Journal Search Engine
Search Advanced Search Adode Reader(link)
Download PDF Export Citaion korean bibliography PMC previewer
ISSN : 1225-1577(Print)
ISSN : 2384-0900(Online)
The Korean Journal of Oral and Maxillofacial Pathology Vol.43 No.6 pp.255-267
DOI : https://doi.org/10.17779/KAOMP.2019.43.6.003

Preliminary Study on Hydrogen Peroxide-Induced Cellular Responses in RAW 264.7 Cells as Determined by IP-HPLC

Yeon Sook Kim
Department of Dental Hygiene, College of Health & Medical Sciences, Cheongju University, Cheongju; Korea
Correspondence: Yeon Sook Kim, Department of Dental Hygiene, College of Health & Medical Sciences, Cheongju University, Cheongju; Korea Tel: +82-43-229-8996, Fax: +82-43-229-8969 E-mail: yeonsookkim1@naver.com
November 10, 2019 November 19, 2019 December 6, 2019

Abstract


Hydrogen peroxide (H2O2) is originally an endogenous small molecule which is reduced into water in cells. In order to know the H2O2-induced oxidative stress in RAW 264.7 cells, first of all, the optimum concentration of exogenous H2O2 which show reactive cellular responses was determined as 40 μM by MTT assay, and followed by 40 μM H2O2 application in RAW 264.7 cells for 30 min, 1, or 2 hours. The expressional changes of essential proteins for cellular proliferation, epigenetic modification, inflammation, apoptosis, survival, and protection were assessed by immunoprecipitation high performance liquid chromatography (IP-HPLC) using 51 antisera. 40 μM H2O2 treatment down-regulated proliferation-related proteins, Ki-67, PCNA, CDK4, cyclin D2, cMyc, and PLK4, induced histone methylation/deacetylation and DNA methylation by increasing levels of HDAC10 and DMAP1 and by decreasing levels of DNMT1 and KDM4D, activated inflammatory reaction by increasing levels of MCP-1, COX-2, CD68, LTA4H, CXCR4, and lysozyme, and dramatically up-regulated cellular apoptosis-, survival-, and protection-related proteins, AIF, PARP-1, caspase 9, c-caspase 9, pAKT1/2/3, SOD-1, HO-1, NF-kB, NRF2, and GSTO1 in RAW 264.7 cells. These observations suggest exogenous 40 μM H2O2-induced oxidative stresses which resulted global cellular responses including not only antioxidant, inflammation, and apoptosis but also proliferation and epigenetic modification. Particularly, 40 μM H2O2-induced apoptosis was mainly derived from PARP-1/AIF signaling leading parthanatos, and 40 μM H2O2-induced suppression of cMyc/MAX/MAD network was relevant to reduction of RAW 264.7 cell proliferation. Accordingly, H2O2 appears to affect RAW 264.7 macrophages in several ways eliciting not only oxidative stresses but also genome-wide DNA damage.



RAW 264.7 세포에서 과산화수소에 의한 세포반응을 IP-HPLC로 확인하는 예비연구

김 연숙
청주대학교 보건의료과학대학 치위생학과

초록


    Cheongju University

    Ⅰ. INTRODUCTION

    Oxidative stress is usually induced by excess accumulation of reactive oxygen and nitrogen species in cells. Reactive oxygen species (ROS), especially hydrogen peroxide (H2O2), are well-known mediators of oxidative damage and cell demise, nevertheless the mechanisms by which they induce specific cell death modalities and the dose-dependent temporal/ spatial molecular signaling are not clearly elucidated. High concentration of H2O2 is known to induce cell death (apoptosis) in a variety of cell types by affecting intracellular signal transduction pathways. In particular, c-Jun N-terminal kinase (JNK) and p38 MAP (mitogen-activated protein) kinase are triggered by extracellular stimuli, e.g., ultraviolet radiation and free radicals including H2O2 and NO.1-5)

    H2O2, the non-radical 2-electron reduction product of oxygen, is normally produced by aerobic metabolic pathways to the level of about 10 nm intracellular concentration in different cells. In liver, it is produced at 50 μmol/min/kg of tissue, which is about 2% of total oxygen absorption at steady condition.6) Metabolically generated H2O2 emerged from recent researches as a central hub in redox signaling and/or oxidative stress. Upon generation by major products, the NADPH oxidases or Complex III of mitochondrial respiratory system, H2O2 is under fine control of peroxiredoxins and glutathione peroxidases with their feedback systems as well as by different catalases.7-10) Of note, H2O2 is recognized as a second messenger in insulin signaling and in several growth factor-induced signaling cascades.11-13) H2O2 transport across membranes is facilitated by aquaporins, also known as peroxiporins.14) As specialized protein cysteines operate as redox switches using H2O2 as thiol oxidant, this reactive oxygen species important for poising the set point of redox proteome, it was suggested that major cellular processes including proliferation, differentiation, apoptosis, tissue repair, inflammation, survival, circadian rhythm, and aging use this low molecular weight oxygen metabolite, H2O2, as signaling compound.6)

    Hydrogen peroxide is known as major redox metabolite for signaling and redox regulation under physiological conditions of H2O2 (1-10nM), whereas higher concentrations induce adaptive stress responses via master signaling switches such as Nrf2/Keap1 or NF-kB.14) Supra-physiological concentrations of H2O2 (>100nM) result in damage of biomolecules, represented as oxidative distress.14)

    It has been known that the p38 and JNK activations by H2O2 are required for apoptosis.1-3,15,16) Furthermore, it has been reported that H2O2 increases the expression of apoptosis-related surface molecule FAS/APO-1 (CD95) and that it induces apoptosis via CD95-mediated signaling.5,17,18)

    In the present in vitro study, the first of all, the optimal biohazard concentration of H2O2 was determined by cell viability analysis, which showed favorable cell survival more than 80% in 3 hours and about 50% cell survival (LD50) in 16 hours. The selected H2O2 concentration, 40 μM, was applied in RAW 264.7 macrophages, and assessed their protein expressions by IP-HPLC to know the dynamic molecular mechanism of H2O2 in cells, which was hardly recognized by ordinary detection methods, e.g., Western blot, RT-PCR (reverse transcription polymerase chain reaction), ELISA (enzyme-linked immunosorbent assay), proteomics, DNA microarray, etc.

    Immunoprecipitation high-performance liquid chromatography (IP-HPLC) has been developed to resolute the previous complication in technique and limited applicability.19,20) A new IP-HPLC protocol was used to assess protein expression levels in various biological fluids, such as urine, blood serum, saliva,21) inflammatory exudates,22-24) and different protein extracts from cells,25,26) liver,27) and cancer tissues.28) IP-HPLC is similar to ELISA, but the former measures protein expression level using protein A/G agarose beads in buffer solution and UV spectroscopy, whereas the latter uses fluorescence-c onjugated antibodies pre-attached on plastic wells and fluoroscopy. Furthermore, multiple trials have shown that IP-HPLC is available to get multiple results of protein levels accurately (±5% standard deviation) and reproducibly.

    Ⅱ. MATERIALS & METHODS

    1. RAW264.7 cell culture in the presence of H2O2

    RAW 264.7 cells, an immortalized murine macrophage cell line (ATCC, USA), were cultured in Dulbecco’s modified Eagle’s medium (DMEM, WelGene Inc. Korea) supplemented with 10% heat-inactivated fetal bovine serum (WelGene Inc. Korea), 100μg/mL streptomycin, 100 unit/mL penicillin, and 250ng/mL amphotericin B (WelGene Inc. Korea) in 5% CO2 at 37.5°C.26) To detect protein expressional changes, RAW 264.7 cells were cultured in antigen-free media.

    About 70% confluent RAW 264.7 cells spreaded on Petri dish surfaces were treated with 40 μM H2O2 (Sigma, USA) for 30 min, 1, and 2 hours; the control cells were treated with 1 mL of normal saline. Cultured cells were gathered with protein lysis buffer (PRO-PREPTM, iNtRON Biotechnology INC, Korea) on ice and immediately preserved in deep freezer (-70°C) until required.

    2. Determination of optimal concentration of H2O2 in RAW 264.7 cell culture by cell viability analysis

    A series of MTT assay were carried out using different concentration of H2O2 in RAW 264.7 cell culture in order to determine the optimal responsible concentration of H2O2 for the cells. Viable cells were detected using MTT dye, which precipitates blue formazan crystals that are reduced by mitochondrial dehydrogenase present in living cells.29)

    Briefly, 5 x 103 cells were seeded in a 96-well plate and cultured overnight. The cells were treated with various concentrations of H2O2 (0-200μM) for 3 and 16 h until treatment with MTT. MTT assays were performed in accordance with general procedures.29) The reduced MTT was measured at 570 nm spectrophotometry in a micro plate reader (BioTek, USA).

    3. Immunoprecipitation high-performance liquid chromatography (IP-HPLC)

    About 100μg of protein extracts were utilized for immunoprecipitation using a protein A/G agarose column (Amicogen, Korea). Each protein A/G agarose columns was separately pre-incubated with 0.25 μg of 51 different antisera for; proliferation-related proteins (n=7), cMyc/MAX/MAD network proteins (n=4(1)), epigenetic modification-related proteins (n=6), inflammatory proteins (n=8), apoptosis and survival-related proteins (n=13), and protection-related proteins (n=14(3)) (( ); overlapped number) (Table 1).

    Briefly, each protein sample was mixed with 5 mL of immunoprecipitation binding buffer (150mM NaCl, 10mM Tris pH 7.4, 1mM EGTA, 1mM EDTA, 0.2mM PMSF, 0.2mM sodium vanadate, and 0.5% NP-40) and incubated in protein A/G agarose (Amicogen, Korea) columns pre-incubated with different antibodies on a rotating stirrer for 1 hour at 4°C.26) After washing columns with Tris-buffer (50 mM Tris, pH 7.5, 0.15M NaCl), target proteins were eluted using 250μL of IgG elution buffer (Pierce, USA). Immunoprecipitated proteins were detected using a HPLC apparatus (1100 series, Agilent, USA) equipped with a reverse phase silicate column and a micro-analytical detector system (SG Highteco, Korea). HPLC analysis was performed using mobile buffer solution (0.15M NaCl/20% acetonitrile) at 0.4 mL/min for 15 min, and proteins were detected using a UV spectrometer at 280 nm. Control and experimental samples were analyzed sequentially to allow comparisons. For IP-HPLC, immunoprecipitated protein peak areas (mAU*s) were mathematically calculated by subtracting negative control antibody peak areas from the whole protein peak areas, and square roots of immunoprecipitated protein peak areas were calculated to normalize concentrations.25,26) Protein percentages in total proteins in experimental and control groups were plotted into a line graph and a star plot. Results were analyzed using the Chi-squared test.25-27)

    The housekeeping proteins, α-tubulin, β-actin, and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) were used as internal controls. Expressional changes of housekeeping proteins were mathematically adjusted to < ±5% using a proportional basal line algorithm. Protein expressional changes of ≤ ±5%, ±5-10%, ±10-20%, and ≥±20% change were defined as minimal, slight, meaningful, or marked, respectively. It was reported that when the IP-HPLC results were compared with the western blot data of cytoplasmic housekeeping protein (β-actin), the IP-HPLC results exhibiting minute error ranges less than ±5% could be analyzed statistically, while the western blot results showed a large error range of more than 20%, thereby it was almost impossible to analyze them statistically.26)

    4. Statistical analysis

    Proportional data (%) of experimental and control groups were plotted into line graphs and star plots, and results were analyzed using the Chi-squared test. The expressions of control housekeeping proteins, that is, α-tubulin, β-actin, and GAPDH were almost non-responsive (≤5%) to 30 min, 1, and 2 hours of 40 μM H2O2 treatment.

    Ⅲ. RESULTS

    1. Determination of optimal concentration of H2O2 in RAW 264.7 cell culture

    In order to know the reactive cellular responses against H2O2-induced oxidative stresses in RAW 264.7 cells, the first of all, the optimal concentration of H2O2 was determined by MTT assay for cell viability test. Among 0 to 200 μM H2O2 applied to RAW 264.7 cells for 3 and 16 hours, 40 μM H2O2 showed 85.7% cell survival in 3 hours and LD50 cell survival (48%) in 16 hours (Fig. 1), therefore, the optimal responsible concentration of H2O2 was determined as 40 μM in this study for the H2O2-induced oxidative stresses in RAW 264.7 cells.

    2. Effects of 40 μM H2O2 on the expressions of proliferation-related proteins in RAW 264.7 cells

    RAW 264.7 cells treated with 40 μM H2O2 for 30 min, 1, or 2 hours exhibited decreases in levels of proliferationactivating proteins, Ki-67 (by 9.7% at 2 hours), proliferation cell nuclear antigen (PCNA, 3.4% at 1 hour), cyclin dependent kinase 4 (CDK4, 9.8% at 2 hours), polo-like kinase 4 (PLK4, 18.9% at 1 hour), and cyclin D2 (9.7% at 30 min), and increases in proliferation-inhibiting proteins, p15/16 (42.9% at 2 hours) and p27 (9.3% at 30 min) levels versus non-treated controls. Particularly, GAPDH necessary for important energy- yielding step in carbohydrate metabolism was downregulated by 40 μM H2O2, 15.7% at 2 hours, contrast to other housekeeping proteins, β-actin and α-tubulin which were minimally changed (< ±7%) (Figs. 2 A1 and A2).

    The expressions of proliferation-activating proteins, Ki-67, PCNA, PLK4, and cyclin D2 were decreased after 30 min of 40 μM H2O2 treatment, while the expressions of proliferationinhibiting proteins, p15/16 and p27 were markedly increased, so that these results suggest 40 μM H2O2 might have a strong anti-proliferative effect on RAW 264.7 cells.

    3. Effects of 40 μM H2O2 on the expressions of cMyc/MAX/MAD network-related proteins in RAW 264.7 cells

    RAW 264.7 cells treated with 40 μM H2O2 for 30 min, 1, or 2 hours exhibited decreases in the expressions of cMyc and MAX by 30.1% and 9.2%, respectively, after 2 hours treatment versus non-treated controls, while MAD-1 expression increased by a maximum of 18.9% at 30 min and maintained by 15.9% at 2 hours of treatment (Figs. 2 B1 and B2). The reduction of cMyc and MAX expressions while elevation of MAD-1 expression in cMyc/MAX/MAD network may contribute to the anti-proliferative effect of 40 μM H2O2 on RAW 264.7 cells. And subsequently, p27 expression was upregulated by 9.3% at 30 min but gradually decreased by 11.6% at 2 hours.

    4. Effects of 40 μM H2O2 on the expressions of epigenetic modification-related proteins in RAW 264.7 cells

    Histone H1 expression slightly decreased in 40 μM H2O2 treated cells to 91.3% at 1 hour and to 94.7% at 2 hours versus non-treated controls. Regarding histone methylation/ deacetylation, the expression of lysine-specific demethylase 4D (KDM4D) was increased by 13.8% at 30 min but gradually decreased by 12% at 2 hours, and that of histone deacetylase 10 (HDAC-10) and DNA methyltransferase 1-associated protein 1 (DMAP1) were similarly increased by 46.7% and 60.7%, respectively, at 1 hours and maintained by 21.1% and 32.1%, respectively, at 2 hours versus non-treated controls. With respect to DNA methylation, DNA (cytosine-5)- methyltransferase 1 (DNMT1) expression was greatly reduced by 43.5% and 34% at 30 min and 1 hour, respectively, but its decrease was recovered by 0.1% at 2 hours. And the expressions of methyl-CpG binding domain 4 (MBD4) was 10.2% higher at 2 hours (Figs. 2 C1 and C2). These results suggest 40 μM H2O2 increased histone methylation/deacetylation and DNA methylation simultaneously, and subsequently repressed DNA transcription in RAW 264.7 cells, and that this epigenetic effect of 40 μM H2O2 might be associated with genome-wide DNA damage.

    5. Effects of 40 μM H2O2 on the expressions of inflammation-related proteins in RAW 264.7 cells

    The most inflammatory proteins were upregulated by 40 μM H2O2 treatment; tumor necrosis factor α (TNFα, 11.9%), lysozyme (6.3%), leukotriene A4 hydrolase (LTA4H, 6.2%), C-X-C chemokine receptor type 4 (CXCR4, 11.9%), macrophage colony-stimulating factor (M-CSF, 6.9%), CD68 (13.4%), cyclooxygenase-2 (COX-2, 20.1%), and monocyte chemotactic proteins (MCP-1, 18.9%) versus non-treated controls. Particularly, the expressions of TNFα, CXCR4, and M-CSF were decreased by 9.2%, 10.3%, and 3.5% at 30 min, respectively, but reactively increased by 11.9%, 11.9%, and 6.9% at 2 hours (Figs. 3 A1 and A2). Resultantly, 40 μM H2O2 markedly upregulated the expressions of M1 macrophage differentiation cytokines, e.g., TNFα, LTA4H, CXCR4, CD68, COX-2, and MCP-1, indicating that RAW 264.7 cells were activated into M1 type macrophages by 40μM H2O2 treatment.

    6. Effects of 40 μM H2O2 on the expressions of apoptosis and survival-related proteins in RAW 264.7 cells

    40 μM H2O2 affected the expressions of p53-mediated apoptosis-related proteins for 2 hours treatment, particularly p53 protein, which was increased by 14% at 1 hour and maintained by 9.2% at 2 hours versus non-treated controls. The expressions of BAX and APAF-1 were increased by 16% at 30 min and 13% at 1 hour, respectively, while BCL2 expression was decreased by 18.4% at 1 hour. These expressions were resulted in the increases of caspase 9 (12.3%) and c-caspase 9 (8.6%) expressions (Figs. 3 B1 and B2). On the other hand, the expressions of PARP-1 and AIF were markedly increased by 27.2% at 1 hour and 48.2% at 2 hours, respectively, which suggest the increase of ssDNA damage and nuclear apoptosis of parthanatos.

    RAW 264.7 cells treated with 40 μM H2O2 showed upregulation of cell survival-related proteins as compared with non-treated controls. The expressions of pAKT1/2/3, telomerase reverse transcriptase (TERT), nuclear factor (erythroid-derived 2)-like 2 (NRF2), and AMP-activated protein kinase (AMPK) were increased by 19.7% at 2 hours, 13.9% at 30 min, 13.7% at 2 hours, and 8.5% at 1 hour, respectively. These results suggest that 40 μM H2O2 induced survival signaling in RAW 264.7 cells.

    7. Effects of 40 μM H2O2 on the expressions of cell protection-related proteins in RAW 264.7 cells

    The expressions of cell protection-related proteins in RAW 264.7 cells were increased by 40 μM H2O2; heat shock protein-70 (HSP-70) by 10.7% at 2 hours, HSP-90 by 14.3% at 30 min, heme oxygenase 1 (HO-1) by 17.8% at 1 hour, glutathione S-transferase ω 1 (GSTO1) by 18.1% at 30 min, Cu-Zn superoxide dismutase-1 (SOD-1) by 43.1% at 30 min, NRF2 by 13.7% at 2 hours, LC by 29.5% at 1 hour, sodiumdependent vitamin C transporter 2 (SVCT2) by 15.1% at 2 hours, leptin by 13.7% at 2 hours, and AMPK by 8.5% at 1 hour as compared with non-treated controls, while the expressions of nitric oxide synthase 1 (NOS1), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB), and mammalian target of rapamycin (mTOR) were decreased by 9.3% at 1 hour, 4.8% at 30 min, and by 9.3% at 30 min, respectively. Particularly, the expressions of antioxidantrelated proteins including heme oxygenase-1 (HO-1), glutathione S-transferase ω 1 (GSTO1), Cu-Zn superoxide dismutase-1 (SOD1), and sodium-dependent vitamin C transporter 2 (SVCT2) were consistently elevated, NRF2, a transcription factor the expression of antioxidant proteins was gradually increased for 2 hours, and the expressions of autophagosome protein (LC3) and anti-obesity protein (leptin) were markedly increased by 40 μM H2O2. (Figs. 3 C1 and C2).

    Ⅳ. DISCUSSION

    RAW 264.7 cells, murine macrophage cell line, are invaluable biological models for mechanistic studies of oxidative stress, because they usually play roles for anti-bacterial and scavenging effects using ROS. RAW 264.7 cells are relatively resistant to the hazard reaction of low H2O2 concentration but eventually succumbed to high H2O2 concentration similar to other cells. In order to know the primitive molecular signaling occurred by H2O2 treatment in RAW 264.7 cells, the optimal H2O2 concentration was determined by cell viability analysis and 40 μM H2O2 produced favorable cell survival over 80% for 3 hours and LD50 survival for 16 hours. Therefore, the present study was performed the H2O2-induced biohazard experiments using 40 μM H2O2 for 2 hours in RAW 264.7 cells. It was estimated that the cells would actively respond to the exogenous ROS derived from H2O2 treatment, survive well for 2 hours of experiment, represent early stages of molecular signaling for cellular proliferation, protection, apoptosis, survival, and adaptation in this study

    It is known that high levels of exogenous H2O2 induce extensive DNA damage, ATP depletion, and severe cytotoxicity, while low levels of exogenous H2O2 trigger a modest drop in ATP activity, delayed toxicity, G2/M arrest, and cell senescence.31) Oxygen free radicals derived from mitochondria induced a reversible ATP drop without affecting cell viability. Instead, cells became senescent after accumulating in the G1/S phase of the cell cycle.32-34) Concomitant inhibition of glycolysis was shown to make cells highly sensitive to death in the presence of different nontoxic concentrations of H2O2, perhaps by inhibiting the ATP-restoring mechanisms.31,35)

    In the present study, 40 μM H2O2 decreased the expressions of proliferation-activating proteins, Ki-67, PCNA, CDK4, cyclin D2, and PLK4, while increased the expressions of proliferation-inhibiting proteins, p15/16 and p27. Concurrently, cMyc and MAX were downregulated by H2O2, while MAD-1 expressions was upregulated. These expressional changes may indicate RAW 264.7 cell proliferation was potently arrested by H2O2 together with diminishment of carbohydrate metabolism in cells. As p27 expression is intimately regulated by cMyc/MAX/MAD network signaling, the increase of p27 expression may closely related to the reduction of MAX homodimer formation but the elevation of MAX-MAD 1 heterodimer formation in cMyc/MAX/MAD network.36,37)

    40 μM H2O2 greatly influenced on the inflammatory protein expressions of RAW 264.7 cells, murine macrophages. Most of inflammatory cytokines were upregulated for 2 hours of H2O2 treatment, among the affected inflammatory proteins, the increases in the expressions of M1 differentiation cytokines, e.g., TNFα (11.9%), LTA4H (6.2%), CXCR4 (11.9%), CD68 (13.4%), COX-2 (20.1%), and MCP-1 (18.9%) more dominant than the minimal increases of M2 differentiation cytokines, e.g., lysozyme (6.3%), and macrophage colony-stimulating factor (M-CSF, 6.9%). These expressional changes may indicate that 40 μM H2O2 potently activate RAW 264.7 cells to differentiate into M1 type macrophages which are utilized for the first line defense of inflammatory reaction.38,39)

    It has been reported that apoptosis is closely associated with oxidative stress in biological systems, and that caspases have been considered to play a pivotal role in the execution phase of apoptosis.3,40,41) In the present study, the expressions of p53-mediated apoptosis proteins, p53, BAX, APAF-1, caspase 9, and c-caspase 9, were increased after 30 min of 40 μM H2O2 treatment, but these expressions were diminished until 2 hours. On the other hands, PARP-1 and AIF were dominantly upregulated at 1 and 2 hours of 40 μM H2O2 treatment.

    H2O2 upregulates Bfl-1, an anti-apoptotic member of the BCL-2 family, which is responsible for the anti-apoptotic activity against ROS. When Jurkat cells and human leukemic T cells, were pretreated with 100 μM H2O2 and then treated with anti-FAS antibody, apoptosis was impaired without change of cell surface Fas expression.18) And 50 μM H2O2 increased telomerase activity, so that this expression was suspected for the BCL-2 increment and the oncogenic reactivation.42) In the present study, 40 μM H2O2 increased TERT expression by 13.9% at 30 min but decreased it by 5.3% at 1 hour and 9.1% at 2 hours, and the expression of BCL-2 was consistently downregulated for 2 hours. Therefore, it was considered that the transient elevation of TERT expression by 40 μM H2O2 would be only reactive to the apoptotic impact of ROS.

    In the present study, 40 μM H2O2 induced the intense expressions of cell protection proteins, including heat shock proteins (HSP-70 and HSP-90), antioxidant-related proteins (HO-1, GSTO1, SOD-1, and SVCT2), autophagosome-related protein (LC3), cellular energy homeostasis-related protein (AMPK), and a transcription factor regulating for the expression of antioxidant proteins (NRF2). Increasing evidence has demonstrated that heme oxygenase-1 (HO-1) is a major enzyme induced by cellular stress, exhibits cellular protective, antioxidant, and anti-inflammatory abilities.43) HO-1 significantly restored the cell viability under H2O2 injury via reducing the generation of ROS and increasing the levels of SOD and glutathione (GSH) activity.43)

    H2O2-stimulated AMPK activation is pro-survival and antiapoptosis in osteoblasts. Autophagy induction and NADPH maintenance are involved in AMPK-mediated pro-survival effects.44) H2O2-induced increases in JNK phosphorylation and cell apoptosis were largely activated by ASK1 overexpression whereas the similar transfection did not affect ROS generation in the cells. Moreover, inhibition of H2O2-increased ROS generation, JNK phosphorylation, and cellular apoptosis by upregulation of heat shock transcription factor-1 HSF1 tended to be disappeared.45) HSP-70 protects cardiomyocytes from apoptosis under oxidative stress by reducing intracellular ROS generation and by inhibiting p38 phosphorylation.46) The present study showed that 40 μM H2O2 induced the expressions of cell protection-related proteins (HSP-70, HSP-90, AMPK, etc.) and phase II type antioxidant proteins (HO-1, GSTO1, and SOD-1), which conjugate drug metabolites or endobiotics via acetylation, glucuronidation, glutathionylation, methylation, and sulphation, making them more hydrophilic; or degrade heme or quinone.47)

    This preliminary study was aimed to determine the optimal concentration of H2O2, which can give minimal ROS impact on RAW 264.7 cells in order to observe the reactive molecular signaling of viable cells maintaining their cellular proliferation, protection, apoptosis, and survival. 40 μM H2O2 determined in this study showed significant ROS injury on RAW 264.7 cells, resulted in the repression of proliferation, inactivation of epigenetic modification, and caspase 3/PARP-1/AIF-associated apoptosis, parthanatos. On the other hand, 40 μM H2O2 activated RAW 264.7 cells to be differentiated into M1 type macrophages, induced cell survival signaling via marked increases in the expressions of heat shock proteins, antioxidantrelated proteins, autophagosome-related protein, cellular energy homeostasis-related protein, and a transcription factor regulating for the expression of antioxidant proteins. Therefore, it is suggested that 40 μM H2O2 is proper concentration to investigate the early stage of H2O2-induced ROS biohazard in RAW 264.7 cells. However, further precise molecular biological investigation should be followed in order to elucidate H2O2-induced molecular signaling mechanisms in cells.

    ACKNOWLEDGEMENTS

    This work was supported by the research grant of Cheongju University (2018.03.01.-2020.02.29.).

    Figure

    KAOMP-43-6-255_F1.gif

    Determination of optimal responsible concentration of H2O2 in RAW 264.7 cells depending on LD50 by MTT assay. 0 to 200 μM H2O2 were applied to RAW 264.7 cells for 3 and 16 hours. 40 μM H2O2 showed 85.7% cell survival in 3 hours and LD50 cell survival (48%) in 16 hours, therefore, 40 μM H2O2 was determined as an optimal responsible concentration for the research of H2O2-induced oxidative stresses in RAW 264.7 cells in this study.

    KAOMP-43-6-255_F2.gif

    Expressions of proliferation-related proteins by 40μM H2O2 treatment in RAW 264.7 cells (A1 and A2), epigenetic modificationrelated proteins (B1 or B2), and epigenetic modification-related proteins (C1 and C2) as determined by IP-HPLC. Line graphs, A1, B1, and C1show protein expressional changes on the same scale (%) versus culture time (30 min, 1, or 2 hours), whereas the star plots (A2, B2, and C2) show the differential expression levels of proteins after 30 min, 1, or 2 hours of treatment on appropriate scales (%).

    KAOMP-43-6-255_F3.gif

    Expressions of inflammatory proteins by 40μM H2O2 treatment in RAW 264.7 cells (A1 and A2), apoptosis and survivalrelated proteins (B1 and B2), and protection-related proteins (B1 and B2) as determined by IP-HPLC. Line graphs, A1, B1, and C1 show protein expressional changes on the same scale (%) versus culture time (30 min, 1, or 2 hours), whereas the star plots (A2, B2, and C2) show the differential expression levels of proteins after 30 min, 1, or 2 hours of treatment on appropriate scales (%).

    Table

    Antibodies used in the study

    Reference

    1. Basu K, Mukhopadhyay A, Ghosh I: Nuclear morphology and c-Jun N-terminal kinase 1 expression differentiate serumstarved oxidative stress signalling from hydrogen peroxideinduced apoptosis in retinal neuronal cell line. Cell Biol Int 2012;36:1021-1027.
    2. Conde de la Rosa L, Schoemaker MH, Vrenken TE: Superoxide anions and hydrogen peroxide induce hepatocyte death by different mechanisms: involvement of JNK and ERK MAP kinases. J Hepatol 2006;44:918-929.
    3. Kim OS, Kim YS, Jang DS: Cytoprotection against hydrogen peroxide-induced cell death in cultured mouse mesangial cells by erigeroflavanone, a novel compound from the flowers of Erigeron annuus. Chem Biol Interact 2009;180:414-420.
    4. Park WH: MAPK inhibitors, particularly the JNK inhibitor, increase cell death effects in H2O2-treated lung cancer cells via increased superoxide anion and glutathione depletion.Oncol Rep 2018;39:860-870.
    5. Pearl-Yafe M, Halperin D, Scheuerman O: The p38 pathway partially mediates caspase-3 activation induced by reactive oxygen species in Fanconi anemia C cells. Biochem Pharmacol 2004;67:539-546.
    6. Sies H: Role of metabolic H2O2 generation: redox signaling and oxidative stress. J Biol Chem 2014;289:8735-8741.
    7. Bast A, Brenninkmeijer JW, Savenije-Chapel EM: Cytochrome P450 oxidase activity and its role in NADPH dependent lipid peroxidation. FEBS Lett 1983;151:185-188.
    8. Chan TS, Moridani M, Siraki A: Hydrogen peroxide supports hepatocyte P450 catalysed xenobiotic/drug metabolic activation to form cytotoxic reactive intermediates. Adv Exp Med Biol 2001;500:233-236.
    9. Golly I, Hlavica P, Schartau W: The functional role of cytochrome b5 reincorporated into hepatic microsomal fractions. Arch Biochem Biophys 1988;260:232-240.
    10. van de Straat R, Vromans RM, Bosman P: Cytochrome P-450-mediated oxidation of substrates by electron-transfer; role of oxygen radicals and of 1- and 2-electron oxidation of paracetamol. Chem Biol Interact 1988;64:267-280.
    11. Bucris E, Beck A, Boura-Halfon S: Prolonged insulin treatment sensitizes apoptosis pathways in pancreatic beta cells. J Endocrinol 2016;230:291-307.
    12. Muller AP, Haas CB, Camacho-Pereira J: Insulin prevents mitochondrial generation of H(2)O(2) in rat brain. Exp Neurol 2013;247:66-72.
    13. Sampson SR, Bucris E, Horovitz-Fried M: Insulin increases H2O2-induced pancreatic beta cell death. Apoptosis 2010; 15:1165-1176.
    14. Sies H: Hydrogen peroxide as a central redox signaling molecule in physiological oxidative stress: Oxidative eustress. Redox Biol 2017;11:613-619.
    15. Kim SH, Kim KH, Yoo BC: Induction of LGR5 by H2O2 treatment is associated with cell proliferation via the JNK signaling pathway in colon cancer cells. Int J Oncol 2012;41: 1744-1750.
    16. Liu GY, Jiang XX, Zhu X: ROS activates JNK-mediated autophagy to counteract apoptosis in mouse mesenchymal stem cells in vitro. Acta Pharmacol Sin 2015;36:1473-1479.
    17. Denning TL, Takaishi H, Crowe SE: Oxidative stress induces the expression of Fas and Fas ligand and apoptosis in murine intestinal epithelial cells. Free Radic Biol Med 2002;33:1641-1650.
    18. Kim H, Kim YN, Kim H: Oxidative stress attenuates Fasmediated apoptosis in Jurkat T cell line through Bfl-1 induction. Oncogene 2005;24:1252-1261.
    19. Clarke NJ, Tomlinson AJ, Ohyagi Y: Detection and quantitation of cellularly derived amyloid beta peptides by immunoprecipitation-HPLC-MS. FEBS Lett 1998;430:419-423.
    20. Luo L, Shen L, Sun F: Immunoprecipitation coupled with HPLC-MS/MS to discover the aromatase ligands from Glycyrrhiza uralensis. Food Chem 2013;138:315-320.
    21. Kim YS, Lee SK: IP-HPLC Analysis of Human Salivary Protein Complexes. Korean Journal of Oral and Maxillofacial Pathology 2015;39:615-622.
    22. Kim SM, Eo MY, Cho YJ: Wound healing protein profiles in the postoperative exudate of bisphosphonate- related osteonecrosis of mandible. Eur Arch Otorhinolaryngol 2017;274:3485-3495.
    23. Kim SM, Eo MY, Cho YJ: Differential protein expression in the secretory fluids of maxillary sinusitis and maxillary retention cyst. Eur Arch Otorhinolaryngol 2017;274:215-222.
    24. Kim SM, Eo MY, Cho YJ: Immunoprecipitation high performance liquid chromatographic analysis of healing process in chronic suppurative osteomyelitis of the jaw. J Craniomaxillofac Surg 2018;46:119-127.
    25. Kim MK, Yoon CS, Kim SG: Effects of 4- Hexylresorcinol on Protein Expressions in RAW 264.7 Cells as Determined by Immunoprecipitation High Performance Liquid Chromatography. Sci Rep 2019;9:3379.
    26. Yoon CS, Kim MK, Kim YS: In vitro protein expression changes in RAW 264.7 cells and HUVECs treated with dialyzed coffee extract by immunoprecipitation high performance liquid chromatography. Sci Rep 2018;8:13841.
    27. Yoon CS, Kim MK, Kim YS: In vivo protein expression changes in mouse livers treated with dialyzed coffee extract as determined by IP-HPLC. Maxillofac Plast Reconstr Surg2018;40:44.
    28. Kim SM, Jeong D, Kim MK: Two different protein expression profiles of oral squamous cell carcinoma analyzed by immunoprecipitation high-performance liquid chromatography.World J Surg Oncol 2017;15:151.
    29. Pagliacci MC, Spinozzi F, Migliorati G: Genistein inhibits tumour cell growth in vitro but enhances mitochondrial reduction of tetrazolium salts: a further pitfall in the use of the MTT assay for evaluating cell growth and survival. Eur J Cancer 1993;29A:1573-1577.
    30. Park MH, Wolff EC: Hypusine, a polyamine-derived amino acid critical for eukaryotic translation. J Biol Chem 2018; 293:18710-18718.
    31. Panieri E, Gogvadze V, Norberg E: Reactive oxygen species generated in different compartments induce cell death, survival, or senescence. Free Radic Biol Med 2013;57:176-187.
    32. Ameziane-El-Hassani R, Dupuy C: Detection of Reactive Oxygen Species in Cells Undergoing Oncogene-Induced Senescence. Methods Mol Biol 2017;1534:139-145.
    33. Ma S, Ochi H, Cui L: Hydrogen peroxide induced downregulation of CD28 expression of Jurkat cells is associated with a change of site alpha-specific nuclear factor binding activity and the activation of caspase-3. Exp Gerontol 2003; 38:1109-1118.
    34. Marazita MC, Dugour A, Marquioni-Ramella MD: Oxidative stress-induced premature senescence dysregulates VEGF and CFH expression in retinal pigment epithelial cells: Implications for Age-related Macular Degeneration. Redox Biol 2016;7:78-87.
    35. Monickaraj F, Aravind S, Nandhini P: Accelerated fat cell aging links oxidative stress and insulin resistance in adipocytes. J Biosci 2013;38:113-122.
    36. Banerjee A, Hu J, Goss DJ: Thermodynamics of proteinprotein interactions of cMyc, Max, and Mad: effect of polyions on protein dimerization. Biochemistry 2006;45: 2333-2338.
    37. Nair SK, Burley SK: Structural aspects of interactions within the Myc/Max/Mad network. Curr Top Microbiol Immunol 2006;302:123-143.
    38. Meyer M, Huaux F, Gavilanes X: Azithromycin reduces exaggerated cytokine production by M1 alveolar macrophages in cystic fibrosis. Am J Respir Cell Mol Biol 2009;41:590-602.
    39. Wijesundera KK, Izawa T, Tennakoon AHl: M1- and M2- macrophage polarization in rat liver cirrhosis induced by thioacetamide (TAA), focusing on Iba1 and galectin-3. Exp Mol Pathol 2014;96:382-392.
    40. Matsura T, Kai M, Fujii Y: Hydrogen peroxide- induced apoptosis in HL-60 cells requires caspase-3 activation. Free Radic Res 1999;30:73-83.
    41. Nakamura T, Sakamoto K: Reactive oxygen species up-regulates cyclooxygenase-2, p53, and Bax mRNA expression in bovine luteal cells. Biochem Biophys Res Commun 2001;284:203-210.
    42. Lopez-Diazguerrero NE, Perez-Figueroa GE, Martinez -Garduno CM: Telomerase activity in response to mild oxidative stress. Cell Biol Int 2012;36:409-413.
    43. Ma T, Chen T, Li P: Heme oxygenase-1 (HO-1) protects human lens epithelial cells (SRA01/04) against hydrogen peroxide (H2O2)-induced oxidative stress and apoptosis. Exp Eye Res 2016;146: 318-329.
    44. She C, Zhu LQ, Zhen YF: Activation of AMPK protects against hydrogen peroxide-induced osteoblast apoptosis through autophagy induction and NADPH maintenance: new implications for osteonecrosis treatment? Cell Signal 2014;26:1-8.
    45. Zhang L, Jiang H, Gao X: Heat shock transcription factor-1 inhibits H2O2-induced apoptosis via down-regulation of reactive oxygen species in cardiac myocytes. Mol Cell Biochem 2011;347:21-28.
    46. Li K, Yang B, Zhao C: Transforming growth factor-betaactivated kinase 1 enhances H2O2-induced apoptosis independently of reactive oxygen species in cardiomyocytes. J Cardiovasc Med (Hagerstown) 2014;15:565-571.
    47. Li Z, Dong X, Liu H: Astaxanthin protects ARPE-19 cells from oxidative stress via upregulation of Nrf2-regulated phase II enzymes through activation of PI3K/Akt. Mol Vis 2013;19: 1656-1666.
    오늘하루 팝업창 안보기 닫기