Nutrition 24 (2008) 187–199 www.elsevier.com/locate/nut
Basic nutritional investigation
Conjugated linoleic acid increases intracellular ROS synthesis and oxygenation of arachidonic acid in macrophages Ewa Stachowska, Ph.D.a,*, Magdalena Bas´kiewicz-Masiuk, M.D., Ph.D.b, Violetta Dziedziejko, Ph.D.c, Izabela Gutowska, Ph.D.a, Irena Baranowska-Bosiacka, Ph.D.c, Mariola Marchlewicz, M.D., Ph.D.d, Barbara Dołe˛gowska, Ph.D.e, Barbara Wiszniewska, Ph.D.d, Bogusław Machalin´ski, M.D., Ph.D.b, and Dariusz Chlubek, M.D., Ph.D.c a
Laboratory of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland b Department of General Pathology, Pomeranian Medical University, Szczecin, Poland c Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland d Department of Histology and Embryology, Pomeranian Medical University, Szczecin, Poland e Department of Laboratory Diagnostics and Molecular Medicine, Pomeranian Medical University, Szczecin, Poland Manuscript received December 22, 2006; accepted October 26, 2007.
Abstract
Objective: Conjugated linoleic acids (CLAs) have potential antiatherosclerotic properties: they may inhibit atherosclerotic processes by reducing the intensity of inflammatory processes. However, in vivo studies have shown that the application of trans-10, cis-12 CLA in obese men increased their oxidative stress. The objective of this study was to determine whether CLA can lead to an increase in oxidative stress and to isoprostane synthesis in macrophages. Methods: Monocytes from peripheral blood and human monocytic leukemia cells were used in this study. Monocytes were differentiated to macrophages, and were incubated with 30 M cis-9, trans-11 CLA and trans-10, cis-12 CLA or linoleic acid for 2 days. In some experiments the inhibitors of peroxisome proliferator-activated receptor-␣ (PPAR-␣) or respiratory chain were added. After incubation, synthesis of reactive oxygen species (ROS), total cellular concentration of adenosine triphosphate, concentration of 8-epi-prostaglandin F2␣, activity of cytoplasolic phospholipase A2 (cPLA2), activity of mitochondria, and expression of mRNA of PPAR-␣ were measured. Results: In cells cultured with CLAs intercellular ROS synthesis increased. In this condition the mitochondrial energy potential was high, and the inhibitors of the respiratory chain and PPAR-␣ reduced ROS concentration. At the same time, the cPLA2 activity was abolished. In contrast, 8-iPF2␣III synthesis increased in CLA cells. Conclusion: Cultivation of cells with CLA leads to an increased ROS synthesis, partly by PPAR-␣ mechanism. An increase in ROS concentration and inhibition of cPLA2 activity can stimulate oxygenation of arachidonic acid and contribute to an increase in 8-epi-PF2␣III level and in the apoptosis process in macrophages. © 2008 Elsevier Inc. All rights reserved.
Keywords:
Macrophages; Conjugated linoleic acid; Reactive oxygen species; Isoprostanes
Introduction Conjugated linolic acid dienes (CLAs) are fatty acids found in food [1,2]. Their most common isomers are cis-9, This study was supported by grant 3 POB 11723 from the State Committee for Scientific Research, Poland. * Corresponding author. Tel.: ⫹48-91466-1516; fax: ⫹48-466-1515. E-mail address:
[email protected] (E. Stachowska). 0899-9007/08/$ – see front matter © 2008 Elsevier Inc. All rights reserved. doi:10.1016/j.nut.2007.10.018
trans-11 C18:2 isomers and trans-10, cis-12 C18:2 isomers [1]. In animals, CLAs have antiatherosclerotic properties; they are responsible for a decrease in the concentration of atherogenic lipoproteins of plasma and for normalization of plasma glucose levels [3–5]. CLAs also inhibit atherosclerotic processes in the body, e.g., by decreasing the intensity of inflammatory processes in cells [6]. The antiatherosclerotic function of CLA, well documented in animals [7], might be useful in the prevention of athero-
188
E. Stachowska et al. / Nutrition 24 (2008) 187–199
sclerosis. However, the application of trans-10 CLA, cis-12 isomer of CLA in the diet of obese men was shown to increase their oxidative stress (measured by the concentration of isoprostane 8-epi-prostaglandin F2␣ [8-epiPGF2␣III] in urine) and increased C-reactive proteins (by 110%) in the body [8]. Trans-10, cis-12 CLA isomer turned out to be the activator of lipid peroxidation and oxidative stress and contributed to an increase in inflammatory marker concentration [8]. Studies on laboratory animals have proved that the metabolic action of CLA in the cell is based on stimulation of receptors activated by peroxisome proliferators [9,10]. As demonstated, CLAs belong to the most potent agonists of such receptors [3]. Peroxisome proliferatoractivated receptors (PPARs) belong to the superfamily of nucleic receptors [11–13]. They are transcription factors that function as regulators of gene transcription related to lipid metabolism [14,15]. Ligands of PPARs are mainly fatty acids and their eicosanoid derivatives [11–13]. The level of expression of individual PPARs differs substantially for different tissue types [11–13]. Three main types of PPAR coded by separate PPAR genes have been identified: ␣, , and ␥. Two of these receptors, PPAR-␣ and PPAR-␥, are particularly active in macrophages [16]. The PPAR-␣ receptor undergoes the most intense expression in tissues characterized by intense catabolism of fatty acids [15]. Its main function is the regulation of genes engaged in mitochondrial and peroxisomal -oxidation of fatty acids [14,15]. PPAR-␣ is required for the induction of genes involved in -oxidation [15]. Both receptor types are activated by different agonists; PPAR-␣ is activated by polyunsaturated fatty acids (including CLA) [15]. CLA is also a natural ligand and activator of PPAR-␣ [3]. CLA isomers appear to be among the most avid fatty acid ligands of PPAR-␣ [3]. Inflammatory reactions underlie the pathogenesis of the atherosclerotic process [17,18] and oxygen free radicals formed during inflammatory reactions contribute to aggravation of atherosclerotic lesions [19]. This is because free radicals synthesized in quantities exceeding the efficiency of antioxidative systems (such as the oxygenated/reduced glutathione system, antioxidant enzymes) lead to damage of biomolecules important for correct functioning of the cell [19]. Reactive oxygen species (ROSs) are toxic products formed as a result of a decrease of molecular oxygen in the cell [17,20]. In monocytes/macrophages, ROS sources are the mitochondrial electron transport chain, cyclo-oxygenases, lipo-oxygenases, cytochrome P-450 and nicotinamide adenosine dinucleotide phosphate oxidase [17,20]. ROSs participate in the regulation of numerous cellular functions, e.g., cellular differentiation, intracellular signaling [21], gene expression regulation, and (through activation of caspases or Fas receptors) in apoptotic stimulation [22]. Macrophages are also a source of free radicals other than ROSs, e.g., reactive nitrogen species [23,24]. In granulocytes ROSs and reactive nitrogen species have a dual function:
they function as signaling molecules and serve as modulators of protein and lipid kinases and phosphatases, receptors, ion channels, and transcription factors. They regulate expression of key cytokines and chemokines that further modulate the inflammatory response. During an inflammatory response, ROSs and reactive nitrogen species modulate phagocytosis, secretion, gene expression, and apoptosis [25]. Isoprostanes are mostly formed during non-enzymatic peroxidation of arachidonic acid (AA), bound to lipid membrane phospholipids, and catalyzed by ROSs [26]. In fact, isoprostanes are chemically stable prostaglandin isomeres [19,26 –28]. In monocytes, cells participating in the inflammatory process, isoprostanes (e.g., 8-epi-PGF2␣III) are synthesized predominantly by non-enzymatic free radical peroxidation of AA in membranes [28]. Macrophages play a key role in the inflammatory process. Thus far reports have indicated an anti-inflammatory function of CLAs in macrophages [10,29]. This report shows for the first time that CLAs are the substances that may increase the syntheses of ROSs and isoprostane 8-epi-PGF2␣III in macrophages. This mechanism may be based on CLA-induced inhibition of cytoplasmic phospholipase A2 (PLA2) [30], which potentially retains AA in membrane phospholipids and increases its susceptibility to ROSs. The objective of the study was to determine whether two isomers of CLA predominant in food (cis-9, trans-11 C18:2 and trans-10, cis-12 C18:2), when administered to macrophages, had an effect on ROS synthesis, mitochondrial activity and isoprostane 8-epi-PGF2␣III production. The cis-9, trans-11 CLA isomer was selected for testing because it is the CLA isomer predominant in the human diet, constituting ⬎80% of all CLAs present in milk and milk-derived products [23]. The second isomer, trans-10, cis-12, is present in milk in trace amounts but was selected on the basis of its high metabolic activity in the cell [31–33].
Materials and methods Reagents and sources RPMI medium, glutamine, and antibiotics (penicillin and streptomycin) were from Sigma-Aldrich (Poznan´, Poland). THP-1 was obtained from American Type Culture Collection (Rockville, MD, USA). Fetal bovine serum was from Gibco (Gibco, Paisley, UK). Phosphate buffered saline (PBS) and Lymphozyten separation medium were from PAP Laboratories (Vienna, Austria). CD14⫹ MicroBeads MS separation columns, nylon filters (30 m), and MACS separator system were from Miltenyi Biotec (Auburn, CA, USA). CLAs were from Nu-CheK Prep (Elysian, MN, USA; 98% purity). The fatty acids for cell culture were dissolved in fatty acid–free albumin from Sigma-Aldrich. All reagents and
E. Stachowska et al. / Nutrition 24 (2008) 187–199
solvents for extraction of 8-epi-PGF2␣III were from SigmaAldrich. C18 reverse-phase columns (JT Baker, Phillipsburg, NJ, USA) and the solid-phase extraction Vacuum Manifold system used for 8-epi-PGF2␣III extraction were from Supelco (Poznan´, Poland). The 8-iPGF2␣ immunoassay kits were from Oxis (Portland, OR, USA). Deionized water was from SigmaAldrich. The Bradford-based protein concentration measured kit was from Sigma-Aldrich. N,N,N=,N=-tetramethyl-pphenylenediamine and 2=,7=-dichlorofluorescein diacetate (DCFH-DA) were obtained from Sigma-Aldrich. An R-phycoerythrin-conjugated mouse anti-human monoclonal antibody set was obtained from BD Biosciences Pharmingen (USA). M-PER mammalian protein extraction reagent was from Pierce (Rockford, IL, USA). An annexin V/fluorescein isothiocyanate (FITC) apoptosis detection kit was obtained from BD Pharmingen. Potassium cyanide was from POCH (Poland), and antimycin A, rotenone, and GW 6471 (inhibitor of PPAR-␣) were from Sigma-Aldrich. MitoTracker green FM was from Molecular Probes (Europe BV, Leiden, The Netherlands). JC-1 fluorochrome (5,5=,6,6=-tetrachloro-1=,13,3=tetraethylbenzimidazol-carbocyane iodide) was from Molecular Probes, Europe BV. Experimental conditions Monocytes (CD14⫹) were isolated from blood from healthy donors. Blood sampling was performed in accordance with the principles outlined in the Declaration of Helsinki [34]. First, peripheral blood mononuclear cells were isolated from anticoagulated blood using Lymphozyten separation medium [35]. Then cells were passed through a 30-m nylon filter to remove clumps and magnetically labeled with CD14⫹ MicroBeads. After incubation (15 min, 6°C) cells were separated in a column that was placed in the magnetic field of a MACS separator. The magnetically labeled CD14⫹ cells were retained in the column, and the unlabeled cells passed through. In the next step, after removal of the column from the magnetic field, the magnetically retained CD14⫹ cells were eluted using a separation buffer (PBS, pH 7.2, supplemented with 0.5% bovine serum albumin [BSA] and 2 mM ethylenediaminetetra-acetic acid). CD14⫹ cells were seeded with RPMI medium with 10% autologous serum. Cells were cultured at 37°C in a 5% CO2 humid atmosphere in RPMI medium containing 2 mM glutamine, antibiotics, and 10% autologous human serum and then cultured for 7 d [17]. THP-1 cells were differentiated into macrophages by administering phorbol myristate acetate [36], and cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum without fatty acid, penicillin (100 U/mL), and streptomycin (100 mg/mL) at 37°C in 5% CO2. The THP-1 monocytes were treated with 100 nM phorbol myristate acetate for 24 h, and then the adherent macrophages were washed three times with PBS and incubated with fatty acids
189
for 48 h at 37°C. Fatty acids were added as a 4-mM stock solution dissolved in 1 mM of fatty acid–free BSA [18]. Isomers of CLA (cis-9, trans-11, and trans-10, cis-12) were used at final concentrations of 30 M. Incubation time and fatty acid concentration were selected on the basis of results obtained in preliminary experiments. Cells with or without fatty acids (incubated with BSA) were incubated for 2 d. The cells were harvested by trypsinization and a pellet was obtained by centrifugation (250g for 5 min). The cell count was determined with a Bright Line Hemacytometer (Sigma-Aldrich). Cell viability was examined using a trypan blue dye exclusion method. Protein concentration was measured by the Bradford method [37]. Differentiation of monocytes to macrophages was estimated by flow cytometry with an anti-CD68 antibody. After 7 d of incubation with fatty acids, the percentage of CD68 cells was assessed by flow cytometry (FACScan) using CellQuest software [38]. CD68 cells were washed with ice-cold PBS, fixed with 4% paraformaldehyde, permeabilized using 0.5% Triton X-100, and incubated with fluorochrome-conjugated monoclonal FITC CD68 antibody for 30 min. Mouse immunoglobulin G2 was used as an isotype control antibody (Becton Dickinson, Oxford, United Kingdom). Macrophages cultured without BSA and fatty acids were estimated in this study as a negative control. Intracellular ROS formation After incubation CLA cells were preloaded (30 min at 37°C) with 5 M DCFH-DA. DCFH-DA becomes fluorescent on oxidation to DCF by hydrogen peroxide within the cell [17,39 – 41]. Cells were harvested and the number of cells exhibiting increased fluorescence of oxidized DCF was measured by flow cytometry (FACSCalibur, Becton Dickinson) as previously described [17]. To determine whether the respiratory chain was the main source of ROSs in this experiment, 1-h incubation of macrophages was carried out (after 2 d of cultivation with CLA). For some cells a KCN (1 mM ) mixture of antimycin A (5 g/mL; a mitochondrial complex III inhibitor) and rotenone (10 M; a mitochondrial complex I inhibitor) was added for 24 h before ROS measurements. In other experiments the PPAR-␣ inhibitor GW 6471 was added (50 M) to cells with fatty acids or BSA 48 h before ROS measurements. Fluorescent microscopy: imaging of mitochondria by Mito Traker Green FM and fluorometric determination of membrane potential by JC-1 MitoTracker Green FM is frequently used as a fluorescent probe in mitochondrial imaging [42]. To stain the mitochondria, macrophages cultivated for 48 h with fatty acids were incubated in a medium containing
190
E. Stachowska et al. / Nutrition 24 (2008) 187–199
MitoTracker (0.2 g of fluorochrome in 1 mL of RPMI medium) for 25 min at 37°C in an incubator. After incubation, the preparations were examined under a fluorescent microscope (Axiskop Zeiss, Germany) using a 09 filter (filter set 09-487909-000; Carl Zeiss, Germany). Mitochondrial membrane potential was estimated with JC-1. The macrophages cultivated with fatty acids were incubated with lipophilic cationic fluorochrome JC-1. The medium containing JC-1 with the final concentration of 10 g/mL was prepared from the initial solution containing 1 mg of JC-1/1 mL of dimethyl sulfoxide. The cells were incubated with fluorochrome in an incubator for 25 min and then examined under a fluorescent microscope. JC-1 exhibits potential dependent accumulation in mitochondria and displays two colors of fluorescence: green and red/orange/yellow [43]. A ⌬psi above 80 –100 mV causes an orange fluorescence emission (emission maximum at 590 nm) due to reversible formation of JC-1 aggregates in polarized mitochondria. The green florescence is characteristic for a monomeric form of the dye that accumulates only in depolarized mitochondria with low mitochondrial membrane potential (⌬⌿ ⬍80 –100 mV). High-performance liquid chromatographic separation of adenosine triphosphate (and other purines) in macrophages cultured with fatty acids After incubation with BSA or fatty acids, the cells were washed with PBS buffer (Sigma-Aldrich) at ⫹4°C and centrifuged (500g) for 10 min at ⫹4°C. The centrifuged cells were suspended in 200 L of PBS and deproteinized with 200 L of 1.3 mol/L of HClO4. The samples were centrifuged (14 000g) for 10 min at ⫹4°C. Two hundred microliters of supernatant was neutralized with 20 L of 3 M K3PO4 (Fluka, Poznan´, Poland) to achieve a pH range of 6.0 –7.0. The samples were centrifuged again (under the same conditions) and the supernatant was frozen at ⫺70°C until further analysis. The concentrations of purines (adenosine-5=triphosphate [ATP], adenosine-5=-diphosphate [ADP], adenosine-5=-monophosphate [AMP], and adenosine) were determined in the prepared samples. For high-performance liquid chromatographic analysis, a Hewlett-Packard chromatographic system (HP 1100, Austria) was used. The analytical column (100 ⫻ 4.6 mm) was packed with 3 m of Hypersil BDS-C18 (Aqilent Technologies, Wilmington, DE, USA). The following buffers were used: buffer A contained 150 mM KH2PO4 and 150 mM KCl adjusted to pH 6.0 with K2HPO4; buffer B was a 15% solution of acetonitrile in buffer A. Peaks were detected by absorption measurements at 254-nm wavelength. The adenylate energy charge (AEC) was evaluated according to the formula by Atkinson and Walton [44]: AEC ⫽ 共关ATP兴 ⫹ 0.5关ADP兴兲/共关ATP兴 ⫹ 关ADP兴 ⫹ 关AMP兴兲
Measurements of non-enzymatic oxidation 8-epi-PGF2␣III After 7 d of incubation with fatty acids, cells and supernatant were collected and frozen at ⫺80°C until 8-epi-PGF2␣III isolation. Lipid fractions were extracted from cells with a Folch mixture [45] (chloroform:methanol 2:1, v/v) with butylated hydroxytoluene (0.05%) as an antioxidant, the mixture was intensively vortex-mixed, and 0.043% MgCl2 was added. The mixture was vortex-mixed again and centrifuged (3824 for 10min). Downer phase was taken and evaporated under N2. The residue after evaporation was saponified and hydrolyzed with 15% KOH and methanol (1:1, v/v). The mixture was vortex-mixed again and incubated at 37°C for 30 min. Thereafter, 1 mM of HCl was added and the mixture was applied under a separation column (Bakerbond spe, RP-18, J.T. Baker) [44]. The 8epi-PGF2␣III was eluted from C18 Sep Pak by a mixture of ethyl acetate:methanol (1:1, v/v) and samples were evaporated under N2, resuspended in dilution buffer, and assayed as described by the Oxis protocol. Measurement of PLA2 activity After incubation, cells (macrophages from blood) with CLA (5 M final concentration) ionophore (A 23187) added to the cells were incubated for 1 h at 37°C with gentle agitation [46]. After incubation, the cells and the medium were used for extraction of total lipids using a Folch mixture (2:1, v/v) of chloroform:methanol containing 0.01% (w/v) butylated hydroxytoluene as an antioxidant, as described previously in detail [30]. Measurement of PLA2 activity in macrophages in THP-1 was described in detail previously [30]. Complement DNA synthesis and quantitative real-time polymerase chain reaction Total RNA was extracted from cell cultures using a Qiagen kit (Gibco). The quality of RNA was confirmed by denaturation gel electrophoresis and analysis on an Agilent 2100 Bioanalyser (Agilent Technologies, Santa Clara, CA, USA). To confirm regulation of the PPARs, a quantitative expression analysis was performed by real-time polymerase chain reaction (PCR) using glyceraldehyde 3-phosphate dehydrogenase (GAPDH) as the reference gene, as described in detail previously [46]. Subsequently, cDNA was subjected to real-time PCR in a reaction mixture containing QuantiTect SYBR Green PCR (Qiagen, Valencia, CA, USA) mix and primers. The following primer sequences were used in this study: PPAR-␣ forward primer: 5=GGACGTGCTTCCTGCTTCAT-3=; reverse primer: 5=CACCATCGCGACCAGATG-3=; GAPDH forward primer: 5=-GCCAGCCGAGCCACATC-3=; reverse primer: 5=GCGCCAATACGACCAAA-3=. All real-time PCR reactions were performed on a DNA Engine Option II
E. Stachowska et al. / Nutrition 24 (2008) 187–199
(MJ Research, GMI Inc., Ramsey, MN, USA). The thermal profile included initial denaturation for 15 min at 95°C followed by 40 amplification cycles of denaturation for 30 s at 72°C. After PCR amplification, melting curve analysis was performed with a temperature profile slope of 1°C/s from 35°C to 95°C. A negative control without the cDNA template was run with every assay to ensure overall specificity. The expression rates were calculated as detailed previously [47,48]. Western blot Antibodies against PPAR-␣ and peroxidase/anti-mouse immunoglobulin G1 were from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Cell monolayers were washed in PBS and lysed with lysing buffer (M-PER mammalian protein extraction reagent, apoprotein 10 mg/mL), pepstatin A 1 mg/mL, sodium orthovanadate 1 M, sodium fluoride 1 M, sodium pyrophosphate 100 mM, leupeptin 10 mg/ mL, ethylene-diaminetetra-acetic acid 500 mM, and phe-
191
nyl methyl sulfonyl fluoride 10 mg/mL). Total protein was separated in 10% sodium dodecylsulfate/polyacrylamide gel electrophoresis, and proteins from gels were blotted onto nitrocellulose membranes at 100 mA for 1 h at 4°C. Membranes were then incubated for 1 h with antibodies direct against PPAR-␣ (1:500) or with a monoclonal anti–-actin (clone AC-74, Sigma-Aldrich). Bound antibody was detected by using the appropriate horseradish peroxidase– conjugated antibody. Signals were visualized by chemiluminescence (Amersham, Buckinghamshire, United Kingdom) [17]. Assay of apoptosis Cells were analyzed by flow cytometry by staining with propidium iodide using FACScan cytofluorometry. The measurement of apoptosis was carried out with a BectonDickinson annexin V/FITC (Oxford, UK) apoptosis detection kit according to the manufacturer’s instructions.
Fig. 1. An intense green fluorescence in the cytoplasm after incubation with MitoTracker Green FM probe in cytoplasm of macrophages cultured with (A) the control bovine serum albumin, (B) trans-10, cis-12 conjugated linoleic acid diene isomer, (C) cis-9, trans-11 conjugated linoleic acid diene isomer, and (D) linoleic acid. THP-1 macrophages were cultured with fatty acids for 48 h as described in MATERIALS AND METHODS.
192
E. Stachowska et al. / Nutrition 24 (2008) 187–199
Fig. 2. Intense yellow-orange fluorescence after incubation macrophages with JC-1 probe. Cells were cultured with (A) the control bovine serum albumin, (B) trans-10, cis-12 conjugated linoleic acid diene isomer, (C) cis-9, trans-11 conjugated linoleic acid diene isomer, and (D) linoleic acid. THP-1 macrophages were cultured with fatty acids for 48 h as described in MATERIALS AND METHODS.
Statistical analysis All results are expressed as mean ⫾ standard error. Because the distribution in most cases deviated from normal (Shapiro-Wilk test), non-parametric tests were used. For related samples, significance was first checked with Friedmann’s analysis of variance, and significant results were subjected to the Wilcoxon matched-pair test. Analysis was done with Statistica 6.1 (Statsoft, Poland). P ⬍ 0.05 was considered statistically significant.
Results Effect of CLA on intracellular ROS synthesis in macrophages Cells cultivated with CLA and LA exhibited bright green fluorescence, indicating the presence of active and intact mitochondria in the cytoplasm (Fig. 1A–D). After incubation of macrophages with the mitochondrial probe JC-1,
yellow-orange fluorescence from the polarized mitochondria was visible, with a correct potential of the intracellular mitochondrial membrane (⌬⌿m 80 –100 mV; Fig. 2A–D). The culture of macrophages with CLA and LA enhanced the synthesis of ROSs in macrophages (P ⱕ 0.02, macrophages from THP-1; P ⱕ 0.04, macrophages from blood; Table 1). In macrophages from THP-1, increased ROS synthesis was found in the environment of the trans-10, cis-12 CLA isomer (⫹38% versus BSA, P ⱕ 0.01) and cis-9, trans-11 CLA (⫹23%, P ⱕ 0.01), with a trend toward increased ROS synthesis in cells cultivated with LA (Table 1). Similarly, in macrophages obtained from blood, increased ROS synthesis was noted in cells cultivated with both CLAs (⫹20% for trans-10, cis-12 CLA, P ⱕ 0.01; ⫹14% for cis-9, trans-11, P ⱕ 0.05; Table 1). To determine whether the respiratory chain is the main source of ROSs in macrophages cultivated with fatty acids, 1-h incubation of macrophages (cultivated earlier for 48 h with fatty acids) with 1 mmol/L of KCN (cytochrome oxydase inhibitor) was performed. In the presence of KCN, ROS synthesis (expressed as percentage of
E. Stachowska et al. / Nutrition 24 (2008) 187–199
193
Table 1 Effect of fatty acids on ROS synthesis in macrophages cultured with fatty acids* Experimental conditions
BSA Trans-10, cis-12 CLA Cis-9, trans-11 CLA Linoleic acid Negative control
Macrophages from THP-1 (n ⫽ 8)
Macrophages from blood (n ⫽ 8)
DCF fluorescence intensity
Percent of control
DCF fluorescence intensity
Percent of control
421 ⫾ 103 589 ⫾ 122‡ 539 ⫾ 116‡ 560 ⫾ 262 400 ⫾ 261
⫹38 ⫹23 ⫹33
100 ⫾ 63 120 ⫾ 68‡ 114 ⫾ 47† 110 ⫾ 57 101 ⫾ 57
⫹20 ⫹14 ⫹10
BSA, bovine serum albumin; CLA, conjugated linoleic acid diene; DCFH-DA, 2=,7=-dichlorofluorescein diacetate; ROS, reactive oxygen species * Macrophages were cultured with fatty acids for 48 h. Cells were the incubated with DCFH-DA (25 mol/L) for 30 min and final fluorescence intensity was measured by flow cytometry. Results are expressed in DCF fluorescence intensity and percentage of control (BSA taken as 100%). Results are shown as mean fluorescence ⫾ SD from eight separate experiments. † P ⬍ 0.05, significant difference versus corresponding control. ‡ P ⬍ 0.01, significant difference versus corresponding control.
BSA control) was decreased in macrophages obtained from THP-1 (Table 2). The reduction of the ROS synthesis was also observed in macrophages cultivated with the mixture of the respiratory chain inhibitors: antimycin A (5 g/mL) and rotenone (10 M; Table 2).
THP-1 monocytes and peripheral blood mononuclear cells; Fig. 4 and Fig. 5A,B).
ROS synthesis in macrophages cultivated with CLA seems to be partly controlled by PPAR-␣
Both isomers of CLA significantly increased the concentration of ATP and other adenylates (adenosine and AMP) in macrophages obtained from THP-1 (Table 3). In the cells cultivated with CLA, the AEC values also increased.
To determine if the ROS synthesis may be the result of processes controlled by PPAR-␣, the cells were cultivated for 48 h with fatty acids and with addition of the inhibitor of the PPAR-␣ receptor, GW 6471 (50 M; Table 2 and Fig. 3a– d). In comparison with the BSA control, ROS synthesis was decreased in the cells cultivated with both isomers of CLA. Yet only one isomer, trans-10, cis-12, resulted in a distinct 28% reduction of the DCF fluorescence; another decreased ROS synthesis by about 10%. In contrast to CLA, the cells cultivated with LA and with GW 6471 tended to show increased ROS synthesis (Table 2, Fig. 3A–D). In this study the mRNA of PPAR-␣ and protein of the receptor were expressed in both types of macrophages (from
CLA changes intracellular concentration of adenylates and value of AEC
Influence of CLA on 8-epi-PGF2␣ concentration In THP-1 macrophages, both CLA isomers increased the concentration of 8-epi-PGF2␣ by 150% (compared with the BSA control, P ⱕ 0.05). In macrophages from peripheral blood, the trans-10, cis-12 CLA isomer increased the concentration of 8-epi-PGF2␣ by 160% (P ⱕ 0.05) and the cis-9, trans-11 CLA isomer increased this value by 120% (compared with the BSA control, P ⱕ 0.05; Table 4).
Table 2 Effect of inhibitors of respiratory chain and PPAR-␣ on ROS synthesis in macrophages cultured with fatty acids Treatment
BSA with inhibitors (control) Trans-10, cis-12 CLA Cis-9, trans-11 CLA Linoleic acid BSA untreated inhibitor
DCF fluorescence intensity KCN (1 mM) treatment (n ⫽ 3)
Antimycin A (5 g/mL) ⫹ rotenone (10 M) mix treatment (n ⫽ 3)
PPAR-␣ inhibitor GW 6471 (50 M, n ⫽ 5)
89 ⫾ 4 74 ⫾ 2 88 ⫾ 5 86 ⫾ 7 402 ⫾ 89
90 ⫾ 9 89 ⫾ 5 84 ⫾ 6 112 ⫾ 3 417 ⫾ 104
100 ⫾ 13 72 ⫾ 5† 94 ⫾ 11 144 ⫾ 18† 392 ⫾ 97
BSA, bovine serum albumin; CLA, conjugated linoleic acid diene; DCFH-DA, 2=,7=-dichlorofluorescein diacetate; PPAR, peroxisome proliferatoractivated receptor; ROS, reactive oxygen species ⴱMacrophages (THP-1) were cultured with fatty acids for 48 h and then incubated with a mixture of antimycin A (5 g/mL) and rotenone (10 M) or with KCN (1 mmol/L). In some experiments macrophages were cultured with GW 6471 (48 h/50 M). Afterward, cells were incubated with DCFH-DA (25 mol/L) for 30 min and the final fluorescence intensity was measured by flow cytometry. Results show tendency and are expressed as fluorescence intensity (mean fluorescence ⫾ SD) from three or five separate experiments. † P ⬍ 0.05, significant difference versus corresponding control.
194
E. Stachowska et al. / Nutrition 24 (2008) 187–199
Fig. 3. Effect of a peroxisome proliferator-activated receptor-␣ inhibitor (GW 6471) on the synthesis of reactive oxygen compounds in macrophages cultured with fatty acids. THP-1 macrophages were cultured with fatty acids and with GW 6471 (50 M) for 48 h. The cells were then incubated with 2=,7=-dichlorofluorescein diacetate (25 mol/L) for 30 min and the final fluorescence intensity was measured by flow cytometry. The plots are samples of flow cytometric analysis. The x axis represents the intensity of fluorescence on the logarithmic scale, and the y axis the number of cells. (A) Control bovine serum albumin, (B) trans-10, cis-12 conjugated linoleic acid diene isomer, (C) cis-9, trans-11 conjugated linoleic acid diene isomer, and (D) linoleic acid. ROS, reactive oxygen species.
CLAs reduce PLA2 activity in macrophages from blood It is estimated that ionophore A12387 is a stimulator of activity of cytoplasmic PLA2 [46]. In our experiment, AA released by PLA2 (as a result of cell stimulation by ionophore A12387) was extracted and separated from the remaining total lipids with the use of thin-layer chromatog-
raphy. Then the concentration of AA methyl esters was measured by gas chromatography. In macrophages from peripheral blood, a trend was observed toward lower secretory PLA2 activity in the CLA environment (Fig. 6). A similar phenomenon was noticed in THP-1 macrophages, as described previously [30].
E. Stachowska et al. / Nutrition 24 (2008) 187–199
Fig. 4. Expression of peroxisome proliferator-activated receptor-␣ gene measurements by quantitative real-time polymerase chain reaction in THP-1 and blood macrophages. Macrophages were cultured with fatty acids for 48 h. Data are expressed as the relative gene expression ratio. Mean values ⫾ SDs (n ⫽ 3) done in triplicate are shown. BSA, bovine serum albumin; CLA, conjugated linoleic acid diene; LA, linoleic acid; PMBC, peripheral blood mononuclear cell.
CLAs in differentiating monocytes can probably increase the number of apoptotic cells
195
in cells (Figs. 1 and 2). Activation of the respiratory chain in macrophages cultivated with CLA is also confirmed by the observed increase in ATP concentration in cells cultivated with these fatty acids (Table 3.). The obtained results indicate that CLAs are used in macrophages as substrates for oxidative phosphorylation, leading to ATP synthesis (Table 3). An additional proof of the ROS mitochondrial origin in cells cultivated with CLA is the inhibition of ROS synthesis observed after application of respiratory chain inhibitors (antimycin, rotenone, and KCN; Table 2). Interestingly, complete inhibition of the respiratory chain (by a mixture of rotenone and antimycin A) did not result in complete inhibition of ROS synthesis in macrophages; this indicates the existence of extramitochondrial sources of ROS in cells cultivated with CLA. Extramitochondrial sources of ROS can be coupled with activity of 15 lipo-oxygenase-1 or cyclo-oxygenase-2, which are not inhibited by respiratory
To investigate whether increased ROS synthesis might have activated the apoptotic process, the cytometric measurement of the early apoptotic phase was performed (with the use of an annexin V kit). In macrophages incubated with CLA, a slight reduction in the number of viable cells was observed (P ⱕ 0.005, macrophages from THP-1; P ⱕ 0.012, macrophages from blood). In macrophages originating from THP-1, both CLA isomers reduced the content of viable cells in the culture by about 10% (P ⱕ 0.05) compared with the BSA control. In macrophages from peripheral blood, the isomers reduced this content by about 6% (P ⱕ 0.05; Table 5). Discussion Long-chain non-esterified fatty acids are the factors that increase ROS synthesis in vivo and in vitro in many types of cells [49 –54]. As shown in this study, fatty acids may increase ROS production in parallel with a partial inhibition of electron transport in the respiratory chain, most likely at the level of complexes I and III, and by changing membrane fluidity [55]. In this report we show for the first time that CLA increases ROS synthesis in macrophages. We believe that, in macrophages cultivated with CLA, the mitochondrial respiratory chain is a source of ROSs, and free radicals are produced as a result of the CLA-induced processes of mitochondrial oxidation. Reduced nicotinamide adenosine dinucleotide (created in the -oxidation process and Krebs cycle) must be oxygenated by the respiratory chain enzyme system [56]. Activation of respiratory chain enzymes results in increased synthesis of ROSs in the cell. The proof for this is a high energy potential of mitochondria and the presence of uncoupled polarized mitochondria
Fig. 5. Analysis of protein peroxisome proliferator-activated receptor-␣ in macrophages from THP-1 (A) and blood (B) cultured with fatty acids. The cell lysates were obtained from macrophages cultured with fatty acids. Lysates were stained with antibody against specified antigens as described in MATERIALS AND METHODS. Each panel represents one of three separate experiments with similar results (mean ⫾ SD). On the diagrams presented below of photography content of protein were compared with -actin. Protein was isolated from macrophages cultured with the control bovine serum albumin (lane a), trans-10, cis-12 conjugated linoleic acid diene isomer (lane b), cis-9, trans-11 conjugated linoleic acid diene isomer (lane c), and linoleic acid (lane d).
196
E. Stachowska et al. / Nutrition 24 (2008) 187–199
Table 3 Content of ATP and AEC values for macrophages cultured with BSA or fatty acids* Treatment
ATP (pmol/mg protein)†
ADP (pmol/mg protein)‡
AMP (pmol/mg protein)§
AEC储
BSA Trans-10, cis-12 CLA Cis-9, trans-11 CLA LA
205.6 ⫾ 5 214.8 ⫾ 4.3¶ 210.2 ⫾ 2.9¶ 208.1 ⫾ 7.7
45.1 ⫾ 1.6 43.4 ⫾ 1.4¶ 41.3 ⫾ 0.76¶ 42.8 ⫾ 2.47¶
22.8 ⫾ 0.15 21.1 ⫾ 0.38¶ 20.3 ⫾ 0.22¶ 24.1 ⫾ 1.36
0.83 ⫾ 0.015 0.85 ⫾ 0.014¶ 0.85 ⫾ 0.015¶ 0.83 ⫾ 0.011
AEC, adenylate energy charge; ADP, adenosine diphosphate; AMP, adenosine monophosphate; ATP, adenosine triphosphate; BSA, bovine serum albumin; CLA, conjugated linoleic acid diene; LA, linoleic acid * AEC values were calculated according to the following formula: AEC ⫽ (ATP ⫹ 0.5 ADP)/(ATP ⫹ ADP ⫹ AMP). Values are means ⫾ SDs of eight independent experiments. † P ⬍ 0.003, Friedmann’s analysis of variance (n ⫽ 8). ‡ P ⬍ 0.016, Friedmann’s analysis of variance (n ⫽ 8). § P ⬍ 0.00006, Friedmann’s analysis of variance (n ⫽ 8). 储 P ⬍ 0.0003, Friedmann’s analysis of variance (n ⫽ 8). ¶ P ⬍ 0.05, significant difference versus corresponding control.
chain inhibitors, or decoupling proteins whose expression is controlled by PPAR-␣. In addition, we have shown in this study that ROS synthesis (in cells cultivated with CLA) is controlled by PPAR-␣; the addition of GW 6471(PPAR-␣ inhibitor) significantly decreased ROS synthesis in the cells cultivated with CLA, but not with LA (Table 2). It seems that CLAs, as strong activators of PPAR-␣ [3,33], stimulate the processes of mitochondrial oxidation by PPAR-␣. PPAR-␣ activation is required for the induction of oxidation enzymes in monocytes/macrophages [3,15]. Such a phenomenon was observed in cells cultivated with ␣-LA and ␥-LA, docosahexaenoic acid and eicosehexaenoic acid. The fatty acids, acting as PPAR-␣ ligands, increased the activity and level of mRNA of enzymes associated with oxygenating fatty acids in the mitochondria. Unlike them, LA, which is preferentially used for phospholipid synthesis, did not have any significant effect on
PPAR-␣ activity [15], and seems to be a weaker ligand for this receptor than ␣-LA and ␥-LA [15]. Importantly, no changes in expression of mRNA of PPAR-␣ were noted, although activation of PPAR-␣ mRNA expression was discovered in cancer cells [3] and in aortas of mice receiving CLA-supplemented feed (in vivo studies) [7]. Activation of ROS production in macrophages cultivated with CLA led to an increase in the synthesis of isoprostane 8-epi-PGF2␣, a non-enzymatic (free radical) product of AA oxygenation (Table 4). Enhanced 8-epi-PGF2␣ synthesis may be explained by two concomitant phenomena: inhibition of PLA2 activity and activation of ROS synthesis in cells. AA “retained” in plasmatic membrane phospholipids (caused by reduced PLA2 activity) provokes the use of this acid as a substrate for free radical synthesis of isoprostanes (including 8-epi-PGF2␣) [28]. Another effect of increased ROS synthesis in macrophages cultivated with CLA was activation of the apoptotic process (Table 5) [15]. When ROSs are produced in quantities exceeding the efficiency of
Table 4 Effect of fatty acids on isoprostane 8-epi-PGF2␣ concentration in macrophages* Treatment
BSA Trans-10, cis-12 CLA Cis-9, trans-11 CLA Linoleic acid Negative control
8-epi-PGF2␣ (ng/g protein) Macrophages from THP-1†
Macrophages from blood‡
0.021 ⫾ 0.02 0.047 ⫾ 0.03§ 0.045 ⫾ 0.04§ 0.021 ⫾ 0.01 0.019 ⫾ 0.019
0.052 ⫾ 0.02 0.126 ⫾ 0.08§ 0.112 ⫾ 0.04§ 0.082 ⫾ 0.06 0.050 ⫾ 0.02
BSA, bovine serum albumin; CLA, conjugated linoleic acid diene; 8-epi-PGF2␣, 8-epi-prostaglandin F2␣ * In macrophages the concentration of 8-epi-PGF2␣ was measured by an immunoenzymatic method. Units are shown as mean ⫾ SD from eight or six separate experiments. † P ⬍ 0.02, Friedmann’s analysis of variance (n ⫽ 8). ‡ P ⬍ 0.05, Friedmann’s analysis of variance (n ⫽ 6). § P ⬍ 0.05, significant difference versus corresponding control.
Fig. 6. Effect of fatty acids on sPLA2 activity in macrophages. In homogenates (prepared based on macrophages cultured with fatty acids), the activity of sPLA2 was measured with spectrophotometry. Data are expressed as unites per microgram of protein and showed as mean ⫾ SD from five separate experiments. BSA, bovine serum albumin; CLA, conjugated linoleic acid diene; LA, linoleic acid; sPLA2, secretory phospholipase A2; t10,c12, trans-10, cis-12.
E. Stachowska et al. / Nutrition 24 (2008) 187–199
197
Table 5 Effect of fatty acids on content of living (non-apoptotic) macrophages cultured with CLA* Treatment
BSA Trans-10, cis-12 CLA Cis-9, trans-11 CLA Linoleic acid Negative control
Macrophages from THP-1†
Macrophages from blood‡
Non-apoptotic cells
Percent of control
Non-apoptotic cells
Percent of control
69.05 ⫾ 22.05 63.53 ⫾ 22.72§ 64.94 ⫾ 23.80§ 58.57 ⫾ 38.79 69.77 ⫾ 16.29
100 ⫺8 ⫺6
93.14 ⫾ 22.91 87.43 ⫾ 14.05§ 87.06 ⫾ 12.2§ 90.89 ⫾ 24.97 95.15 ⫾ 24.97
100 ⫺6 ⫺6
BSA, bovine serum albumin; CLA, conjugated linoleic acid diene * Macrophages were cultured with fatty acids for 48 h. Cells were then harvested, stained with annexin V/fluorescein isothiocyanate and propidium iodide, and analyzed by flow cytometry. Data show content (percentage) of non-apoptotic cells and the difference in content of non-apoptotic cells observed between control (BSA) and fatty acids (mean ⫾ SD from five or eight separate experiments). † P ⬍ 0.005, Friedmann’s analysis of variance (n ⫽ 5). ‡ P ⬍ 0.012, Friedmann’s analysis of variance (n ⫽ 8). § P ⬍ 0.05, significant difference versus corresponding control.
cellular antioxidant systems (such as glutathione, antioxidant enzymes), the structures important for correct functioning of the cell are damaged and the apoptotic process is activated [17,19,21]. Thus far, proapoptotic properties have been demonstrated for polyunsaturated long-chain fatty acids (e.g., ␥-linolenic acid and eicosehexaenoic acid) [57], trans-isomers of fatty acids [58], and the trans-10, cis-12 CLA isomer [18]. An enhanced apoptotic process was observed also in aortas of mice receiving CLA-supplemented feed [7]. In the prophylaxis of ischemic heart disease, great importance is attached to the use of products containing substances that limit the intensity of inflammatory processes. They also include fatty acids appropriately selected in the diet [59]. The phenomenon of inhibition of inflammatory reactions after food supplementation with CLAs suggested that these acids might be used in the prophylaxis of ischemic heart disease in humans [10]. However, the intensity of free radical reactions in monocytes cultivated with CLAs, noted in these studies, warrants a critical view of the possibility of including CLAs in prophylaxis of ischemic heart disease. This opinion is also supported by the existing results of tests conducted on endothelial cells where inhibition of nitric oxide and prostaglandin E2 synthesis was observed in cultivation with CLAs (the cis-9, trans-11 CLA isomer and the trans-10, cis-12 CLA isomer). Both substances (nitric oxide and prostaglandin E2) are of key significance for vasodilation [33]. In addition, in in vivo studies in obese men using the trans-10, cis-12 CLA isomer in their diet, a concern-raising phenomenon of an increase in the concentration of 8-isoPGF2␣ isoprostane in urine was observed [8].
[2]
[3]
[4]
[5]
[6]
[7]
[8]
[9] [10]
[11] [12]
[13]
References [14] [1] Chin SF, Storkson JM, Liu W, Albright KJ, Parizia J. Conjugated linoleic acid (9,11- and 10,12-octadecadienoic acid) is produced in
conventional but not germ-free rats fed linoleic acid. J Nutr 1994;124:694 –701. Lin H, Boylston TD, Chang MJ, Luedecke LO, Shultz TD. Survey of the conjugated linoleic acid contents of dairy products. J Dairy Sci 1995;78:2358 – 65. Moya-Camarena SY, Vanden Heuvel JP, Blanchard SG, Leesnitzer LA, Belury MA. Conjugated linoleic acid is a potent naturally occurring ligand and activator of PPARalpha. J Lipid Res 1999;40:1426 –33. Nicolosi RJ, Rogers EJ, Kritchevsky D, Scimeca JA, Huth PJ. Dietary conjugated linoleic acid reduces plasma lipoproteins and early aortic atherosclerosis in hypercholesterolemic hamsters. Artery 1997;22: 266 –77. Houseknecht KL, Vanden Heuvel JP, Moya-Camarena SY, Portocarrero CP, Peck LW, Nickel KP, Belury MA. Dietary conjugated linoleic acid normalizes impaired glucose tolerance in the Zucker diabetic fatty fa/fa rat. Biochem Biophys Res Commun 1998;244: 678 – 82 (erratum Biochem Biophys Res Commun 1998;247:911). Whigham LD, Cook EB, Stahl JL, Saban R, Bjorling DE, Pariza MW, Cook ME. CLA reduces antigen-induced histamine and PGE(2) release from sensitized guinea pig tracheae. Am J Physiol Regul Integr Comp Physiol 2001;280:R908 –12. Toomey S, Roche H, Fitzgerald D, Belton O. Regression of preestablished atherosclerosis in the apoE-/-mouse by conjugated linoleic acid. Biochem Soc Trans 2003;31:1075–9. Riserus U, Basu S, Jovinge S, Fredrikson GN, Arnlov J, Vessby B. Supplementation with conjugated linoleic acid causes isomerdependent oxidative stress and elevated C-reactive protein: a potential link to fatty acid-induced insulin resistance. Circulation 2002;106:1925–9. Sampath H, Ntambi JM. Polyunsaturated fatty acid regulation of gene expression. Nutr Rev 2004;62:333–9. Yu Y, Correll PH, Vanden Heuvel JP. Conjugated linoleic acid decreases production of pro-inflammatory products in macrophages: evidence for a PPAR gamma-dependent mechanism. Biochim Biophys Acta 2002;15:89 –99. Kota BP, Huang TH, Roufogalis BD. An overview on biological mechanisms of PPARs. Pharmacol Res 2005;51:85–94. Ricote M, Li AC, Willson TM, Kelly CJ, Glass CK. The peroxisome proliferator-activated receptor-gamma is a negative regulator of macrophage activation. Nature 1998;391:79 – 82. Tontonoz P, Nagy L, Alvarez JG, Thomazy VA, Evans RM. PPARgamma promotes monocyte/macrophage differentiation and uptake of oxidized LDL. Cell 1998;93:241–52. Lemberger T, Desvergne B, Wahli W. Peroxisome proliferatoractivated receptors: a nuclear receptor signaling pathway in lipid physiology. Annu Rev Cell Dev Biol 1996;12:335– 63.
198
E. Stachowska et al. / Nutrition 24 (2008) 187–199
[15] Nakamura MT, Cheon Y, Li Y, Nara TY. Mechanisms of regulation of gene expression by fatty acids. Lipids 2004;39:1077– 83. [16] Marx N, Sukhova G, Murphy C, Libby P, Plutzky J. Macrophages in human atheroma contain PPARgamma: differentiation-dependent peroxisomal proliferator-activated receptor gamma (PPARgamma) expression and reduction of MMP-9 activity through PPARgamma activation in mononuclear phagocytes in vitro. Am J Pathol 1998; 153:17–23. [17] Barbieri SS, Eligini S, Brambilla M, Tremoli E, Colli S. Reactive oxygen species mediate cyclooxygenase-2 induction during monocyte to macrophage differentiation: critical role of NADPH oxidase. Cardiovasc Res 2003;60:187–97. [18] Evans M, Geigerman C, Cook J, Curtis L, Kuebler B, McIntosh M. Conjugated linoleic acid suppresses triglyceride accumulation and induces apoptosis in 3T3-L1 preadipocytes. Lipids 2000;35:899 –910. [19] Montuschi P, Barnes PJ, Roberts LJ II. Isoprostanes: markers and mediators of oxidative stress. FASEB J 2004;18:1791– 800. [20] Chinetti G, Griglio S, Antonucci M, Torra IP, Delerive P, Majd Z, et al. Activation of proliferator-activated receptors alpha and gamma induces apoptosis of human monocyte-derived macrophages. J Biol Chem 1998;273:25573– 80. [21] Irani K. Oxidant signaling in vascular cell growth, death, and survival: a review of the roles of reactive oxygen species in smooth muscle and endothelial cell mitogenic and apoptotic signaling. Circ Res 2000;87:179 – 83. [22] Geronikaki AA, Gavalas AM. Antioxidants and inflammatory disease: synthetic and natural antioxidants with anti-inflammatory activity. Comb Chem High Throughput Screen 2006;9:425–32. [23] Swindle EJ, Metcalfe DD, Coleman JW. Rodent and human mast cells produce functionally significant intracellular reactive oxygen species but not nitric oxide. J Biol Chem 2004;279:48751–9. [24] Cadenas E. Mitochondrial free radical production and cell signaling. Mol Aspects Med 2004;25:17–26. [25] Fialkow L, Wang Y, Downey GP. Reactive oxygen and nitrogen species as signaling molecules regulating neutrophil function. Free Radic Biol Med 2007;42:153– 64. [26] Morrow JD, Roberts LJ. The isoprostanes: unique bioactive products of lipid peroxidation. Prog Lipid Res 1997;36:1–21. [27] Dillon SA, Lowe GM, Billington D, Rahman K. Dietary supplementation with aged garlic extract reduces plasma and urine concentrations of 8-iso-prostaglandin F(2 alpha) in smoking and nonsmoking men and women. J Nutr 2002;132:168 –71. [28] Pratico D, FitzGerald GA. Generation of 8-epiprostaglandin F2alpha by human monocytes. Discriminate production by reactive oxygen species and prostaglandin endoperoxide synthase-2. J Biol Chem 1996;271:8919 –24. [29] Stachowska E, Dziedziejko V, Safranow K, Jakubowska K, Olszewska M, Machalinski B, Chlubek D. Effect of conjugated linoleic acids on the activity and mRNA expression of 5- and 15lipoxygenases in human macrophages. J Agric Food Chem 2007;55: 5335– 42. [30] Stachowska E, Dziedziejko V, Safranow K, Gutowska I, Adler G, Ciechanowicz A, et al. Inhibition of phospholipase A(2) activity by conjugated linoleic acids in human macrophages. Eur J Nutr 2007; 46:28 –33. [31] Fritsche J, Steinhart H. Analysis, occurrence and physiological properties of trans fatty acids (TFA)with particular emphasis on conjugated linoleic acid isomers (CLA). Lipid 1998;100:190 –121. [32] Choi Y, Park Y, Pariza MW, Ntambi JM. Regulation of stearoyl-CoA desaturase activity by the trans-10,cis-12 isomer of conjugated linoleic acid in HepG2 cells. Biochem Biophys Res Commun 2001;284: 689 –93. [33] Eder K, Schleser S, Becker K, Korting R. Conjugated linoleic acids lower the release of eicosanoids and nitric oxide from human aortic endothelial cells. J Nutr 2003;133:4083–9.
[34] World Medical Association Declaration of Helsinki. Recommendations guiding physicians in biomedical research involving human subjects. Cardiovasc Res 1997;35:2–3. [35] Eligini S, Brambilla M, Banfi C, Camera M, Sironi L, Barbieri SS, et al. Oxidized phospholipids inhibit cyclooxygenase-2 in human macrophages via nuclear factor-kappaB/IkappaB- and ERK2-dependent mechanisms. Cardiovasc Res 2002;55:406 –9. [36] Fu Y, Luo N, Lopes-Virella MF, Garvey WT. The adipocyte lipid binding protein (ALBP/aP2) gene facilitates foam cell formation in human THP-1 macrophages. Atherosclerosis 2002;165:259 – 69. [37] Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 1976;72:248 –54. [38] Umino T, Skold CM, Pirruccello SJ, Spurzem JR, Rennard SI. Twocolour flow-cytometric analysis of pulmonary alveolar macrophages from smokers. Eur Respir J 1999;13:894 –9. [39] Bartosz G. Use of spectroscopic probes for detection of reactive oxygen species. Clin Chim Acta 2006;368:53–76. [40] Aam BB, Fonnum F. ROS scavenging effects of organic extract of diesel exhaust particles on human neutrophil granulocytes and rat alveolar macrophages. Toxicology 2007;230:207–18. [41] Labro MT, Ollivier V, Babin-Chevaye C. Interaction of rifalazil with oxidant-generating systems of human polymorphonuclear neutrophils. Antimicrob Agents Chemother 2005;49:5018 –23. [42] Bowser DN, Minamikawa T, Nagley P, Williams DA. Role of mitochondria in calcium regulation of spontaneously contracting cardiac muscle cells. Biophys J 1998;75:2004 –14. [43] Elmore SP, Nishimura Y, Qian T, Herman B, Lemasters JJ. Discrimination of depolarized from polarized mitochondria by confocal fluorescence resonance energy transfer. Arch Biochem Biophys 2004; 422:145–52. [44] Atkinson DE, Walton GM. Adenosine triphosphate conservation in metabolic regulation. Rat liver citrate cleavage enzyme. J Biol Chem 1967;242:3239 – 41. [45] Folch J, Lees M, Stanley-Sloane V. A simple method for the isolation and purification of total lipids from animal tissues. J Biol Chem 1957;226:497–9. [46] Winkler JD, Bolognese BJ, Roshak AK, Sung CM, Marshall LA. Evidence that 85 kDa phospholipase A2 is not linked to CoAindependent transacylase-mediated production of platelet-activating factor in human monocytes. Biochim Biophys Acta 1997;1346: 173– 84. [47] Dembinska-Kiec A, Polus A, Kiec-Wilk B, Grzybowska J, Mikolajczyk M, Hartwich J, et al. Proangiogenic activity of beta-carotene is coupled with the activation of endothelial cell chemotaxis. Biochim Biophys Acta 2005;1740:222–39. [48] Pfaffl MW. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res 2001;29:e45. [49] Aitken RJ, Wingate JK, De Iuliis GN, Koppers AJ, McLaughlin EA. Cis-unsaturated fatty acids stimulate reactive oxygen species generation and lipid peroxidation in human spermatozoa. J Clin Endocrinol Metab 2006;91:4154 – 63. [50] Liu H, Zhang D, Zhao B, Zhao J. Superoxide anion, the main species of ROS in the development of ARDS induced by oleic acid. Free Radic Res 2004;38:1281–7. [51] Sanders LM, Henderson CE, Hong MY, Barhoumi R, Burghardt RC, Wang N, et al. An increase in reactive oxygen species by dietary fish oil coupled with the attenuation of antioxidant defenses by dietary pectin enhances rat colonocyte apoptosis. J Nutr 2004;134:3233– 8. [52] Cury-Boaventura MF, Curi R. Regulation of reactive oxygen species (ROS) production by C18 fatty acids in Jurkat and Raji cells. Clin Sci (Lond) 2005;108:245–53. [53] Tripathy D, Mohanty P, Dhindsa S, Syed T, Ghanim H, Aljada A, Dandona P. Elevation of free fatty acids induces inflammation and
E. Stachowska et al. / Nutrition 24 (2008) 187–199 impairs vascular reactivity in healthy subjects. Diabetes 2003; 52:2882–7. [54] Hatanaka E, Levada-Pires AC, Pithon-Curi TC, Curi R. Systematic study on ROS production induced by oleic, linoleic, and gammalinolenic acids in human and rat neutrophils. Free Radic Biol Med 2006;41:1124 –32. [55] Schonfeld P, Wojtczak L. Fatty acids decrease mitochondrial generation of reactive oxygen species at the reverse electron transport but increase it at the forward transport. Biochim Biophys Acta 2007;1767:1032– 40. [56] Murray RD, Granner DK, Mayes PA, Rodwell VW. Biochemistry. 24th ed. East Norwalk, CT: Appleton & Lange; 2003.
199
[57] Phoon MC, Desbordes C, Howe J, Chow VT. Linoleic and linolelaidic acids differentially influence proliferation and apoptosis of MOLT-4 leukaemia cells. Cell Biol Int 2001;25:777– 84. [58] Zapolska-Downar D, Zapolska-Downar A, Bukawska H, Gałka H, Naruszewicz M. Ibuprofen protects low density lipoproteins against oxidative modification. Life Sci 1999;65:2289 –303. [59] Expert Panel on Detection and Treatment of High Blood Cholesterol in Adults. Summary of the second report of the National Cholesterol Education Program (NCEP). Expert panel on detection, evaluation, and treatment of high blood cholesterol in adults (Adult Treatment Panel II). JAMA 1993;16:3015–23.