear damage, release of cytochrome c and activation of caspase-3, which were reduced by N-acetylcysteine. The depletion of GSH due to L-butionine-sulfoximine enhanced the MMC-induced cell death and formation of reactive oxygen species in SCLC cells, whereas the oxidation of GSH due to diamide or $NH_2Cl$ did not affect cytotoxicity of MMC. The results show that MMC may cause cell death in SCLC cells by inducing mitochondrial dysfunction, leading to activation of caspase-9 and -3. The MMC-induced change in the mitochondrial membrane permeability, followed by cell death, in SCLC cells may be significantly enhanced by the depletion of GSH. In contrast, the oxidation of GSH may not affect cytotoxicity of MMC.

Mouse Melanoma Cell Migration is Dependent on Production of Reactive Oxygen Species under Normoxia Condition

  • Im, Yun-Sun;Ryu, Yun-Kyoung;Moon, Eun-Yi
    • Biomolecules & Therapeutics
    • /
    • v.20 no.2
    • /
    • pp.165-170
    • /
    • 2012
  • Cell migration plays a role in many physiological and pathological processes. Reactive oxygen species (ROS) produced in mammalian cells influence intracellular signaling processes which in turn regulate various biological activities. Here, we investigated whether melanoma cell migration could be controlled by ROS production under normoxia condition. Cell migration was measured by wound healing assay after scratching confluent monolayer of B16F10 mouse melanoma cells. Cell migration was enhanced over 12 h after scratching cells. In addition, we found that ROS production was increased by scratching cells. ERK phosphorylation was also increased by scratching cells but it was decreased by the treatment with ROS scavengers, N-acetylcysteine (NAC). Tumor cell migration was inhibited by the treatment with PD98059, ERK inhibitor, NAC or DPI, well-known ROS scavengers. Tumor cell growth as judged by succinate dehydrogenase activity was inhibited by NAC treatment. When mice were intraperitoneally administered with NAC, the intracellular ROS production was reduced in peripheral blood mononuclear cells. In addition, B16F10 tumor growth was significantly inhibited by in vivo treatment with NAC. Collectively, these findings suggest that tumor cell migration and growth could be controlled by ROS production and its downstream signaling pathways, in vitro and in vivo.

Curcumin-induced Growth Inhibitory Effects on HeLa Cells Altered by Antioxidant Modulators

  • Hong, Jung-Il
    • Food Science and Biotechnology
    • /
    • v.16 no.6
    • /
    • pp.1029-1034
    • /
    • 2007
  • Curcumin (diferuloyl methane), originated rhizomes of Curcuma longa L. has been suggested as an anti-inflammatory and anti-carcinogenic agent. In the present study, modulation of cytotoxic effects of curcumin on HeLa cells by different types of antioxidants was investigated. Cytotoxic effects of curcumin were significantly enhanced in the presence of superoxide dismutase (SOD) by decreasing $IC_{50}$ to 15.4 from $26.0\;{\mu}M$ after 24 hr incubation; the activity was not altered by catalase. The effect of curcumin was significantly less pronounced in the presence of 4 mM N-acetylcysteine (NAC). Low concentration (<1 mM) of NAC, however, increased the efficacy of curcumin. Cysteine and ${\beta}$-mercaptoethanol that have a thiol group, showed the similar biphasic patterns as NAC for modulating curcumin cytotoxicity, which was, however, constantly enhanced by ascorbic acid, a non-thiol antioxidant. In the presence of SOD, ascorbic acid, and 0.5 mM NAC, cellular levels of curcumin were significantly increased by 31-66%, whereas 4 mM NAC decreased the level. The present results indicate that thiol reducing agents showed a biphasic effect in modulating cytotoxicity of curcumin; it is likely that their thiol group is reactive with curcumin especially at high concentrations.

6-Hydroxydopamine-induced Adaptive Increase in GSH Is Dependent on Reactive Oxygen Species and Ca2+ but not on Extracellular Signal-regulated Kinase in SK-N-SH Human Neuroblastoma Cells

  • JIN Da-Qing;Park Byung CHUL;KIM Jung-Ae
    • Biomolecules & Therapeutics
    • /
    • v.13 no.4
    • /
    • pp.256-262
    • /
    • 2005
  • We examined the signaling molecules involved in the 6-hydroxydopamine (6-OHDA)-induced neuronal cell death and increase in cellular glutathione (GSH) level in SK-N-SH cells. The 6-OH-DA-induced cell death was significantly prevented by the pretreatment with N-acetylcysteine (NAC), a thiol antioxidant, and BAPTA, an intracellular $Ca^{2+}$ chelator. Although 6-OHDA induced ERK phosphorylation, the pretreatment with PD98059, an ERK inhibitor, did not block 6-OHDA-induced cell death. In addition, the 6-OHDA-induced activation of caspase-3, a key signal for apoptosis, was blocked by the pretreatment with NAC and BAPTA. While the level of reactive oxygen species (ROS) was significantly increased in the 6-OHDA-treated cells, the cellular GSH level was not altered for the first 6-hr exposure to 6-OHDA, but after then, the level was significantly increased, which was also blocked by the pretreatment with NAC and BAPTA, but not by PD98059. Depletion of GSH by pretreating the cells with DL-buthionine-(S,R)-sulfoximine (BSO), a glutathione synthesis inhibitor, rather significantly potentiated the 6-OHDA-induced death. In contrast to the pretreatment with NAC, 6-OHDA-induced cell death was not prevented by the post-treatment with NAC 30 min after 6-OHDA treatment. The results indicate that the GSH level which is increased adaptively by the 6-OHDA-induced ROS and intracellular $Ca^{2+}$ is not enough to overcome the death signal mediated through ROS-$Ca^{2+}$ -caspase pathway.

Rosuvastatin Induces ROS-mediated Apoptosis in Human Prostate Cancer PC-3 Cells (Rosuvastatin이 유도하는 ROS가 전립선암 PC-3 세포주의 세포사멸 유도에 미치는 영향)

  • Choi, Hyeun Deok;Baik, Jong Jin;Kim, Sang Hun;Yu, Sun Nyoung;Chun, Sung Hak;Kim, Young Wook;Nam, Hyo Won;Kim, Kwang Youn;Ahn, Soon Cheol
    • Journal of Life Science
    • /
    • v.26 no.4
    • /
    • pp.398-405
    • /
    • 2016
  • Statins, the inhibitors of 3-hydroxy 3-methylglutaryl coenzyme A (HMG-CoA) reductase, are widely used in treatments of hypercholesterolemia and newly known as anti-cancer effect of various cancer cells. Recently, several studies suggested that reactive oxygen species (ROS) play a critical role on cell death signaling. However, mechanism of ROS by rosuvastatin is currently unclear. This study aimed to explore the molecular mechanism of apoptosis by rosuvastatin in human prostate cancer PC-3 cells. Cell viability and apoptosis-related protein expression were measured by MTT assay and western blotting, respectively. In addition, the levels of apoptosis and ROS were analyzed. The results showed that rosuvastatin dramatically reduced cell viability in a dose- and time-dependent manner. We confirmed that rosuvastatin induced apoptosis through reduction of procaspase-3 and cleavage of poly (ADP-ribose) polymerase (PARP) in PC-3 cells. In addition, rosuvastatin stimulated ROS production in a dose-dependent manner and pre-treatment with N-acetylcysteine (NAC), a ROS scavenger, significantly recovered rosuvastatin-induced ROS and apoptosis. Thus, we concluded that rosuvastain induces apoptosis through generation of ROS in human prostate cancer PC-3 cells and provides a promising approach to improve the efficacy of cancer therapy.