ketamine inhibits tumor necrosis factor-α and interleukin-6 gene expression.pdf

Upload: huplala-hypocrate

Post on 03-Apr-2018

219 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/28/2019 Ketamine inhibits tumor necrosis factor- and interleukin-6 gene expression.pdf

    1/9

    Ketamine inhibits tumor necrosis factor- and interleukin-6 gene

    expressions in lipopolysaccharide-stimulated macrophages through

    suppression of toll-like receptor 4-mediated c-Jun N-terminal kinase

    phosphorylation and activator protein-1 activation

    Gone-Jhe Wu a,b,c, Ta-Liang Chen c,d, Yune-Fang Ueng e, Ruei-Ming Chen a,

    a Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwanb Department of Anesthesiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan

    c Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwand Department of Anesthesiology, Taipei Medical University Hospital, Taipei Medi cal University, Taipei, Taiwan

    e National Research Institute of Chinese Medicine, Taipei, Taiwan

    Received 9 October 2007; revised 16 November 2007; accepted 28 November 2007

    Available online 8 December 2007

    Abstract

    Our previous study showed that ketamine, an intravenous anesthetic agent, has anti-inflammatory effects. In this study, we further evaluated

    the effects of ketamine on the regulation of tumor necrosis factor- (TNF-) and interlukin-6 (IL-6) gene expressions and its possible signal-

    transducing mechanisms in lipopolysaccharide (LPS)-activated macrophages. Exposure of macrophages to 1, 10, and 100 M ketamine, 100 ng/ml

    LPS, or a combination of ketamine and LPS for 1, 6, and 24 h was not cytotoxic to macrophages. A concentration of 1000M of ketamine alone or in

    combined treatment with LPS caused significant cell death. Administration of LPS increased cellular TNF- and IL-6 protein levelsin concentration-

    and time-dependent manners. Meanwhile, treatment with ketamine concentration- and time-dependently alleviated the enhanced effects. LPS

    induced TNF- and IL-6 mRNA syntheses. Administration of ketamine at a therapeutic concentration (100 M) significantly inhibited LPS-induced

    TNF- and IL-6 mRNA expressions. Application of toll-like receptor 4 (TLR4) small interfering (si)RNA into macrophages decreased cellular TLR4

    levels. Co-treatment of macrophages with ketamine and TLR4 siRNA decreased the LPS-induced TNF- and IL-6 productions more than alone

    administration of TLR4 siRNA. LPS stimulated phosphorylation of c-Jun N-terminal kinase and translocation of c-Jun and c-Fos from the cytoplasm

    to nuclei. However, administration of ketamine significantly decreased LPS-induced activation of c-Jun N-terminal kinase and translocation of c-Jun

    and c-Fos. LPS increased the binding of nuclear extracts to activator protein-1 consensus DNA oligonucleotides. Administration of ketamine

    significantly ameliorated LPS-induced DNA binding activity of activator protein-1. Therefore, a clinically relevant concentration of ketamine can

    inhibitTNF- andIL-6gene expressions in LPS-activated macrophages. The suppressive mechanisms occur through suppression of TLR4-mediated

    sequential activations of c-Jun N-terminal kinase and activator protein-1.

    2007 Elsevier Inc. All rights reserved.

    Keywords: Ketamine; Macrophages; LPS; TNF-; IL-6; TLR4; JNK/AP-1

    Introduction

    Ketamine, a widely used intravenous anesthetic agent, has

    more-stable hemodynamics than other anesthetic agents so it is

    often applied as an inducer of anesthesia in critically ill patients

    (White et al., 1982; Bourgoin et al., 2003). Ketamine can also be

    a competitive N-methyl-D-aspartate-receptor antagonist and has

    been shown to have therapeutic potential in preventing anoxic

    damage from stroke in humans (Rothman et al., 1987).

    Clinically, induction of anesthesia with ketamine is usually

    associated with increases in cardiac output, arterial blood pres-

    sure, and heart rate (Reich and Silvay, 1989; Chen et al., 2005b).

    In addition, ketamine has also been shown to possess anti-

    Available online at www.sciencedirect.com

    Toxicology and Applied Pharmacology 228 (2008) 105 113www.elsevier.com/locate/ytaap

    Corresponding author. Graduate Institute of Medical Sciences, College of

    Medicine, Taipei Medical University, Taipei, Taiwan, No. 250 Wu-Xing St.,

    Taipei 110, Taiwan. Fax: +886 2 23778620.

    E-mail address: [email protected] (R.-M. Chen).

    0041-008X/$ - see front matter 2007 Elsevier Inc. All rights reserved.doi:10.1016/j.taap.2007.11.027

    mailto:[email protected]://dx.doi.org/10.1016/j.taap.2007.11.027http://dx.doi.org/10.1016/j.taap.2007.11.027mailto:[email protected]
  • 7/28/2019 Ketamine inhibits tumor necrosis factor- and interleukin-6 gene expression.pdf

    2/9

    inflammatory effects in both patients and experimental animals

    (Rofael et al., 2003; Yang et al., 2005; Suliburk and Mercer,

    2007). Previous studies provided in vitro data demonstrating

    that ketamine can induce dysfunction of lymphocytes, natural

    killer cells, and neutrophils (Hofbauer et al., 1998; Weigand

    et al., 2000; Melamed et al., 2003). A recent study performed in

    our lab further showed that ketamine at a therapeuticconcentration selectively suppressed macrophage functions of

    phagocytosis, oxidative ability, and cytokine production (Chang

    et al., 2005).

    In the innate immunity system, macrophages play pivotal

    roles in cellular host defense against infection and tissue injury

    (Calandra and Roger, 2003). In response to stimuli, macro-

    phages can produce and release a variety of inflammatory

    cytokines, which induce serial inflammatory reactions (Nathan,

    1987; Froidevaux et al., 2001). Tumor necrosis factor- (TNF-

    ) and interleukin-6 (IL-6), two typical and critical inflamma-

    tory cytokines predominantly produced by macrophages, are

    reported to have pleiotropic effects on regulating the immuneresponse, acute-phase reaction, and hematopoiesis (Bendtzen,

    1998). Previous studies showed that TNF- is involved in the

    progression of myocardial infarction, alcohol-induced liver

    disease, rheumatoid arthritis, human systemic lupus erythema-

    tosus, and macrophage-mediated tumor cytotoxicity (Jacob,

    1992; Wilder and Elenkov, 1999; Frangogiannis et al., 2002;

    Nagy, 2004). IL-6 has been implicated as a regulator which

    modulates macrophage antitumor activities, myeloid cell dif-

    ferentiation, and endometriosis (Sachs et al., 1989; Bonta and

    Ben-Efraim, 1993; Sidell et al., 2002). Levels of TNF- and IL-

    6 in macrophages can be altered by endogenous or exogenous

    factors, which then lead to immunomodulation (Scott and

    Kingsley, 2006).Toll-like receptors (TLRs) are type I transmembrane pro-

    teins with extracellular domains comprised largely of leucine-

    rich repeats and intracellular signaling domains (Akira et al.,

    2006). Mammalian TLRs contain at least 12 member proteins

    which trigger host resistance to infection (Akira et al., 2006;

    Trinchieri and Sher, 2007). In macrophages, lipopolysacchar-

    ide (LPS), an important contributing factor to the pathogenesis

    of septic syndromes, can specifically activate TLR4, then

    stimulate translocation of the transcription factors, nuclear

    factor kappa B (NFB) and activator protein-1 (AP-1), from

    the cytoplasm to nuclei, ultimately inducing gene expression

    of inflammatory cytokines (Jones et al., 2001; Fan et al.,2004). Previous studies demonstrated that ketamine can inhibit

    TNF- gene expression through suppression of NFB acti-

    vation (Sun et al., 2004; Yang et al., 2005). C-Jun N-terminal

    kinase (JNK)-mediated AP-1 activation is critical for cell

    differentiation, apoptosis, and tumorigenesis (Potapova et al.,

    2001; Paik et al., 2003). Meanwhile, the role of AP-1 in

    ketamine-induced immunosuppression is still unknown. Thus,

    this study was aimed to evaluate the effects of ketamine on the

    biosyntheses of TNF- and IL-6 in LPS-activated macro-

    phages and its possible signal-transducing mechanisms from

    the viewpoints of cell viability, protein and RNA expressions,

    TLR4 knockdown, JNK phosphorylation, and AP-1 transloca-

    tion and transactivation.

    Materials and methods

    Cell culture and drug treatment. Macrophage-like Raw 264.7 cells,

    purchased from American Type Culture Collection (Rockville, MD, USA),

    were cultured in RPMI 1640 medium (Gibco-BRL, Grand Island, NY, USA)

    supplemented with 10% fetal calf serum, L-glutamine, penicillin (100 IU/ml),

    and streptomycin (100 g/ml) in 75-cm2 flasks at 37 C in a humidified

    atmosphere of 5% CO2.Ketamine and LPS were dissolved in phosphate-buffered saline (PBS)

    (0.14 M NaCl, 2.6 mM KCl, 8 mMNa2HPO4, and 1.5 mM KH2PO4). According

    to the clinical application, concentrations of ketamine at 1, 10, 100, and

    1000 M, which correspond to 0.01, 0.1, 1, and 10 times the clinical plasma

    concentration (Domino et al., 1982; Grant et al., 1983), were chosen as the

    administered concentrations in this study. Prior to the addition of ketamine,

    macrophages were washed with PBS buffer, and nonadherent cells were

    removed. Control macrophages received PBS only.

    Assay of cell viability. Cell viability was determined by a colorimetric 3-(4,5-

    dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay as described

    previously (Chen et al., 2007). Briefly, macrophages (2 104 cells per well) were

    seeded in 96-well tissue culture plates overnight. After drug treatment,

    macrophages were cultured with new medium containing 0.5 mg/ml MTT for a

    further 3 h. Theblueformazanproducts in macrophages were dissolvedin dimethylsulfoxide and spectrophotometrically measured at a wavelength of 550 nm.

    Enzyme-linked immunosorbent assay (ELISA). Levels of TNF- and IL-6 in

    the culture medium of macrophages were determined according to a previously

    described method (Chen et al., 2005a). Briefly, macrophages (2 104 cells/well)

    were seeded in 96-well tissue culture plates overnight. After drug treatment, the

    medium was collected and centrifuged. The amounts of TNF- and IL-6 were

    quantified following the standard protocols of the ELISA kits purchased from

    Endogen (Woburn, MA, USA).

    Reverse-transcription polymerase chain reaction (RT-PCR) assay. Messen-

    ger RNA from macrophages exposed to LPS, ketamine, or a combination of

    ketamine and LPS was prepared for RT-PCR analyses of TNF-, IL-6, and -

    actin mRNA. Oligonucleotides for the PCR analyses of TNF-, IL-6, and -

    actin mRNA were designed and synthesized by Clontech Laboratories (Palo

    Alto, CA, USA). The oligonucleotide sequences of the respective upstream and

    downstream primers for these mRNA analyses were 5-ATGAGCACA-

    GAAAGCATGATCCGC-3 and 3-CTCAGGCCCGTCCAGATGAAACC-5

    for TNF-, 5-ATGAAGTTCCTCTCTGCAAGAGACT-3 and 3-CACTAG-

    GTTTGCCGAGTAGATCTC-5 for IL-6, and 5-GTGGGCCGCTCTAGG-

    CACCAA-3 and 5-CTCTTTGATGTCACGCACGATTTC-3 for -actin (Wu

    et al., 2003). The PCR reaction was carried out using 35 cycles of 94 C for 45 s,

    60 C for 45 s, and 72 C for 2 min. The PCR products were loaded onto a 1.8%

    agarose gel containing 0.1 g/ml ethidium bromide and electrophoretically

    separated. DNA bands were visualized and photographed under UVlight

    exposure. The intensities of the DNA bands in the agarose gel were quantified

    with the aid of the UVIDOCMW ver. 99.03 digital imaging system (Uvtec,

    Cambridge, UK).

    TLR4 knockdown. Translation of TLR4 mRNA in macrophages was

    knocked-down using an RNA interference (RNAi) method following a small

    interfering (si)RNA transfection protocol provided by Santa Cruz Biotechnol-

    ogy (Santa Cruz, CA, USA). TLR4 siRNA was purchased from Santa Cruz

    Biotechnology, and is a pool of 3 target-specific 2025-nt siRNAs designed to

    knock-down TLR4's expression. Briefly, after culturing macrophages in

    antibiotic-free RPMI medium at 37 C in a humidified atmosphere of 5%

    CO2 for 24 h, the siRNA duplex solution, which was diluted in the siRNA

    transfection medium (Santa Cruz Biotechnology), was added to the macro-

    phages. After transfecting for 24 h, the medium was replaced with normal RPMI

    medium, and macrophages were treated with ketamine, LPS, or a combination

    of ketamine and LPS.

    Immunoblotting analyses of JNK, phosphorylated JNK, TLR4, and

    -actin. Protein analyses were carried out according to a previously describedmethod (Tai et al., 2007). After drug treatment, cell lysates were prepared in ice-

    106 G.-J. Wu et al. / Toxicology and Applied Pharmacology 228 (2008) 105113

  • 7/28/2019 Ketamine inhibits tumor necrosis factor- and interleukin-6 gene expression.pdf

    3/9

    cold radioimmunoprecipitation assay buffer (25 mM TrisHCl (pH 7.2), 0.1%

    SDS, 1% Triton X-100, 1% sodium deoxycholate, 0.15 M NaCl, and 1 mM

    EDTA). Protein concentrations were quantified using a bicinchonic acid protein

    assay kit (Pierce, Rockford, IL, USA). The proteins (50 g/well) were subjected

    to sodium dodecylsulfate polyacrylamide gel electrophoresis (SDS-PAGE), and

    transferred to nitrocellulose membranes. After blocking, the phosphorylated

    JNKs were immunodetected using a rabbit polyclonal antibody with a synthetic

    phospho-peptide corresponding to residues Thr183/Tyr185 of the human JNK(Cell Signaling, Danvers, MA, USA). The JNK was detected using a mouse

    monoclonal antibody against the human JNK (Cell Signaling) as the internal

    standard. TLR4 was immunodetected using a goat polyclonal antibody against

    mouse TLR4 (Santa Cruz Biotechnology). Cellular -actin protein was

    immunodetected using a mouse monoclonal antibody against mouse -actin

    (Sigma, St. Louis, MO, USA) as the internal standard. These protein bands were

    quantified using a digital imaging system (UVtec).

    Extraction of nuclear proteins and immunodetection. Nuclear components

    were extracted, and an immunodetection was carried out following the method

    ofWu et al. (2007). After drug treatment, nuclear extracts of macrophages were

    prepared. Protein concentrations were quantified by a bicinchonic acid protein

    assay kit (Pierce). Nuclear proteins (50 g/well) were subjected to SDS-PAGE

    and transferred to nitrocellulose membranes. After blocking, nuclear c-Jun and

    c-Fos were immunodetected using rabbit polyclonal antibodies against mouse c-Jun or human c-Fos (Santa Cruz Biotechnology). Total cellular c-Jun and c-Fos

    were immunodetected as internal standards. Intensities of the immunoreactive

    bands were determined using a digital imaging system (UVtec).

    Electrophoretic mobility shift assa y (EMSA). An EMSAwas performed using

    a Dig gel shift kit (Roche Diagnostics, Mannheim, Germany). Briefly, AP-1

    consensus oligonucleotides (Santa Cruz Biotechnology) were labeled with Dig.

    The nuclear extracts (10 ng) were reacted with Dig-labeled oligonucleotides at

    room temperature for 25 min. The complex was subjected to nondenatured

    polyacrylamide gel electrophoresis, and transferred to positively charged nylon

    membranes. After cross-linking at 120 mJ and blocking with the blocking buffer

    (Santa Cruz Biotechnology) at room temperature for 30 min, the membranes

    were immunoreacted with the anti-Dig-AP. Following washing and chemilu-

    minescent detection, the membranes were exposed to X-ray film. Intensities of

    the immunoreactive bands were determined using a digital imaging system(UVtec).

    Statistical analysis. The statistical significance of differences among control,

    ketamine-, LPS-, and ketamine+ LPS-treated macrophages was evaluated using

    nonparametric ANOVA followed by Duncan's multiple-range test, and

    differences were considered statistically significant at p values ofb0.05.

    Results

    To evaluate the toxic effects of ketamine and LPS on macro-

    phages, cell viability was assayed (Fig. 1). Treatment with 1, 10,

    and 100 M ketamine for 1, 6, and 24 h was not cytotoxic to

    macrophages (Fig. 1A). In 1- and 6-h-treated macrophages,ketamine at 1000 M did not affect the cell viability. However,

    after exposure to 1000 M ketamine for 24 h, the cell viability

    was significantly reduced by 48%. Administration of 100 ng/ml

    LPS to macrophages for 1, 6, and 24 h did not influence cell

    viability (Fig. 1B). The combination of LPS and ketamine at 1,

    10, and 100 M for 1, 6, and 24 h was not toxic to macrophages.

    When the administered time interval reached 24 h, exposure to

    the combination of LPS and 1000 M ketamine caused 50% of

    macrophages to die (Fig. 1B).

    Amounts of TNF- and IL-6 were detected to determine the

    effects of ketamine on the syntheses of these two cytokines in

    LPS-activated macrophages (Fig. 2). Exposure of macrophages

    to LPS for 24 h caused a 7-fold increase in the levels of cellular

    TNF- (Fig. 2A). Ketamine at 1 M did not change LPS-

    induced TNF- synthesis. Treatment with 10, 100, and 1000M

    ketamine significantly decreased cellular TNF- by 23%, 53%,

    and 74%, respectively. Administration of 100 ng/ml LPS to

    macrophages for 1, 6, and 24 h caused significant increases of

    2.3-, 3.8-, and 7.3-fold, respectively, in cellular TNF- levels

    (Fig. 2B). Treatment with ketamine at a therapeutic concentra-

    tion of 100 M for 1, 6, and 24 h did not affect TNF-

    production. Exposure to 100 M ketamine for 1 h did not

    influence LPS-induced TNF- synthesis. After being adminis-

    tered for 6 and 24 h, a clinically relevant concentration ofketamine significantly decreased LPS-induced augmentation in

    the levels of TNF- by 42% and 58%, respectively (Fig. 2B).

    Administration of 100 ng/ml LPS to macrophages for 24 h

    increased cellular IL-6 amounts by 30-fold (Fig. 2C). Ketamine

    at 1 M did not change the LPS-induced IL-6 production.

    Meanwhile, when the concentrations reached 10, 100, and

    1000 M, ketamine significantly reduced LPS-induced IL-6

    synthesis by 33%, 61%, and 83%, respectively. Exposure to

    100 ng/ml LPS for 1, 6, and 24 h augmented cellular IL-6 levels

    by 8-, 20-, and 34-fold, respectively (Fig. 2D). Treatment of

    macrophages with 100 M ketamine for 1, 6, and 24 h did not

    change the cellular IL-6 amounts. In 1-h-treated macrophages,

    ketamine did not influence LPS-induced IL-6 production. After

    Fig. 1. Effects of ketamine (KTM) and lipopolysaccharide (LPS) on macrophage

    viability. Macrophages were exposed to 1, 10, 100, and 1000 M KTM (A), or

    to 100 ng/ml LPS or a combination of LPS and KTM at 1, 10, 100, and 1000M

    (B) for 1, 6, and 24 h. Cell viability was determined using a colorimetric method.

    Each value represents the meanSEM for n =6. The asterisk () indicates that

    the value significantly differs from the respective control, pb0.05. O.D., optical

    density.

    107G.-J. Wu et al. / Toxicology and Applied Pharmacology 228 (2008) 105113

  • 7/28/2019 Ketamine inhibits tumor necrosis factor- and interleukin-6 gene expression.pdf

    4/9

    Fig. 2. Concentration- and time-dependent effects of ketamine (KTM) on lipopolysaccharide (LPS)-induced TNF- and IL-6 productions. Macrophages were exposed

    to either 100 ng/ml LPS or a combination of LPS and 1, 10, 100, and 1000M KTM for 24 h (A and C), or to 100 ng/ml LPS, 100M KTM, or a combination of KTM

    and LPS for 1, 6, and 24 h (B and D). The levels of TNF- and IL-6 in the culture medium of macrophages were quantified using enzyme-linked immunosorbent

    assays. Each value represents the meanSEM for n =6. The symbols, and #, indicate that a value significantly (pb0.05) differed from the control or LPS-treated

    groups, respectively.

    Fig. 3. Effects of ketamine (KTM) on lipopolysaccharide (LPS)-induced TNF- and IL-6 mRNA syntheses. Macrophages were exposed to either 100ng/ml LPS, 100M

    KTM, or a combination of KTM and LPS for 6 h. Messenger RNA from control and drug-treated macrophages was prepared for RT-PCR analyses of TNF- and IL-6

    mRNA(A and C, top panels).

    -Actin mRNAwas detected as an internal standard (bottom panels). These DNAbands were quantifiedand analyzed (B andD). Each valuerepresents the meanSEM forn =4. The symbols, and #, indicate that a value significantly (pb0.05) differed from the control or LPS-treated groups, respectively.

    108 G.-J. Wu et al. / Toxicology and Applied Pharmacology 228 (2008) 105113

  • 7/28/2019 Ketamine inhibits tumor necrosis factor- and interleukin-6 gene expression.pdf

    5/9

    Fig. 4. Effects of TLR4 small interfering (si)RNA on TNF- and IL-6 productions. TLR4 siRNAwas applied to macrophages for 24 and 48 h. The amounts of TLR4

    were immunodetected using a goat polyclonal antibody against mouse TLR4 (A, top panel). -Actin was quantified as an internal standard (bottom panel). These

    immunoreactive protein bands were quantified and analyzed (B). After application of TLR4 siRNA for 48 h, macrophages were exposed to either 100 ng/ml

    lipopolysaccharide (LPS), 100 M ketamine (KTM), or a combination of KTM and LPS for another 24 h. Enzyme-linked immunosorbent assays were performed to

    determine the levels of TNF- and IL-6 in the culture medium of macrophages. Each value represents the mean SEM forn =6. The symbols, , #, and indicate that a

    value significantly (pb0.05) differed from the control, LPS-, or KTM+LPS-treated groups, respectively.

    Fig. 5. Effects of ketamine (KTM) on lipopolysaccharide (LPS)-induced phosphorylation of JNK and the translocation of c-Jun and c-Fos. Macrophages were exposed to

    either 100ng/mlLPS, 100M KTM, or a combination of KTMand LPSfor 1 h. Phosphorylated JNK(P-JNK) wasimmunodetectedusinga rabbit polyclonal antibody with

    a synthetic phospho-peptidecorresponding to residues Thr183/Tyr185 of human JNK(A, top panel). JNKwas analyzed using a mouse monoclonal antibody against human

    JNK as an internal standard (bottom panel). Nuclear c-Jun (Nc-Jun) and c-Fos (Nc-Fos) were immunodetected using rabbit polyclonal antibodies against mouse c-Jun or

    human c-Fos (C). Total c-Jun (Tc-Jun) and c-Fos (Tc-Fos) were detected as internal standards. These protein bands were quantified and analyzed (B and D). Each valuerepresents the meanSEM for n =4. The symbols, and #, indicate that a value significantly (pb0.05) differed from the control or LPS-treated groups, respectively.

    109G.-J. Wu et al. / Toxicology and Applied Pharmacology 228 (2008) 105113

  • 7/28/2019 Ketamine inhibits tumor necrosis factor- and interleukin-6 gene expression.pdf

    6/9

    exposure for 6 and 24 h, ketamine at a therapeutic concentration

    (100 M) significantly decreased LPS-induced IL-6 synthesis

    by 40% and 53%, respectively (Fig. 2D).

    To determine the mechanism of ketamine-caused suppres-

    sion of TNF- and IL-6 syntheses in LPS-activated macro-

    phages, the RNA levels of these two cytokines were quantified

    (Fig. 3). In untreated macrophages, low levels of TNF- and IL-6 were detected (Figs. 3A, C, top panels, lane 1). After exposure

    to 100 ng/ml LPS for 6 h, TNF- and IL-6 mRNA were

    obviously induced (Figs. 3A, C, top panels, lane 2). Treatment

    with ketamine alone did not affect the synthesis of TNF- or IL-

    6 mRNA (Figs. 3A, C, top panels, lane 3). Meanwhile, ketamine

    at a clinically relevant concentration (100 M) inhibited LPS-

    induced TNF- and IL-6 mRNA productions (Figs. 3A, C, top

    panels, lane 4). The amounts of -actin mRNA were detected as

    internal controls (Figs. 3A, C, bottom panels). These DNA

    bands were quantified and analyzed (Figs. 3B, D). Administra-

    tion of LPS caused significant 63- and 20-fold increases in

    cellular TNF- and IL-6 mRNA levels, respectively. A thera-peutic concentration (100 M) of ketamine significantly in-

    hibited LPS-induced TNF- and IL-6 mRNA levels by 73%

    and 79%, respectively (Figs. 3B, D).

    TLR4 siRNA was applied to macrophages to determine the

    roles of this membrane receptor in ketamine-induced suppres-

    sion of TNF- and IL-6 syntheses in LPS-activated macro-

    phages (Fig. 4). After treatment with TLR4 siRNA for 24 and

    48 h, levels of TLR4 protein in macrophages were obviously

    downregulated (Fig. 4A, top panel). The amounts of -actin in

    macrophages were detected as internal standards (bottom

    panel). These protein bands were quantified and analyzed

    (Fig. 4B). Application of TLR4 siRNA to macrophages for 24

    and 48 h significantly decreased cellular TLR4 levels by 47%and 87%, respectively. Administration of TLR4 siRNA for 48 h

    slightly affected the basal levels of TNF- or IL-6 (Figs. 4C, D).

    After application of TLR4 siRNA in macrophages for 48 h, the

    LPS-induced increases in the amounts of TNF- and IL-6

    decreased by 66% and 70%, respectively. Administration of

    ketamine at a therapeutic concentration (100 M) significantly

    decreased LPS-induced syntheses of TNF- and IL-6 by 70%

    and 72%, respectively. By comparison with alone administra-

    tion of TLR4 siRNA, co-treatment of macrophages with

    ketamine and TLR4 siRNA caused more suppression in the

    LPS-induced TNF- and IL-6 syntheses (Figs. 4C, D).

    To determine the signal-transducing mechanisms of keta-mine-caused inhibition of TNF- and IL-6 gene expressions in

    LPS-activated macrophages, phosphorylation of JNK1/2 and

    the translocations of c-Jun and c-Fos from the cytoplasm to

    nuclei were evaluated (Fig. 5). Exposure of macrophages to

    LPS obviously increased the phosphorylation of JNK1/2

    (Fig. 5A, top panel, lane 2). Ketamine did not influence

    JNK1/2 phosphorylation (lane 3). However, treatment with

    ketamine decreased LPS-induced JNK1/2 phosphorylation

    (lane 4). Nonphosphorylated JNK was immunodetected as the

    internal standard (Fig. 5A, bottom panel). These protein bands

    were quantified and analyzed (Fig. 5B). Administration of LPS

    increased phosphorylation of JNK1 and JNK2 by 2.5- and 2.2-

    fold, respectively. After exposure to a therapeutic concentration

    of ketamine, LPS-induced phosphorylation of JNK1 and JNK2

    was significantly ameliorated by 37% and 46%, respectively

    (Fig. 5B).

    Exposure to LPS increased nuclear levels of c-Jun and c-Fos

    (Fig. 5C, top 1 and 3 panels, lane 2). Treatment with ketamine

    did not affect nuclear c-Jun or c-Fos levels (lane 3). After

    exposure to a clinically relevant concentration of ketamine, theLPS-caused increases in nuclear c-Jun and c-Fos were

    significantly reduced (lane 4). Total levels of c-Jun and c-Fos

    in macrophages were immunodetected as internal standards

    (Fig. 5C, top 2 and bottom panels). These immunoreactive

    protein bands were quantified and analyzed (Fig. 5D). Admin-

    istration of LPS caused significant 2.8- and 2.2-fold enhance-

    ments in the nuclear c-Jun and c-Fos levels, respectively. After

    treatment with ketamine, the LPS-induced increases in the

    amounts of nuclear c-Jun and c-Fos significantly suppressed

    (Fig. 5D).

    Analysis by EMSA was performed to evaluate the down-

    stream events of ketamine-caused suppression of the phosphor-ylation of JNK and the sequential translocation of c-Jun and

    c-Fos (Fig. 6). Administration of LPS to macrophages increased

    the DNA binding activity of nuclear extracts to AP-1 consensus

    oligonucleotides (Fig. 6A, top panel, lane 2). Treatment with

    ketamine alone did not influence the binding activity of AP-1

    (lane 3). However, the LPS-induced enhancement in the DNA

    Fig. 6. Effects of ketamine (KTM) on lipopolysaccharide (LPS)-induced DNA

    binding activity of the AP-1 transcriptional factor. Macrophages were exposed

    to either 100 ng/ml LPS, 100 M KTM, or a combination of KTM and LPS for

    1 h. AP-1 consensus oligonucleotides were labeled with digoxigenin, and then

    reacted with 10 g of nuclear extracts. The complex was electrophoretically

    separated and blotted onto nylon membranes. After cross-linking and blocking,

    the membranes were immunoreacted with the anti-digoxigenin-AP. Following

    washing and chemiluminescent detection, the membranes wereexposedto X-ray

    films, and the amounts of free probes were detected as an internal standard

    (A). These DNAprotein binding bands were quantified and analyzed (B). Each

    value represents the meanSEM forn = 4. The symbols, and #, indicate that a

    value significantly (pb

    0.05) differed from the control or LPS-treated groups,respectively.

    110 G.-J. Wu et al. / Toxicology and Applied Pharmacology 228 (2008) 105113

  • 7/28/2019 Ketamine inhibits tumor necrosis factor- and interleukin-6 gene expression.pdf

    7/9

    binding activity of nuclear extracts to AP-1 consensus oligo-

    nucleotides was obviously ameliorated following administration

    of a clinically relevant concentration of ketamine (100 M; lane

    4). The free probes were detected as internal standards (Fig. 6A,

    bottom panel). These protein-DNA bands were quantified and

    analyzed (Fig. 6B). Exposure of macrophages to LPS increased

    the DNA-binding activity of nuclear extracts to AP-1 consensusoligonucleotides by 12.6-fold. Meanwhile, treatment with

    ketamine significantly decreased LPS-induced increases in the

    association of nuclear extracts with AP-1 DNA oligonucleotides

    (Fig. 6B).

    Discussion

    Ketamine at a therapeutic concentration (100 M) can

    downregulate TNF- and IL-6 syntheses. This study showed

    that exposure of macrophages to LPS significantly increased

    cellular TNF- and IL-6 protein levels. Administration of

    ketamine caused concentration- and time-dependent decreasesin LPS-induced TNF- and IL-6 productions. Ketamine is an

    intravenous anesthetic agent usually used for critically ill

    patients (White et al., 1982; Bourgoin et al., 2003). Ketamine

    at 100 M is within the range of clinically relevant concentra-

    tions (Domino et al., 1982; Grant et al., 1983). After exposure to

    a therapeutic concentration of 100 M, ketamine was not

    cytotoxic to macrophages but significantly ameliorated bio-

    syntheses of TNF- and IL-6 induced by LPS. A previous study

    reported that ketamine suppressed cytokine production in rats

    which were suffering acute lung injury, but only at a large dose

    (Yang et al., 2005). Thus, this study further showed that

    ketamine at a clinically relevant concentration could inhibit

    TNF- and IL-6 productions in LPS-activated macrophages.LPS has been implicated as a critical factor in the process of

    septic syndrome (Raetz et al., 1994). TNF- and IL-6 are two

    major inflammatory cytokines produced by macrophages, and

    have been reported to participate in regulating the immune

    response, acute-phase reaction, and hematopoiesis (Bendtzen,

    1998). Decreases in the levels of these two cytokines can lead to

    immunosuppression. Our previous study showed that ketamine

    can specifically reduce phagocytosis and the oxidative ability of

    macrophages (Chang et al., 2005). In this study, we found that

    the ketamine-involved suppression of TNF- and IL-6 bio-

    syntheses contributed to its anti-inflammatory effects in LPS-

    activated macrophages.The mechanism of ketamine-induced suppression of TNF-

    and IL-6 syntheses occurs at the transcriptional level. Exposure

    of macrophages to LPS increased the amounts of TNF- and IL-

    6 proteins in concentration- and time-dependent manners. After

    administration of ketamine, the LPS-induced enhancements of

    TNF- and IL-6 proteins were significantly ameliorated. In rats

    injected with endotoxin, ketamine was shown to attenuate septic

    syndrome through downregulating TNF- and IL-6 biosynth-

    eses at the protein level (Taniguchi et al., 2001). Meanwhile,

    RNA levels of TNF- and IL-6 in macrophages were augmented

    following LPS administration. Administration of ketamine at a

    therapeutic concentration inhibited LPS-induced TNF- and IL-

    6 mRNA productions. Analysis of transcription factors further

    revealed that exposure to LPS increased the translocation and

    transactivation of c-Jun and c-Fos, but ketamine significantly

    alleviated the augmentation. Jun and Fos are two critical

    components for constructing the heterodimers of the transcrip-

    tion factor, AP-1 (Hess et al., 2004). AP-1 consensus DNA

    motifs have been found in the promoter regions of both TNF-

    and IL-6 genes (Luet al., 2005). Therefore, ketamine inhibits thebiosyntheses of TNF- and IL-6 through a transcriptional

    mechanism. The ketamine-involved suppression of TNF- and

    IL-6 productions may be TLR4-dependent. Mammalian TLRs

    mediate a host's resistance to infection (Akira et al., 2006;

    Trinchieri and Sher, 2007).

    This study showed that application of TLR4 siRNA into

    macrophages decreased cellular TLR4 protein expression. In

    parallel with TLR4 knockdown, the LPS-induced enhancements

    of TNF- and IL-6 syntheses were simultaneously reduced. A

    previous study used targeted disruption of genomic TLR4 in

    mice to also confirm that the TLR4 receptor is necessary for

    sensitive responses to LPS (Hoshino et al., 1999).Thus, the LPS-caused enhancements of TNF- and IL-6 syntheses are TLR4-

    dependent. Co-treatment of macrophages with ketamine and

    TLR4 siRNA caused more suppression of the LPS-induced

    TNF- and IL-6 expressions than alone treatment with TLR4

    siRNA. Therefore, TLR4 could be used as an effector involved

    in the ketamine-caused suppression of TNF- and IL-6

    productions in LPS-activated macrophages.

    Ketamine can decrease JNK phosphorylation to inhibitTNF-

    and IL-6 gene expressions and macrophage activation. Expo-

    sure of macrophages to LPS increased JNK phosphorylation.

    Meanwhile, administration of ketamine at a clinically relevant

    concentration (100 M) significantly decreased LPS-induced

    JNK phosphorylation. In response to LPS stimulation, proteinkinase JNK is activated via a TLR4-dependent mechanism

    (Jones et al., 2001; Fan et al., 2004). Our results revealed that

    ketamine can attenuate TLR4-mediated signals in LPS-

    activated macrophages. Thus, the ketamine-induced reduction

    of JNK phosphorylation may be due to suppression of TLR4-

    involved signal transduction by this intravenous anesthetic.

    JNK is an upstream protein kinase for activating AP-1

    (Potapova et al., 2001). Phosphorylation of JNK can activate

    AP-1 and induce certain gene expression. For example, a

    previous study showed that the JNK pathway participates in

    upregulating LPS-induced inducible nitric oxide (NO) synthase

    expression and NO production (Zhang et al., 2006). In the rathippocampus, pretreatment with ketamine was shown to

    decrease JNK phosphorylation induced by cerebral ischemia-

    reperfusion (Lahti et al., 2003). In this study, we further

    demonstrated that ketamine can decrease LPS-induced JNK

    phosphorylation. In addition, stress-induced JNK activation has

    been reported to be important in cell differentiation, apoptosis,

    and tumorigenesis (Potapova et al., 2001; Paik et al., 2003).

    Therefore, ketamine can decrease JNK phosphorylation to

    inhibit TNF- and IL-6 gene expressions, which leads to

    suppression of LPS-induced macrophage activation.

    Ketamine ameliorates LPS-induced translocation of c-Jun

    and c-Fos from the cytoplasm to nuclei via suppression of

    JNK activation. Exposure of macrophages to LPS significantly

    111G.-J. Wu et al. / Toxicology and Applied Pharmacology 228 (2008) 105113

  • 7/28/2019 Ketamine inhibits tumor necrosis factor- and interleukin-6 gene expression.pdf

    8/9

    increased the nuclear levels of c-Jun and c-Fos. After admin-

    istration of a therapeutic concentration of ketamine, the LPS-

    caused enhancements significantly decreased. JNK activation

    has been reported to phosphorylate c-Jun and c-Fos, which

    triggers the translocation of these two DNA-binding proteins

    from the cytoplasm to nuclei (Potapova et al., 2001). Treatment

    with ketamine can reduce LPS-induced JNK phosphorylation.The total protein levels of c-Jun and c-Fos were not influenced

    by the administration of ketamine, LPS, or a combination of

    ketamine and LPS. Thus, one of the major reasons explaining

    how ketamine can decrease nuclear c-Jun and c-Fos levels is

    that this intravenous anesthetic agent can suppress the

    translocation of c-Jun and c-Fos from the cytoplasm to nuclei

    via downregulation of LPS-stimulated JNK phosphorylation.

    Extracellular signal-regulated kinases can contribute to activa-

    tion of AP-1 (Kolch, 2005). Therefore, the other possible reason

    explaining the ketamine-caused reduction in AP-1 activation is

    that ketamine may modulate extracellular signal-regulated

    kinase phosphorylation to downregulate activation of this tran-scription factor.

    The AP-1 transcription factor is involved in ketamine-

    induced suppression of TNF- and IL-6 gene expressions. Jun

    and Fos are two major components in the heterodimeric structure

    of AP-1 (Hess et al., 2004). The ketamine-induced suppression

    of c-Jun and c-Fos translocation from the cytoplasm to nuclei

    means that this anesthetic agent can decrease AP-1 activation in

    LPS-stimulated macrophages. The AP-1 consensus DNA motifs

    are found in the promoter regions of the TNF- and IL-6 genes

    (Lu et al., 2005). Analysis by EMSA revealed that exposure

    of macrophages to LPS increased the DNA binding activity of

    AP-1. The LPS-induced increases in AP-1 DNA binding activity

    were significantly alleviated following administration ofketamine at a clinically relevant concentration (100 M). In

    response to stimulation by LPS, there are at least two different

    mechanisms reported to respectively induce TLR-dependent

    activation of NFB and AP-1 to regulate the expressions of

    inflammatory cytokine genes (Jones et al., 2001, Fan et al.,

    2004). In the intestine of rats, ketamine was shown to inhibit

    endotoxin-induced TNF- and IL-6 productions via suppression

    of NFB activity (Sun et al., 2004). Yang et al. (2005) reported

    that a large dose of ketamine can reduce NFB activation and

    inhibit expressions of TNF- and IL-6 genes in endotoxin-

    inducedrat acute lung injury (Yang et al., 2005). In this study, we

    further showed that a therapeutic concentration of ketamine canlower LPS-induced TNF- and IL-6 gene expressions through

    suppressing the translocation and transactivation of AP-1 in

    LPS-activated macrophages. Meanwhile, this study can not rule

    out the roles ofNFB in ketamine-involved suppression of TNF-

    and IL-6 gene expressions in LPS-activated macrophages.

    In conclusion, the present study shows that a clinically

    relevant concentration of ketamine (100 M) can decrease the

    biosyntheses of TNF- and IL-6 in LPS-activated macrophages.

    In parallel with decreases in protein levels, administration of

    ketamine significantly inhibited TNF- and IL-6 mRNA pro-

    ductions. Analysis of RNA interference further revealed that the

    ketamine-involved suppressions of TNF- and IL-6 syntheses

    were TLR4-dependent. After exposure to ketamine, the LPS-

    induced phosphorylation of JNK and sequent translocation of

    AP-1 from the cytoplasm to nuclei was significantly amelio-

    rated. Consequently, ketamine can suppress AP-1 DNA binding

    activity induced by LPS. According to our present data, we

    suggest that ketamine reduces the biosyntheses of TNF- and

    IL-6 in LPS-activated macrophages through suppressing TLR4-

    dependent JNK activation and AP-1 translocation and transac-tivation. The ketamine-induced suppression of inflammatory

    cytokine syntheses can partially explain its anti-inflammatory

    and immunosuppressive effects in clinical applications. In the

    future study, we will evaluate the signal-transducing effects of

    ketamine on the downstream molecules of TLR4 to further

    determine the roles of TLR4 in suppression of LPS-induced

    TNF- and IL-6 gene expressions induced by this intravenous

    anesthetic agent.

    Acknowledgments

    This study was supported by the National Science Council(NSC95-2745-B-038-001-URD) and Shin Kong Wu Ho-Su

    Memorial Hospital (SKH-TMU-94-04), Taipei, Taiwan. The

    authors express their gratitude to Ms. Yi-Ling Lin for her

    technical support and data collection during the experiments.

    References

    Akira, S., Uematsu, S., Takeuchi, O., 2006. Pathogen recognition and innate

    immunity. Cell 124, 783801.

    Bendtzen, K., 1998. Interleukin 1, interleukin 6 and tumor necrosis factor in

    infection, inflammation and immunity. Immunol. Lett. 19, 183191.

    Bonta, I.L., Ben-Efraim, S., 1993. Involvement of inflammatory mediators in

    macrophage antitumor activity. J. Leukoc. Biol. 54, 613626.

    Bourgoin, A., Albanese, J., Wereszczynski, N., Charbit, M., Vialet, R., Martin,

    C., 2003. Safety of sedation with ketamine in severe head injury patients:

    comparison with sufentanil. Crit. Care Med. 31, 711717.

    Calandra, T., Roger, T., 2003. Macrophage migration inhibitory factor: a

    regulator of innate immunity. Nat. Rev., Immunol. 3, 791800.

    Chang, Y., Chen, T.L., Sheu, J.R., Chen, R.M., 2005. Suppressive effects of

    ketamine on macrophage functions. Toxicol. Appl. Pharmacol. 204, 2735.

    Chen, R.M., Chen, T.G.,Chen, T.L., Lin,L.L., Chang, C.C., Chang, H.C., Wu,C.H.,

    2005a. Anti-inflammatory and antioxidative effects of propofol on lipopoly-

    saccharide-activated macrophages. Ann. N.Y. Acad. Sci. 1042, 262271.

    Chen, R.M., Chen, T.L., Lin, Y.L., Chen, T.G., Tai, Y.T., 2005b. Ketamine

    reduces nitric oxide biosynthesis in human umbilical vein endothelial cells

    through downregulating endothelial nitric oxide synthase expression and

    intracellular calcium levels. Crit. Care Med. 33, 10441049.

    Chen, T.G., Chen, T.L., Chang, H.C., Tai, Y.T., Cherng, Y.G., Chang, Y.T.,

    Chen, R.M., 2007. Oxidized low-density lipoprotein induces apoptotic

    insults to mouse cerebral endothelial cells via a bax-mitochondria-caspase

    protease pathway. Toxicol. Appl. Pharmacol. 219, 4253.

    Domino, E.F., Zsigmond, E.K., Domino, L.E., Domino, K.E., Kothary, S.P.,

    Domino, S.E., 1982. Plasma levels of ketamine and two of its metabolites in

    surgical patients using a gas chromatographic mass fragmentographic assay.

    Anesth. Analg. 61, 8792.

    Fan, H., Peck, O.M., Tempel, G.E., Halushka, P.V., Cook, J.A., 2004. Toll-like

    receptor 4 coupled GI protein signaling pathways regulate extracellular

    signal-regulated kinase phosphorylation and AP-1 activation independent of

    NFkappaB activation. Shock 22, 5762.

    Frangogiannis, N.G., Smith, C.W., Entman, M.L., 2002. The inflammatory

    response in myocardial infarction. Cardiovasc. Res. 53, 3147.

    Froidevaux, C., Roger, T., Martin, C., Glauser, M.P., Calandra, T., 2001.

    Macrophage migration inhibitory factor and innate immune responses tobacterial infections. Crit. Care Med. 29, S13S15.

    112 G.-J. Wu et al. / Toxicology and Applied Pharmacology 228 (2008) 105113

  • 7/28/2019 Ketamine inhibits tumor necrosis factor- and interleukin-6 gene expression.pdf

    9/9

    Grant, I.S., Nimmo, W.S., McNicol, L.R., Clements, J.A., 1983. Ketamine

    disposition in children and adults. Br. J. Anaesth. 55, 11071110.

    Hess, J., Angel, P., Schorpp-Kistner, M., 2004. AP-1 subunits: quarrel and

    harmony among siblings. J. Cell Sci. 117, 59655973.

    Hofbauer, R., Moser, D., Hammerschmidt, V., Kapiotis, S., Frass, M., 1998.

    Ketamine significantly reduces the migration of leukocytes through

    endothelial cell monolayers. Crit. Care Med. 26, 15451549.

    Hoshino, K., Takeuchi, O., Kawai, T., Sanjo, H., Ogawa, T., Takeda, Y., Takeda,K., Akira, S., 1999. Toll-like receptor 4 (TLR4)-deficient mice are hypo-

    responsive to lipopolysaccharide: evidence for TLR 4 as the lps gene

    product. J. Immunol. 162, 37493752.

    Jacob, C.O., 1992. Studies on the role of tumor necrosis factor in murine and

    human autoimmunity. J. Autoimmun. 5, 1331343.

    Jones, B.W., Means, T.K., Heldwein, K.A., Keen, M.A., Hill, P.J., Belisle, J.T.,

    Fenton, M.J., 2001. Different Toll-like receptor agonists induce distinct

    macrophage responses. J. Leukoc. Biol. 69, 10361044.

    Kolch, W., 2005. Coordinating ERK/MAPK signalling through scaffolds and

    inhibitors. Nat. Rev., Mol. Cell Biol. 6, 827837.

    Lahti, A., Jalonen, U., Kankaanranta, H., Moilanen, E., 2003. c-Jun NH2-

    terminal kinase inhibitor anthra(1,9-cd)pyrazol-6(2H)-one reduces inducible

    nitric-oxide synthase expression by destabilizing mRNA in activated

    macrophages. Mol. Pharmacol. 64, 308315.

    Lu, H., Wu, J.Y., Kudo, T., Graham, D.Y., Yamaoka, Y., 2005. Regulation ofinterleukin-6 promoter activation in gastric epithelial cells infected with

    Helicobacter pylori. Mol. Biol. Cell 16, 49544966.

    Melamed, R., Bar-Yosef, S., Shakhar, G., Shakhar, K., Ben-Eliyahu, S., 2003.

    Suppression of natural killer cell activity and promotion of tumor metastasis

    by ketamine, thiopental, and halothane, but not by propofol: mediating

    mechanisms and prophylactic measures. Anesth. Analg. 97, 13311339.

    Nagy, L.E., 2004. Stabilization of tumor necrosis factor- mRNA in

    macrophages in response to chronic ethanol exposure. Alcohol 33, 229233.

    Nathan, C.F., 1987. Neutrophil activation on biological surfaces. Massive

    secretion of hydrogen peroxide in response to products of macrophages and

    lymphocytes. J. Clin. Invest. 80, 15501560.

    Paik, Y.H., Schwabe, R.F., Bataller, R., Russo, M.P., Jobin, C., Brenner, D.A.,

    2003. Toll-like receptor 4 mediates inflammatory signaling by bacterial

    lipopolysaccharide in human hepatic stellate cells. Hepatology 37,

    10431055.Potapova, O., Basu, S., Mercola, D., Holbrook, N.J., 2001. Protective role

    for c-Jun in the cellular response to DNA damage. J. Biol. Chem. 276,

    2854628553.

    Raetz, C.R., Ulevitch, R.J., Wright, S.D., Sibley, C.H., Ding, A., Nathan, C.F.,

    1994. Gram-negative endotoxin: an extraordinary lipid with profound

    effects on eukaryotic signal transduction. FASEB J. 5, 26522660.

    Reich, D.L., Silvay, G., 1989. Ketamine: an update on the first twenty-five years

    of clinical experience. Can. J. Anaesth. 36, 186197.

    Rofael, H.Z., Turkall, R.M., Abdel-Rahman, M.S., 2003. Effect of ketamine on

    cocaine-induced immunotoxicity in rats. Int. J. Toxicol. 22, 343358.

    Rothman, S.M., Thurston, J.H., Hauhart, R.E., Clark, G.D., Solomon, J.S.,

    1987. Ketamine protects hippocampal neurons from anoxia in vitro.

    Neuroscience 21, 673678.

    Sachs, L., Lotem, J., Shabo, Y., 1989. The molecular regulators of macrophage

    and granulocyte development. Ann. N.Y. Acad. Sci. 557, 417435.

    Scott, D.L., Kingsley, G.H., 2006. Tumor necrosis factor inhibitors for

    rheumatoid arthritis. N. Engl. J. Med. 355, 704712.

    Sidell, N., Han, S.W., Parthasarathy, S., 2002. Regulation and modulation ofabnormal immune responses in endometriosis. Ann. N.Y. Acad. Sci. 955,

    159173.

    Suliburk, J.W., Mercer, D.W., 2007. Ketamine attenuates early l ipopolysacchar-

    ide-induced gastric dysfunction: role of stress-inducible phosphoproteins.

    J. Trauma 62, 316319.

    Sun, J., Wang, X.D., Liu, H., 2004. Ketamine suppresses endotoxin-induced

    NF-kappaB activation and cytokines production in the intestine. Acta

    Anaesthesiol. Scand. 48, 317321.

    Tai, Y.T., Chen, T.L., Cherng, Y.G., Chang, C.C., Hwang, Y.P., Chen, J.T., Chen,

    R.M., 2007. Pretreatment with low nitric oxide protects osteoblasts from

    high nitric oxide-induced apoptotic insults through regulation of c-Jun

    N-terminal kinase/c-Jun-mediated Bcl-2 gene expression and protein trans-

    location. J. Orthop. Res. 25, 625635.

    Taniguchi, T., Shibata, K., Yamamoto, K., 2001. Ketamine inhibits endotoxin-

    induced shock in rats. Anesthesiology 95, 928932.Trinchieri, G., Sher, A., 2007. Cooperation of Toll-like receptor signals in innate

    immune defence. Nat. Rev., Immunol. 7, 179190.

    Weigand, M.A., Schmidt, H., Zhao, Q., Plaschke, K., Martin, E., Bardenheuer,

    H.J., 2000. Ketamine modulates the stimulated adhesion molecule

    expression on human neutrophils in vitro. Anesth. Analg. 90, 206212.

    White, P.F., Way, W.L., Trevor, A.J., 1982. Ketamine: its pharmacology and

    therapeutic uses. Anesthesiology 56, 119136.

    Wilder, R.L., Elenkov, I.J., 1999. Hormonal regulation of tumor necrosis factor-

    alpha, interleukin-12 and interleukin-10 production by activated macro-

    phages. A disease-modifying mechanism in rheumatoid arthritis and

    systemic lupus erythematosus? Ann. N.Y. Acad. Sci. 876, 1431.

    Wu, C.H., Chen, T.L., Chen, T.G., Ho, W.P., Chiu, W.T., Chen, R.M., 2003.

    Nitric oxide modulates pro- and anti-inflammatory cytokines in lipopoly-

    saccharide-activated macrophages. J. Trauma 55, 540545.

    Wu, G.J., Chen, T.G., Chang, H.C., Chiu, W.T., Chang, C.C., Chen, R.M., 2007.Nitric oxide from both exogenous and endogenous sources activates

    mitochondria-dependent events and induces insults to human chondrocytes.

    J. Cell. Biochem. 101, 15201531.

    Yang, J., Li, W., Duan, M., Zhou, Z., Lin, N., Wang, Z., Sun, J., Xu, J., 2005.

    Large dose ketamine inhibits lipopolysaccharide-induced acute lung injury

    in rats. Inflamm. Res. 54, 133137.

    Zhang, Q.G., Xu, Y.L., Li, H.C., Han, D., Zhang, G.Y., 2006. NMDA receptor/

    L-VGCC-dependent expression and AMPA/KA receptor-dependent activa-

    tion of c-Jun induced by cerebral ischemia in rat hippocampus. Neurosci.

    Lett. 398, 268273.

    113G.-J. Wu et al. / Toxicology and Applied Pharmacology 228 (2008) 105113