Background/Aims: The phenolic abietane diterpene component of rosemary and sage, carnosic acid, may either induce or inhibit apoptosis of nucleated cells. The mechanisms involved in the effects of carnosic acid include altered mitochondrial function and gene expression. Human erythrocytes lack mitochondria and nuclei but are nevertheless able to enter suicidal death or eryptosis, which is characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine translocation to the erythrocyte surface. Cellular mechanisms involved in the stimulation of eryptosis include oxidative stress, increase of cytosolic Ca2+ activity ([Ca2+]i), and ceramide formation. The present study explored, whether and how carnosic acid induces eryptosis. Methods: Phosphatidylserine exposure at the cell surface was estimated from annexin V binding, cell volume from forward scatter, [Ca2+]i from Fluo3-fluorescence, ROS formation from DCFDA dependent fluorescence and ceramide abundance utilizing specific antibodies. Results: A 48 hours exposure of human erythrocytes to carnosic acid significantly increased the percentage of annexin-V-binding cells (2.5 µg/ml), significantly decreased forward scatter (10 µg/ml), significantly increased Fluo3 fluorescence (10 µg/ml), significantly increased ceramide abundance (10 µg/ml), significantly increased hemolysis (10 µg/ml), but significantly decreased DCFDA fluorescence (10 µg/ml). The effect of carnosic acid on annexin-V-binding was significantly blunted, but not abolished by removal of extracellular Ca2+. Conclusion: Carnosic acid triggers cell shrinkage and phospholipid scrambling of the human erythrocyte cell membrane, an effect paralleled by and/or in part due to Ca2+ entry and increased ceramide abundance.

Carnosic acid, a phenolic abietane diterpene component of rosemary and sage [1,2,3,4,5,6,7], has been shown to exert anti-oxidative [1,2,8,9,10,11,12,13,14,15,16,17,18], anti-inflammatory [9,19,20,21,22,23], anti-adipogenic [2,22,23,24,25,26,27,28,29,30,31], anti-diabetic [24,27,29,32], anti-fatty liver [33], anti-tumor [6,20,32,34,35,36,37,38,39,40], anti-angiogenic [20,41], anti-microbial [1,6,32,42,43], pro-apoptotic [19,26,37,44,45,46,47], pro-autophagic [48], anti-apoptotic [5,13,14,16], gastroprotective [15], and neuroprotective [5,8,9,11,16,32,49,50,51,52,53,54,55] effects. Cellular mechanisms involved in the effects of carnosic acid include up-regulation of Nuclear factor (erythroid-derived 2)-like 2 (Nfr2) activity [8,13,55,56], preservation of mitochondrial respiratory function [11,56], mitochondrial depolarization [47], cytochrome-c release [36], increase in Bax:Bcl-2 ratio [36], caspase activation [36,37], up-regulation of NAD+-dependent deacetylase SIRT1 [2], glutathione S-transferase [8,57], death receptor (DR)5 [19], Bcl-2 interacting mediator of cell death (Bim) [19] and p53 up-regulated modulator of apoptosis (PUMA) [19] expression, down-regulation of c-FLIP and Bcl-2 expression [19] as well as stimulation of PGE2 synthesis [15]. Signaling involved in the effect of carnosic acid further includes phosphatidylinositol 3-kinase (PI3K)/Akt [26,46,47,48,49], nuclear factor-kappa B (NF-κB) [21,49], and p38 kinase [8,16,45,57].

To the best of our knowledge, nothing is known about effects of carnosic acid on survival of erythrocytes, cells lacking mitochondria and nuclei. Similar to apoptosis of nucleated cells, erythrocytes may enter eryptosis [58], the suicidal erythrocyte death characterized by cell shrinkage [59] and cell membrane scrambling with phosphatidylserine translocation to the cell surface [58]. Eryptosis may be triggered by activation of Ca2+ permeable unselective cation channels with subsequent Ca2+ entry and increase of cytosolic Ca2+ activity ([Ca2+]i) [58]. Mechanisms fostering eryptosis further include ceramide [60], energy depletion [58], activated caspases [58,61,62], activation of casein kinase 1α, Janus-activated kinase JAK3, protein kinase C, and p38 kinase [58], as well as inhibition or lack of AMP activated kinase AMPK, cGMP-dependent protein kinase, PAK2 kinase, and sorafenib/sunitinib sensitive kinases [58]. Eryptosis could be stimulated by a wide variety of xenobiotics [58,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87]. Moreover, enhanced eryptosis is observed in several clinical conditions, including dehydration [76], hyperphosphatemia [86], chronic kidney disease (CKD) [68,88,89,90], hemolytic-uremic syndrome [91], diabetes [92], hepatic failure [93], malignancy [58], sepsis [94], Sickle-cell disease [58], beta-thalassemia [58], Hb-C deficiency, G6PD-deficiency [58] and Wilsons disease [95].

The present study explored whether and how carnosic acid could trigger or inhibit eryptosis. To this end, human erythrocytes from healthy volunteers were treated with carnosic acid and phosphatidylserine surface abundance, cell volume, [Ca2+]i, ROS formation, and ceramide abundance determined by flow cytometry.

Erythrocytes, solutions and chemicals

Fresh Li-Heparin-anticoagulated blood samples were kindly provided by the blood bank of the University of Tübingen. The study is approved by the ethics committee of the University of Tübingen (184/2003 V). The blood was centrifuged at 120 x g for 20 min at 21 °C and the platelets and leukocytes-containing supernatant was disposed. Erythrocytes were incubated in vitro at a hematocrit of 0.4% in Ringer solution containing (in mM) 125 NaCl, 5 KCl, 1 MgSO4, 32 N-2-hydroxyethylpiperazine-N-2-ethanesulfonic acid (HEPES; pH 7.4), 5 glucose, 1 CaCl2, at 37°C for 48 hours. Where indicated, erythrocytes were exposed to carnosic acid (Sigma Aldrich, Hamburg, Germany) at the indicated concentrations.

Annexin-V-binding and forward scatter

After incubation under the respective experimental condition, a 150 µl cell suspension was washed in Ringer solution containing 5 mM CaCl2 and then stained with Annexin-V-FITC (1:200 dilution; ImmunoTools, Friesoythe, Germany) in this solution at 37°C for 15 min under protection from light. The annexin V abundance at the erythrocyte surface was subsequently determined on a FACS Calibur (BD, Heidelberg, Germany). Annexin V binding was measured with an excitation wavelength of 488 nm and an emission wavelength of 530 nm. A marker (M1) was placed to set an arbitrary threshold between annexin V-binding cells and control cells. The same threshold was used for untreated and carnosic acid treated erythrocytes. A dot plot of forward scatter (FSC) vs. side scatter (SSC) was set to linear scale for both parameters. The threshold of forward scatter was set at the default value of “52”.

Intracellular Ca2+

After incubation, a 150 µl cell suspension was centrifuged at 1600 rpm for 3 mins and, after trashing the supernatant, the erythrocytes were stained with Fluo-3/AM (Biotium, Hayward, USA) in Ringer solution containing 5 mM CaCl2 and 5 µM Fluo-3/AM for 30 min. Then, Ca2+-dependent fluorescence intensity was measured with an excitation wavelength of 488 nm and an emission wavelength of 530 nm on a FACS Calibur.

Reactive oxidant species (ROS)

Oxidative stress was determined utilizing 2',7'-dichlorodihydrofluorescein diacetate (DCFDA). After incubation, a 150 µl suspension of erythrocytes was centrifuged at 1600 rpm for 3 min and, after trashing the supernatant, the erythrocytes were stained with DCFDA (Sigma, Schnelldorf, Germany) in Ringer solution containing DCFDA at a final concentration of 10 µM. Erythrocytes were incubated at 37°C for 30 min in the dark and then washed in Ringer solution. The DCFDA-loaded erythrocytes were resuspended in 200 µl Ringer solution, and ROS-dependent fluorescence intensity was measured at an excitation wavelength of 488 nm and an emission wavelength of 530 nm on a FACS Calibur (BD).

Ceramide abundance

For the determination of ceramide, a monoclonal antibody-based assay was used. After incubation, a 100 µl cell suspension was centrifuged at 1600 rpm for 3 min, and, after trashing the supernatant, the erythrocytes were stained for 1 hour at 37°C with 1 µg/ml anti ceramide antibody (clone MID 15B4, Alexis, Grünberg, Germany) in PBS containing 0.1% bovine serum albumin (BSA) at a dilution of 1:10. After two washing steps with PBS-BSA, cells were stained for 30 min with polyclonal fluorescein- isothiocyanate (FITC)-conjugated goat anti-mouse IgG and IgM specific antibody (BD Pharmingen, Hamburg, Germany) diluted 1:50 in PBS-BSA. Unbound secondary antibody was removed by repeated washing with PBS-BSA. Samples were then analyzed by flow cytometric analysis at an excitation wavelength of 488 nm and an emission wavelength of 530 nm.

Hemolysis

In order to determine hemolysis, the samples were centrifuged (3 mins at 1600 rpm, room temperature) after incubation under the respective experimental conditions and the supernatants were harvested. As a measure of hemolysis, the hemoglobin (Hb) concentration of the supernatant was determined photometrically at 405 nm. The absorption of the supernatant of erythrocytes lysed in distilled water was defined as 100 % hemolysis. Hemolysis is expressed in % in order to allow comparison with % annexin V binding cells.

Statistics

Data are expressed as arithmetic means ± SEM. As indicated in the figure legends, statistical analysis was made using ANOVA with Tukey's test as post-test and t test as appropriate. n denotes the number of different erythrocyte specimens studied. Since different erythrocyte specimens used in distinct experiments are differently susceptible to triggers of eryptosis, the same erythrocyte specimens have been used for control and experimental conditions.

The present study explored the influence of carnosic acid on eryptosis, the suicidal erythrocyte death characterized by cell membrane scrambling with phosphatidylserine translocation to the cell surface and by cell shrinkage. In order to identify phosphatidylserine exposing erythrocytes, phosphatidylserine was stained with FITC-labelled annexin and annexin-V-binding quantified by flow cytometry. The erythrocytes were analyzed following incubation for 48 hours in Ringer solution without or with carnosic acid (1-10 µg/ml). As illustrated in Fig. 1, a 48 hours exposure to carnosic acid increased the percentage of phosphatidylserine exposing erythrocytes, an effect reaching statistical significance at 2.5 µg/ml carnosic acid.

The suicidal erythrocyte death could involve hemolysis, a cell death distinct from eryptosis. In order to estimate the effect of carnosic acid on hemolysis, the hemoglobin concentration in the supernatant was determined. As a result, a 48 hours incubation with carnosic acid resulted in hemolysis, an effect reaching statistical significance at 10 µg/ml carnosic acid (Fig. 2).

In order to estimate erythrocyte volume, forward scatter was determined utilizing flow cytometry following a 48 hours incubation in Ringer solution without or with carnosic acid (1- 10 µg/ml). As illustrated in Fig. 3, carnosic acid decreased erythrocyte forward scatter, an effect reaching statistical significance at 10 µg/ml carnosic acid concentration.

In order to estimate cytosolic Ca2+ activity ([Ca2+]i), the erythrocytes were loaded with the Ca2+ sensitive dye Fluo3 and the Fluo3 fluorescence taken as measure of [Ca2+]i. As illustrated in Fig. 4, a 48 hours exposure to carnosic acid increased the Fluo3 fluorescence, an effect reaching statistical significance at 10 µg/ml carnosic acid.

A further series of experiments tested whether the carnosic acid-induced translocation of phosphatidylserine or erythrocyte shrinkage required entry of extracellular Ca2+. To this end, erythrocytes were incubated for 48 hours in the absence or presence of 10 µg/ml carnosic acid in the presence or nominal absence of extracellular Ca2+. As illustrated in Fig. 5, removal of extracellular Ca2+ significantly blunted the effect of carnosic acid on annexin-V-binding. However, carnosic acid significantly increased the percentage of annexin-V-binding erythrocytes even in the absence of extracellular Ca2+. Thus, entry of extracellular Ca2+ accounted for a large part but not for the full effect of carnosic acid on cell membrane scrambling. Removal of extracellular Ca2+ further tended to blunt the effect of carnosic acid on forward scatter, an effect, however not reaching statistical significance. Again, even in the absence of extracellular Ca2+, carnosic acid significantly decreased the erythrocyte forward scatter (Fig. 6). Thus, entry of extracellular Ca2+ presumably contributed to but did not account for the effect of carnosic acid on cell volume.

Stimulators of Ca2+ entry and subsequent eryptosis include oxidative stress. Reactive oxygen species (ROS) was thus quantified utilizing 2′,7′-dichlorodihydrofluorescein diacetate (DCFDA). As shown in Fig. 7, a 48 hours exposure to carnosic acid (10 µg/ml) significantly decreased the DCFDA fluorescence. Accordingly, carnosic acid decreased oxidative stress.

Eryptosis could be stimulated without increase of [Ca2+]i by ceramide. Thus, the effect of carnosic acid on ceramide abundance in the cell membrane was estimated by flow cytometry utilizing specific antibodies. As shown in Fig. 8, a 48 hours exposure to carnosic acid (10 µg/ml) significantly increased the ceramide abundance at the erythrocyte surface.

The present observations reveal a novel effect of carnosic acid, i.e. the stimulation of suicidal erythrocyte death or eryptosis. Exposure of human erythrocytes to carnosic acid is followed by cell shrinkage and cell membrane scrambling with phosphatidylserine translocation to the erythrocyte surface.

The effect of 10 µg/ml carnosic acid on cell membrane scrambling and cell shrinkage was paralleled by an increase of cytosolic Ca2+ activity ([Ca2+]i), which is known to trigger cell membrane scrambling by activating an illdefined scramblase [58]. An increase of [Ca2+]i is further known to trigger erythrocyte shrinkage by activation of Ca2+ sensitive K+ channels with subsequent cell shrinkage due to K+ exit, cell membrane hyperpolarization, Cl- exit and thus cellular loss of KCl with water [59]. Comparison of Fig. 1 with Fig. 3 and Fig. 4 reveals that lower concentrations of carnosic acid (2.5 µg/ml) are required to trigger cell membrane scrambling than those required for cell shrinkage and increase of [Ca2+]i. It thus appears that, unlike cell shrinkage, cell membrane scrambling involves a mechanism other than increase of [Ca2+]i.

The effect of high concentrations of carnosic acid was, however, at least in part due to triggering of Ca2+ entry from the extracellular space, as removal of extracellular Ca2+ significantly blunted the effect of carnosic acid on annexin-V-binding. Ca2+ entered presumably through Ca2+ permeable cation channels [58]. The channels may be activated by oxidative stress [58]. However, DCFDA fluorescence reveals that carnosic acid treatment rather decreased the abundance of reactive oxidant species. Thus, the mechanism stimulating the channels remained elusive. The Ca2+ independent triggering of cell membrane scrambling and forward scatter is presumably in part the result of increased abundance of ceramide at the erythrocyte surface.

Besides its effect on eryptosis, carnosic acid also triggered hemolysis. Consequences of eryptosis and hemolysis include the clearance of the defective erythrocytes from circulating blood with potential development of anemia [58]. To the extend that eryptosis precedes hemolysis, eryptosis could prevent or attenuate release of hemoglobin, which otherwise passes the renal glomerular filter, subsequently precipitates in the acidic lumen of renal tubules and thus occludes nephrons [96].

Eryptosis may favourably influence the clinical course of malaria. Following infection with the malaria pathogen Plasmodium, the pathogen imposes oxidative stress on the infected host erythrocyte thus activating Ca2+-permeable erythrocyte cation channels [58,97]. By accelerating eryptosis, sickle-cell trait, beta-thalassemia-trait, Hb-C deficiency and G6PD-deficiency expedite the clearance of infected erythrocytes, thus decreasing parasitemia and protecting against a severe course of malaria [58,98,99,100]. Enhanced eryptosis further decreases parasitemia in iron deficiency [101] and following treatment with lead [101], chlorpromazine [102] or NO synthase inhibitors [102]. It is tempting to speculate that carnosic acid may similarly accelerate eryptosis of Plasmodium infected erythrocytes and thus have the potential to favourably influence the clinical course of the disease.

Besides fostering the development of anemia, eryptosis may lead to adherence of phosphatidylserine exposing erythrocytes to the vascular wall [103], stimulation of blood clotting and triggering of thrombosis [104,105,106]. Eryptosis may thus compromize microcirculation [60,104,107,108,109,110].

Carnosic acid triggers eryptosis with cell shrinkage and cell membrane scrambling, an effect paralleled by and in part due to increase of cytosolic Ca2+ activity and ceramide formation.

The authors acknowledge the meticulous preparation of the manuscript by Tanja Loch. The study was supported by the Deutsche Forschungsgemeinschaft and Open Access Publishing Fund of Tuebingen University.

The authors of this manuscript state that they have no conflicts of interest to declare.

1.
Birtic S, Dussort P, Pierre FX, Bily AC, Roller M: Carnosic acid. Phytochemistry 2015;115:9-19.
2.
Hu Y, Zhang N, Fan Q, Lin M, Zhang C, Fan G, Zhai X, Zhang F, Chen Z, Yao J: Protective efficacy of carnosic acid against hydrogen peroxide induced oxidative injury in HepG2 cells through the SIRT1 pathway. Can J Physiol Pharmacol 2015;10.1139/cjpp-2014-05131-7.
3.
Kracun-Kolarevic M, Dmitrovic S, Filipovic B, Peric M, Misic D, Simonovic A, Todorovic S: Influence of sodium salicylate on rosmarinic acid, carnosol and carnosic acid accumulation by Salvia officinalis L. shoots grown in vitro. Biotechnol Lett 2015;10.1007/s10529-015-1825-1.
4.
Labbe C, Faini F, Calderon D, Molina J, Arredondo S: Variations of carnosic acid and carnosol concentrations in ethanol extracts of wild Lepechinia salviae in Spring (2008-2011). Nat Prod Commun 2014;9:1413-1416.
5.
Meng P, Yoshida H, Tanji K, Matsumiya T, Xing F, Hayakari R, Wang L, Tsuruga K, Tanaka H, Mimura J, Kosaka K, Itoh K, Takahashi I, Kawaguchi S, Imaizumi T: Carnosic acid attenuates apoptosis induced by amyloid-beta 1-42 or 1-43 in SH-SY5Y human neuroblastoma cells. Neurosci Res 2015;94:1-9.
6.
Pertino MW, Theoduloz C, Butassi E, Zacchino S, Schmeda-Hirschmann G: Synthesis, antiproliferative and antifungal activities of 1,2,3-triazole-substituted carnosic Acid and carnosol derivatives. Molecules 2015;20:8666-8686.
7.
Song Y, Yan H, Chen J, Wang Y, Jiang Y, Tu P: Characterization of in vitro and in vivo metabolites of carnosic acid, a natural antioxidant, by high performance liquid chromatography coupled with tandem mass spectrometry. J Pharm Biomed Anal 2014;89:183-196.
8.
Chen JH, Ou HP, Lin CY, Lin FJ, Wu CR, Chang SW, Tsai CW: Carnosic acid prevents 6-hydroxydopamine-induced cell death in SH-SY5Y cells via mediation of glutathione synthesis. Chem Res Toxicol 2012;25:1893-1901.
9.
Hou CW, Lin YT, Chen YL, Wang YH, Chou JL, Ping LY, Jeng KC: Neuroprotective effects of carnosic acid on neuronal cells under ischemic and hypoxic stress. Nutr Neurosci 2012;10.1179/1476830512Y.0000000021.
10.
Jordan MJ, Castillo J, Banon S, Martinez-Conesa C, Sotomayor JA: Relevance of the carnosic acid/carnosol ratio for the level of rosemary diterpene transfer and for improving lamb meat antioxidant status. Food Chem 2014;151:212-218.
11.
Miller DM, Singh IN, Wang JA, Hall ED: Nrf2-ARE activator carnosic acid decreases mitochondrial dysfunction, oxidative damage and neuronal cytoskeletal degradation following traumatic brain injury in mice. Exp Neurol 2015;264:103-110.
12.
Raes K, Doolaege EH, Deman S, Vossen E, De Smet S: Effect of carnosic acid, quercetin and alpha-tocopherol on lipid and protein oxidation in an in vitro simulated gastric digestion model. Int J Food Sci Nutr 2015;66:216-221.
13.
Sahu BD, Putcha UK, Kuncha M, Rachamalla SS, Sistla R: Carnosic acid promotes myocardial antioxidant response and prevents isoproterenol-induced myocardial oxidative stress and apoptosis in mice. Mol Cell Biochem 2014;394:163-176.
14.
Sahu BD, Rentam KK, Putcha UK, Kuncha M, Vegi GM, Sistla R: Carnosic acid attenuates renal injury in an experimental model of rat cisplatin-induced nephrotoxicity. Food Chem Toxicol 2011;49:3090-3097.
15.
Theoduloz C, Pertino MW, Schmeda-Hirschmann G: Gastroprotective mechanisms of action of semisynthetic carnosic acid derivatives in human cells. Molecules 2013;19:581-594.
16.
Wu CR, Tsai CW, Chang SW, Lin CY, Huang LC, Tsai CW: Carnosic acid protects against 6-hydroxydopamine-induced neurotoxicity in in vivo and in vitro model of Parkinson's disease: involvement of antioxidative enzymes induction. Chem Biol Interact 2015;225:40-46.
17.
Xiang Q, Liu Z, Wang Y, Xiao H, Wu W, Xiao C, Liu X: Carnosic acid attenuates lipopolysaccharide-induced liver injury in rats via fortifying cellular antioxidant defense system. Food Chem Toxicol 2013;53:1-9.
18.
Park M, Han J, Lee CS, Soo BH, Lim KM, Ha H: Carnosic acid, a phenolic diterpene from rosemary, prevents UV-induced expression of matrix metalloproteinases in human skin fibroblasts and keratinocytes. Exp Dermatol 2013;22:336-341.
19.
Jung KJ, Min KJ, Bae JH, Kwon TK: Carnosic acid sensitized TRAIL-mediated apoptosis through down-regulation of c-FLIP and Bcl-2 expression at the post translational levels and CHOP-dependent up-regulation of DR5, Bim, and PUMA expression in human carcinoma caki cells. Oncotarget 2015;6:1556-1568.
20.
Rajasekaran D, Manoharan S, Silvan S, Vasudevan K, Baskaran N, Palanimuthu D: Proapoptotic, anti-cell proliferative, anti-inflammatory and anti-angiogenic potential of carnosic acid during 7,12 dimethylbenz[a] anthracene-induced hamster buccal pouch carcinogenesis. Afr J Tradit Complement Altern Med 2012;10:102-112.
21.
Tsai CW, Liu KL, Lin YR, Kuo WC: The mechanisms of carnosic acid attenuates tumor necrosis factor-alpha-mediated inflammation and insulin resistance in 3T3-L1 adipocytes. Mol Nutr Food Res 2014;58:654-664.
22.
Park MY, Mun ST: Carnosic acid inhibits TLR4-MyD88 signaling pathway in LPS-stimulated 3T3-L1 adipocytes. Nutr Res Pract 2014;8:516-520.
23.
Romo-Vaquero M, Larrosa M, Yanez-Gascon MJ, Issaly N, Flanagan J, Roller M, Tomas-Barberan FA, Espin JC, Garcia-Conesa MT: A rosemary extract enriched in carnosic acid improves circulating adipocytokines and modulates key metabolic sensors in lean Zucker rats: Critical and contrasting differences in the obese genotype. Mol Nutr Food Res 2014;58:942-953.
24.
Dickmann LJ, VandenBrink BM, Lin YS: In vitro hepatotoxicity and cytochrome P450 induction and inhibition characteristics of carnosic acid, a dietary supplement with antiadipogenic properties. Drug Metab Dispos 2012;40:1263-1267.
25.
Gaya M, Repetto V, Toneatto J, Anesini C, Piwien-Pilipuk G, Moreno S: Antiadipogenic effect of carnosic acid, a natural compound present in Rosmarinus officinalis, is exerted through the C/EBPs and PPARgamma pathways at the onset of the differentiation program. Biochim Biophys Acta 2013;1830:3796-3806.
26.
Kim YJ, Kim JS, Seo YR, Park JH, Choi MS, Sung MK: Carnosic acid suppresses colon tumor formation in association with antiadipogenic activity. Mol Nutr Food Res 2014;58:2274-2285.
27.
Lipina C, Hundal HS: Carnosic acid stimulates glucose uptake in skeletal muscle cells via a PME-1/PP2A/PKB signalling axis. Cell Signal 2014;26:2343-2349.
28.
Romo-Vaquero M, Selma MV, Larrosa M, Obiol M, Garcia-Villalba R, Gonzalez-Barrio R, Issaly N, Flanagan J, Roller M, Tomas-Barberan FA, Garcia-Conesa MT: A rosemary extract rich in carnosic acid selectively modulates caecum microbiota and inhibits beta-glucosidase activity, altering fiber and short chain fatty acids fecal excretion in lean and obese female rats. PLoS One 2014;9:e94687.
29.
Zhao Y, Sedighi R, Wang P, Chen H, Zhu Y, Sang S: Carnosic Acid as a major bioactive component in rosemary extract ameliorates high-fat-diet-induced obesity and metabolic syndrome in mice. J Agric Food Chem 2015;63:4843-4852.
30.
Park MY, Sung MK: Carnosic Acid Inhibits Lipid Accumulation in 3T3-L1 Adipocytes Through Attenuation of Fatty Acid Desaturation. J Cancer Prev 2015;20:41-49.
31.
Park MY, Sung MK: Carnosic acid attenuates obesity-induced glucose intolerance and hepatic fat accumulation by modulating genes of lipid metabolism in C57BL/6J-ob/ob mice. J Sci Food Agric 2015;95:828-835.
32.
Bozic D, Papaefthimiou D, Bruckner K, de Vos RC, Tsoleridis CA, Katsarou D, Papanikolaou A, Pateraki I, Chatzopoulou FM, Dimitriadou E, Kostas S, Manzano D, Scheler U, Ferrer A, Tissier A, Makris AM, Kampranis SC, Kanellis AK: Towards Elucidating Carnosic Acid Biosynthesis in Lamiaceae: Functional Characterization of the Three First Steps of the Pathway in Salvia fruticosa and Rosmarinus officinalis. PLoS One 2015;10:e0124106.
33.
Park MY, Mun ST: Dietary carnosic acid suppresses hepatic steatosis formation via regulation of hepatic fatty acid metabolism in high-fat diet-fed mice. Nutr Res Pract 2013;7:294-301.
34.
Barni MV, Carlini MJ, Cafferata EG, Puricelli L, Moreno S: Carnosic acid inhibits the proliferation and migration capacity of human colorectal cancer cells. Oncol Rep 2012;27:1041-1048.
35.
Einbond LS, Wu HA, Kashiwazaki R, He K, Roller M, Su T, Wang X, Goldsberry S: Carnosic acid inhibits the growth of ER-negative human breast cancer cells and synergizes with curcumin. Fitoterapia 2012;83:1160-1168.
36.
Kar S, Palit S, Ball WB, Das PK: Carnosic acid modulates Akt/IKK/NF-kappaB signaling by PP2A and induces intrinsic and extrinsic pathway mediated apoptosis in human prostate carcinoma PC-3 cells. Apoptosis 2012;17:735-747.
37.
Pesakhov S, Khanin M, Studzinski GP, Danilenko M: Distinct combinatorial effects of the plant polyphenols curcumin, carnosic acid, and silibinin on proliferation and apoptosis in acute myeloid leukemia cells. Nutr Cancer 2010;62:811-824.
38.
Steiner M, Priel I, Giat J, Levy J, Sharoni Y, Danilenko M: Carnosic acid inhibits proliferation and augments differentiation of human leukemic cells induced by 1,25-dihydroxyvitamin D3 and retinoic acid. Nutr Cancer 2001;41:135-144.
39.
Wang R, Li H, Guo G, Li X, Yu X, Li H, Wang J, Liu F, Chen X: Augmentation by carnosic acid of apoptosis in human leukaemia cells induced by arsenic trioxide via upregulation of the tumour suppressor PTEN. J Int Med Res 2008;36:682-690.
40.
Park SY, Song H, Sung MK, Kang YH, Lee KW, Park JH: Carnosic acid inhibits the epithelial-mesenchymal transition in B16F10 melanoma cells: a possible mechanism for the inhibition of cell migration. Int J Mol Sci 2014;15:12698-12713.
41.
Lopez-Jimenez A, Garcia-Caballero M, Medina MA, Quesada AR: Anti-angiogenic properties of carnosol and carnosic acid, two major dietary compounds from rosemary. Eur J Nutr 2013;52:85-95.
42.
Exarchou V, Kanetis L, Charalambous Z, Apers S, Pieters L, Gekas V, Goulas V: HPLC-SPE-NMR characterization of major metabolites in Salvia fruticosa Mill. extract with antifungal potential: relevance of carnosic acid, carnosol, and hispidulin. J Agric Food Chem 2015;63:457-463.
43.
Shin HB, Choi MS, Ryu B, Lee NR, Kim HI, Choi HE, Chang J, Lee KT, Jang DS, Inn KS: Antiviral activity of carnosic acid against respiratory syncytial virus. Virol J 2013;10:303.
44.
Min KJ, Jung KJ, Kwon TK: Carnosic Acid Induces Apoptosis Through Reactive Oxygen Species-mediated Endoplasmic Reticulum Stress Induction in Human Renal Carcinoma Caki Cells. J Cancer Prev 2014;19:170-178.
45.
Tsai CW, Lin CY, Lin HH, Chen JH: Carnosic acid, a rosemary phenolic compound, induces apoptosis through reactive oxygen species-mediated p38 activation in human neuroblastoma IMR-32 cells. Neurochem Res 2011;36:2442-2451.
46.
Wang R, Cong WH, Guo G, Li XX, Chen XL, Yu XN, Li H: Synergism between carnosic acid and arsenic trioxide on induction of acute myeloid leukemia cell apoptosis is associated with modulation of PTEN/Akt signaling pathway. Chin J Integr Med 2012;18:934-941.
47.
Xiang Q, Ma Y, Dong J, Shen R: Carnosic acid induces apoptosis associated with mitochondrial dysfunction and Akt inactivation in HepG2 cells. Int J Food Sci Nutr 2015;66:76-84.
48.
Gao Q, Liu H, Yao Y, Geng L, Zhang X, Jiang L, Shi B, Yang F: Carnosic acid induces autophagic cell death through inhibition of the Akt/mTOR pathway in human hepatoma cells. J Appl Toxicol 2015;35:485-492.
49.
Lin CY, Chen JH, Fu RH, Tsai CW: Induction of Pi form of glutathione S-transferase by carnosic acid is mediated through PI3K/Akt/NF-kappaB pathway and protects against neurotoxicity. Chem Res Toxicol 2014;27:1958-1966.
50.
Meng P, Yoshida H, Matsumiya T, Imaizumi T, Tanji K, Xing F, Hayakari R, Dempoya J, Tatsuta T, Aizawa-Yashiro T, Mimura J, Kosaka K, Itoh K, Satoh K: Carnosic acid suppresses the production of amyloid-beta 1-42 by inducing the metalloprotease gene TACE/ADAM17 in SH-SY5Y human neuroblastoma cells. Neurosci Res 2013;75:94-102.
51.
Merad M, Soufi W, Ghalem S, Boukli F, Baig MH, Ahmad K, Kamal MA: Molecular interaction of acetylcholinesterase with carnosic acid derivatives: a neuroinformatics study. CNS Neurol Disord Drug Targets 2014;13:440-446.
52.
Park JA, Kim S, Lee SY, Kim CS, Kim do K, Kim SJ, Chun HS: Beneficial effects of carnosic acid on dieldrin-induced dopaminergic neuronal cell death. Neuroreport 2008;19:1301-1304.
53.
Rasoolijazi H, Azad N, Joghataei MT, Kerdari M, Nikbakht F, Soleimani M: The protective role of carnosic acid against beta-amyloid toxicity in rats. ScientificWorldJournal 2013;2013:917082.
54.
Yoshida H, Meng P, Matsumiya T, Tanji K, Hayakari R, Xing F, Wang L, Tsuruga K, Tanaka H, Mimura J, Kosaka K, Itoh K, Takahashi I, Imaizumi T: Carnosic acid suppresses the production of amyloid-beta 1-42 and 1-43 by inducing an alpha-secretase TACE/ADAM17 in U373MG human astrocytoma cells. Neurosci Res 2014;79:83-93.
55.
Zhang D, Lee B, Nutter A, Song P, Dolatabadi N, Parker J, Sanz-Blasco S, Newmeyer T, Ambasudhan R, McKercher SR, Masliah E, Lipton SA: Protection from cyanide-induced brain injury by the Nrf2 transcriptional activator carnosic acid. J Neurochem 2015;133:898-908.
56.
Miller DM, Singh IN, Wang JA, Hall ED: Administration of the Nrf2-ARE activators sulforaphane and carnosic acid attenuates 4-hydroxy-2-nonenal-induced mitochondrial dysfunction ex vivo. Free Radic Biol Med 2013;57:1-9.
57.
Lin CY, Wu CR, Chang SW, Wang YJ, Wu JJ, Tsai CW: Induction of the pi class of glutathione S-transferase by carnosic acid in rat Clone 9 cells via the p38/Nrf2 pathway. Food Funct 2015;6:1936-1943.
58.
Lang F, Qadri SM: Mechanisms and significance of eryptosis, the suicidal death of erythrocytes. Blood Purif 2012;33:125-130.
59.
Lang PA, Kaiser S, Myssina S, Wieder T, Lang F, Huber SM: Role of Ca2+-activated K+ channels in human erythrocyte apoptosis. Am J Physiol Cell Physiol 2003;285:C1553-C1560.
60.
Abed M, Towhid ST, Mia S, Pakladok T, Alesutan I, Borst O, Gawaz M, Gulbins E, Lang F: Sphingomyelinase-induced adhesion of eryptotic erythrocytes to endothelial cells. Am J Physiol Cell Physiol 2012;303:C991-999.
61.
Lau IP, Chen H, Wang J, Ong HC, Leung KC, Ho HP, Kong SK: In vitro effect of CTAB- and PEG-coated gold nanorods on the induction of eryptosis/erythroptosis in human erythrocytes. Nanotoxicology 2012;6:847-856.
62.
Maellaro E, Leoncini S, Moretti D, Del Bello B, Tanganelli I, De Felice C, Ciccoli L: Erythrocyte caspase-3 activation and oxidative imbalance in erythrocytes and in plasma of type 2 diabetic patients. Acta Diabetol 2013;50:489-495.
63.
Jilani K, Lang F: Carmustine-induced phosphatidylserine translocation in the erythrocyte membrane. Toxins (Basel) 2013;5:703-716.
64.
Vota DM, Maltaneri RE, Wenker SD, Nesse AB, Vittori DC: Differential erythropoietin action upon cells induced to eryptosis by different agents. Cell Biochem Biophys 2013;65:145-157.
65.
Zappulla D: Environmental stress, erythrocyte dysfunctions, inflammation, and the metabolic syndrome: adaptations to CO2 increases? J Cardiometab Syndr 2008;3:30-34.
66.
Lupescu A, Jilani K, Zbidah M, Lang F: Patulin-induced suicidal erythrocyte death. Cell Physiol Biochem 2013;32:291-299.
67.
Abed M, Zoubi KA, Theurer M, Lang F: Effect of dermaseptin on erythrocytes. Basic Clin Pharmacol Toxicol 2013;113:347-352.
68.
Ahmed MS, Langer H, Abed M, Voelkl J, Lang F: The uremic toxin acrolein promotes suicidal erythrocyte death. Kidney Blood Press Res 2013;37:158-167.
69.
Ghashghaeinia M, Cluitmans JC, Toulany M, Saki M, Koberle M, Lang E, Dreischer P, Biedermann T, Duszenko M, Lang F, Bosman GJ, Wieder T: Age Sensitivity of NFkappaB Abundance and Programmed Cell Death in Erythrocytes Induced by NFkappaB Inhibitors. Cell Physiol Biochem 2013;32:801-813.
70.
Alzoubi K, Honisch S, Abed M, Lang F: Triggering of Suicidal Erythrocyte Death by Penta-O-galloyl-beta-d-glucose. Toxins (Basel) 2014;6:54-65.
71.
Jilani K, Enkel S, Bissinger R, Almilaji A, Abed M, Lang F: Fluoxetine induced suicidal erythrocyte death. Toxins (Basel) 2013;5:1230-1243.
72.
Lupescu A, Bissinger R, Jilani K, Lang F: Triggering of suicidal erythrocyte death by celecoxib. Toxins (Basel) 2013;5:1543-1554.
73.
Arnold M, Lang E, Modicano P, Bissinger R, Faggio C, Abed M, Lang F: Effect of nitazoxanide on erythrocytes. Basic Clin Pharmacol Toxicol 2014;114:421-426.
74.
Oswald G, Alzoubi K, Abed M, Lang F: Stimulation of suicidal erythrocyte death by ribavirin. Basic Clin Pharmacol Toxicol 2014;114:311-317.
75.
Lupescu A, Bissinger R, Herrmann T, Oswald G, Jilani K, Lang F: Induction of suicidal erythrocyte death by novobiocin. Cell Physiol Biochem 2014;33:670-680.
76.
Abed M, Feger M, Alzoubi K, Pakladok T, Frauenfeld L, Geiger C, Towhid ST, Lang F: Sensitization of erythrocytes to suicidal erythrocyte death following water deprivation. Kidney Blood Press Res 2013;37:567-578.
77.
Alzoubi K, Calabròa S, Bissinger R, Abed M, Faggio C, Lang F: Stimulation of Suicidal Erythrocyte Death by Artesunate. Cell Physiol Biochem 2014;34:2232-2244.
78.
Arnold M, Bissinger R, Lang F: Mitoxantrone-induced suicidal erythrocyte death. Cell Physiol Biochem 2014;34:1756-1767.
79.
Bissinger R, Fischer S, Jilani K, Lang F: Stimulation of Erythrocyte Death by Phloretin. Cell Physiol Biochem 2014;34:2256-2265.
80.
Bissinger R, Lupescu A, Zelenak C, Jilani K, Lang F: Stimulation of eryptosis by cryptotanshinone. Cell Physiol Biochem 2014;34:432-442.
81.
Bissinger R, Modicano P, Frauenfeld L, Lang E, Jacobi J, Faggio C, Lang F: Estramustine-induced suicidal erythrocyte death. Cell Physiol Biochem 2013;32:1426-1436.
82.
Jacobi J, Lang E, Bissinger R, Frauenfeld L, Modicano P, Faggio C, Abed M, Lang F: Stimulation of erythrocyte cell membrane scrambling by mitotane. Cell Physiol Biochem 2014;33:1516-1526.
83.
Lupescu A, Bissinger R, Warsi J, Jilani K, Lang F: Stimulation of erythrocyte cell membrane scrambling by gedunin. Cell Physiol Biochem 2014;33:1838-1848.
84.
Malik A, Bissinger R, Calabro S, Faggio C, Jilani K, Lang F: Aristolochic Acid Induced Suicidal Erythrocyte Death. Kidney Blood Press Res 2014;39:408-419.
85.
Tesoriere L, Attanzio A, Allegra M, Cilla A, Gentile C, Livrea MA: Oxysterol mixture in hypercholesterolemia-relevant proportion causes oxidative stress-dependent eryptosis. Cell Physiol Biochem 2014;34:1075-1089.
86.
Voelkl J, Alzoubi K, Mamar AK, Ahmed MS, Abed M, Lang F: Stimulation of suicidal erythrocyte death by increased extracellular phosphate concentrations. Kidney Blood Press Res 2013;38:42-51.
87.
Zhang R, Xiang Y, Ran Q, Deng X, Xiao Y, Xiang L, Li Z: Involvement of calcium, reactive oxygen species, and ATP in hexavalent chromium-induced damage in red blood cells. Cell Physiol Biochem 2014;34:1780-1791.
88.
Abed M, Artunc F, Alzoubi K, Honisch S, Baumann D, Foller M, Lang F: Suicidal erythrocyte death in end-stage renal disease. J Mol Med (Berl) 2014;92:871-879.
89.
Polak-Jonkisz D, Purzyc L: Ca(2+) influx versus efflux during eryptosis in uremic erythrocytes. Blood Purif 2012;34:209-210; author reply 210.
90.
Calderon-Salinas JV, Munoz-Reyes EG, Guerrero-Romero JF, Rodriguez-Moran M, Bracho-Riquelme RL, Carrera-Gracia MA, Quintanar-Escorza MA: Eryptosis and oxidative damage in type 2 diabetic mellitus patients with chronic kidney disease. Mol Cell Biochem 2011;357:171-179.
91.
Lang PA, Beringer O, Nicolay JP, Amon O, Kempe DS, Hermle T, Attanasio P, Akel A, Schafer R, Friedrich B, Risler T, Baur M, Olbricht CJ, Zimmerhackl LB, Zipfel PF, Wieder T, Lang F: Suicidal death of erythrocytes in recurrent hemolytic uremic syndrome. J Mol Med (Berl) 2006;84:378-388.
92.
Nicolay JP, Schneider J, Niemoeller OM, Artunc F, Portero-Otin M, Haik G, Jr., Thornalley PJ, Schleicher E, Wieder T, Lang F: Stimulation of suicidal erythrocyte death by methylglyoxal. Cell Physiol Biochem 2006;18:223-232.
93.
Lang E, Gatidis S, Freise NF, Bock H, Kubitz R, Lauermann C, Orth HM, Klindt C, Schuier M, Keitel V, Reich M, Liu G, Schmidt S, Xu HC, Qadri SM, Herebian D, Pandyra AA, Mayatepek E, Gulbins E, Lang F, Haussinger D, Lang KS, Foller M, Lang PA: Conjugated bilirubin triggers anemia by inducing erythrocyte death. Hepatology 2015;61:275-284.
94.
Kempe DS, Akel A, Lang PA, Hermle T, Biswas R, Muresanu J, Friedrich B, Dreischer P, Wolz C, Schumacher U, Peschel A, Gotz F, Doring G, Wieder T, Gulbins E, Lang F: Suicidal erythrocyte death in sepsis. J Mol Med (Berl) 2007;85:273-281.
95.
Lang PA, Schenck M, Nicolay JP, Becker JU, Kempe DS, Lupescu A, Koka S, Eisele K, Klarl BA, Rubben H, Schmid KW, Mann K, Hildenbrand S, Hefter H, Huber SM, Wieder T, Erhardt A, Haussinger D, Gulbins E, Lang F: Liver cell death and anemia in Wilson disease involve acid sphingomyelinase and ceramide. Nat Med 2007;13:164-170.
96.
Harrison HE, Bunting H, Ordway NK, Albrink WS: The Pathogenesis of the Renal Injury Produced in the Dog by Hemoglobin or Methemoglobin. J Exp Med 1947;86:339-356.
97.
Kirk K: Membrane transport in the malaria-infected erythrocyte. Physiol Rev 2001;81:495-537.
98.
Ayi K, Giribaldi G, Skorokhod A, Schwarzer E, Prendergast PT, Arese P: 16alpha-bromoepiandrosterone, an antimalarial analogue of the hormone dehydroepiandrosterone, enhances phagocytosis of ring stage parasitized erythrocytes: a novel mechanism for antimalarial activity. Antimicrob Agents Chemother 2002;46:3180-3184.
99.
Ayi K, Turrini F, Piga A, Arese P: Enhanced phagocytosis of ring-parasitized mutant erythrocytes: a common mechanism that may explain protection against falciparum malaria in sickle trait and beta-thalassemia trait. Blood 2004;104:3364-3371.
100.
Cappadoro M, Giribaldi G, O'Brien E, Turrini F, Mannu F, Ulliers D, Simula G, Luzzatto L, Arese P: Early phagocytosis of glucose-6-phosphate dehydrogenase (G6PD)-deficient erythrocytes parasitized by Plasmodium falciparum may explain malaria protection in G6PD deficiency. Blood 1998;92:2527-2534.
101.
Koka S, Huber SM, Boini KM, Lang C, Foller M, Lang F: Lead decreases parasitemia and enhances survival of Plasmodium berghei-infected mice. Biochem Biophys Res Commun 2007;363:484-489.
102.
Koka S, Lang C, Niemoeller OM, Boini KM, Nicolay JP, Huber SM, Lang F: Influence of NO synthase inhibitor L-NAME on parasitemia and survival of Plasmodium berghei infected mice. Cell Physiol Biochem 2008;21:481-488.
103.
Borst O, Abed M, Alesutan I, Towhid ST, Qadri SM, Foller M, Gawaz M, Lang F: Dynamic adhesion of eryptotic erythrocytes to endothelial cells via CXCL16/SR-PSOX. Am J Physiol Cell Physiol 2012;302:C644-C651.
104.
Andrews DA, Low PS: Role of red blood cells in thrombosis. Curr Opin Hematol 1999;6:76-82.
105.
Chung SM, Bae ON, Lim KM, Noh JY, Lee MY, Jung YS, Chung JH: Lysophosphatidic acid induces thrombogenic activity through phosphatidylserine exposure and procoagulant microvesicle generation in human erythrocytes. Arterioscler Thromb Vasc Biol 2007;27:414-421.
106.
Zwaal RF, Comfurius P, Bevers EM: Surface exposure of phosphatidylserine in pathological cells. Cell Mol Life Sci 2005;62:971-988.
107.
Closse C, Dachary-Prigent J, Boisseau MR: Phosphatidylserine-related adhesion of human erythrocytes to vascular endothelium. Br J Haematol 1999;107:300-302.
108.
Gallagher PG, Chang SH, Rettig MP, Neely JE, Hillery CA, Smith BD, Low PS: Altered erythrocyte endothelial adherence and membrane phospholipid asymmetry in hereditary hydrocytosis. Blood 2003;101:4625-4627.
109.
Pandolfi A, Di Pietro N, Sirolli V, Giardinelli A, Di Silvestre S, Amoroso L, Di Tomo P, Capani F, Consoli A, Bonomini M: Mechanisms of uremic erythrocyte-induced adhesion of human monocytes to cultured endothelial cells. J Cell Physiol 2007;213:699-709.
110.
Wood BL, Gibson DF, Tait JF: Increased erythrocyte phosphatidylserine exposure in sickle cell disease: flow-cytometric measurement and clinical associations. Blood 1996;88:1873-1880.
Open Access License / Drug Dosage / Disclaimer
This article is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License (CC BY-NC-ND). Usage and distribution for commercial purposes as well as any distribution of modified material requires written permission. Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug. Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.