Hostname: page-component-8448b6f56d-m8qmq Total loading time: 0 Render date: 2024-04-23T20:34:14.127Z Has data issue: false hasContentIssue false

Dietary indicaxanthin from cactus pear (Opuntia ficus-indica L. Mill) fruit prevents eryptosis induced by oxysterols in a hypercholesterolaemia-relevant proportion and adhesion of human erythrocytes to endothelial cell layers

Published online by Cambridge University Press:  14 July 2015

Luisa Tesoriere
Affiliation:
Department of Biological Chemical and Pharmaceutical Science and Technologies (STEBICEF), Università di Palermo, Via M. Cipolla 74, 90123Palermo, Italy
Alessandro Attanzio
Affiliation:
Department of Biological Chemical and Pharmaceutical Science and Technologies (STEBICEF), Università di Palermo, Via M. Cipolla 74, 90123Palermo, Italy
Mario Allegra
Affiliation:
Department of Biological Chemical and Pharmaceutical Science and Technologies (STEBICEF), Università di Palermo, Via M. Cipolla 74, 90123Palermo, Italy
Maria A. Livrea*
Affiliation:
Department of Biological Chemical and Pharmaceutical Science and Technologies (STEBICEF), Università di Palermo, Via M. Cipolla 74, 90123Palermo, Italy
*
*Corresponding author: M. A. Livrea, email maria.livrea@unipa.it
Rights & Permissions [Opens in a new window]

Abstract

Toxic oxysterols in a hypercholesterolaemia-relevant proportion cause suicidal death of human erythrocytes or eryptosis. This process proceeds through early production of reactive oxygen species (ROS), release of prostaglandin (PGE2) and opening of PGE2-dependent Ca channels, membrane phosphatidylserine (PS) externalisation, and cell shrinkage. The present study was the first to reveal that a bioavailable phytochemical, indicaxanthin (Ind) from cactus pear fruit, in a concentration range (1·0–5·0 μM) consistent with its plasma level after a fruit meal, prevents PS externalisation and cell shrinkage in a dose-dependent manner when incubated with isolated healthy human erythrocytes exposed to an oxysterol mixture for 48 h. Dietary Ind inhibited ROS production, glutathione (GSH) depletion, PGE2 release and Ca2+ entry. Ind alone did not modify the erythrocyte redox environment or affect other parameters. Ex vivo spiking of normal human blood with the oxysterol mixture for 48 h induced eryptosis, resulting in the production of ROS and decreased levels of GSH, which was prevented by concurrent exposure to 5 μm-Ind. The adherence of eryptotic erythrocytes to the endothelium causes vascular tissue injury. Erythrocytes isolated from blood incubated with the oxysterol mixture plus 5 μm-Ind did not adhere to endothelial cell monolayers. Eryptotic erythrocytes may contribute to thrombotic complications in hypercholesterolaemia. Our findings suggest the positive effects of diets containing Ind on erythrocytes in hypercholesterolaemic subjects.

Type
Full Papers
Copyright
Copyright © The Authors 2015 

A physiological rate of programmed death, or eryptosis( Reference Lang, Gulbins and Lerche 1 ), of erythrocytes allows clearance of aged cells; however, an increased number of circulating eryptotic erythrocytes, as observed in various diseases( Reference Bonomini, Sirolli and Settefrati 2 Reference Calderon-Salinas, Munoz-Reyes and Guerrero-Romero 5 ), is dangerous and leads to complications. Because of phosphatidylserine (PS) exposed at the erythrocyte surface, eryptotic cells may activate coagulant enzymes( Reference Chung, Bae and Lim 6 ), causing thrombosis and thrombo-occlusive disease( Reference Wood, Gibson and Tait 3 , Reference Chung, Bae and Lim 6 Reference Pandolfi, Di Pietro and Sirolli 9 ), and adhere to endothelial cells( Reference Closse, Dachary-Prigent and Boisseau 8 , Reference Borst, Abed and Alesutan 10 ), promoting vascular damage. Our recent studies have for the first time demonstrated that a mixture of oxysterols, known to be elevated in the serum of hypercholesterolaemic subjects( Reference Addis, Emanuel and Bergmann 11 Reference Chang, Abdalla and Sevanian 13 ), exerts a remarkable eryptotic activity compared with healthy erythrocytes( Reference Tesoriere, Attanzio and Allegra 14 ), an event that enhances the pathogenic potential of these cholesterol derivatives in the initiation and promotion of atherogenic processes( Reference Poli, Biasi and Leonarduzzi 15 , Reference Brown and Jessup 16 ).

Evidence on the impact of dietary vegetable intake on the prevention of chronic diseases, including cancer, neurodegenerative disorders and atherosclerosis, has emerged from various studies( Reference Slavin and Lloyd 17 ). Polyphenol phytochemicals have long been considered as protective agents; however, recent attention has been focused on betalains( Reference Stintzing and Carle 18 ), a group of pigments sharing the common structure of betalamic acid. Indicaxanthin (Ind; Fig. 1), the characteristic yellow pigment of cactus pear (Opuntia ficus-indica) fruit, is the immonium derivative of proline with betalamic acid( Reference Piattelli and Conn 19 ). Ind is a reducing and amphipathic molecule, interacts with and partitions in membranes, enters various cells, including erythrocytes, and counteracts oxidative damage induced by various agents( Reference Butera, Tesoriere and Di Gaudio 20 Reference Tesoriere, Allegra and Butera 26 ). Moreover, it has been shown to modulate specific redox-driven signalling pathways involved in the inflammatory response in cultured endothelial and intestinal cells, and interfere with molecular mechanisms involved in the apoptotic activity of 7-ketocholesterol (7-KC) in a human monocyte/macrophage cell line( Reference Gentile, Tesoriere and Allegra 27 29 ). Dietary Ind that may be absorbed by passive diffusion through the intestinal epithelium has been found to be unmodified in human blood and exhibits high bioavailability, reaching plasma concentrations of several micromolars after a cactus pear fruit meal( Reference Tesoriere, Allegra and Butera 30 Reference Tesoriere, Gentile and Angileri 32 ). Therefore, the aim of the present study was to investigate whether dietary Ind affected oxysterol-induced eryptosis in human erythrocytes. For this purpose, a mixture of oxysterols in hypercholesterolaemia-relevant proportions( Reference Addis, Emanuel and Bergmann 11 Reference Chang, Abdalla and Sevanian 13 ) and Ind concentrations consistent with the plasma levels achieved after a dietary intake of cactus pear fruits( Reference Tesoriere, Gentile and Angileri 32 ) were considered. The study was performed with either isolated erythrocytes or after ex vivo spiking of whole blood with oxysterols in the presence or absence of Ind. Finally, the effect of dietary Ind on the adherence of oxysterol-treated erythrocytes to endothelial cell monolayers (human umbilical vein endothelial cells; HUVEC) was explored.

Fig. 1 Molecular structure of indicaxanthin.

Materials and methods

7-KC, cholestan-3β,5α,6β-triol (TRIOL), 5α,6α-epoxycholesterol (α-epox), 5β,6β-epoxy-cholesterol (β-epox), 7α-hydroxy-cholesterol (7α-OH) and 7β-hydroxy-cholesterol (7β-OH) were obtained from Avanti Polar Lipids, Inc. All other reagents and chemicals were obtained from Sigma Chemical Co., unless otherwise indicated.

Preparation of indicaxanthin

Ind was isolated from cactus pear (Opuntia ficus-indica L. Mill) fruit pulp (yellow cultivar) by methanol extraction, purified and quantified as reported previously( 29 ).

Cells and incubation conditions

Erythrocytes collected from the blood of healthy volunteers (n 5; age 25–68 years; normal BMI range), with informed consent, were isolated by centrifugation (2000  g , 4°C, 20 min) on a Ficoll gradient (Biochrom KG). Erythrocytes (0·4 % haematocrit) were incubated at 37°C, 5 % CO2 and 90 % humidity in Ringer solution containing 125 mm-NaCl, 5 mm-KCl, 1 mm-MgSO4, 32 mm-N-2-hydroxyethylpiperazine-N-2-ethanesulfonic acid (HEPES)/NaOH, 5 mm-glucose, 1 mm-CaCl2 (pH 7·4). The cells were pre-incubated for 1 h in the absence or presence of Ind before adding a mixture of oxysterols, followed by incubation for 48 h. The mixture of oxysterols at final concentrations of 7 μm-7-KC, 2 μm-TRIOL, 4 μm-α-epox, 1 μm-7α-OH, 2 μm-7β-OH and 4 μm-β-epox (20 μmof total oxysterols) was added to the cells using tetrahydrofuran at a 0·1 % (v/v) final concentration. Preliminary experiments showed that under these conditions, tetrahydrofuran did not have any effect on erythrocytes; therefore, erythrocytes incubated with the solvent were used as the control.

Measurement of phosphatidylserine externalisation and forward scatter

Erythrocytes washed in Ringer solution were adjusted at 1·0 × 106 cells/ml with the combining buffer according to the manufacturer's protocol (eBioscience, Inc.). Cell suspension (100 μl) was added to a new tube, and incubated with 5 μl Annexin V-FITC at room temperature for 15 min in the dark. Fluorescence-activated cell sorting (FACS) analysis was carried out using an Epics XL™ flow cytometer with Expo32 software (Beckman Coulter). Cells were analysed by forward scatter, and annexin V fluorescence intensity was measured in the fluorescence channel FL-1 with an excitation wavelength of 488 nm and an emission wavelength of 530 nm. At least 1 × 104 cells were examined.

Measurement of cytosolic calcium

Intracellular Ca2+ concentration was measured by the cell-permeable probe fluo-3 AM, whose fluorescence directly represents the ion concentration, as reported previously( Reference Tesoriere, Attanzio and Allegra 14 ). Fluorescent intensities were analysed by FACS analysis. Ca2+-dependent fluorescence intensity was measured by FACS analysis in the fluorescence channel FL-1 with an excitation wavelength of 488 nm and an emission wavelength of 530 nm. At least 1 × 104 cells were examined.

Measurement of PGE2

After incubation, as indicated above, PGE2 released (in pg/ml) from erythrocytes (1 × 109 cells/ml) was quantified using a Prostaglandin E2 Enzyme Immunoassay Kit (Cayman Chemical Corporation, Inc.) in accordance with the manufacturer's protocol. Briefly, after incubation, the cells were pelleted by centrifugation at 4°C, for 5 min, at 450  g . Samples of the supernatant were diluted at 1:2·5 with assay buffer. Then, a 100 μl sample, a 50 μl alkaline phosphatase PGE2 conjugate and a 50 μl monoclonal anti-PGE2 EIA antibody were applied to goat anti-mouse IgG microtitre plates, and incubated at room temperature for 2 h. After washing, 200 μl of p-nitrophenyl phosphate substrate solution were added and incubated at room temperature for 45 min. Finally, optical density at 405 nm was measured on a microplate reader. PGE2 concentrations in the samples were calculated from a PGE2 standard curve (25–5000 pg/ml), which was run in parallel.

Measurement of intracellular reactive oxygen species

Reactive oxygen species (ROS) production was monitored by measuring fluorescence changes arising from intracellular oxidation of dichloro-dihydro-fluorescein diacetate (DCFDA, 10 μm). A final concentration was added to the cell medium 30 min before the end of the treatment. Erythrocytes were collected by centrifugation (5 min, 2000 rpm, 4°C), washed, suspended in PBS and subjected to FACS analysis. At least 1 × 104 cells were examined.

Measurement of glutathione

Butylated hydroxytoluene (40 mm, 25 μl) was added to 3·0 ml of incubation mixture to stop oxidative reactions. Then, the cells were precipitated and lysed with 0·5 ml H2O, and proteins were precipitated by the addition of 0·5 ml of metaphosphoric acid solution (1·67 g metaphosphoric acid, 0·20 g EDTA and 30 g NaCl in 100 ml H2O). After centrifugation (3000  g , 10 min), glutathione (GSH) was determined by titration with 5,5'-dithiobis-(2-nitrobenzoic acid) and spectrophotometric quantification at 412 nm using a molar extinction coefficient of 13 600 cm− 1, as reported previously( Reference Hu and Packer 33 ).

Partition of indicaxanthin in erythrocytes

Erythrocytes (2 ml of incubation mixture) were extracted with three volumes of chloroform–methanol (2:1, v/v). The methanol phase was dried under N2, resuspended in 1 % acetic acid in water, and analysed by HPLC using a Gilson modular liquid chromatography system (Gilson, Inc.) equipped with M 302 and 305 pumps and an injector (Model 77-25; Rheodyne) with a 20 μl injector loop and an M 802 manometric module. The chromatographic column was a Varian Microsorb C-18 column (250 × 4·6 mm inner diameter, 5 mm; Varian, Inc.) with a guard cartridge (5 × 3·9 mm; Varian, Inc.). Elution was performed with a 10 min linear gradient of solvent A (1 % acetic acid in water) to 10 % solvent B (1 % acetic acid in acetonitrile), at a flow of 1·5 ml/min. Detection was performed at 482 nm using an M 118 UV–Vis detector with the Gilson 712 HPLC system controller software. Sensitivity was 0·05 % absorbance units. The retention time of Ind was 4·2 min. Ind concentration was quantified by reference to standard curves constructed with 1–50 ng of the purified compound, and by relating the amount of the compound under analysis to the peak area.

Ex vivo spiking of blood with oxysterols and/or indicaxanthin

Whole blood collected from healthy volunteers (n 5, 39–45 % haematocrit) after an overnight fast was individually incubated with the oxysterol mixture either in the absence or presence of 5 μm-Ind (37°C, 5 % CO2, 95 % humidity, 48 h). Erythrocytes were isolated by centrifugation on a Ficoll gradient, washed and resuspended in PBS to obtain a haematocrit of 0·4 %. Cytofluorometric determination of PS exposure and ROS production were as described previously. The experiments were replicated two times. The present study was conducted according to the guidelines laid down in the Declaration of Helsinki.

Endothelial cell culture and gravity adherence assays

HUVEC (American Type Culture Collection) were only used for up to eight additional passages. Cells were grown in 75 mm2 flasks with endothelial cell basal medium MV2 (PromoCell) containing 100 pg/ml of heparin, 10 % fetal bovine serum (GIBCO), penicillin/streptomycin (120 U/ml (72 μg/ml) and 120 pg/ml) and 30 pg/ml of endothelial cell growth supplement. At 85 % confluence, the cells were subcultured in ninety-six-well flat-bottom Costar plates, and used for adherence assays within 24 h. The adherence of erythrocytes to HUVEC was measured according to the method of Sugihara et al. ( Reference Sugihara, Sugihara and Mohandas 34 ), with some modification. Briefly, erythrocytes in Ringer solution (300 μl, 0·4 % haematocrit) were layered on confluent HUVEC that were previously washed with the same solution, and incubated at 37°C for 30 min. The plate was sealed with a packing tape, and inverted at 37°C for an additional 30 min. While still inverted, the occlusive sheet was removed, bulk fluid was taken out, and the sides and rim of each well were quickly swabbed to eliminate any fluid without disturbing the endothelial layer. SDS (0·5 %, 200 μl) was added and Hb in each well was determined at 405 nm, after incubation for 1 h at room temperature with stirring. Adherence was expressed as the percentage of erythrocytes adhering to HUVEC, calculated from the ratio of Hb absorbance after incubation to Hb absorbance of erythrocytes layered on HUVEC (average Hb absorbance of replicate wells), with correction for the absorbance of wells containing only HUVEC.

Statistical analysis

Data are presented as means and standard deviations, with independent experiments being carried out in triplicate. Statistical comparisons were made by the one-way ANOVA test followed by Bonferroni's correction for multiple comparisons using Instat-3 Statistical Software (GraphPad Software, Inc.). In all cases, significance was accepted if the null hypothesis was rejected at the P< 0·05 level.

Results

Indicaxanthin and oxysterol-induced eryptosis in isolated cells

Eryptosis was stimulated by a 48 h treatment of isolated erythrocytes with a mixture of oxysterols mimicking their plasma combination in hypercholesterolaemia (final concentration 20 μm)( Reference Addis, Emanuel and Bergmann 11 Reference Chang, Abdalla and Sevanian 13 ). Hallmarks of eryptosis, including erythrocyte membrane scrambling with PS exposure and cell shrinkage, were investigated by cytofluorometry. Pre-incubation of erythrocytes with 1 to 5 μm-Ind decreased the oxysterol-induced increase in annexin V-binding fluorescent cells (42·9 % v. 2·0 % of the control) in a dose-dependent manner, with total protection conferred at the concentration of 5 μm. This concentration did not show any appreciable effect in the absence of oxysterols (Fig. 2(a) and (b)). Dietary Ind (5 μm) also prevented the loss of cell volume, estimated from forward scatter (Fig. 2(c) and (d)). Eryptosis is associated with Ca2+ entry and cytosolic Ca activity( Reference Lang, Gulbins and Lerche 1 ). Cytosolic Ca2+ was evaluated by a fluorescent probe. Dietary Ind dose-dependently reduced the oxysterol-induced increase in Fluo3-AM fluorescence intensity, returning to control values at the concentration of 5 μm (Fig. 3(a)). Formation of PGE2 controls the opening of Ca channels after treatment of erythrocytes with oxysterols( Reference Lang, Kempe and Myssina 35 ). Immunodetection measurements showed that Ind dose-dependently reduced the release of PGE2 in the medium (Fig. 3(b)). Ind alone did not modify cytosolic Ca levels or influence the release of PGE2 (Fig. 3(a) and (b)). Oxysterol-induced eryptosis is controlled by intracellular ROS generation and accompanied by the depletion of GSH( Reference Tesoriere, Attanzio and Allegra 14 ). Cytofluorometric analysis with DCFDA showed that 1 to 5 μm-Ind inhibited ROS production in a dose-dependent manner (Fig. 4(a) and (b)). The depletion of GSH was also prevented by Ind in a dose-dependent manner (Fig. 4(c)). Ind alone did not have any effect on ROS production or modify the redox tone of the cells at the concentration of 5 μm (Fig. 4(a) and (c)).

Fig. 2 Inhibition of oxysterol-induced eryptosis by indicaxanthin (Ind). (a) Percentage of phosphatidylserine-exposing erythrocytes or (c) their forward scatter, after 48 h incubation with the oxysterol mixture in the presence of 0–5 μm-Ind. Histograms of (b) annexin V-binding cells or (d) their forward scatter in a typical experiment in the absence () or presence () of 5 μm-Ind. Cells incubated with vehicle were used as the control. Values are means of six independent experiments carried out in triplicate, with their standard deviations represented by vertical bars. *Each value was significantly different from that of the control (P< 0·0001; ANOVA with Bonferroni's test). † Each value was significantly different from that of the oxysterol mixture in the absence of indicaxanthin (P< 0·05; ANOVA with Bonferroni's test).

Fig. 3 Inhibition of oxysterol-induced (a) Ca2+ entry and (b) PGE2 release by indicaxanthin (Ind) in human erythrocytes. Cells were incubated in the absence or presence of 0–5 μm-Ind for 48 h. Cells incubated with vehicle were used as the control. Values are means of six independent experiments carried out in triplicate, with their standard deviations represented by vertical bars. * Each value was significantly different from that of the control (P< 0·0001; ANOVA with Bonferroni's test). † Each value was significantly different from that of the oxysterol mixture in the absence of indicaxanthin (P< 0·05; ANOVA with Bonferroni's test).

Fig. 4 Inhibition of oxysterol-induced (a, b) reactive oxygen species (ROS) production and (c) glutathione (GSH) depletion by indicaxanthin (Ind) in human erythrocytes. (a) Dichloro-dihydro-fluorescein diacetate (DCFDA)-associated mean fluorescence intensity (MFI) and (c) GSH levels, after 48 h incubation of erythrocytes with the oxysterol mixture in the absence or presence of 0–5 μm-Ind. (b) Histogram of DCFDA-stained cells in a typical experiment without () or with () 5 μm-Ind. Cells incubated with vehicle were used as the control. Values are means of six independent experiments carried out in triplicate, with their standard deviations represented by vertical bars. * Each value was significantly different from that of the control (P< 0·0001; ANOVA with Bonferroni's test). † Each value was significantly different from that of the oxysterol mixture in the absence of indicaxanthin (P< 0·05; ANOVA with Bonferroni's test).

Previous studies have indicated that ROS generation controlled the upstream production of PGE2, followed by the opening of PGE2-dependent Ca channels and Ca2+ entry, in the oxysterol-induced eryptotic signalling axis( Reference Tesoriere, Attanzio and Allegra 14 ). The intracellular ROS level was monitored from 60 min to 48 h in erythrocytes incubated with a mixture of oxysterols, either in the absence or presence of 5 μm-Ind. ROS were rapidly generated within 60 min; thereafter, a steady-state level was maintained until the end of the observation at 48 h, in the absence of Ind. ROS production appeared to be completely prevented during the entire observation time following 1 h pre-incubation with Ind (Fig. 5). Separate experiments were carried out to investigate the incorporation and fate of Ind in oxysterol-treated erythrocytes. Ind extracted from erythrocytes after 1 h pre-incubation amounted to 0·5 nmol/107 cells, and a progressive decline in the level was observed 1 h after the addition of oxysterols, with disappearance being observed at 12 h (Fig. 5). Taken together, the data suggest an early activity of Ind against ROS production, resulting in a long-lasting protection of the cell redox environment, even after Ind consumption.

Fig. 5 Time-dependent reactive oxygen species (ROS) production in erythrocytes treated with the oxysterol mixture in the absence () or presence () of 5 μm-indicaxanthin (Ind) and incorporation of the phytochemical in the cells. Dichloro-dihydro-fluorescein diacetate-associated mean fluorescence intensity (MFI). Values are means of six independent experiments carried out in triplicate, with their standard deviations represented by vertical bars. , ROS production (MFI); , Ind (nmol/107 cells).

Indicaxanthin and oxysterol-induced eryptosis ex vivo

Ex vivo spiking of blood with the oxysterol mixture was carried out to simulate a pathophysiological condition of hypercholesterolaemia. Whole blood from healthy human subjects was incubated with a mixture of oxysterols, either in the absence or presence of 5 μm-Ind, and then erythrocytes were isolated and subjected to annexin V-binding and DCFDA-associated fluorescence measurements. With respect to the cells isolated from homologous blood incubated in the absence of Ind, oxysterols induced an increase in PS exposure (Fig. 6(a)) and intracellular ROS production (Fig. 6(b)), with the depletion of cellular GSH content (Fig. 6(c)). All these effects were completely prevented by Ind (Fig. 6(a)–(c)). PS-exposing erythrocytes have been shown to adhere to the cells of the vascular wall, playing a role in vaso-occlusive events( Reference Chung, Bae and Lim 6 , Reference Andrews and Low 7 ). Additional experiments were performed to investigate the activity of Ind against the adherence of oxysterol-treated erythrocytes to endothelial cells. HUVEC monolayers were exposed to erythrocytes, isolated after the ex vivo spiking of blood with a mixture of oxysterols in the absence or presence of 5 μm-Ind for 48 h. After gentle washing of wells and treatment with SDS, the adherence of erythrocytes was evaluated from the ratio of Hb of lysed HUVEC to Hb of erythrocytes applied to the wells. Following incubation with a mixture of oxysterols, Ind completely prevented the adhesion of erythrocytes to HUVEC isolated after the ex vivo spiking of blood (Fig. 7). Ex vivo spiking with 5 μm-Ind alone did not affect the adherence of erythrocytes (Fig. 7).

Fig. 6 (a) Phosphatidylserine exposure, (b) reactive oxygen species (ROS) production and (c) glutathione (GSH) level in erythrocytes isolated after a 48 h ex vivo spiking of fresh human blood with the oxysterol mixture in the absence or presence of 5 μm-indicaxanthin (Ind). Cells isolated from homologous blood incubated in the presence of vehicle were used as the control. Values are means of independent experiments carried out in duplicate with blood collected from five volunteers (each contributing two samples (n 10)), with their standard deviations represented by vertical bars. * Each value was significantly different from that of the control (P< 0·0001; ANOVA with Bonferroni's test).

Fig. 7 Adherence of erythrocytes to human umbilical vein endothelial cell monolayers isolated after a 48 h ex vivo spiking of fresh human blood with the oxysterol mixture in the absence or presence of 5 μm-indicaxanthin (Ind). Cells isolated from homologous blood incubated in the presence of vehicle were used as the control. Values are means of independent experiments carried out in duplicate with blood collected from five volunteers (each contributing two samples (n 10)), with their standard deviations represented by vertical bars. * Each value was significantly different from that of the control (P< 0·0001; ANOVA with Bonferroni's test).

Discussion

Oxysterols of pathophysiological interest have recently been revealed as powerful inducers of suicidal erythrocyte death or eryptosis( Reference Tesoriere, Attanzio and Allegra 14 ). The present study investigating for the first time the anti-eryptotic activity of a dietary phytochemical shows that Ind from cactus pear (Opuntia ficus-indica) fruit, in a concentration range including the amounts measured in human plasma after a fruit meal( Reference Tesoriere, Allegra and Butera 30 ), prevents eryptosis of healthy human erythrocytes exposed to a mixture of oxysterols mimicking the plasma composition of subjects with hypercholesterolaemia( Reference Addis, Emanuel and Bergmann 11 Reference Chang, Abdalla and Sevanian 13 ). The effect was observed in either isolated cells or after the ex vivo spiking of whole blood.

Mechanistic aspects of eryptosis promoted by various oxysterols, including the mixture used in the present study, have for the first time been investigated in our laboratory( Reference Tesoriere, Attanzio and Allegra 14 ). The process that appeared entirely controlled by early ROS generation was totally abrogated by antioxidants and substantially independent of Ca ions. A number of studies have characterised Ind as a radical scavenger( Reference Butera, Tesoriere and Di Gaudio 20 , Reference Tesoriere, Allegra and Butera 22 ), and antioxidant activity and protective effects of the pigment against chemically induced oxidative stress have been shown in biological environments( Reference Tesoriere, Allegra and Butera 26 , Reference Tesoriere, Butera and D'Arpa 36 ). It has also been observed that erythrocytes of healthy human subjects incorporate Ind after consumption of a cactus pear fruit meal, which results in the resistance of erythrocytes to an oxidative stress induced ex vivo ( Reference Tesoriere, Butera and Allegra 25 ). Given the pivotal role of ROS and oxidative stress in oxysterol-induced eryptosis( Reference Tesoriere, Attanzio and Allegra 14 ), the fact that Ind prevents ROS production and preserves the cell redox balance could easily explain its anti-eryptotic effect. Our kinetic study monitoring the level of ROS generated in oxysterol-treated erythrocytes and the level of incorporated Ind, between 1 and 48 h, suggests the activity of Ind occurring at a very early step, preventing the incidence of the eryptotic signal and downstream events. Apparently, this is enough to definitely inhibit the eryptotic cascade even after consumption of Ind. ROS-controlled PGE2 formation followed by the opening of plasma membrane Ca channels are the characteristic features of oxysterol-induced eryptosis( Reference Tesoriere, Attanzio and Allegra 14 ). Indeed, we observed that while preventing ROS production, Ind also prevented PGE2 release and Ca2+ entry, which supports its primary activity at the level of ROS scavenging and/or production. Our experiments were designed to mimic a human pathophysiological condition of hypercholesterolaemia. In this context, eventual activity of Ind in the absence of Ca was not investigated.

The molecular mechanism underlying the activity of Ind could not be assessed here; however, previous studies and present findings allow some speculation. Because of their chemico-physical characteristics, oxysterols and Ind may share the property of perturbing membrane arrangement( Reference Turco Liveri, Sciascia and Lombardo 23 , Reference Turco Liveri, Sciascia and Allegra 24 , Reference Olkkonen and Hynynen 37 , Reference Massey and Pownall 38 ) and possibly interfering with membrane lipids and functional proteins. The anti-apoptotic activity of Ind recently observed in human macrophages exposed to the highly toxic 7-KC( 29 ) has been accounted for by interference with early 7-KC-dependent activation of NADPH oxidase 4, which preceded Ca2+ entry and cytosolic Ca activity. The presence of NADPH oxidase at the erythrocyte membrane( Reference George, Pushkaran and Kostantinidis 39 ) and the observed inhibition of oxysterol-induced ROS production may suggest the occurrence of Ind activity at this level, which deserves to be investigated. Moreover, the activity of Ind as an oxyradical scavenger( Reference Butera, Tesoriere and Di Gaudio 20 , Reference Tesoriere, Allegra and Butera 22 , Reference Tesoriere, Allegra and Butera 26 , Reference Tesoriere, Butera and D'Arpa 36 ) cannot be ruled out.

According to current knowledge on the redox regulation of signalling pathways involved in cell survival and death( Reference Forman, Davies and Ursini 40 , Reference Leonarduzzi, Sottero and Poli 41 ), disturbance of the cellular redox homeostatic system and potentially harmful effects induced by redox-active phytochemicals may emerge in normal cells compared with, for example, tumour cells or cells under various pathophysiological conditions( Reference Nair, Li and Kong 42 ). It is important to mention that our experiments provide evidence that dietary-compatible concentrations of Ind that counteracted oxysterol activity did not modify the redox environment of erythrocytes in the absence of oxysterols, ruling out interference with redox-dependent physiological functions of healthy cells.

A pathophysiological condition of hypercholesterolaemia was simulated by treating whole blood with a mixture of oxysterols. A remarkable amount of eryptotic erythrocytes, ROS formation and depletion of cellular GSH content were observed after 48 h, with all these effects being prevented by the concurrent exposure of erythrocytes to dietary-compatible amounts of Ind. Finally, PS exposure in eryptotic erythrocytes is a key event that promotes the adherence of erythrocytes to endothelial cells( Reference Borst, Abed and Alesutan 10 ). We observed that Ind completely inhibited the adhesion of erythrocytes to HUVEC monolayers, isolated after the spiking of blood with a mixture of oxysterols. Ind is a membrane-active compound that interacts with and is located in phospholipid bilayers( Reference Turco Liveri, Sciascia and Lombardo 23 , Reference Turco Liveri, Sciascia and Allegra 24 ). Our findings that Ind did not exhibit adhesive effects when alone indicated that while preventing oxysterol-promoted, PS exposure-dependent adhesive events, Ind did not create or unmask other adherence sites( Reference Borst, Abed and Alesutan 10 ).

Cytotoxic oxysterols are implicated in the pathophysiology of atherosclerosis, where they induce apoptosis in the cells of the vascular wall and in monocytes/macrophages( Reference Palozza, Serini and Verdecchia 43 Reference Larsson, Baird and Nyhalah 45 ), thus contributing to the development of an atheromatous plaque( Reference Tabas 46 ). The eryptotic activity of these compounds in hypercholesterolaemia may cause additional injury because of the adhesion of eryptotic erythrocytes to endothelial cells and eventual thrombo-occlusive complications. Our previous( 29 ) and present findings showing that nutritionally relevant amounts of Ind protect against toxicity induced by oxysterols in immunocompetent cells and erythrocytes suggest the potential health benefits from consumption of cactus pear fruits in preventing atherogenesis-related pathologies.

Acknowledgements

The present study was supported by Grant from Italy's Ministry of University and Research, MIUR (University of Palermo, ex 60 %, 2012). The MIUR had no role in the design and analysis of the study or in the writing of this article.

The authors' contributions are as follows: L. T. and M. A. formulated the research questions and designed the study; M. A. L. coordinated the work and wrote the paper; A. A. carried out the experiments and analysed the data.

None of the authors has any conflict of interest to declare.

References

1 Lang, F, Gulbins, E, Lerche, H, et al. (2008) Eryptosis, a window to systemic disease. Cell Physiol Biochem 22, 373380.CrossRefGoogle ScholarPubMed
2 Bonomini, M, Sirolli, V, Settefrati, N, et al. (1999) Increased erythrocyte phosphatidylserine exposure in chronic renal failure. J Am Soc Nephrol 10, 19821990.Google Scholar
3 Wood, BL, Gibson, DF & Tait, JF (1996) Increased erythrocyte phosphatidylserine exposure in sickle cell disease: flow-cytometric measurement and clinical associations. Blood 88, 8731880.CrossRefGoogle ScholarPubMed
4 Basu, S, Banerjee, D, Chandra, S, et al. (2010) Eryptosis in hereditary spherocytosis and thalassemia: role of glycoconjugates. Glycoconj J 27, 717722.Google Scholar
5 Calderon-Salinas, JV, Munoz-Reyes, EG, Guerrero-Romero, JF, et al. (2011) Eryptosis and oxidative damage in type 2 diabetic mellitus patients with chronic kidney disease. Mol Cell Biochem 357, 171179.CrossRefGoogle ScholarPubMed
6 Chung, SM, Bae, ON, Lim, KM, et al. (2007) Lysophosphatidic acid induces thrombogenic activity through phosphatidylserine exposure and procoagulant microvesicle generation in human erythrocytes. Arterioscler Thromb Vasc Biol 27, 414421.Google Scholar
7 Andrews, DA & Low, PS (1999) Role of red blood cells in thrombosis. Curr Opin Hematol 6, 7682.Google Scholar
8 Closse, C, Dachary-Prigent, J & Boisseau, MR (1999) Phosphatidylserine-related adhesion of human erythrocytes to vascular endothelium. Br J Haematol 107, 300302.CrossRefGoogle ScholarPubMed
9 Pandolfi, A, Di Pietro, N, Sirolli, V, et al. (2007) Mechanisms of uremic erythrocyte-induced adhesion of human monocytes to cultured endothelial cells. J Cell Physiol 213, 699709.Google Scholar
10 Borst, O, Abed, M, Alesutan, I, et al. (2012) Dynamic adhesion of eryptotic erythrocytes to endothelial cells via CXCL16/SR-PSOX. Am J Physiol Cell Physiol 302, C644C651.Google Scholar
11 Addis, PB, Emanuel, HA, Bergmann, SD, et al. (1989) Capillary quantification of cholesterol oxidation products in plasma lipoproteins of fasted humans. Free Radic Biol Med 7, 179182.Google Scholar
12 Sevanian, A, Seraglia, R, Traldi, P, et al. (1994) Analysis of plasma cholesterol oxidation products using gas- and high performance liquid chromatography/mass spectrometry. Free Radic Biol Med 17, 397409.Google Scholar
13 Chang, YH, Abdalla, DS & Sevanian, A (1997) Characterization of cholesterol oxidation products formed by oxidative modification of low density lipoprotein. Free Radic Biol Med 23, 202214.Google Scholar
14 Tesoriere, L, Attanzio, A, Allegra, M, et al. (2014) Oxysterol mixture in hypercholesterolemia-relevant proportion causes oxidative stress-dependent eryptosis. Cell Physiol Biochem 34, 10751079.CrossRefGoogle ScholarPubMed
15 Poli, G, Biasi, F & Leonarduzzi, G (2013) Oxysterols in the pathogenesis of major chronic diseases. Redox Biol 1, 125130.CrossRefGoogle ScholarPubMed
16 Brown, AJ & Jessup, W (1999) Oxysterols and atherosclerosis. Atherosclerosis 142, 128.CrossRefGoogle ScholarPubMed
17 Slavin, JL & Lloyd, B (2012) Health benefits of fruits and vegetables. Adv Nutr 3, 506511.Google Scholar
18 Stintzing, FC & Carle, R (2007) Betalains – emerging prospects for food scientists. Trends in Food Sci Technol 18, 514525.Google Scholar
19 Piattelli, M (1981) The betalains: Structure, biosynthesis, and chemical taxonomy. In The Biochemistry of Plants, Vol. 7, pp. 557575 [Conn, EE, editor]. New York: Academic Press, Inc.Google Scholar
20 Butera, D, Tesoriere, L, Di Gaudio, F, et al. (2002) Antioxidant activities of Sicilian prickly pear (Opuntia ficus indica) fruit extracts and reducing properties of its betalains: betanin and indicaxanthin. J Agric Food Chem 50, 68956901.Google Scholar
21 Allegra, M, Furtmuller, PG, Jantschko, W, et al. (2005) Mechanism of interaction of betanin and indicaxanthin with human myeloperoxidase and hypochlorous acid. Biochem Biophys Res Commun 332, 837844.Google Scholar
22 Tesoriere, L, Allegra, M, Butera, D, et al. (2007) Kinetics of the lipoperoxyl radical-scavenging activity of indicaxanthin in solution and unilamellar liposomes. Free Radic Res 41, 226233.Google Scholar
23 Turco Liveri, ML, Sciascia, L, Lombardo, R, et al. (2007) Spectrophotometric evidence for the solubilization site of betalain pigments in membrane biomimetic systems. J Agric Food Chem 55, 28362840.Google Scholar
24 Turco Liveri, ML, Sciascia, L, Allegra, M, et al. (2009) Partition of indicaxanthin in membrane biomimetic systems. A kinetic and modeling approach. J Agric Food Chem 57, 1095910963.CrossRefGoogle ScholarPubMed
25 Tesoriere, L, Butera, D, Allegra, M, et al. (2005) Distribution of betalain pigments in red blood cells after consumption of cactus pear fruits and increased resistance of the cells to ex vivo induced oxidative hemolysis in humans. J Agric Food Chem 53, 12661270.Google Scholar
26 Tesoriere, L, Allegra, M, Butera, D, et al. (2006) Cytoprotective effects of the antioxidant phytochemical indicaxanthin in β-thalassemia red blood cells. Free Radic Res 40, 753761.Google Scholar
27 Gentile, C, Tesoriere, L, Allegra, M, et al. (2004) Antioxidant betalains from cactus pear (Opuntia ficus-indica) inhibit endothelial ICAM-1 expression. Ann N Y Acad Sci 1028, 481486.Google Scholar
28 Tesoriere, L, Attanzio, A, Allegra, M, et al. (2014) Indicaxanthin inhibits NADPH oxidase (NOX)-1 activation and NF-κB-dependent release of inflammatory mediators and prevents the increase of epithelial permeability in IL-1β-exposed Caco-2 cells. Br J Nutr 111, 415423.Google Scholar
29 Tesoriere. L., Attanzio. A., Allegra, M., et al (2013) Phytochemical indicaxanthin suppresses 7-ketocholesterol-induced THP-1 cell apoptosis by preventing cytosolic Ca+2 increase and oxidative stress. Br J Nutr 110, 230240.Google Scholar
30 Tesoriere, L, Allegra, M, Butera, D, et al. (2004) Absorption, excretion, and distribution in low density lipoproteins of dietary antioxidant betalains. Potential health effects of betalains in humans. Am J Clin Nutr 80, 941945.CrossRefGoogle ScholarPubMed
31 Tesoriere, L, Fazzari, M, Angileri, F, et al. (2008) In vitro digestion of betalainic foods. Stability and bioaccessibility of betaxanthins and betacyanins and antioxidative potential of food digesta. J Agric Food Chem 56, 1048710492.Google Scholar
32 Tesoriere, L, Gentile, C, Angileri, F, et al. (2013) Trans-epithelial transport of the betalain pigments indicaxanthin and betanin across Caco-2 cell monolayers and influence of food matrix. Eur J Nutr 52, 10771087.CrossRefGoogle ScholarPubMed
33 Hu, ML (1994) Measurement of protein thiol groups and glutathione in plasma. In Methods in Enzymology, Vol. 233, pp. 380385 [Packer, L, editor]. San Diego, CA: Academic Press.Google Scholar
34 Sugihara, K, Sugihara, T, Mohandas, N, et al. (1992) Thrombospondin mediates adherence of CD38+ sickle reticulocytes to endothelial cells. Blood 80, 26342642.Google Scholar
35 Lang, PA, Kempe, DS, Myssina, S, et al. (2005) PGE2 in the regulation of programmed erythrocyte death. Cell Death Differ 12, 415428.Google Scholar
36 Tesoriere, L, Butera, D, D'Arpa, D, et al. (2003) Increased resistance to oxidation of betalain-enriched human low density lipoproteins. Free Radic Res 37, 689696.Google Scholar
37 Olkkonen, VM & Hynynen, R (2009) Interactions of oxysterols with membranes and proteins. Mol Aspects Med 30, 123133.CrossRefGoogle ScholarPubMed
38 Massey, JB & Pownall, HJ (2006) Structures of biologically active oxysterols determine their differential effects on phospholipid membranes. Biochemistry 45, 1074710758.Google Scholar
39 George, A, Pushkaran, S, Kostantinidis, DG, et al. (2013) Erythrocyte NADPH oxidase activity modulated by Rac GTPases, PKC, and plasma cytokines contributes to oxidative stress in sickle cell disease. Blood 121, 20992107.Google Scholar
40 Forman, HJ, Davies, KJA & Ursini, F (2014) How do nutritional antioxidants really work: nucleophilic tone and para-hormesis versus free radical scavenging in vivo . Free Radic Biol Med 66, 2435.Google Scholar
41 Leonarduzzi, G, Sottero, B & Poli, G (2010) Targeting tissue oxidative damage by means of cell signaling modulators: the antioxidant concept revisited. Pharmacol Ther 128, 336374.Google Scholar
42 Nair, S, Li, W & Kong, AT (2007) Natural dietary anti-cancer chemopreventive compounds: redox-mediated differential signaling mechanisms in cytoprotection of normal cells versus cytotoxicity in tumor cells. Acta Pharmacol Sin 28, 459472.Google Scholar
43 Palozza, P, Serini, S, Verdecchia, S, et al. (2007) Redox regulation of 7-ketocholesterol-induced apoptosis by β-carotene in human macrophages. Free Radic Biol Med 42, 15791590.Google Scholar
44 Biasi, F, Leonarduzzi, G, Vizio, B, et al. (2004) Oxysterol mixtures prevent proapoptotic effects of 7-ketocholesterol in macrophages: implications for proatherogenic gene modulation. FASEB J 18, 693695.CrossRefGoogle ScholarPubMed
45 Larsson, DA, Baird, S, Nyhalah, JD, et al. (2006) Oxysterol mixtures, in atheroma-relevant proportions, display synergistic and proapoptotic effects. Free Radic Biol Med 41, 902910.Google Scholar
46 Tabas, I (2010) Macrophage death and defective inflammation resolution in atherosclerosis. Nature 10, 3645.Google Scholar
Figure 0

Fig. 1 Molecular structure of indicaxanthin.

Figure 1

Fig. 2 Inhibition of oxysterol-induced eryptosis by indicaxanthin (Ind). (a) Percentage of phosphatidylserine-exposing erythrocytes or (c) their forward scatter, after 48 h incubation with the oxysterol mixture in the presence of 0–5 μm-Ind. Histograms of (b) annexin V-binding cells or (d) their forward scatter in a typical experiment in the absence () or presence () of 5 μm-Ind. Cells incubated with vehicle were used as the control. Values are means of six independent experiments carried out in triplicate, with their standard deviations represented by vertical bars. *Each value was significantly different from that of the control (P< 0·0001; ANOVA with Bonferroni's test). † Each value was significantly different from that of the oxysterol mixture in the absence of indicaxanthin (P< 0·05; ANOVA with Bonferroni's test).

Figure 2

Fig. 3 Inhibition of oxysterol-induced (a) Ca2+ entry and (b) PGE2 release by indicaxanthin (Ind) in human erythrocytes. Cells were incubated in the absence or presence of 0–5 μm-Ind for 48 h. Cells incubated with vehicle were used as the control. Values are means of six independent experiments carried out in triplicate, with their standard deviations represented by vertical bars. * Each value was significantly different from that of the control (P< 0·0001; ANOVA with Bonferroni's test). † Each value was significantly different from that of the oxysterol mixture in the absence of indicaxanthin (P< 0·05; ANOVA with Bonferroni's test).

Figure 3

Fig. 4 Inhibition of oxysterol-induced (a, b) reactive oxygen species (ROS) production and (c) glutathione (GSH) depletion by indicaxanthin (Ind) in human erythrocytes. (a) Dichloro-dihydro-fluorescein diacetate (DCFDA)-associated mean fluorescence intensity (MFI) and (c) GSH levels, after 48 h incubation of erythrocytes with the oxysterol mixture in the absence or presence of 0–5 μm-Ind. (b) Histogram of DCFDA-stained cells in a typical experiment without () or with () 5 μm-Ind. Cells incubated with vehicle were used as the control. Values are means of six independent experiments carried out in triplicate, with their standard deviations represented by vertical bars. * Each value was significantly different from that of the control (P< 0·0001; ANOVA with Bonferroni's test). † Each value was significantly different from that of the oxysterol mixture in the absence of indicaxanthin (P< 0·05; ANOVA with Bonferroni's test).

Figure 4

Fig. 5 Time-dependent reactive oxygen species (ROS) production in erythrocytes treated with the oxysterol mixture in the absence () or presence () of 5 μm-indicaxanthin (Ind) and incorporation of the phytochemical in the cells. Dichloro-dihydro-fluorescein diacetate-associated mean fluorescence intensity (MFI). Values are means of six independent experiments carried out in triplicate, with their standard deviations represented by vertical bars. , ROS production (MFI); , Ind (nmol/107 cells).

Figure 5

Fig. 6 (a) Phosphatidylserine exposure, (b) reactive oxygen species (ROS) production and (c) glutathione (GSH) level in erythrocytes isolated after a 48 h ex vivo spiking of fresh human blood with the oxysterol mixture in the absence or presence of 5 μm-indicaxanthin (Ind). Cells isolated from homologous blood incubated in the presence of vehicle were used as the control. Values are means of independent experiments carried out in duplicate with blood collected from five volunteers (each contributing two samples (n 10)), with their standard deviations represented by vertical bars. * Each value was significantly different from that of the control (P< 0·0001; ANOVA with Bonferroni's test).

Figure 6

Fig. 7 Adherence of erythrocytes to human umbilical vein endothelial cell monolayers isolated after a 48 h ex vivo spiking of fresh human blood with the oxysterol mixture in the absence or presence of 5 μm-indicaxanthin (Ind). Cells isolated from homologous blood incubated in the presence of vehicle were used as the control. Values are means of independent experiments carried out in duplicate with blood collected from five volunteers (each contributing two samples (n 10)), with their standard deviations represented by vertical bars. * Each value was significantly different from that of the control (P< 0·0001; ANOVA with Bonferroni's test).