Background/Aims: The Polo-like kinase 1 (Plk1) inhibitor volasertib is used in the treatment of malignancy. Volasertib is partially effective by triggering suicidal death or apoptosis of tumor cells. Similar to apoptosis of nucleated cells, erythrocytes may enter suicidal cell death or eryptosis, which is characterized by cell membrane scrambling with phosphatidylserine translocation to the cell surface and by cell shrinkage. Stimulators of eryptosis include energy depletion, hyperosmotic shock, oxidative stress and excessive increase of cytosolic Ca2+ activity ([Ca2+]i). The present study explored, whether volasertib impacts on eryptosis. Methods: Human erythrocytes have been exposed to energy depletion (glucose withdrawal for 48 hours), hyperosmotic shock (addition of 550 mM sucrose for 6 hours), oxidative stress (addition of 0.3 mM tert-butylhydroperoxide [tBOOH] for 50 min) or Ca2+ ionophore ionomycin (1 µM for 60 min) in absence and presence of volasertib (0.5-1.5 µg/ml) and flow cytometry was employed to quantify phosphatidylserine exposure at the cell surface from annexin-V-binding, cell volume from forward scatter, [Ca2+]i from Fluo3 fluorescence, reactive oxygen species from 2’,7’-dichlorodihydrofluorescein diacetate (DCFDA) fluorescence and ceramide abundance utilizing antibodies. For comparison, annexin-V-binding and forward scatter were determined following a 48 hours exposure of human leukemic K562 cells in RPMI-1640 medium to volasertib. Results: Treatment with volasertib alone did not significantly modify annexin-V-binding or forward scatter in mature erythrocytes. Energy depletion, hyperosmotic shock, oxidative stress and ionomycin, all markedly and significantly increased the percentage of annexin-V-binding erythrocytes, and decreased the forward scatter. Volasertib significantly blunted the effect of energy depletion and hyperosmotic shock, but not of oxidative stress and ionomycin on annexin-V-binding. Volasertib did not significantly influence the effect of any maneuver on forward scatter. In K562 cells, volasertib enhanced annexin-V-binding and decreased the forward scatter. Conclusions: Volasertib is a novel inhibitor of erythrocyte cell membrane scrambling following energy depletion and hyperosmotic shock, effects contrasting the stimulation of K562 cell apoptosis.

The Polo-like kinase 1 (Plk1) inhibitor volasertib (BI6727) [1-10] has been successfully used in preclinical and clinical studies for the treatment of several malignancies [1, 2, 7-14] including acute myeloid leukaemia (AML) [2-9, 11, 15-20]. Volasertib is in part effective by triggering suicidal death or apoptosis of tumor cells [1, 7]. Reported side effects of treatment with volasertib or combinations of volasertib with other cytostatic drugs include anemia [12, 13, 21].

At least in theory, the anemia following volasertib treatment could result from stimulation of suicidal erythrocyte death or eryptosis [22, 23], which is characterized by cell membrane scrambling with phosphatidylserine translocation to the cell surface [22]. Another hallmark of eryptosis is cell shrinkage [24]. Eryptosis could be stimulated by cell stress, e.g. by energy depletion [22], hyperosmotic shock [22], and oxidative stress [22]. Signaling mechanisms triggering eryptosis include increase of cytosolic Ca2+ activity ([Ca2+]i) [22], ceramide [25], caspases [22, 26, 27], G-protein Gαi2 [28], casein kinase 1α [22], Janus-activated kinase JAK3 [22], protein kinase C [22], and p38 kinase [22]. Signaling mechanisms inhibiting eryptosis include the kinases AMPK [22], cGMP-dependent protein kinase [22], MSK1/2 [29], PAK2 [22] and sorafenib/sunitinib sensitive kinases [22]. Eryptosis is stimulated by a myriad of xenobiotics [22, 29-83] and enhanced eryptosis is observed in a variety of clinical conditions including iron deficiency [22], dehydration [84], hyperphosphatemia [74], vitamin D excess [38], chronic kidney disease (CKD) [85-89] hemolytic-uremic syndrome [90], diabetes [91], hepatic failure [92, 93], malignancy [94, 95], arteritis [96], sepsis [97], sickle-cell disease [22], beta-thalassemia [22], Hb-C and G6PD-deficiency [22], Wilsons disease [98], as well as advanced age [99]. Eryptosis further increases following erythrocyte storage for transfusion [100].

The present study explored, whether volasertib impacts on eryptosis. To this end, human erythrocytes from healthy volunteers were exposed to volasertib in the absence and presence of energy depletion, hyperosmotic shock, oxidative stress, as well as Ca2+ loading. Phosphatidylserine surface abundance and cell volume were determined by flow cytometry. For comparison, the effect of volasertib was tested on annexin-V-binding and forward scatter of human leukemic K562 cells.

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 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 for 48 hours to glucose depleted Ringer solution, for 6 hours to hypertonic Ringer (addition of 550 mM sucrose, Sigma Aldrich, Hamburg, Germany), for 50 minutes to the oxidant tert-butyl-hydroperoxide (0.3 mM, Sigma Aldrich, Hamburg, Germany), or for 60 minutes to Ca2+ ionophore ionomycin (1 µM, Merck Millipore, Darmstadt, Germany), each in the absence and presence of volasertib (MedChem Express, Princeton, USA).

K562 cell culture and treatments

The K562 human leukaemia cells (Sigma Aldrich, Hamburg, Germany) were cultured using RPMI-1640 medium (Biochrom GmbH, Berlin, Germany) supplemented with 2.0 g/l NaHCO3, without L-glutamine. The cells were cultured at 37°C in a moistened incubator with 5% CO2. Where indicated, volasertib (0.5 – 1.5 μg/ ml) was added to the cell culture medium.

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 volasertib 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, 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. Afterwards, the geomean of the Ca2+ dependent fluorescence was determined.

Reactive oxygen species (ROS)

Oxidative stress was determined utilizing 2’,7’-dichlorodihydrofluorescein diacetate (DCFDA). After incubation, a 150 µl suspension of erythrocytes was washed in Ringer solution and stained with DCFDA (Sigma, Aldrich, Germany) in Ringer solution containing 10 µM DCFDA. Erythrocytes were incubated at 37°C for 30 min in the dark and washed two times 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). Subsequently, the geomean of the DCFDA dependent fluorescence was determined.

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 analysed by flow cytometric analysis with an excitation wavelength of 488 nm and an emission wavelength of 530 nm. Finally, the geomean of the ceramide-dependent fluorescence was determined.

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. 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 volasertib stimulates or interferes with eryptosis, the suicidal erythrocyte death. The two hallmarks of eryptosis, i.e. cell membrane scrambling and cell shrinkage were determined by flow cytometry. Phosphatidylserine exposing erythrocytes were identified utilizing annexin-V-binding to phosphatidylserine, cell shrinkage was quantified utilizing forward scatter.

The percentage of human erythrocytes binding annexin-V after 48 hours incubation in standard, glucose containing, Ringer was low (2.4 ± 0.3%, n = 14) and not significantly modified by the presence of 1.5 µg/ml volasertib (2.5 ± 0.4%, n = 14). Thus, in standard glucose containing Ringer solution, volasertib did not appreciably modify erythrocyte cell membrane scrambling. As illustrated in Fig. 1, glucose removal was followed by a marked increase of the percentage of annexin-V-binding erythrocytes. The addition of volasertib (0.5-1.5 µg/ml) blunted the increase of the percentage of annexin-V-binding erythrocytes following glucose deprivation, an effect reaching statistical significance at 1 and 1.5 µg/ml volasertib. Nevertheless, even in the presence of volasertib, energy depletion significantly increased the percentage of phosphatidylserine exposing erythrocytes (Fig. 1). Volasertib thus partially blunted cell membrane scrambling following energy depletion.

Fig. 1.

Volasertib sensitivity of phosphatidylserine exposure following energy depletion. A. Original histograms of annexin-V-binding of erythrocytes following exposure for 48 hours to glucose containing Ringer solution (grey areas), Ringer solution without glucose (green areas) and Ringer solution without glucose and presence of volasertib (1.5 µg/ml) (blue areas). B. Arithmetic means ± SEM (n = 14) of the percentage of annexin-V-binding erythrocytes after a 48 hours treatment with Ringer solution with glucose (grey bar) or without glucose in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in absence of glucose and presence of DMSO. ***(p<0.001) indicates significant difference from the presence of glucose, ##(p<0.01), ###(p<0.001) indicates significant difference from the absence of volasertib (ANOVA).

Fig. 1.

Volasertib sensitivity of phosphatidylserine exposure following energy depletion. A. Original histograms of annexin-V-binding of erythrocytes following exposure for 48 hours to glucose containing Ringer solution (grey areas), Ringer solution without glucose (green areas) and Ringer solution without glucose and presence of volasertib (1.5 µg/ml) (blue areas). B. Arithmetic means ± SEM (n = 14) of the percentage of annexin-V-binding erythrocytes after a 48 hours treatment with Ringer solution with glucose (grey bar) or without glucose in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in absence of glucose and presence of DMSO. ***(p<0.001) indicates significant difference from the presence of glucose, ##(p<0.01), ###(p<0.001) indicates significant difference from the absence of volasertib (ANOVA).

Close modal

Erythrocyte forward scatter was, following a 48 hours exposure to Ringer solution in the presence of glucose, again similar in the absence (517 ± 6%, n = 14) and presence (522 ± 5.4%, n = 14) of volasertib (1.5 µg/ml). Accordingly, in standard glucose containing Ringer solution, volasertib did not appreciably modify erythrocyte volume. As illustrated in Fig. 2, a 48 hours exposure to glucose-depleted Ringer solution was followed by a marked decrease of forward scatter, reflecting erythrocyte shrinkage. The decline of erythrocyte forward scatter was, following a 48 hours exposure to Ringer solution in the absence of glucose, virtually identical in the absence and presence of volasertib (0.5–1.5 µg/ml). Accordingly, neither in energy repleted nor in energy depleted erythrocytes did volasertib significantly modify erythrocyte volume.

Fig. 2.

Volasertib sensitivity of erythrocyte shrinkage following energy depletion. A. Original histograms of forward scatter of erythrocytes following exposure for 48 hours to glucose containing Ringer solution (grey areas), Ringer solution without glucose (green areas) and Ringer solution without glucose and presence of volasertib (1.5 µg/ml) (blue areas). B. Arithmetic means ± SEM (n = 14) of the erythrocyte forward scatter after a 48 hours treatment with Ringer solution with glucose (grey bar) or without glucose in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in absence of glucose and presence of DMSO. ***(p<0.001) indicates significant difference from the presence of glucose (ANOVA).

Fig. 2.

Volasertib sensitivity of erythrocyte shrinkage following energy depletion. A. Original histograms of forward scatter of erythrocytes following exposure for 48 hours to glucose containing Ringer solution (grey areas), Ringer solution without glucose (green areas) and Ringer solution without glucose and presence of volasertib (1.5 µg/ml) (blue areas). B. Arithmetic means ± SEM (n = 14) of the erythrocyte forward scatter after a 48 hours treatment with Ringer solution with glucose (grey bar) or without glucose in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in absence of glucose and presence of DMSO. ***(p<0.001) indicates significant difference from the presence of glucose (ANOVA).

Close modal

As volasertib did not, in contrast to previous observations in nucleated cells, significantly stimulate eryptosis, further experiments were performed exploring whether volasertib stimulated apoptosis of K562 cells. As shown in Fig. 3, a 48 hours exposure to volasertib increased the percentage of phosphatidylserine exposing K562 cells at each of the concentrations applied (0.5–1.5 µg/ml) and as illustrated in Fig. 4, volasertib triggered cell shrinkage, an effect reaching statistical significance at 0.5 µg/ml and 1.5 µg/ml volasertib.

Fig. 3.

Effect of Volasertib on phosphatidylserine exposure of K562 cells. A. Original histograms of annexin-V-binding of K562 cells following exposure for 48 hours to RPMI 1640 medium without (grey area) and with presence of volasertib (1.5 µg/ml) (blue areas). B. Arithmetic means ± SEM (n =10) of the percentage of annexin-V-binding K562 cells following incubation for 48 hours to RPMI 1640 medium without (grey bar) or with (blue bars) Volasertib (0.5 – 1.5 µg/ml). For comparison, the effect of the solvent DMSO is shown (Striped bar). ***(p<0.001) indicates significant difference from the absence of volasertib (ANOVA).

Fig. 3.

Effect of Volasertib on phosphatidylserine exposure of K562 cells. A. Original histograms of annexin-V-binding of K562 cells following exposure for 48 hours to RPMI 1640 medium without (grey area) and with presence of volasertib (1.5 µg/ml) (blue areas). B. Arithmetic means ± SEM (n =10) of the percentage of annexin-V-binding K562 cells following incubation for 48 hours to RPMI 1640 medium without (grey bar) or with (blue bars) Volasertib (0.5 – 1.5 µg/ml). For comparison, the effect of the solvent DMSO is shown (Striped bar). ***(p<0.001) indicates significant difference from the absence of volasertib (ANOVA).

Close modal
Fig. 4.

Effect of Volasertib on K562 cell forward scatter (FSC). A. Original histograms of forward scatter reflecting cell volume of K562 cells following exposure for 48 hours to RPMI 1640 medium without (grey area) and with presence of volasertib (1.5 µg/ml) (blue areas). B. Arithmetic means ± SEM (n =10) of K562 cell forward scatter (FSC) following incubation for 48 hours to RPMI 1640 medium without (grey bar) or with (blue bars) volasertib (0.5 – 1.5 µg/ml). For comparison, the effect of the solvent DMSO is shown (Striped bar). *(p<0.05) indicates significant difference from the absence of volasertib (ANOVA).

Fig. 4.

Effect of Volasertib on K562 cell forward scatter (FSC). A. Original histograms of forward scatter reflecting cell volume of K562 cells following exposure for 48 hours to RPMI 1640 medium without (grey area) and with presence of volasertib (1.5 µg/ml) (blue areas). B. Arithmetic means ± SEM (n =10) of K562 cell forward scatter (FSC) following incubation for 48 hours to RPMI 1640 medium without (grey bar) or with (blue bars) volasertib (0.5 – 1.5 µg/ml). For comparison, the effect of the solvent DMSO is shown (Striped bar). *(p<0.05) indicates significant difference from the absence of volasertib (ANOVA).

Close modal

Further experiments addressed the influence of volasertib on cytosolic Ca2+ activity ([Ca2+]i), which was estimated from Fluo3 fluorescence. As illustrated in Fig. 5, a 48 hours exposure to glucose depleted Ringer solution was followed by a marked significant increase of [Ca2+]i both, in the absence and presence of volasertib. The addition of volasertib (0.5-1.5 µg/ml) did not significantly modify [Ca2+]i in erythrocytes without or with energy depletion.

Fig. 5.

Volasertib sensitivity of cytosolic Ca2+ activity following energy depletion. A. Original histograms of Fluo3 fluorescence reflecting cytosolic Ca2+ activity of erythrocytes following exposure for 48 hours to glucose containing Ringer solution (grey areas), Ringer solution without glucose (green areas) and Ringer solution without glucose and presence of volasertib (1.5 µg/ml) (blue areas). B. Arithmetic means ± SEM (n = 14) of erythrocyte Fluo3 fluorescence after a 48 hours treatment with Ringer solution with glucose (grey bar) or without glucose in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in absence of glucose and presence of DMSO. ***(p<0.001) indicates significant difference from the presence of glucose.

Fig. 5.

Volasertib sensitivity of cytosolic Ca2+ activity following energy depletion. A. Original histograms of Fluo3 fluorescence reflecting cytosolic Ca2+ activity of erythrocytes following exposure for 48 hours to glucose containing Ringer solution (grey areas), Ringer solution without glucose (green areas) and Ringer solution without glucose and presence of volasertib (1.5 µg/ml) (blue areas). B. Arithmetic means ± SEM (n = 14) of erythrocyte Fluo3 fluorescence after a 48 hours treatment with Ringer solution with glucose (grey bar) or without glucose in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in absence of glucose and presence of DMSO. ***(p<0.001) indicates significant difference from the presence of glucose.

Close modal

Reactive oxygen species (ROS) was determined utilizing 2’,7’-dichlorodihydrofluorescein diacetate (DCFDA). After 48 hours incubation in standard glucose containing Ringer, the ROS was similar in the absence (19.07 ± 0.64, n = 5) and presence (18.63 ± 0.46, n = 5) of 1.5 µg/ml volasertib. As illustrated in Fig. 6, removal of glucose was followed by a marked significant increase of DCFDA fluorescence in the absence and presence of volasertib (1.5 µg/ml). The addition of volasertib did not significantly modify DCFDA fluorescence following glucose depletion.

Fig. 6.

Volasertib sensitivity of reactive oxygen species following energy depletion. A. Original histograms of DCFDA fluorescence reflecting reactive oxygen species in erythrocytes following exposure for 48 hours to glucose containing Ringer solution (grey areas), Ringer solution without glucose (green areas) and Ringer solution without glucose and presence of volasertib (1.5 µg/ml) (blue areas). B. Arithmetic means ± SEM (n = 5) of DCFDA fluorescence in erythrocytes following a 48 hours treatment with Ringer solution with glucose (grey bar) or without glucose in the absence (green bar) and presence (blue bars) of volasertib (1.5 µg/ml). **(p<0.01) indicates significant difference from the presence of glucose.

Fig. 6.

Volasertib sensitivity of reactive oxygen species following energy depletion. A. Original histograms of DCFDA fluorescence reflecting reactive oxygen species in erythrocytes following exposure for 48 hours to glucose containing Ringer solution (grey areas), Ringer solution without glucose (green areas) and Ringer solution without glucose and presence of volasertib (1.5 µg/ml) (blue areas). B. Arithmetic means ± SEM (n = 5) of DCFDA fluorescence in erythrocytes following a 48 hours treatment with Ringer solution with glucose (grey bar) or without glucose in the absence (green bar) and presence (blue bars) of volasertib (1.5 µg/ml). **(p<0.01) indicates significant difference from the presence of glucose.

Close modal

The abundance of ceramide at the erythrocyte surface was quantified utilizing specific antibodies. The abundance of ceramide was similar following a 48 hours incubation in standard glucose containing Ringer (10.11 ± 0.24, n = 5) and in glucose depleted Ringer without (10.1 ± 0.2, n = 5) and with (9.8 ± 0.2, n = 5) addition of volasertib.

A next series of experiments addressed the effect of hyperosmotic shock (addition of 550 mM sucrose) for 6 hours. As illustrated in Fig. 7, hyperosmotic shock was followed by a marked increase of the percentage of annexin-V-binding erythrocytes. The addition of volasertib (0.5-1.5 µg/ml) blunted the increase of the percentage of annexin-V-binding erythrocytes following hyperosmotic shock, an effect reaching statistical significance at 1 and 1.5 µg/ml volasertib. Again, even in the presence of volasertib, hyperosmotic shock significantly increased the percentage of phosphatidylserine exposing erythrocytes (Fig. 7). Volasertib thus partially blunted cell membrane scrambling following hyperosmotic shock.

Fig. 7.

Volasertib sensitivity of phosphatidylserine exposure following hyperosmotic shock. A. Original histograms of annexin-V-binding of erythrocytes following exposure for 6 hours to isotonic Ringer solution (grey area), or hypertonic Ringer solution (550 mM sucrose added) in absence (green areas) or presence (blue areas) of volasertib (1.5 µg/ml). B. Arithmetic means ± SEM (n = 10) of the percentage of annexin-V-binding erythrocytes after a 6 hours treatment with isotonic Ringer solution (grey bar), or hypertonic Ringer solution (550 mM sucrose added) in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in presence of tert-butylhydroperoxide and DMSO. ***(p<0.001) indicates significant difference from isotonic Ringer, ##(p<0.01) indicates significant difference from the absence of volasertib (ANOVA).

Fig. 7.

Volasertib sensitivity of phosphatidylserine exposure following hyperosmotic shock. A. Original histograms of annexin-V-binding of erythrocytes following exposure for 6 hours to isotonic Ringer solution (grey area), or hypertonic Ringer solution (550 mM sucrose added) in absence (green areas) or presence (blue areas) of volasertib (1.5 µg/ml). B. Arithmetic means ± SEM (n = 10) of the percentage of annexin-V-binding erythrocytes after a 6 hours treatment with isotonic Ringer solution (grey bar), or hypertonic Ringer solution (550 mM sucrose added) in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in presence of tert-butylhydroperoxide and DMSO. ***(p<0.001) indicates significant difference from isotonic Ringer, ##(p<0.01) indicates significant difference from the absence of volasertib (ANOVA).

Close modal

Hyperosmotic shock was further followed by a marked decrease of forward scatter, reflecting erythrocyte shrinkage. The erythrocyte shrinkage following hyperosmotic shock was virtually identical in the absence and presence of volasertib (0.5-1.5 µg/ml) (Fig. 8). Accordingly, volasertib did not significantly interfere with the effect of hyperosmotic shock on erythrocyte volume.

Fig. 8.

Volasertib sensitivity of erythrocyte shrinkage following hyperosmotic shock. A. Original histograms of erythrocyte forward scatter following exposure for 6 hours to isotonic Ringer solution (grey area), or hypertonic Ringer solution (550 mM sucrose added) in absence (green areas) or presence (blue areas) of volasertib (1.5 µg/ml). B. Arithmetic means ± SEM (n = 10) of erythrocyte forward scatter after a 6 hours treatment with isotonic Ringer solution (grey bar), or hypertonic Ringer solution (550 mM sucrose added) in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in presence of tert-butylhydroperoxide and DMSO. ***(p<0.001) indicates significant difference from isotonic Ringer (ANOVA).

Fig. 8.

Volasertib sensitivity of erythrocyte shrinkage following hyperosmotic shock. A. Original histograms of erythrocyte forward scatter following exposure for 6 hours to isotonic Ringer solution (grey area), or hypertonic Ringer solution (550 mM sucrose added) in absence (green areas) or presence (blue areas) of volasertib (1.5 µg/ml). B. Arithmetic means ± SEM (n = 10) of erythrocyte forward scatter after a 6 hours treatment with isotonic Ringer solution (grey bar), or hypertonic Ringer solution (550 mM sucrose added) in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in presence of tert-butylhydroperoxide and DMSO. ***(p<0.001) indicates significant difference from isotonic Ringer (ANOVA).

Close modal

A next series of experiments addressed the effect of oxidative stress due to addition of 0.3 mM tert-butylhydroperoxide (tBOOH) for 50 minutes. As illustrated in Fig. 9, oxidative stress was followed by a marked increase of the percentage of annexin-V-binding erythrocytes. The addition of volasertib (0.5-1.5 µg/ml) did not significantly interfere with the increase of the percentage of annexin-V-binding erythrocytes following oxidative stress. Instead, oxidative stress significantly increased the percentage of phosphatidylserine exposing erythrocytes both, in the absence and presence of volasertib (Fig. 9).

Fig. 9.

Volasertib sensitivity of phosphatidylserine exposure following oxidative stress. A. Original histograms of annexin-V-binding of erythrocytes following exposure for 50 min to Ringer solution without tert-butylhydroperoxide (grey area), or Ringer solution with tert-butylhydroperoxide (0.3 mM) and absence (green areas) or presence (blue areas) of volasertib (1.5 µg/ml). B. Arithmetic means ± SEM (n = 15) of the percentage of annexin-V-binding erythrocytes after a 50 min treatment with Ringer solution without tert-butylhydroperoxide (grey bar) or with tert-butylhydroperoxide (0.3 mM) in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in presence of tert-butylhydroperoxide and DMSO. ***(p<0.001) indicates significant difference from the absence of tert-butylhydroperoxide (ANOVA).

Fig. 9.

Volasertib sensitivity of phosphatidylserine exposure following oxidative stress. A. Original histograms of annexin-V-binding of erythrocytes following exposure for 50 min to Ringer solution without tert-butylhydroperoxide (grey area), or Ringer solution with tert-butylhydroperoxide (0.3 mM) and absence (green areas) or presence (blue areas) of volasertib (1.5 µg/ml). B. Arithmetic means ± SEM (n = 15) of the percentage of annexin-V-binding erythrocytes after a 50 min treatment with Ringer solution without tert-butylhydroperoxide (grey bar) or with tert-butylhydroperoxide (0.3 mM) in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in presence of tert-butylhydroperoxide and DMSO. ***(p<0.001) indicates significant difference from the absence of tert-butylhydroperoxide (ANOVA).

Close modal

Oxidative stress was further followed by a marked decrease of forward scatter, reflecting erythrocyte shrinkage. The erythrocyte shrinkage following oxidative stress was virtually identical in the absence and presence of volasertib (0.5-1.5 µg/ml). Accordingly, volasertib did not significantly interfere with the effect of oxidative stress on erythrocyte volume (Fig. 10).

Fig. 10.

Volasertib sensitivity of erythrocyte shrinkage following oxidative stress. A. Original histograms of erythrocyte forward scatter following exposure for 50 min to Ringer solution without tert-butylhydroperoxide (grey area), or Ringer solution with tert-butylhydroperoxide (0.3 mM) and absence (green areas) or presence (blue areas) of volasertib (1.5 µg/ml). B. Arithmetic means ± SEM (n = 15) of erythrocyte forward scatter after a 50 min treatment with Ringer solution without tert-butylhydroperoxide (grey bar) or with tert-butylhydroperoxide (0.3 mM) in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in presence of tert-butylhydroperoxide and DMSO. ***(p<0.001) indicates significant difference from absence of tert-butylhydroperoxide (ANOVA).

Fig. 10.

Volasertib sensitivity of erythrocyte shrinkage following oxidative stress. A. Original histograms of erythrocyte forward scatter following exposure for 50 min to Ringer solution without tert-butylhydroperoxide (grey area), or Ringer solution with tert-butylhydroperoxide (0.3 mM) and absence (green areas) or presence (blue areas) of volasertib (1.5 µg/ml). B. Arithmetic means ± SEM (n = 15) of erythrocyte forward scatter after a 50 min treatment with Ringer solution without tert-butylhydroperoxide (grey bar) or with tert-butylhydroperoxide (0.3 mM) in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in presence of tert-butylhydroperoxide and DMSO. ***(p<0.001) indicates significant difference from absence of tert-butylhydroperoxide (ANOVA).

Close modal

A next series of experiments explored the effect of Ca2+ entry, which was achieved by a 60 min exposure of erythrocytes to Ca2+ ionophore ionomycn (1 µM). As illustrated in Fig. 11, exposure of the erythrocytes for 60 minutes to 1 µM ionomycin was followed by a sharp increase of the percentage of annexin-V-binding erythrocytes. The effect tended to be slightly blunted by volasertib, an effect, however, not reaching statistical significance. Instead, even in the presence of volasertib, ionomycin significantly increased the percentage of phosphatidylserine exposing erythrocytes (Fig. 11).

Fig. 11.

Volasertib sensitivity of phosphatidylserine exposure following Ca2+ loading. A. Original histograms of annexin-V-binding of erythrocytes following exposure for 60 min to Ringer solution without Ca2+ ionophore ionomycin (grey area), or Ringer solution with ionomycin (1 µM) and absence (green areas) or presence (blue areas) of volasertib (1.5 µg/ml). B. Arithmetic means ± SEM (n = 10) of the percentage of annexin-V-binding erythrocytes after a 60 min treatment with Ringer solution without ionomycin (grey bar) or with ionomycin (1 µM) in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in presence of ionomycin and DMSO. ***(p<0.001) indicates significant difference from the absence of ionomycin (ANOVA).

Fig. 11.

Volasertib sensitivity of phosphatidylserine exposure following Ca2+ loading. A. Original histograms of annexin-V-binding of erythrocytes following exposure for 60 min to Ringer solution without Ca2+ ionophore ionomycin (grey area), or Ringer solution with ionomycin (1 µM) and absence (green areas) or presence (blue areas) of volasertib (1.5 µg/ml). B. Arithmetic means ± SEM (n = 10) of the percentage of annexin-V-binding erythrocytes after a 60 min treatment with Ringer solution without ionomycin (grey bar) or with ionomycin (1 µM) in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in presence of ionomycin and DMSO. ***(p<0.001) indicates significant difference from the absence of ionomycin (ANOVA).

Close modal

Exposure of the erythrocytes for 60 minutes to 1 µM ionomycin was followed by a sharp decrease of forward scatter (Fig. 12). The cell shrinkage was not significantly modified by volasertib (Fig. 12).

Fig. 12.

Volasertib sensitivity of erythrocyte shrinkage following Ca2+ loading. A. Original histograms of erythrocyte forward scatter following exposure for 60 min to Ringer solution without ionomycin (grey area), or Ringer solution with ionomycin (1 µM) and absence (green areas) or presence (blue areas) of volasertib (1.5 µg/ml). B. Arithmetic means ± SEM (n = 10) of erythrocyte forward scatter after a 60 min treatment with Ringer solution without ionomycin (grey bar) or with ionomycin (1 µM) in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in presence of ionomycin and DMSO. ***(p<0.001) indicates significant difference from the absence of ionomycin (ANOVA).

Fig. 12.

Volasertib sensitivity of erythrocyte shrinkage following Ca2+ loading. A. Original histograms of erythrocyte forward scatter following exposure for 60 min to Ringer solution without ionomycin (grey area), or Ringer solution with ionomycin (1 µM) and absence (green areas) or presence (blue areas) of volasertib (1.5 µg/ml). B. Arithmetic means ± SEM (n = 10) of erythrocyte forward scatter after a 60 min treatment with Ringer solution without ionomycin (grey bar) or with ionomycin (1 µM) in the absence (green bar) and presence (blue bars) of volasertib (0.5-1.5 µg/ml). Striped bar is in presence of ionomycin and DMSO. ***(p<0.001) indicates significant difference from the absence of ionomycin (ANOVA).

Close modal

The present observations reveal a novel, unexpected effect of volasertib on erythrocyte cell membrane scrambling, which is a hallmark of eryptosis, the suicidal erythrocyte death. Treatment with volasertib alone did not significantly modify annexin-V-binding or forward scatter, but volasertib significantly blunted the stimulating effect of energy depletion and hyperosmotic shock on cell membrane scrambling. The volasertib concentrations required for this effect were in the range of concentrations (up to 1.45 µg/ml) encountered in the plasma of patients [21]. Volasertib did not significantly interfere with the stimulating effect of oxidative stress and ionomycin on cell membrane scrambling and did not significantly interfere with the cell shrinkage following energy depletion, hyperosmotic shock, oxidative stress and ionomycin.

The failure of volasertib to interfere with cell shrinkage could be explained by its inability to interfere with increase of cytosolic Ca2+ activity ([Ca2+]i). An increase of [Ca2+]i activates Ca2+ sensitive K+ channels leading to K+ exit, cell membrane hyperpolarization, Cl- exit and thus cellular loss of KCl with water [22].

The inhibitory effect of volasertib on cell membrane scrambling following energy depletion and hyperosmotic shock contrasts the stimulating effect of volasertib on apoptosis of nucleated cells, as shown here for K562 cells. While apoptosis and eryptosis share the eventual stimulation of cell membrane scrambling, the signaling triggering cell membrane scrambling differs considerably [22, 101]. Along those lines, apoptosis following inhibition of polo-like kinases may involve mitochondria [102], which are absent in mature erythrocytes. In view of the stimulation of tumor cell death and the simultaneous protection against eryptosis of energy or shrunken erythrocytes, volasertib may be particularly useful in the treatment of malignancies associated with anemia [103].

The present study did not uncover the signaling involved in the effect of volasertib on cell membrane scrambling following energy depletion and hyperosmotic shock. As shown for energy depletion, volasertib did not significantly interfere with the increase of cytosolic Ca2+ activity and did not modify ceramide formation. In theory, eryptosis could involve Pololike kinase 1 (Plk1), wich is specifically inhibited by volasertib [1-10]. However, the effects of volasertib are not restricted to the polo-like kinase 1 (Plk1) isoform but the substance inhibits, albeit to a lesser extent, the isoforms Plk2 and Plk3 [8]. Moreover, other effects unrelated to Polo-like kinases cannot be ruled out. Further experimental effort is required to identify the mechanisms accomplishing the inhibitory effect of volasertib on eryptosis following energy depletion and hyperosmotic shock. Moreover, additional analysis may be required to understand why volasertib is effective following energy depletion and hyperosmotic shock, but not following oxidative stress and ionomycin. The discrepancy points to volasertib sensitive signaling triggering cell membrane scrambling, which is shared by energy depletion and hyperosmotic shock but not by oxidative stress and Ca2+ overload.

Whatever mechanism involved, volasertib is inhibiting rather than stimulating erythrocyte cell membrane scrambling. The observed enhanced incidence of anemia in volasertib-combination treatments may be attributed to the effects of the combined drug. Alternatively, volasertib triggers apoptosis of progenitor cells thus compromizing formation of new erythrocytes. If so, administration of the drug should decrease reticulocyte numbers in circulating blood. To the best of our knowledge, nothing is known about an effect of volasertib on reticulocytes. In any case, inhibition of eryptosis counteracts anemia. Phosphatidylserine exposing erythrocytes are rapidly cleared from circulating blood and eryptosis leads to anemia as soon as the loss of eryptotic erythrocytes outcasts the formation of new erythrocytes by erythropoiesis [22].

As volasertib did not interfere with cell shrinkage, it presumably does not foster hemolysis. Hemoglobin released following hemolysis may otherwise pass the renal glomerular filter, precipitate in the acidic lumen of renal tubules, occlude nephrons and may thus lead to renal failure [104].

As phosphatidylserine exposing erythrocytes further adhere to the vascular wall [105], stimulate blood clotting/thrombosis [106-108], and interfere with microcirculation [25, 106, 109-112], the inhibition of eryptosis may decrease cardiovascular events in tumor patients.

In conclusion, volasertib inhibits erythrocyte cell membrane scrambling following energy depletion and hyperosmotic shock and thus rather counteracts suicidal erythrocyte death and development of anemia.

The authors acknowledge the meticulous preparation of the manuscript by Lejla Subasic. 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.
Gutteridge RE, Ndiaye MA, Liu X, Ahmad N: Plk1 Inhibitors in Cancer Therapy: From Laboratory to Clinics. Mol Cancer Ther 2016; 15: 1427-1435.
2.
Van den Bossche J, Lardon F, Deschoolmeester V, De Pauw I, Vermorken JB, Specenier P, Pauwels P, Peeters M, Wouters A: Spotlight on Volasertib: Preclinical and Clinical Evaluation of a Promising Plk1 Inhibitor. Med Res Rev 2016; 36: 749-786.
3.
Talati C, Griffiths EA, Wetzler M, Wang ES: Polo-like kinase inhibitors in hematologic malignancies. Crit Rev Oncol Hematol 2016; 98: 200-210.
4.
Prochazkova I, Vojtesek B: [Polo-like Kinase 1 as a Target for Anti-tumor Therapy]. Klin Onkol 2015; 28 Suppl 2: 2s32-39.
5.
Liu X: Targeting Polo-Like Kinases: A Promising Therapeutic Approach for Cancer Treatment. Transl Oncol 2015; 8: 185-195.
6.
Helmke C, Becker S, Strebhardt K: The role of Plk3 in oncogenesis. Oncogene 2016; 35: 135-147.
7.
Brandwein JM: Targeting polo-like kinase 1 in acute myeloid leukemia. Ther Adv Hematol 2015; 6: 80-87.
8.
Janning M, Fiedler W: Volasertib for the treatment of acute myeloid leukemia: a review of preclinical and clinical development. Future Oncol 2014; 10: 1157-1165.
9.
Yim H: Current clinical trials with polo-like kinase 1 inhibitors in solid tumors. Anticancer Drugs 2013; 24: 999-1006.
10.
Schoffski P: Polo-like kinase (PLK) inhibitors in preclinical and early clinical development in oncology. Oncologist 2009; 14: 559-570.
11.
Gjertsen BT, Schoffski P: Discovery and development of the Polo-like kinase inhibitor volasertib in cancer therapy. Leukemia 2015; 29: 11-19.
12.
Awada A, Dumez H, Aftimos PG, Costermans J, Bartholomeus S, Forceville K, Berghmans T, Meeus MA, Cescutti J, Munzert G, Pilz K, Liu D, Schoffski P: Phase I trial of volasertib, a Polo-like kinase inhibitor, plus platinum agents in solid tumors: safety, pharmacokinetics and activity. Invest New Drugs 2015; 33: 611-620.
13.
Stadler WM, Vaughn DJ, Sonpavde G, Vogelzang NJ, Tagawa ST, Petrylak DP, Rosen P, Lin CC, Mahoney J, Modi S, Lee P, Ernstoff MS, Su WC, Spira A, Pilz K, Vinisko R, Schloss C, Fritsch H, Zhao C, Carducci MA: An open-label, single-arm, phase 2 trial of the Polo-like kinase inhibitor volasertib (BI 6727) in patients with locally advanced or metastatic urothelial cancer. Cancer 2014; 120: 976-982.
14.
Lin CC, Su WC, Yen CJ, Hsu CH, Su WP, Yeh KH, Lu YS, Cheng AL, Huang DC, Fritsch H, Voss F, Taube T, Yang JC: A phase I study of two dosing schedules of volasertib (BI 6727), an intravenous polo-like kinase inhibitor, in patients with advanced solid malignancies. Br J Cancer 2014; 110: 2434-2440.
15.
Shafer D, Grant S: Update on rational targeted therapy in AML. Blood Rev 2016; 30: 275-283.
16.
Podoltsev NA, Stahl M, Zeidan AM, Gore SD: Selecting initial treatment of acute myeloid leukaemia in older adults. Blood Rev 2017; 31: 43-62.
17.
Hao Z, Kota V: Volasertib for AML: clinical use and patient consideration. Onco Targets Ther 2015; 8: 1761-1771.
18.
Erba HP: Finding the optimal combination therapy for the treatment of newly diagnosed AML in older patients unfit for intensive therapy. Leuk Res 2015; 39: 183-191.
19.
Sasine JP, Schiller GJ: Emerging strategies for high-risk and relapsed/refractory acute myeloid leukemia: novel agents and approaches currently in clinical trials. Blood Rev 2015; 29: 1-9.
20.
Montalban-Bravo G, Garcia-Manero G: Novel drugs for older patients with acute myeloid leukemia. Leukemia 2015; 29: 760-769.
21.
Schoffski P, Awada A, Dumez H, Gil T, Bartholomeus S, Wolter P, Taton M, Fritsch H, Glomb P, Munzert G: A phase I, dose-escalation study of the novel Polo-like kinase inhibitor volasertib (BI 6727) in patients with advanced solid tumours. Eur J Cancer 2012; 48: 179-186.
22.
Lang E, Lang F: Mechanisms and pathophysiological significance of eryptosis, the suicidal erythrocyte death. Semin Cell Dev Biol 2015; 39: 35-42.
23.
Dermaku-Sopjani M, Abazi S, Faggio C, Kolgeci J, Sopjani M: AMPK-sensitive cellular transport. J Biochem 2014; 155: 147-158.
24.
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.
25.
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.
26.
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.
27.
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.
28.
Bissinger R, Lang E, Ghashghaeinia M, Singh Y, Zelenak C, Fehrenbacher B, Honisch S, Chen H, Fakhri H, Umbach AT, Liu G, Rexhepaj R, Liu G, Schaller M, Mack AF, Lupescu A, Birnbaumer L, Lang F, Qadri SM: Blunted apoptosis of erythrocytes in mice deficient in the heterotrimeric G-protein subunit Galphai2. Sci Rep 2016; 6: 30925.
29.
Lang E, Bissinger R, Fajol A, Salker MS, Singh Y, Zelenak C, Ghashghaeinia M, Gu S, Jilani K, Lupescu A, Reyskens KM, Ackermann TF, Foller M, Schleicher E, Sheffield WP, Arthur JS, Lang F, Qadri SM: Accelerated apoptotic death and in vivo turnover of erythrocytes in mice lacking functional mitogen- and stress-activated kinase MSK1/2. Sci Rep 2015; 5: 17316.
30.
Alzoubi K, Egler J, Abed M, Lang F: Enhanced Eryptosis Following Auranofin Exposure. Cell Physiol Biochem 2015; 37: 1018-1028.
31.
Bissinger R, Bouguerra G, Stockinger K, Abbes S, Lang F: Triggering of Suicidal Erythrocyte Death by Topotecan. Cell Physiol Biochem 2015; 37: 1607-1618.
32.
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.
33.
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.
34.
Briglia M, Fazio A, Signoretto E, Faggio C, Lang F: Edelfosine Induced Suicidal Death of Human Erythrocytes. Cell Physiol Biochem 2015; 37: 2221-2230.
35.
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.
36.
Egler J, Lang F: Licochalcone A Induced Suicidal Death of Human Erythrocytes. Cell Physiol Biochem 2015; 37: 2060-2070.
37.
Fazio A, Briglia M, Faggio C, Alzoubi K, Lang F: Stimulation of Suicidal Erythrocyte Death by Garcinol. Cell Physiol Biochem 2015; 37: 805-815.
38.
Lang E, Jilani K, Bissinger R, Rexhepaj R, Zelenak C, Lupescu A, Lang F, Qadri SM: Vitamin D-Rich Diet in Mice Modulates Erythrocyte Survival. Kidney Blood Press Res 2015; 40: 403-412.
39.
Officioso A, Alzoubi K, Manna C, Lang F: Clofazimine Induced Suicidal Death of Human Erythrocytes. Cell Physiol Biochem 2015; 37: 331-341.
40.
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.
41.
Stockinger K, Bissinger R, Bouguerra G, Abbes S, Lang F: Enhanced Eryptosis Following Exposure to Carnosic Acid. Cell Physiol Biochem 2015; 37: 1779-1791.
42.
Waibel S, Bissinger R, Bouguerra G, Abbes S, Lang F: Saquinavir Induced Suicidal Death of Human Erythrocytes. Cell Physiol Biochem 2015; 37: 1973-1982.
43.
Zierle J, Bissinger R, Egler J, Lang F: Lapatinib Induced Suicidal Death of Human Erythrocytes. Cell Physiol Biochem 2015; 37: 2275-2287.
44.
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.
45.
Briglia M, Fazio A, Faggio C, Lang F: Triggering of Suicidal Erythrocyte Death by Zosuquidar. Cell Physiol Biochem 2015; 37: 2355-2365.
46.
Fazio A, Briglia M, Faggio C, Alzoubi K, Lang F: Oxaliplatin Induced Suicidal Death of Human Erythrocytes. Cell Physiol Biochem 2015; 37: 2393-2404.
47.
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.
48.
Officioso A, Manna C, Alzoubi K, Lang F: Bromfenvinphos induced suicidal death of human erythrocytes. Pestic Biochem Physiol 2016; 126: 58-63.
49.
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
50.
Zierle J, Bissinger R, Bouguerra G, Abbes S, Lang F: Triggering of Suicidal Erythrocyte Death by Regorafenib. Cell Physiol Biochem 2016; 38: 160-172.
51.
Pagano M, Faggio C: The use of erythrocyte fragility to assess xenobiotic cytotoxicity. Cell Biochem Funct 2015; 33: 351-355.
52.
Al Mamun Bhuyan A, Signoretto E, Bissinger R, Lang F: Enhanced Eryptosis Following Exposure to Dolutegravir. Cell Physiol Biochem 2016; 39: 639-650.
53.
Al Mamun Bhuyan A, Signoretto E, Lang F: Triggering of Suicidal Erythrocyte Death by Psammaplin A. Cell Physiol Biochem 2016; 39: 908-918.
54.
Almasry M, Jemaa M, Mischitelli M, Faggio C, Lang F: Stimulation of Suicidal Erythrocyte Death by Phosphatase Inhibitor Calyculin A. Cell Physiol Biochem 2016; 40: 163-171.
55.
Bissinger R, Bhuyan AA, Signoretto E, Lang F: Stimulating Effect of Elvitegravir on Suicidal Erythrocyte Death. Cell Physiol Biochem 2016; 38: 1111-1120.
56.
Egler J, Zierle J, Lang F: Stimulating Effect of Manumycin A on Suicidal Erythrocyte Death. Cell Physiol Biochem 2016; 38: 1147-1156.
57.
Mischitelli M, Jemaa M, Almasry M, Faggio C, Lang F: Stimulation of Erythrocyte Cell Membrane Scrambling by Quinine. Cell Physiol Biochem 2016; 40: 657-667.
58.
Mischitelli M, Jemaa M, Almasry M, Faggio C, Lang F: Stimulation of Suicidal Erythrocyte Death by Rottlerin. Cell Physiol Biochem 2016; 40: 558-566.
59.
Mischitelli M, Jemaa M, Almasry M, Faggio C, Lang F: Triggering of Suicidal Erythrocyte Death by Fascaplysin. Cell Physiol Biochem 2016; 39: 1638-1647.
60.
Mischitelli M, Jemaa M, Almasry M, Faggio C, Lang F: Ca2+ Entry, Oxidative Stress, Ceramide and Suicidal Erythrocyte Death Following Diosgenin Treatment. Cell Physiol Biochem 2016; 39: 1626-1637.
61.
Mischitelli M, Jemaa M, Almasry M, Faggio C, Lang F: Triggering of Erythrocyte Cell Membrane Scrambling by Emodin. Cell Physiol Biochem 2016; 40: 91-103.
62.
Peter T, Bissinger R, Lang F: Stimulation of Eryptosis by Caspofungin. Cell Physiol Biochem 2016; 39: 939-949.
63.
Peter T, Bissinger R, Liu G, Lang F: Anidulafungin-Induced Suicidal Erythrocyte Death. Cell Physiol Biochem 2016; 38: 2272-2284.
64.
Peter T, Bissinger R, Signoretto E, Mack AF, Lang F: Micafungin-Induced Suicidal Erythrocyte Death. Cell Physiol Biochem 2016; 39: 584-595.
65.
Pretorius E, du Plooy JN, Bester J: A Comprehensive Review on Eryptosis. Cell Physiol Biochem 2016; 39: 1977-2000.
66.
Shan F, Yang R, Ji T, Jiao F: Vitamin C Inhibits Aggravated Eryptosis by Hydrogen Peroxide in Glucose-6-Phosphated Dehydrogenase Deficiency. Cell Physiol Biochem 2016; 39: 1453-1462.
67.
Signoretto E, Honisch S, Briglia M, Faggio C, Castagna M, Lang F: Nocodazole Induced Suicidal Death of Human Erythrocytes. Cell Physiol Biochem 2016; 38: 379-392.
68.
Signoretto E, Laufer SA, Lang F: Stimulating Effect of Sclareol on Suicidal Death of Human Erythrocytes. Cell Physiol Biochem 2016; 39: 554-564.
69.
Signoretto E, Zierle J, Bissinger R, Castagna M, Bossi E, Lang F: Triggering of Suicidal Erythrocyte Death by Pazopanib. Cell Physiol Biochem 2016; 38: 926-938.
70.
Wesseling MC, Wagner-Britz L, Huppert H, Hanf B, Hertz L, Nguyen DB, Bernhardt I: Phosphatidylserine Exposure in Human Red Blood Cells Depending on Cell Age. Cell Physiol Biochem 2016; 38: 1376-1390.
71.
Zierle J, Bissinger R, Lang F: Inhibition by Teriflunomide of Erythrocyte Cell Membrane Scrambling Following Energy Depletion, Oxidative Stress and Ionomycin. Cell Physiol Biochem 2016; 39: 1877-1890.
72.
Bissinger R, Modicano P, Alzoubi K, Honisch S, Faggio C, Abed M, Lang F: Effect of saponin on erythrocytes. Int J Hematol 2014; 100: 51-59.
73.
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.
74.
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.
75.
Al Mamun Bhuyan A, Bissinger R, Cao H, Lang F: Triggering of Suicidal Erythrocyte Death by bexaroten. Cell Physiol Biochem 2016;in press.:
76.
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.
77.
Bissinger R, Bouguerra G, Al Mamun Bhuyan A, Waibel S, Abbes S, Lang F: Efavirenz Induced Suicidal Death of Human Erythrocytes. Cell Physiol Biochem 2015; 37: 2496-2507.
78.
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.
79.
Faggio C, Alzoubi K, Calabro S, Lang F: Stimulation of suicidal erythrocyte death by PRIMA-1. Cell Physiol Biochem 2015; 35: 529-540.
80.
Jemaa M, Mischitelli M, Fezai M, Almasry M, Faggio C, Lang F: Stimulation of Suicidal Erythrocyte Death by the CDC25 Inhibitor NSC-95397. Cell Physiol Biochem 2016; 40: 597-607.
81.
Lang E, Modicano P, Arnold M, Bissinger R, Faggio C, Abed M, Lang F: Effect of thioridazine on erythrocytes. Toxins (Basel) 2013; 5: 1918-1931.
82.
Signoretto E, Bissinger R, Castagna M, Lang F: Stimulation of Eryptosis by Combretastatin A4 Phosphate Disodium (CA4P). Cell Physiol Biochem 2016; 38: 969-981.
83.
Bouguerra G, Bissinger R, Abbes S, Lang F: Stimulation of Eryptosis by Narasin. Cell Physiol Biochem 2015; 37: 1807-1816.
84.
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.
85.
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.
86.
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.
87.
Polak-Jonkisz D, Purzyc L: Ca(2+) influx versus efflux during eryptosis in uremic erythrocytes. Blood Purif 2012; 34: 209-210; author reply 210.
88.
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.
89.
Bissinger R, Artunc F, Qadri SM, Lang F: Reduced Erythrocyte Survival in Uremic Patients Under Hemodialysis or Peritoneal Dialysis. Kidney Blood Press Res 2016; 41: 966-977.
90.
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.
91.
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.
92.
Lang E, Pozdeev VI, Gatidis S, Qadri SM, Haussinger D, Kubitz R, Herebian D, Mayatepek E, Lang F, Lang KS, Lang PA: Bile Acid-Induced Suicidal Erythrocyte Death. Cell Physiol Biochem 2016; 38: 1500-1509.
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.
Bissinger R, Schumacher C, Qadri SM, Honisch S, Malik A, Gotz F, Kopp HG, Lang F: Enhanced eryptosis contributes to anemia in lung cancer patients. Oncotarget 2016; 7: 14002-14014.
95.
Qadri SM, Mahmud H, Lang E, Gu S, Bobbala D, Zelenak C, Jilani K, Siegfried A, Foller M, Lang F: Enhanced suicidal erythrocyte death in mice carrying a loss-of-function mutation of the adenomatous polyposis coli gene. J Cell Mol Med 2012; 16: 1085-1093.
96.
Bissinger R, Kempe-Teufel DS, Honisch S, Qadri SM, Randrianarisoa E, Haring HU, Henes J, Lang F: Stimulated Suicidal Erythrocyte Death in Arteritis. Cell Physiol Biochem 2016; 39: 1068-1077.
97.
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.
98.
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.
99.
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.
100.
Lang E, Pozdeev VI, Xu HC, Shinde PV, Behnke K, Hamdam JM, Lehnert E, Scharf RE, Lang F, Haussinger D, Lang KS, Lang PA: Storage of Erythrocytes Induces Suicidal Erythrocyte Death. Cell Physiol Biochem 2016; 39: 668-676.
101.
Kiraz Y, Adan A, Kartal Yandim M, Baran Y: Major apoptotic mechanisms and genes involved in apoptosis. Tumour Biol 2016; 37: 8471-8486.
102.
Hugle M, Belz K, Fulda S: Identification of synthetic lethality of PLK1 inhibition and microtubule-destabilizing drugs. Cell Death Differ 2015; 22: 1946-1956.
103.
Lang E, Bissinger R, Qadri SM, Lang F: Suicidal death of erythrocytes in cancer and its chemotherapy: A potential target in the treatment of tumor-associated anemia. Int J Cancer 2017; 141: 1522-1528.
104.
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.
105.
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.
106.
Andrews DA, Low PS: Role of red blood cells in thrombosis. Curr Opin Hematol 1999; 6: 76-82.
107.
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.
108.
Zwaal RF, Comfurius P, Bevers EM: Surface exposure of phosphatidylserine in pathological cells. Cell Mol Life Sci 2005; 62: 971-988.
109.
Closse C, Dachary-Prigent J, Boisseau MR: Phosphatidylserine-related adhesion of human erythrocytes to vascular endothelium. Br J Haematol 1999; 107: 300-302.
110.
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.
111.
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.
112.
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.