Background/Aims: The phytochemical polyphenol rottlerin is a potent activator of diverse Ca2+ -sensitive K+ channels. Those channels play a decisive role in the execution of eryptosis, the suicidal death of erythrocytes, which is characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine translocation to the erythrocyte surface. Signaling involved in the stimulation of eryptosis includes increase of cytosolic Ca2+ activity ([Ca2+]i) and ceramide. The present study explored, whether rottlerin induces eryptosis and, if so, to test for the involvement of Ca2+ entry and ceramide. Methods: Flow cytometry was employed to estimate phosphatidylserine exposure at the cell surface from annexin-V-binding, cell volume from forward scatter, [Ca2+]i from Fluo3-fluorescence, and ceramide abundance utilizing specific antibodies. Hemolysis was quantified by determination of haemoglobin concentration in the supernatant. Results: A 48 hours exposure of human erythrocytes to rottlerin (1 - 5 µM) significantly increased the percentage of annexin-V-binding cells, an effect paralleled by significant decrease of forward scatter. Up to 5 µM rottlerin failed to significantly increase average Fluo3-fluorescence. Rottlerin (5 µM) did, however, significantly increase the ceramide abundance. Rottlerin (5 µM) further significantly increased hemolysis. The effect of rottlerin (5 µM) on annexin-V-binding was virtually abolished by removal of extracellular Ca2+. Conclusions: Rottlerin stimulates eryptosis with erythrocyte shrinkage and phospholipid scrambling of the erythrocyte cell membrane, an effect paralleled by and at least in part due to Ca2+ entry and ceramide.

The natural polyphenol rottlerin has antioxidant, anti-inflammatory, antipsoriatic, antiamyloid and anti-cancer potency [1,2,3]. The substance is partially effective by influencing the activity and/or expression of several enzymes, such as kinases, heme oxygenase, DNA methyltransferase, cyclooxygenase, and lipoxygenase, as well as transcription factors including NF-κB and STAT [1]. Moreover, rottlerin may prevent aggregation of different amyloid precursors, such as α-synuclein, amyloid Aβ, prion proteins, and lysozyme [1,2,3]. Rottlerin has further been shown to stimulate Ca2+ -sensitive K+ channels [4].

Activation of Ca2+ -sensitive K+ channels is a key event in eryptosis [5], the suicidal death of erythrocytes characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine translocation to the cell surface [6]. Eryptosis is stimulated by increase of cytosolic Ca2+ activity ([Ca2+]i) [6] and ceramide [7]. Further stimulators of eryptosis include oxidative stress [6] and energy depletion [6]. Eryptosis may or may not involve activation of caspases depending on the respective trigger of eryptosis [6,8,9]. Kinases fostering eryptosis include casein kinase 1α, Janus-activated kinase JAK3, protein kinase C, and/or p38 kinase [6]. Kinases inhibiting eryptosis include AMP activated kinase AMPK, cGMP-dependent protein kinase, PAK2 kinase and sorafenib/sunitinib sensitive kinases [6]. Eryptosis is stimulated by a myriad of xenobiotics [6,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43]. Enhanced eryptosis is observed in several clinical conditions including dehydration [44], hyperphosphatemia [45], chronic kidney disease (CKD) [46,47,48,49], hemolytic-uremic syndrome [50], diabetes [51], hepatic failure [52], malignancy [6], sepsis [53], sickle-cell disease [6], beta-thalassemia [6], Hb-C and G6PD-deficiency [6], as well as Wilsons disease [54].

The present study explored, whether rottlerin stimulates eryptosis. To this end, human erythrocytes drawn from healthy volunteers were exposed to rottlerin and phosphatidylserine abundance at the erythrocyte surface, erythrocyte volume, [Ca2+]i, and ceramide determined by flow cytometry. For compasison, hemolysis was quantified from hemoglobin release.

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 g for 20 min at 21 °C and the platelets and leukocytes-containing supernatant was disposed. Erythrocytes were incubated in vitro for 48 hours at 37°C and 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. without or with rottlerin (Santa Cruz Biotechnology, USA). Where indicated, CaCl2 was removed and 1 mM EGTA added.

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 rottlerin 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”.

Hemolysis

Following incubation, the erythrocyte suspension was centrifuged for 3 min at 1600 rpm, 4°C, and the supernatant harvested. As a measure of hemolysis, the hemoglobin (Hb) concentration in the supernatant was determined photometrically at 405 nm. The absorption of the supernatant of erythrocytes lysed in distilled water was defined as 100% hemolysis.

Intracellular Ca2+

After incubation, erythrocytes were washed in Ringer solution and loaded with Fluo-3/AM (Biotium, Hayward, USA) in Ringer solution containing 5 mM CaCl2 and 5 µM Fluo-3/AM. The cells were incubated at 37°C for 30 min. Ca2+-dependent fluorescence intensity was measured with an excitation wavelength of 488 nm and an emission wavelength of 530 nm on a FACS Calibur.

Ceramide abundance

For the determination of ceramide, a monoclonal antibody-based assay was used. To this end, cells 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. The samples were washed twice with PBS-BSA. The cells were stained for 30 minutes with polyclonal fluorescein isothiocyanate (FITC) conjugated goat anti-mouse IgG and IgM specific antibody (Pharmingen, Hamburg, Germany) diluted 1:50 in PBS-BSA. Unbound secondary antibody was removed by repeated washing with PBS-BSA. The samples were analyzed by flow cytometric analysis with an excitation wavelength of 488 nm and an emission wavelength of 530 nm. As a control, secondary antibody alone was used.

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 whether rottlerin is capable to trigger eryptosis, the suicidal erythrocyte death. Hallmarks of eryptosis are cell shrinkage and phospholipid scrambling of the cell membrane with phosphatidylserine translocation to the cell surface.

Erythrocyte volume was estimated from forward scatter which was determined utilizing flow cytometry. Prior to measurements, the erythrocytes were incubated for 48 hours in Ringer solution without or with rottlerin (1 - 5 µM). As illustrated in Fig. 1, rottlerin increased average erythrocyte forward scatter, an effect reaching statistical significance at 10 µM rottlerin concentration. A closer analysis of the histograms revealed that treatment with rottlerin increased the percentage of both, swollen and shrunken erythrocytes.

Fig. 1

Effect of rottlerin on erythrocyte forward scatter. A. Original histograms of forward scatter of erythrocytes following exposure for 48 hours to Ringer solution without (grey area) and with (black line) presence of 5 µM rottlerin. B. Arithmetic means ± SEM (n = 10) of the erythrocyte forward scatter (FSC) following incubation for 48 hours to Ringer solution without (white bar) or with (black bars) rottlerin (1 - 5 µM) or solvant DMSO alone (grey bar). *(P<0.05), **(p<0.01), ***(p<0.001) indicate significant difference from the absence of rottlerin (ANOVA).

Fig. 1

Effect of rottlerin on erythrocyte forward scatter. A. Original histograms of forward scatter of erythrocytes following exposure for 48 hours to Ringer solution without (grey area) and with (black line) presence of 5 µM rottlerin. B. Arithmetic means ± SEM (n = 10) of the erythrocyte forward scatter (FSC) following incubation for 48 hours to Ringer solution without (white bar) or with (black bars) rottlerin (1 - 5 µM) or solvant DMSO alone (grey bar). *(P<0.05), **(p<0.01), ***(p<0.001) indicate significant difference from the absence of rottlerin (ANOVA).

Close modal

Phosphatidylserine exposing erythrocytes were identified utilizing annexin-V-binding, as determined by flow cytometry. Prior to measurements, the erythrocytes were again incubated for 48 hours in Ringer solution without or with rottlerin (1 - 5 µM). As shown in Fig. 2, a 48 hours exposure to rottlerin significantly increased the percentage of phosphatidylserine exposing erythrocytes at all concentrations tested.

Fig. 2

Effect of rottlerin on phosphatidylserine exposure. A. Original histogram of annexin-V-binding of erythrocytes following exposure for 48 hours to Ringer solution without (grey area) and with (black line) presence of 5 µM rottlerin. B. Arithmetic means ± SEM (n = 10) of erythrocyte annexin-V-binding following incubation for 48 hours to Ringer solution without (white bar) or with (black bars) rottlerin (1 - 5 µM) or solvant DMSO alone (grey bar). ***(p<0.001) indicates significant difference from the absence of rottlerin (ANOVA).

Fig. 2

Effect of rottlerin on phosphatidylserine exposure. A. Original histogram of annexin-V-binding of erythrocytes following exposure for 48 hours to Ringer solution without (grey area) and with (black line) presence of 5 µM rottlerin. B. Arithmetic means ± SEM (n = 10) of erythrocyte annexin-V-binding following incubation for 48 hours to Ringer solution without (white bar) or with (black bars) rottlerin (1 - 5 µM) or solvant DMSO alone (grey bar). ***(p<0.001) indicates significant difference from the absence of rottlerin (ANOVA).

Close modal

Hemoglobin concentration in the supernatant was taken as a measure of hemolysis. Prior to measurements, the erythrocytes were again incubated for 48 hours in Ringer solution without or with rottlerin (5 µM). As illustrated in Fig. 3, a 48 hours exposure to 5 µM rottlerin significantly increased the percentage of hemolytic erythrocytes.

Fig. 3

Effect of rottlerin on hemolysis. Arithmetic means ± SEM (n = 15) of the percentage hemolytic erythrocytes following incubation for 48 hours to Ringer solution without (white bar) or with (black bar) rottlerin (5 µM) or solvant DMSO alone (grey bar). **(p<0.01), indicates significant difference from the absence of rottlerin (ANOVA).

Fig. 3

Effect of rottlerin on hemolysis. Arithmetic means ± SEM (n = 15) of the percentage hemolytic erythrocytes following incubation for 48 hours to Ringer solution without (white bar) or with (black bar) rottlerin (5 µM) or solvant DMSO alone (grey bar). **(p<0.01), indicates significant difference from the absence of rottlerin (ANOVA).

Close modal

Fluo3 fluorescence was taken as a measure of cytosolic Ca2+ activity ([Ca2+]i). Prior to measurements, the erythrocytes were again incubated for 48 hours in Ringer solution without or with rottlerin (1 - 5 µM). As illustrated in Fig. 4A,B, a 48 hours incubation with rottlerin did not significantly modify the average Fluo3 fluorescence. Higher concentrations of rottlerin (10 µM) significantly (p<0.05) increased Fluo-3 fluorescence (from 13.4 ± 1.3 to 18.2 ± 3.1 n = 10).

Fig. 4

Effect of rottlerin on cytosolic Ca2+ activity. A. Original histograms of Fluo-3 fluorescence in erythrocytes following exposure for 48 hours to Ringer solution without (grey area) and with (black line) presence of 5 µM rottlerin. B. Arithmetic means ± SEM (n = 10) of Fluo-3 fluorescence in erythrocytes following incubation for 48 hours to Ringer solution without (white bar) or with (black bars) rottlerin (1 - 5 µM) or solvant DMSO alone (grey bar).

Fig. 4

Effect of rottlerin on cytosolic Ca2+ activity. A. Original histograms of Fluo-3 fluorescence in erythrocytes following exposure for 48 hours to Ringer solution without (grey area) and with (black line) presence of 5 µM rottlerin. B. Arithmetic means ± SEM (n = 10) of Fluo-3 fluorescence in erythrocytes following incubation for 48 hours to Ringer solution without (white bar) or with (black bars) rottlerin (1 - 5 µM) or solvant DMSO alone (grey bar).

Close modal

A next series of experiments explored whether the rottlerin-induced translocation of phosphatidylserine required entry of extracellular Ca2+. To this end, erythrocytes were incubated for 48 hours in the absence or presence of 5 µM rottlerin in the presence or nominal absence of extracellular Ca2+. As illustrated in Fig. 5, removal of extracellular Ca2+ significantly blunted the effect of rottlerin on annexin-V-binding. In the absence of extracellular Ca2+, rottlerin did not significantly increase the percentage annexin-V-binding erythrocytes. Thus, rottlerin-induced cell membrane scrambling was in large part dependent on entry of extracellular Ca2+.

Fig. 5

Ca2+ sensitivity of rottlerin -induced phosphatidylserine exposure. A,B. Original histogram of annexin-V-binding of erythrocytes following exposure for 48 hours to Ringer solution without (grey area) and with (black line) rottlerin (5 µM) in the presence (A) and absence (B) of extracellular Ca2+. C. Arithmetic means ± SEM (n = 10) of annexin-V-binding of erythrocytes after a 48 hours treatment with Ringer solution without (white bars) or with (black bars) rottlerin (5 µM) in the presence (left bars, +Ca2+) and absence (right bars, -Ca2+) of Ca2+. ***(p<0.001) indicates significant difference from the absence of rottlerin, ###(p<0.001) indicates significant difference from the presence of Ca2+ (ANOVA).

Fig. 5

Ca2+ sensitivity of rottlerin -induced phosphatidylserine exposure. A,B. Original histogram of annexin-V-binding of erythrocytes following exposure for 48 hours to Ringer solution without (grey area) and with (black line) rottlerin (5 µM) in the presence (A) and absence (B) of extracellular Ca2+. C. Arithmetic means ± SEM (n = 10) of annexin-V-binding of erythrocytes after a 48 hours treatment with Ringer solution without (white bars) or with (black bars) rottlerin (5 µM) in the presence (left bars, +Ca2+) and absence (right bars, -Ca2+) of Ca2+. ***(p<0.001) indicates significant difference from the absence of rottlerin, ###(p<0.001) indicates significant difference from the presence of Ca2+ (ANOVA).

Close modal

Ca2+ sensitivity of cell membrane scrambling could be enhanced by ceramide. Ceramide abundance at the erythrocyte surface was thus quantified utilizing specific antibodies. As shown in Fig. 6, a 48 hours exposure to 5 µM rottlerin significantly increased the ceramide abundance.

Fig. 6

Effect of rottlerin on ceramide abundance. A. Original histograms of ceramide abundance at the erythrocyte surface following exposure for 48 hours to Ringer solution without (grey area) and with (black line) presence of 5 µM rottlerin. B. Arithmetic means ± SEM (n = 10) of ceramide abundance at the erythrocyte surface following incubation for 48 hours to Ringer solution without (white bar) or with (black bars) rottlerin (5 µM). ***(p<0.001) indicates significant difference from the absence of rottlerin (ANOVA).

Fig. 6

Effect of rottlerin on ceramide abundance. A. Original histograms of ceramide abundance at the erythrocyte surface following exposure for 48 hours to Ringer solution without (grey area) and with (black line) presence of 5 µM rottlerin. B. Arithmetic means ± SEM (n = 10) of ceramide abundance at the erythrocyte surface following incubation for 48 hours to Ringer solution without (white bar) or with (black bars) rottlerin (5 µM). ***(p<0.001) indicates significant difference from the absence of rottlerin (ANOVA).

Close modal

The present study discloses a novel effect of rottlerin, i.e. the stimulation of eryptosis, the suicidal erythrocyte death characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine translocation to the erythrocyte surface. The rottlerin concentrations required for stimulation of eryptosis were in the range of plasma concentrations (2.9 ± 0.4 µM) observed in mice following rottlerin treatment [55]. Similar concentrations have previously been shown to stimulate apoptosis of tumor cells [56].

The effect of rottlerin on cell membrane scrambling was in large part dependent on Ca2+ entry from the extracellular space, as it was markedly decreased by removal of extracellular Ca2+. Up to 5 µM rottlerin did not significantly increase the average Fluo3 fluorescence suggesting that rottlerin did not increase average cytosolic Ca2+ activity ([Ca2+]i). Only higher concentrations of rottlerin did increase average Fluo3 fluorescence. It thus appears that rottlerin enhances the Ca2+ sensitivity of cell membrane scrambling rather than increasing [Ca2+]i. Rottlerin treatment increased the abundance of ceramide, which is known to enhance the Ca2+ sensitivity of erythrocyte cell membrane scrambling [6].

Rottlerin treatment increased both, the percentage of shrunken and swollen cells. The cell shrinkage is presumably due to Ca2+ entry from the extracellular space with subsequent increase of [Ca2+]i, activation of Ca2+ sensitive K+ channels, K+ exit, cell membrane hyperpolarization, Cl- exit and thus cellular loss of KCl with water [6]. The mechanisms accounting for swelling of a subset of erythrocytes remained elusive.

The cell swelling presumably accounts for the increase of hemolysis following rottlerin treatment. Hemolysis leads to release of hemoglobin, which may pass the renal glomerular filter, precipitate in the acidic lumen of renal tubules, occlude nephrons and thus cause renal failure [57]. Eryptosis may accomplish clearance of defective erythrhocytes prior to hemolysis thus preventing release of hemoglobin. Eryptosis may further trigger clearance of erythrocytes, which are infected with the malaria pathogen Plasmodium. Along those lines, stimulation of eryptosis may favourably influence the clinical course of malaria [6].

The clearance of eryptotic erythrocytes from circulating blood may, on the other hand, result in anemia to the extent that the loss of erythrocytes outcasts the formation of new erythrocytes by erythropoiesis [6]. Phosphatidylserine exposing erythrocytes may further compromise microcirculation [7,58,59,60,61,62] by adherence to the vascular wall [63], stimulation of blood clotting and triggering of thrombosis [58,64,65].

In conclusion, rottlerin triggers eryptosis with cell shrinkage and cell membrane scrambling, an effect involving Ca2+ entry and ceramide.

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.

None.

1.
Maioli E, Torricelli C, Valacchi G: Rottlerin and curcumin: a comparative analysis. Ann N Y Acad Sci 2012;1259:65-76.
2.
Maioli E, Torricelli C, Valacchi G: Rottlerin and cancer: novel evidence and mechanisms. ScientificWorldJournal 2012;2012:350826.
3.
Maioli E, Valacchi G: Rottlerin: bases for a possible usage in psoriasis. Curr Drug Metab 2010;11:425-430.
4.
Soltoff SP: Rottlerin: an inappropriate and ineffective inhibitor of PKCdelta. Trends Pharmacol Sci 2007;28:453-458.
5.
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.
6.
Lang E, Lang F: Mechanisms and pathophysiological significance of eryptosis, the suicidal erythrocyte death. Semin Cell Dev Biol 2015;39:35-42.
7.
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.
8.
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.
9.
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.
10.
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.
11.
Alzoubi K, Egler J, Abed M, Lang F: Enhanced Eryptosis Following Auranofin Exposure. Cell Physiol Biochem 2015;37:1018-1028.
12.
Arnold M, Bissinger R, Lang F: Mitoxantrone-induced suicidal erythrocyte death. Cell Physiol Biochem 2014;34:1756-1767.
13.
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.
14.
Bissinger R, Barking S, Alzoubi K, Liu G, Liu G, Lang F: Stimulation of Suicidal Erythrocyte Death by the Antimalarial Drug Mefloquine. Cell Physiol Biochem 2015;36:1395-1405.
15.
Bissinger R, Bouguerra G, Stockinger K, Abbes S, Lang F: Triggering of Suicidal Erythrocyte Death by Topotecan. Cell Physiol Biochem 2015;37:1607-1618.
16.
Bouguerra G, Aljanadi O, Bissinger R, Abbes S, Lang F: Embelin-Induced Phosphatidylserine Translocation in the Erythrocyte Cell Membrane. Cell Physiol Biochem 2015;37:1629-1640.
17.
Bouguerra G, Bissinger R, Abbes S, Lang F: Zopolrestat Induced Suicidal Death of Human Erythrocytes. Cell Physiol Biochem 2015;37:1537-1546.
18.
Briglia M, Fazio A, Faggio C, Laufer S, Alzoubi K, Lang F: Triggering of Suicidal Erythrocyte Death by Ruxolitinib. Cell Physiol Biochem 2015;37:768-778.
19.
Briglia M, Fazio A, Signoretto E, Faggio C, Lang F: Edelfosine Induced Suicidal Death of Human Erythrocytes. Cell Physiol Biochem 2015;37:2221-2230.
20.
Calabro S, Alzoubi K, Faggio C, Laufer S, Lang F: Triggering of Suicidal Erythrocyte Death Following Boswellic Acid Exposure. Cell Physiol Biochem 2015;37:131-142.
21.
Egler J, Lang F: Licochalcone A Induced Suicidal Death of Human Erythrocytes. Cell Physiol Biochem 2015;37:2060-2070.
22.
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.
23.
Faggio C, Alzoubi K, Calabro S, Lang F: Stimulation of suicidal erythrocyte death by PRIMA-1. Cell Physiol Biochem 2015;35:529-540.
24.
Lupescu A, Bissinger R, Goebel T, Salker MS, Alzoubi K, Liu G, Chirigiu L, Mack AF, Qadri SM, Lang F: Enhanced suicidal erythrocyte death contributing to anemia in the elderly. Cell Physiol Biochem 2015;36:773-783.
25.
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.
26.
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.
27.
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.
28.
Officioso A, Alzoubi K, Manna C, Lang F: Clofazimine Induced Suicidal Death of Human Erythrocytes. Cell Physiol Biochem 2015;37:331-341.
29.
Peter T, Bissinger R, Enkel S, Alzoubi K, Oswald G, Lang F: Programmed erythrocyte death following in vitro Treosulfan treatment. Cell Physiol Biochem 2015;35:1372-1380.
30.
Stockinger K, Bissinger R, Bouguerra G, Abbes S, Lang F: Enhanced Eryptosis Following Exposure to Carnosic Acid. Cell Physiol Biochem 2015;37:1779-1791.
31.
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.
32.
Waibel S, Bissinger R, Bouguerra G, Abbes S, Lang F: Saquinavir Induced Suicidal Death of Human Erythrocytes. Cell Physiol Biochem 2015;37:1973-1982.
33.
Zierle J, Bissinger R, Egler J, Lang F: Lapatinib Induced Suicidal Death of Human Erythrocytes. Cell Physiol Biochem 2015;37:2275-2287.
34.
Bissinger R, Waibel S, Bouguerra G, Al Mamun Bhuyan A, Abbes S, Lang F: Enhanced Eryptosis Following Exposure to Lopinavir. Cell Physiol Biochem 2015;37:2486-2495.
35.
Briglia M, Calabro S, Signoretto E, Alzoubi K, Laufer S, Faggio C, Lang F: Fucoxanthin Induced Suicidal Death of Human Erythrocytes. Cell Physiol Biochem 2015;37:2464-2475.
36.
Briglia M, Fazio A, Faggio C, Lang F: Triggering of Suicidal Erythrocyte Death by Zosuquidar. Cell Physiol Biochem 2015;37:2355-2365.
37.
Fazio A, Briglia M, Faggio C, Alzoubi K, Lang F: Oxaliplatin Induced Suicidal Death of Human Erythrocytes. Cell Physiol Biochem 2015;37:2393-2404.
38.
Macczak A, Cyrkler M, Bukowska B, Michalowicz J: Eryptosis-inducing activity of bisphenol A and its analogs in human red blood cells (in vitro study). J Hazard Mater 2016;307:328-335.
39.
Officioso A, Alzoubi K, Lang F, Manna C: Hydroxytyrosol inhibits phosphatidylserine exposure and suicidal death induced by mercury in human erythrocytes: Possible involvement of the glutathione pathway. Food Chem Toxicol 2016;89:47-53.
40.
Officioso A, Manna C, Alzoubi K, Lang F: Bromfenvinphos induced suicidal death of human erythrocytes. Pestic Biochem Physiol 2016;126:58-63.
41.
Qadri SM, Donkor DA, Bhakta V, Eltringham-Smith LJ, Dwivedi DJ, Moore JC, Pepler L, Ivetic N, Nazi I, Fox-Robichaud AE, Liaw PC, Sheffield WP: Phosphatidylserine externalization and procoagulant activation of erythrocytes induced by Pseudomonas aeruginosa virulence factor pyocyanin. J Cell Mol Med 2016;10.1111/jcmm.12778
42.
Zierle J, Bissinger R, Bouguerra G, Abbes S, Lang F: Triggering of Suicidal Erythrocyte Death by Regorafenib. Cell Physiol Biochem 2016;38:160-172.
43.
Pagano M, Faggio C: The use of erythrocyte fragility to assess xenobiotic cytotoxicity. Cell Biochem Funct 2015;33:351-355.
44.
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.
45.
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.
46.
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.
47.
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.
48.
Polak-Jonkisz D, Purzyc L: Ca(2+) influx versus efflux during eryptosis in uremic erythrocytes. Blood Purif 2012;34:209-210; author reply 210.
49.
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.
50.
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.
51.
Nicolay J P, 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.
52.
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.
53.
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.
54.
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.
55.
Lu QY, Zhang L, Lugea A, Moro A, Edderkaoui M, Eibl G, Pandol SJ, Go VL: Determination of Rottlerin, a Natural Protein Kinases C Inhibitor, in Pancreatic Cancer Cells and Mouse Xenografts by RP-HPLC Method. J Chromatogr Sep Tech 2013;4:
56.
Ohno I, Eibl G, Odinokova I, Edderkaoui M, Damoiseaux RD, Yazbec M, Abrol R, Goddard WA, 3rd, Yokosuka O, Pandol SJ, Gukovskaya AS: Rottlerin stimulates apoptosis in pancreatic cancer cells through interactions with proteins of the Bcl-2 family. Am J Physiol Gastrointest Liver Physiol 2010;298:G63-73.
57.
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.
58.
Andrews DA, Low PS: Role of red blood cells in thrombosis. Curr Opin Hematol 1999;6:76-82.
59.
Closse C, Dachary-Prigent J, Boisseau MR: Phosphatidylserine-related adhesion of human erythrocytes to vascular endothelium. Br J Haematol 1999;107:300-302.
60.
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.
61.
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.
62.
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.
63.
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.
64.
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.
65.
Zwaal RF, Comfurius P, Bevers EM: Surface exposure of phosphatidylserine in pathological cells. Cell Mol Life Sci 2005;62:971-988.
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.