Next Article in Journal
Epidemiology and Clinical Features of Ciguatera Fish Poisoning in Hong Kong
Previous Article in Journal
Milk Thistle Extract and Silymarin Inhibit Lipopolysaccharide Induced Lamellar Separation of Hoof Explants in Vitro
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Piperlongumine-Induced Phosphatidylserine Translocation in the Erythrocyte Membrane

1
Department of Physiology, Eberhard-Karls-University of Tuebingen, Gmelinstr. 5, 72076 Tuebingen, Germany
2
Department of Biochemistry, University of Agriculture, 38040 Faisalabad, Pakistan
*
Author to whom correspondence should be addressed.
Toxins 2014, 6(10), 2975-2988; https://doi.org/10.3390/toxins6102975
Submission received: 3 July 2014 / Revised: 2 September 2014 / Accepted: 24 September 2014 / Published: 14 October 2014

Abstract

:
Background: Piperlongumine, a component of Piper longum fruit, is considered as a treatment for malignancy. It is effective by inducing apoptosis. Mechanisms involved in the apoptotic action of piperlongumine include oxidative stress and activation of p38 kinase. In analogy to apoptosis of nucleated cells, erythrocytes may undergo eryptosis, the suicidal death of erythrocytes characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine-exposure at the erythrocyte surface. Signaling involved in eryptosis include increase of cytosolic Ca2+-activity ([Ca2+]i), formation of ceramide, oxidative stress and activation of p38 kinase. Methods: Cell volume was estimated from forward scatter, phosphatidylserine-exposure from annexin V binding, [Ca2+]i from Fluo3 fluorescence, reactive oxygen species from 2',7'-dichlorodihydrofluorescein-diacetate fluorescence, and ceramide abundance from binding of fluorescent antibodies in flow cytometry. Results: A 48 h exposure to piperlongumine (30 µM) was followed by significant decrease of forward scatter and increase of annexin-V-binding. Piperlongumine did not significantly modify [Ca2+]i and the effect was not dependent on presence of extracellular Ca2+. Piperlongumine significantly increased ROS formation and ceramide abundance. Conclusions: Piperlongumine triggers cell membrane scrambling, an effect independent from entry of extracellular Ca2+ but at least partially due to ROS and ceramide formation.

1. Introduction

Piper longum fruit has been and is used for the treatment of multiple disorders including chronic bronchitis, asthma, constipation, gonorrhea, paralysis of the tongue, diarrhea, cholera, chronic malaria, viral hepatitis, respiratory infections, stomachache, bronchitis, diseases of the spleen, cough, and tumors [1]. Piperlongumine (piplartine), an amide alkaloid component of Piper longum fruit [1], has particularly strong cytotoxic activity both, in vitro and in vivo [2,3,4,5] and enhances the sensitivity of tumor cells to cytostatic treatment [6]. Piperlongumine is at least partially effective by inducing apoptosis of tumor cells [2,7,8,9,10,11,12,13,14]. Mechanisms involved in the triggering of apoptosis by piperlongumine include induction of oxidative stress [6,7,9,12,15,16,17]. Piperlongumine may bind in tumor cells to the active sites of several key cellular antioxidants including glutathione S transferase and carbonyl reductase 1 [18]. Beyond that, piperlongumine inhibits PI3K/Akt/mTOR signalling leading to down-regulation of the NF-kB pathway, altered gene expression and activation of the mitochondrial apoptotic pathway [3,4,8,14,19,20]. Piperlongumine further activates p38 kinase [15,17], inhibits proteasomal protein degradation [10], and suppresses the transcription factor signal transducer and activator of transcription (STAT) 3 thus modulating expression of multiple Stat3-regulated genes [2].
Similar to apoptosis of nucleated cells, erythrocytes could enter eryptosis, the suicidal erythrocyte death characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine exposure to the erythrocyte surface [21]. Eryptosis could be triggered by increase of cytosolic Ca2+ concentration ([Ca2+]i) [22], ceramide formation [21], caspase activation [23,24,25,26,27], lack of AMP activated kinase AMPK [28], stimulation of casein kinase 1α [29,30], lack of cGMP-dependent protein kinase [31], stimulation of Janus-activated kinase JAK3 [32], stimulation of protein kinase C [33], stimulation of p38 kinase [34], inhibition of PAK2 kinase [35] as well as inhibition of sorafenib [36] and sunitinib [37] sensitive kinases.
To the best of our knowledge, an effect of piperlongumine on suicidal erythrocyte death has never been reported before. The purpose of the present study was to explore whether piperlongumine triggers eryptosis, i.e., the suicidal death of cells devoid of mitochondria and nuclei. Moreover, the study explored the involvement of [Ca2+]i, ROS formation and ceramide abundance.

2. Results and Discussion

The present study explored the putative effect of piperlongumine on eryptosis, the suicidal erythrocyte death. Hallmarks of eryptosis include breakdown of phosphatidylserine asymmetry of the erythrocyte cell membrane with subsequent translocation of phosphatidylserine to the cell surface. Phosphatidylserine exposing erythrocytes were identified by annexin-V-binding in flow cytometry. As illustrated in Figure 1, a 48 h exposure to piperlongumine increased the percentage of annexin-V-binding erythrocytes, an effect reaching statistical significance at 15 µM piperlongumine concentration.
A further hallmark of eryptosis is cell shrinkage. Cell volume was thus estimated from forward scatter in flow cytometry. As shown in Figure 2, a 48 h exposure to piperlongumine slightly decreased average forward scatter, an effect reaching statistical significance at 15 µM piperlongumine concentration. Closer inspection of the histogram reveals that the forward scatter decreases substantially in a minor portion of erythrocytes, whereas the forward scatter of most erythrocytes does not appreciably change. Thus, the average forward scatter underestimates the effect of piperlongumine on those erythrocytes which undergo eryptosis.
Figure 1. Effect of piperlongumine on phosphatidylserine exposure (A) Original histogram of annexin V binding of erythrocytes following exposure for 48 h to Ringer solution without (grey shadow) and with (black line) presence of 30 µM piperlongumine; (B) Arithmetic means ± SEM (n = 15) of erythrocyte annexin-V-binding following incubation for 48 h to Ringer solution without (white bar) or with (black bars) presence of piperlongumine (5–30 µM) or, for comparison, DMSO (0.1%) alone. * (p < 0.05), *** (p < 0.001) indicate significant differences from the absence of piperlongumine (ANOVA).
Figure 1. Effect of piperlongumine on phosphatidylserine exposure (A) Original histogram of annexin V binding of erythrocytes following exposure for 48 h to Ringer solution without (grey shadow) and with (black line) presence of 30 µM piperlongumine; (B) Arithmetic means ± SEM (n = 15) of erythrocyte annexin-V-binding following incubation for 48 h to Ringer solution without (white bar) or with (black bars) presence of piperlongumine (5–30 µM) or, for comparison, DMSO (0.1%) alone. * (p < 0.05), *** (p < 0.001) indicate significant differences from the absence of piperlongumine (ANOVA).
Toxins 06 02975 g001
Figure 2. Effect of piperlongumine on erythrocyte forward scatter (A) Original histogram of forward scatter of erythrocytes following exposure for 48 h to Ringer solution without (grey shadow) and with (black line) presence of 30 µM piperlongumine; (B) Arithmetic means ± SEM (n = 15) of the normalized erythrocyte forward scatter (FSC) following incubation for 48 h to Ringer solution without (white bar) or with (black bars) piperlongumine (5–30 µM) or, for comparison, DMSO (0.1%) alone. ** (p < 0.01), *** (p < 0.001) indicate significant differences from the absence of piperlongumine (ANOVA).
Figure 2. Effect of piperlongumine on erythrocyte forward scatter (A) Original histogram of forward scatter of erythrocytes following exposure for 48 h to Ringer solution without (grey shadow) and with (black line) presence of 30 µM piperlongumine; (B) Arithmetic means ± SEM (n = 15) of the normalized erythrocyte forward scatter (FSC) following incubation for 48 h to Ringer solution without (white bar) or with (black bars) piperlongumine (5–30 µM) or, for comparison, DMSO (0.1%) alone. ** (p < 0.01), *** (p < 0.001) indicate significant differences from the absence of piperlongumine (ANOVA).
Toxins 06 02975 g002
Both, cell membrane scrambling and cell shrinkage could have resulted from increase of cytosolic Ca2+ activity ([Ca2+]i). In order to estimate [Ca2+]i, erythrocytes were loaded with Fluo3-AM and Fluo3 fluorescence determined by flow cytometry. As illustrated in Figure 3A,B, a 48 h exposure of human erythrocytes to piperlongumine tended to slightly increase Fluo3 fluorescence, an effect, however, not reaching statistical significance even at the highest concentration (30 µM) employed.
Figure 3. Effect of piperlongumine on erythrocyte cytosolic Ca2+ concentration (A) Original histogram of Fluo3 fluorescence in erythrocytes following exposure for 48 h to Ringer solution without (grey shadow) and with (black line) presence of 30 µM piperlongumine; (B) Arithmetic means ± SEM (n = 15) of the Fluo3 fluorescence (arbitrary units) in erythrocytes exposed for 48 h to Ringer solution without (white bar) or with (black bars) piperlongumine (5–30 µM) or, for comparison, DMSO (0.1%) alone; (C) Arithmetic means ± SEM (n = 5) of the percentage of annexin-V-binding erythrocytes after a 48 h treatment with Ringer solution without (white bars) or with (black bars) 30 µM piperlongumine in the presence (left bars, +Calcium) and absence (right bars, −Calcium) of calcium. *** (p < 0.001) indicates significant difference from the respective values in the absence of piperlongumine (ANOVA).
Figure 3. Effect of piperlongumine on erythrocyte cytosolic Ca2+ concentration (A) Original histogram of Fluo3 fluorescence in erythrocytes following exposure for 48 h to Ringer solution without (grey shadow) and with (black line) presence of 30 µM piperlongumine; (B) Arithmetic means ± SEM (n = 15) of the Fluo3 fluorescence (arbitrary units) in erythrocytes exposed for 48 h to Ringer solution without (white bar) or with (black bars) piperlongumine (5–30 µM) or, for comparison, DMSO (0.1%) alone; (C) Arithmetic means ± SEM (n = 5) of the percentage of annexin-V-binding erythrocytes after a 48 h treatment with Ringer solution without (white bars) or with (black bars) 30 µM piperlongumine in the presence (left bars, +Calcium) and absence (right bars, −Calcium) of calcium. *** (p < 0.001) indicates significant difference from the respective values in the absence of piperlongumine (ANOVA).
Toxins 06 02975 g003
Additional experiments explored, whether the piperlongumine induced cell membrane scrambling required entry of extracellular Ca2+. To this end, erythrocytes were exposed for 48 h to 30 µM piperlongumine in the presence or nominal absence of extracellular Ca2+. As shown in Figure 3C, the effect of piperlongumine on annexin-V-binding tended to be lower in the nominal absence than in the presence of Ca2+, an effect, however, not reaching statistical significance. In the nominal absence of extracellular Ca2+, piperlongumine still significantly increased the percentage of annexin V binding erythrocytes. Thus, the effect of piperlongumine on cell membrane scrambling did not critically depend on Ca2+ entry.
In order to determine the effect of piperlongumine exposure on hemolysis, the percentage of hemolysed erythrocytes was quantified from hemoglobin concentration in the supernatant. According to the hemoglobin concentration, a 48 h incubation with 0, 5, 10, 15 and 30 µM piperlongumine resulted in hemolysis of 2.4% ± 0.4%, 2.6% ± 0.3%, 2.5% ± 0.5%, 2.5% ± 0.2% and 4.6% ± 1.5% (n = 4), respectively.
In search for further mechanisms involved in the effect of piperlongumine on eryptosis, reactive oxygen species (ROS) were determined utilizing 2',7'-dichlorodihydrofluorescein diacetate (DCFDA). As shown in Figure 4, a 48 h exposure to 30 µM piperlongumine markedly and significantly increased the DCFDA fluorescence, a finding pointing to induction of oxidative stress. The percentage of oxidized erythrocytes was higher than the percentage of erythrocytes with translocated phosphatidylserine. Accordingly, strong ROS formation was apparently required to stimulate cell membrane scrambling.
Figure 4. Effect of piperlongumine on reactive oxygen species (A) Original histogram of 2',7'-dichlorodihydrofluorescein diacetate (DCFDA) fluorescence in erythrocytes following exposure for 48 h to Ringer solution without (grey shadow) and with (black line) presence of 30 µM piperlongumine; (B) Arithmetic means ± SEM (n = 5) of erythrocyte DCFDA fluorescence following incubation for 48 h to Ringer solution without (white bar) or with (black bar) presence of piperlongumine (30 µM). *** (p < 0.001) indicates significant difference from the absence of piperlongumine (ANOVA).
Figure 4. Effect of piperlongumine on reactive oxygen species (A) Original histogram of 2',7'-dichlorodihydrofluorescein diacetate (DCFDA) fluorescence in erythrocytes following exposure for 48 h to Ringer solution without (grey shadow) and with (black line) presence of 30 µM piperlongumine; (B) Arithmetic means ± SEM (n = 5) of erythrocyte DCFDA fluorescence following incubation for 48 h to Ringer solution without (white bar) or with (black bar) presence of piperlongumine (30 µM). *** (p < 0.001) indicates significant difference from the absence of piperlongumine (ANOVA).
Toxins 06 02975 g004
As cell membrane scrambling could be further elicited even at constant [Ca2+]i by ceramide, additional experiments explored, whether piperlongumine increases ceramide formation. The abundance of ceramide at the erythrocyte surface was visualized with an anti-ceramide antibody. As shown in Figure 5, a 48 h exposure to 30 µM piperlongumine significantly increased the ceramide abundance at the erythrocyte surface.
Figure 5. Effect of piperlongumine on ceramide abundance (A) Original histogram of anti-ceramide fluorescein isothiocyanate (FITC) fluorescence in erythrocytes after exposure for 48 h to Ringer solution without (grey shadow) and with (black line) presence of 30 µM piperlongumine; (B) Arithmetic means ± SEM (n = 9) of ceramide abundance at the erythrocyte surface following incubation for 48 h to Ringer solution without (white bar) or with (black bar) presence of piperlongumine (30 µM). ** (p < 0.01) indicates significant difference from the absence of piperlongumine (ANOVA).
Figure 5. Effect of piperlongumine on ceramide abundance (A) Original histogram of anti-ceramide fluorescein isothiocyanate (FITC) fluorescence in erythrocytes after exposure for 48 h to Ringer solution without (grey shadow) and with (black line) presence of 30 µM piperlongumine; (B) Arithmetic means ± SEM (n = 9) of ceramide abundance at the erythrocyte surface following incubation for 48 h to Ringer solution without (white bar) or with (black bar) presence of piperlongumine (30 µM). ** (p < 0.01) indicates significant difference from the absence of piperlongumine (ANOVA).
Toxins 06 02975 g005
The present study reveals that exposure of human erythrocytes to piperlongumine is followed by eryptosis, the suicidal erythrocyte death characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine translocation to the erythrocyte surface. The piperlongumine concentrations required for the triggering of eryptosis are within the range of piperlongumine concentrations employed to elicit apoptosis of tumor cells [20].
Piperlongumine did not significantly increase cytosolic Ca2+ activity ([Ca2+]i), a major trigger of eryptosis [21]. Instead, piperlongumine increased the abundance of ceramide and induced oxidative stress, both well known triggers of eryptosis [21]. In tumor cells, the apoptotic effect of piperlongumine similarly involves oxidative stress [6,7,9,12,15,16,17]. To the best of our knowledge, an effect of piperlongumine on ceramide formation has never been shown before. It would thus be interesting to learn whether piperlongumine triggers ceramide formation as well in nucleated cells.
The phosphatidylserine exposing erythrocytes attach to respective receptors of phagocytes with subsequent engulfment and degradation of the defective erythrocytes [21]. Thus, eryptotic phosphatidylserine exposing erythrocytes are rapidly cleared from circulating blood [21]. The eryptotic cell shrinkage [22] counteracts swelling of the defective cells, which would otherwise lead to rupture of the cell membrane with release of cellular hemoglobin. Released hemoglobin may be filtered in renal glomeruli, subsequently precipitate in the acidic lumen of renal tubules and thus occlude the tubules [38].
Excessive eryptosis may become pathophysiologically relevant. The clearance of phosphatidylserine exposing erythrocytes from circulating blood may lead to anemia as soon as the loss of eryptotic erythrocytes fails to be outweighed by similar formation of new erythrocytes [21]. Treatment of erythrocytes with 15 or 30 µM piperlongumine approximately doubles the percentage of phosphatidylserine exposing erythrocytes. In the absence of compensatory increase of erythropoiesis, the effect would halve the number of erythrocytes in circulating blood and thus lead to severe anemia. Phosphatidylserine exposing erythrocytes may further adhere to endothelial receptors [39]. The attachment of eryptotic erythrocytes to the vascular wall compromises microcirculation [39,40,41,42,43,44]. Moreover, phosphatidylserine exposing erythrocytes stimulate blood clotting and thus thrombosis [40,45,46].
The sensitivity of erythrocytes to piperlongumine may be augmented by additional exposure to further eryptosis triggering xenobiotics [21,37,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80] or in patients suffering from disorders facilitating eryptosis [21], such as diabetes [27,81,82], renal insufficiency [83], hemolytic uremic syndrome [84], sepsis [85], malaria [86], sickle cell disease [86], Wilson’s disease [87], iron deficiency [88], malignancy [89], phosphate depletion [90], and metabolic syndrome [65].

3. Experimental Section

3.1. 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 (184/2003V). The blood was centrifuged at 120 rcf for 20 min at 23 °C and the platelets and leukocytes-containing supernatant were disposed. Erythrocytes were washed 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, and 1 CaCl2. For the experiments, erythrocytes were incubated in vitro at a hematocrit of 0.4% at 37 °C for 48 h. Where indicated, erythrocytes were exposed to piperlongumine (Sigma-Aldrich, Hamburg, Germany) at the indicated concentrations. In Ca2+-free Ringer solution, 1 mM CaCl2 was substituted by 1 mM glycol-bis(2-aminoethylether)-N,N,N',N'-tetraacetic acid (EGTA).

3.2. FACS Analysis of Annexin-V-Binding and Forward Scatter

After incubation under the respective experimental condition, 50 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 20 min under protection from light. In the following, the forward scatter (FSC) of the cells was determined, and annexin-V fluorescence intensity was measured with an excitation wavelength of 488 nm and an emission wavelength of 530 nm on a FACS Calibur (BD, Heidelberg, Germany).

3.3. Measurement of Intracellular Ca2+

After incubation, erythrocytes were washed in Ringer solution and then loaded with Fluo-3/AM (Biotium, Hayward, CA, USA) in Ringer solution containing 5 mM CaCl2 and 5 µM Fluo-3/AM. The cells were incubated at 37 °C for 30 min and washed twice in Ringer solution containing 5 mM CaCl2. The Fluo-3/AM-loaded erythrocytes were resuspended in 200 µL Ringer. Then, Ca2+-dependent fluorescence intensity was measured with an excitation wavelength of 488 nm and an emission wavelength of 530 nm on a FACS Calibur. The adequacy of Fluo-3 fluorescence as a tool to estimate cytosolic Ca2+ activity has been tested in several previous studies [29,91,92,93,94].

3.4. Determination of ROS Production

ROS production was determined utilizing 2',7'-dichlorodihydrofluorescein diacetate (DCFDA) [95]. Briefly, the cells were suspended in FACS buffer and the fluorescence was analysed with flow cytometry (FACS-calibur from Becton Dickinson; Heidelberg, Germany). DCFDA fluorescence intensity was measured in FL-1 with an excitation wavelength of 488 nm and an emission wavelength of 530 nm.

3.5. Determination of Ceramide Formation

For the determination of ceramide, a monoclonal antibody-based assay was used. After incubation, cells were stained for 1 h 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. Subsequently, the cells were stained for 30 min 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 then analyzed by flow cytometric analysis with an excitation wavelength of 488 nm and an emission wavelength of 530 nm.

3.6. Measurement of Hemolysis

For the determination of hemolysis, the samples were centrifuged (3 min at 1600 RPM, room temperature) after incubation, and the supernatants were harvested. As a measure of hemolysis, the hemoglobin (Hb) concentration of the supernatant was determined photometrically at 405 nm. The absorption of the supernatant of erythrocytes lysed in distilled water was defined as 100% hemolysis.

3.7. 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. The number of different erythrocyte specimens studied is given as n. 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.

4. Conclusions

In conclusion, the present paper unequivocally and significantly demonstrates that piperlongumine stimulates eryptosis with erythrocyte shrinkage and cell membrane scrambling. Thus, piperlongumine is able to trigger suicidal death in cells lacking mitochondria and nuclei. The cellular mechanisms involved include oxidative stress and ceramide formation. To the best of our knowledge, an effect of piperlongumine on suicidal erythrocyte death or of ceramide formation in any cell type has never been reported before.

Acknowledgments

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

Author Contributions

Rosi Bissinger, Abaid Malik and Jamshed Warsi performed the experiments. Rosi Bissinger executed the statistical analysis. Kashif Jilani supervised the study and prepared the figures. Florian Lang designed the study and drafted the manuscript. All authors carefully read and approved the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kumar, S.; Kamboj, J.; Suman; Sharma, S. Overview for various aspects of the health benefits of piper longum linn. Fruit. J. Acupunct. Meridian Stud. 2011, 4, 134–140. [Google Scholar]
  2. Bharadwaj, U.; Eckols, T.K.; Kolosov, M.; Kasembeli, M.M.; Adam, A.; Torres, D.; Zhang, X.; Dobrolecki, L.E.; Wei, W.; Lewis, M.T.; et al. Drug-repositioning screening identified piperlongumine as a direct stat3 inhibitor with potent activity against breast cancer. Oncogene 2014. [Google Scholar] [CrossRef]
  3. Makhov, P.; Golovine, K.; Teper, E.; Kutikov, A.; Mehrazin, R.; Corcoran, A.; Tulin, A.; Uzzo, R.G.; Kolenko, V.M. Piperlongumine promotes autophagy via inhibition of akt/mtor signalling and mediates cancer cell death. Br. J. Cancer 2014, 110, 899–907. [Google Scholar]
  4. Shrivastava, S.; Kulkarni, P.; Thummuri, D.; Jeengar, M.K.; Naidu, V.G.; Alvala, M.; Redddy, G.B.; Ramakrishna, S. Piperlongumine, an alkaloid causes inhibition of pi3 k/akt/mtor signaling axis to induce caspase-dependent apoptosis in human triple-negative breast cancer cells. Apoptosis 2014, 19, 1148–1164. [Google Scholar]
  5. Wang, Y.H.; Morris-Natschke, S.L.; Yang, J.; Niu, H.M.; Long, C.L.; Lee, K.H. Anticancer principles from medicinal (hu jiao) plants. J. Tradit. Complement. Med. 2014, 4, 8–16. [Google Scholar]
  6. Yao, J.X.; Yao, Z.F.; Li, Z.F.; Liu, Y.B. Radio-sensitization by piper longumine of human breast adenoma mda-mb-231 cells in vitro. Asian Pac. J. Cancer Prev. 2014, 15, 3211–3217. [Google Scholar]
  7. Adams, D.J.; Dai, M.; Pellegrino, G.; Wagner, B.K.; Stern, A.M.; Shamji, A.F.; Schreiber, S.L. Synthesis, cellular evaluation, and mechanism of action of piperlongumine analogs. Proc. Natl. Acad. Sci. USA 2012, 109, 15115–15120. [Google Scholar]
  8. Farrand, L.; Kim, J.Y.; Im-Aram, A.; Suh, J.Y.; Lee, H.J.; Tsang, B.K. An improved quantitative approach for the assessment of mitochondrial fragmentation in chemoresistant ovarian cancer cells. PLoS One 2013, 8, e74008. [Google Scholar]
  9. Gong, L.H.; Chen, X.X.; Wang, H.; Jiang, Q.W.; Pan, S.S.; Qiu, J.G.; Mei, X.L.; Xue, Y.Q.; Qin, W.M.; Zheng, F.Y.; et al. Piperlongumine induces apoptosis and synergizes with cisplatin or paclitaxel in human ovarian cancer cells. Oxid. Med. Cell. Longev. 2014, 2014, 906804. [Google Scholar] [CrossRef]
  10. Halasi, M.; Wang, M.; Chavan, T.S.; Gaponenko, V.; Hay, N.; Gartel, A.L. Ros inhibitor n-acetyl-l-cysteine antagonizes the activity of proteasome inhibitors. Biochem. J. 2013, 454, 201–208. [Google Scholar]
  11. Maund, S.L.; Nolley, R.; Peehl, D.M. Optimization and comprehensive characterization of a faithful tissue culture model of the benign and malignant human prostate. Lab. Investig. 2014, 94, 208–221. [Google Scholar]
  12. Raj, L.; Ide, T.; Gurkar, A.U.; Foley, M.; Schenone, M.; Li, X.; Tolliday, N.J.; Golub, T.R.; Carr, S.A.; Shamji, A.F.; et al. Selective killing of cancer cells by a small molecule targeting the stress response to ros. Nature 2011, 475, 231–234. [Google Scholar]
  13. Tomasini-Johansson, B.R.; Johnson, I.A.; Hoffmann, F.M.; Mosher, D.F. Quantitative microtiter fibronectin fibrillogenesis assay: Use in high throughput screening for identification of inhibitor compounds. Matrix Biol. 2012, 31, 360–367. [Google Scholar]
  14. Han, S.S.; Tompkins, V.S.; Son, D.J.; Kamberos, N.L.; Stunz, L.L.; Halwani, A.; Bishop, G.A.; Janz, S. Piperlongumine inhibits lmp1/myc-dependent mouse b-lymphoma cells. Biochem. Biophys. Res. Commun. 2013, 436, 660–665. [Google Scholar]
  15. Liu, J.M.; Pan, F.; Li, L.; Liu, Q.R.; Chen, Y.; Xiong, X.X.; Cheng, K.; Yu, S.B.; Shi, Z.; Yu, A.C.; et al. Piperlongumine selectively kills glioblastoma multiforme cells via reactive oxygen species accumulation dependent jnk and p38 activation. Biochem. Biophys. Res. Commun. 2013, 437, 87–93. [Google Scholar]
  16. Randhawa, H.; Kibble, K.; Zeng, H.; Moyer, M.P.; Reindl, K.M. Activation of erk signaling and induction of colon cancer cell death by piperlongumine. Toxicol. In Vitro 2013, 27, 1626–1633. [Google Scholar]
  17. Wang, Y.; Wang, J.W.; Xiao, X.; Shan, Y.; Xue, B.; Jiang, G.; He, Q.; Chen, J.; Xu, H.G.; Zhao, R.X.; et al. Piperlongumine induces autophagy by targeting p38 signaling. Cell Death Dis. 2013, 4, e824. [Google Scholar] [CrossRef]
  18. Saeidnia, S.; Abdollahi, M. Antioxidants: Friends or foe in prevention or treatment of cancer: The debate of the century. Toxicol. Appl. Pharmacol. 2013, 271, 49–63. [Google Scholar]
  19. Ginzburg, S.; Golovine, K.V.; Makhov, P.B.; Uzzo, R.G.; Kutikov, A.; Kolenko, V.M. Piperlongumine inhibits nf-kappab activity and attenuates aggressive growth characteristics of prostate cancer cells. Prostate 2014, 74, 177–186. [Google Scholar]
  20. Han, S.S.; Son, D.J.; Yun, H.; Kamberos, N.L.; Janz, S. Piperlongumine inhibits proliferation and survival of burkitt lymphoma in vitro. Leuk. Res. 2013, 37, 146–154. [Google Scholar]
  21. Lang, E.; Qadri, S.M.; Lang, F. Killing me softly—Suicidal erythrocyte death. Int. J. Biochem. Cell Biol. 2012, 44, 1236–1243. [Google Scholar]
  22. Lang, P.A.; Kaiser, S.; Myssina, S.; Wieder, T.; Lang, F.; Huber, S.M. Role of Ca2+-activated k+ channels in human erythrocyte apoptosis. Am. J. Physiol. Cell Physiol. 2003, 285, C1553–C1560. [Google Scholar]
  23. Bhavsar, S.K.; Bobbala, D.; Xuan, N.T.; Foller, M.; Lang, F. Stimulation of suicidal erythrocyte death by alpha-lipoic acid. Cell. Physiol. Biochem. 2010, 26, 859–868. [Google Scholar]
  24. Foller, M.; Huber, S.M.; Lang, F. Erythrocyte programmed cell death. IUBMB Life 2008, 60, 661–668. [Google Scholar]
  25. Foller, M.; Mahmud, H.; Gu, S.; Wang, K.; Floride, E.; Kucherenko, Y.; Luik, S.; Laufer, S.; Lang, F. Participation of leukotriene c(4) in the regulation of suicidal erythrocyte death. J. Physiol. Pharmacol. 2009, 60, 135–143. [Google Scholar]
  26. Lau, I.P.; Chen, H.; Wang, J.; Ong, H.C.; Leung, K.C.; Ho, H.P.; Kong, S.K. In vitro effect of ctab- and peg-coated gold nanorods on the induction of eryptosis/erythroptosis in human erythrocytes. Nanotoxicology 2012, 6, 847–856. [Google Scholar]
  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. [Google Scholar]
  28. Foller, M.; Sopjani, M.; Koka, S.; Gu, S.; Mahmud, H.; Wang, K.; Floride, E.; Schleicher, E.; Schulz, E.; Munzel, T.; et al. Regulation of erythrocyte survival by amp-activated protein kinase. FASEB J. 2009, 23, 1072–1080. [Google Scholar]
  29. Kucherenko, Y.; Zelenak, C.; Eberhard, M.; Qadri, S.M.; Lang, F. Effect of casein kinase 1alpha activator pyrvinium pamoate on erythrocyte ion channels. Cell. Physiol. Biochem. 2012, 30, 407–417. [Google Scholar]
  30. Zelenak, C.; Eberhard, M.; Jilani, K.; Qadri, S.M.; Macek, B.; Lang, F. Protein kinase ck1alpha regulates erythrocyte survival. Cell. Physiol. Biochem. 2012, 29, 171–180. [Google Scholar]
  31. Foller, M.; Feil, S.; Ghoreschi, K.; Koka, S.; Gerling, A.; Thunemann, M.; Hofmann, F.; Schuler, B.; Vogel, J.; Pichler, B.; et al. Anemia and splenomegaly in cgki-deficient mice. Proc. Natl. Acad. Sci. USA 2008, 105, 6771–6776. [Google Scholar]
  32. Bhavsar, S.K.; Gu, S.; Bobbala, D.; Lang, F. Janus kinase 3 is expressed in erythrocytes, phosphorylated upon energy depletion and involved in the regulation of suicidal erythrocyte death. Cell. Physiol. Biochem. 2011, 27, 547–556. [Google Scholar]
  33. Klarl, B.A.; Lang, P.A.; Kempe, D.S.; Niemoeller, O.M.; Akel, A.; Sobiesiak, M.; Eisele, K.; Podolski, M.; Huber, S.M.; Wieder, T.; et al. Protein kinase c mediates erythrocyte “programmed cell death” following glucose depletion. Am. J. Physiol. Cell Physiol. 2006, 290, C244–C253. [Google Scholar]
  34. Gatidis, S.; Zelenak, C.; Fajol, A.; Lang, E.; Jilani, K.; Michael, D.; Qadri, S.M.; Lang, F. P38 mapk activation and function following osmotic shock of erythrocytes. Cell. Physiol. Biochem. 2011, 28, 1279–1286. [Google Scholar]
  35. Zelenak, C.; Foller, M.; Velic, A.; Krug, K.; Qadri, S.M.; Viollet, B.; Lang, F.; Macek, B. Proteome analysis of erythrocytes lacking amp-activated protein kinase reveals a role of pak2 kinase in eryptosis. J. Proteome Res. 2011, 10, 1690–1697. [Google Scholar]
  36. Lupescu, A.; Shaik, N.; Jilani, K.; Zelenak, C.; Lang, E.; Pasham, V.; Zbidah, M.; Plate, A.; Bitzer, M.; Foller, M.; et al. Enhanced erythrocyte membrane exposure of phosphatidylserine following sorafenib treatment: An in vivo and in vitro study. Cell. Physiol. Biochem. 2012, 30, 876–888. [Google Scholar]
  37. Shaik, N.; Lupescu, A.; Lang, F. Sunitinib-sensitive suicidal erythrocyte death. Cell. Physiol. Biochem. 2012, 30, 512–522. [Google Scholar]
  38. Harrison, H.E.; Bunting, H.; Ordway, N.K.; Albrink, W.S. The pathogenesis of the renal injury produced in the dog by hemoglobin or methemoglobin. J. Exp. Med. 1947, 86, 339–356. [Google Scholar]
  39. Borst, O.; Abed, M.; Alesutan, I.; Towhid, S.T.; Qadri, S.M.; 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. [Google Scholar]
  40. Andrews, D.A.; Low, P.S. Role of red blood cells in thrombosis. Curr. Opin. Hematol. 1999, 6, 76–82. [Google Scholar]
  41. Closse, C.; Dachary-Prigent, J.; Boisseau, M.R. Phosphatidylserine-related adhesion of human erythrocytes to vascular endothelium. Br. J. Haematol. 1999, 107, 300–302. [Google Scholar]
  42. Gallagher, P.G.; Chang, S.H.; Rettig, M.P.; Neely, J.E.; Hillery, C.A.; Smith, B.D.; Low, P.S. Altered erythrocyte endothelial adherence and membrane phospholipid asymmetry in hereditary hydrocytosis. Blood 2003, 101, 4625–4627. [Google Scholar]
  43. 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. [Google Scholar]
  44. Wood, B.L.; Gibson, D.F.; Tait, J.F. Increased erythrocyte phosphatidylserine exposure in sickle cell disease: Flow-cytometric measurement and clinical associations. Blood 1996, 88, 1873–1880. [Google Scholar]
  45. Chung, S.M.; Bae, O.N.; Lim, K.M.; Noh, J.Y.; Lee, M.Y.; Jung, Y.S.; Chung, J.H. Lysophosphatidic acid induces thrombogenic activity through phosphatidylserine exposure and procoagulant microvesicle generation in human erythrocytes. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 414–421. [Google Scholar]
  46. Zwaal, R.F.; Comfurius, P.; Bevers, E.M. Surface exposure of phosphatidylserine in pathological cells. Cell. Mol. Life Sci. 2005, 62, 971–988. [Google Scholar]
  47. Abed, M.; Towhid, S.T.; 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–C999. [Google Scholar]
  48. Bottger, E.; Multhoff, G.; Kun, J.F.; Esen, M. Plasmodium falciparum-infected erythrocytes induce granzyme b by nk cells through expression of host-hsp70. PLoS One 2012, 7, e33774. [Google Scholar]
  49. Firat, U.; Kaya, S.; Cim, A.; Buyukbayram, H.; Gokalp, O.; Dal, M.S.; Tamer, M.N. Increased caspase-3 immunoreactivity of erythrocytes in stz diabetic rats. Exp. Diabetes Res. 2012, 2012, 316384. [Google Scholar] [CrossRef]
  50. Ganesan, S.; Chaurasiya, N.D.; Sahu, R.; Walker, L.A.; Tekwani, B.L. Understanding the mechanisms for metabolism-linked hemolytic toxicity of primaquine against glucose 6-phosphate dehydrogenase deficient human erythrocytes: Evaluation of eryptotic pathway. Toxicology 2012, 294, 54–60. [Google Scholar]
  51. Gao, M.; Cheung, K.L.; Lau, I.P.; Yu, W.S.; Fung, K.P.; Yu, B.; Loo, J.F.; Kong, S.K. Polyphyllin d induces apoptosis in human erythrocytes through ca(2)(+) rise and membrane permeabilization. Arch. Toxicol. 2012, 86, 741–752. [Google Scholar]
  52. Ghashghaeinia, M.; Cluitmans, J.C.; Akel, A.; Dreischer, P.; Toulany, M.; Koberle, M.; Skabytska, Y.; Saki, M.; Biedermann, T.; Duszenko, M.; et al. The impact of erythrocyte age on eryptosis. Br. J. Haematol. 2012, 157, 606–614. [Google Scholar]
  53. Jilani, K.; Lupescu, A.; Zbidah, M.; Abed, M.; Shaik, N.; Lang, F. Enhanced apoptotic death of erythrocytes induced by the mycotoxin ochratoxin A. Kidney Blood Press. Res. 2012, 36, 107–118. [Google Scholar]
  54. Jilani, K.; Lupescu, A.; Zbidah, M.; Shaik, N.; Lang, F. Withaferin a-stimulated Ca2+ entry, ceramide formation and suicidal death of erythrocytes. Toxicol. In Vitro 2013, 27, 52–58. [Google Scholar]
  55. Kucherenko, Y.V.; Lang, F. Inhibitory effect of furosemide on non-selective voltage-independent cation channels in human erythrocytes. Cell. Physiol. Biochem. 2012, 30, 863–875. [Google Scholar]
  56. Lang, E.; Jilani, K.; Zelenak, C.; Pasham, V.; Bobbala, D.; Qadri, S.M.; Lang, F. Stimulation of suicidal erythrocyte death by benzethonium. Cell. Physiol. Biochem. 2011, 28, 347–354. [Google Scholar]
  57. Lang, E.; Qadri, S.M.; Jilani, K.; Zelenak, C.; Lupescu, A.; Schleicher, E.; Lang, F. Carbon monoxide-sensitive apoptotic death of erythrocytes. Basic Clin. Pharmacol. Toxicol. 2012, 111, 348–355. [Google Scholar]
  58. Lupescu, A.; Jilani, K.; Zbidah, M.; Lang, E.; Lang, F. Enhanced ca(2+) entry, ceramide formation, and apoptotic death of erythrocytes triggered by plumbagin. J. Nat. Prod. 2012, 75, 1956–1961. [Google Scholar]
  59. Lupescu, A.; Jilani, K.; Zbidah, M.; Lang, F. Induction of apoptotic erythrocyte death by rotenone. Toxicology 2012, 300, 132–137. [Google Scholar]
  60. Polak-Jonkisz, D.; Purzyc, L. Ca influx vs. efflux during eryptosis in uremic erythrocytes. Blood Purif. 2012, 34, 209–210. [Google Scholar]
  61. Qian, E.W.; Ge, D.T.; Kong, S.K. Salidroside protects human erythrocytes against hydrogen peroxide-induced apoptosis. J. Nat. Prod. 2012, 75, 531–537. [Google Scholar]
  62. Shaik, N.; Zbidah, M.; Lang, F. Inhibition of ca(2+) entry and suicidal erythrocyte death by naringin. Cell. Physiol. Biochem. 2012, 30, 678–686. [Google Scholar]
  63. Vota, D.M.; Maltaneri, R.E.; Wenker, S.D.; Nesse, A.B.; Vittori, D.C. Differential erythropoietin action upon cells induced to eryptosis by different agents. Cell Biochem. Biophys. 2013, 65, 145–157. [Google Scholar]
  64. Weiss, E.; Cytlak, U.M.; Rees, D.C.; Osei, A.; Gibson, J.S. Deoxygenation-induced and Ca(2+) dependent phosphatidylserine externalisation in red blood cells from normal individuals and sickle cell patients. Cell Calcium 2012, 51, 51–56. [Google Scholar]
  65. Zappulla, D. Environmental stress, erythrocyte dysfunctions, inflammation, and the metabolic syndrome: Adaptations to co2 increases? J. Cardiometab. Syndr. 2008, 3, 30–34. [Google Scholar]
  66. Zbidah, M.; Lupescu, A.; Jilani, K.; Lang, F. Stimulation of suicidal erythrocyte death by fumagillin. Basic Clin. Pharmacol. Toxicol. 2013, 112, 346–351. [Google Scholar]
  67. Zbidah, M.; Lupescu, A.; Shaik, N.; Lang, F. Gossypol-induced suicidal erythrocyte death. Toxicology 2012, 302, 101–105. [Google Scholar]
  68. Zelenak, C.; Pasham, V.; Jilani, K.; Tripodi, P.M.; Rosaclerio, L.; Pathare, G.; Lupescu, A.; Faggio, C.; Qadri, S.M.; Lang, F. Tanshinone iia stimulates erythrocyte phosphatidylserine exposure. Cell. Physiol. Biochem. 2012, 30, 282–294. [Google Scholar]
  69. Abed, M.; Herrmann, T.; Alzoubi, K.; Pakladok, T.; Lang, F. Tannic acid induced suicidal erythrocyte death. Cell. Physiol. Biochem. 2013, 32, 1106–1116. [Google Scholar]
  70. Ahmed, M.S.; Langer, H.; Abed, M.; Voelkl, J.; Lang, F. The uremic toxin acrolein promotes suicidal erythrocyte death. Kidney Blood Press. Res. 2013, 37, 158–167. [Google Scholar]
  71. Ghashghaeinia, M.; Cluitmans, J.C.; Toulany, M.; Saki, M.; Koberle, M.; Lang, E.; Dreischer, P.; Biedermann, T.; Duszenko, M.; Lang, F.; et al. Age sensitivity of nfkappab abundance and programmed cell death in erythrocytes induced by nfkappab inhibitors. Cell. Physiol. Biochem. 2013, 32, 801–813. [Google Scholar]
  72. Lupescu, A.; Jilani, K.; Zbidah, M.; Lang, F. Patulin-induced suicidal erythrocyte death. Cell. Physiol. Biochem. 2013, 32, 291–299. [Google Scholar]
  73. Alzoubi, K.; Alktifan, B.; Oswald, G.; Fezai, M.; Abed, M.; Lang, F. Breakdown of phosphatidylserine asymmetry following treatment of erythrocytes with lumefantrine. Toxins (Basel) 2014, 6, 650–664. [Google Scholar]
  74. Alzoubi, K.; Honisch, S.; Abed, M.; Lang, F. Triggering of suicidal erythrocyte death by penta-O-galloyl-beta-d-glucose. Toxins (Basel) 2014, 6, 54–65. [Google Scholar]
  75. Frauenfeld, L.; Alzoubi, K.; Abed, M.; Lang, F. Stimulation of erythrocyte cell membrane scrambling by mushroom tyrosinase. Toxins (Basel) 2014, 6, 1096–1108. [Google Scholar]
  76. Jilani, K.; Enkel, S.; Bissinger, R.; Almilaji, A.; Abed, M.; Lang, F. Fluoxetine induced suicidal erythrocyte death. Toxins (Basel) 2013, 5, 1230–1243. [Google Scholar]
  77. Jilani, K.; Lang, F. Carmustine-induced phosphatidylserine translocation in the erythrocyte membrane. Toxins (Basel) 2013, 5, 703–716. [Google Scholar]
  78. 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. [Google Scholar]
  79. Lupescu, A.; Bissinger, R.; Jilani, K.; Lang, F. Triggering of suicidal erythrocyte death by celecoxib. Toxins (Basel) 2013, 5, 1543–1554. [Google Scholar]
  80. Lupescu, A.; Bissinger, R.; Jilani, K.; Lang, F. In vitro induction of erythrocyte phosphatidylserine translocation by the natural naphthoquinone shikonin. Toxins (Basel) 2014, 6, 1559–1574. [Google Scholar]
  81. Calderon-Salinas, J.V.; Munoz-Reyes, E.G.; Guerrero-Romero, J.F.; Rodriguez-Moran, M.; Bracho-Riquelme, R.L.; Carrera-Gracia, M.A.; Quintanar-Escorza, M.A. Eryptosis and oxidative damage in type 2 diabetic mellitus patients with chronic kidney disease. Mol. Cell Biochem. 2011, 357, 171–179. [Google Scholar]
  82. Nicolay, J.P.; Schneider, J.; Niemoeller, O.M.; Artunc, F.; Portero-Otin, M.; Haik, G., Jr.; Thornalley, P.J.; Schleicher, E.; Wieder, T.; Lang, F. Stimulation of suicidal erythrocyte death by methylglyoxal. Cell. Physiol. Biochem. 2006, 18, 223–232. [Google Scholar]
  83. Myssina, S.; Huber, S.M.; Birka, C.; Lang, P.A.; Lang, K.S.; Friedrich, B.; Risler, T.; Wieder, T.; Lang, F. Inhibition of erythrocyte cation channels by erythropoietin. J. Am. Soc. Nephrol. 2003, 14, 2750–2757. [Google Scholar]
  84. Lang, P.A.; Beringer, O.; Nicolay, J.P.; Amon, O.; Kempe, D.S.; Hermle, T.; Attanasio, P.; Akel, A.; Schafer, R.; Friedrich, B.; et al. Suicidal death of erythrocytes in recurrent hemolytic uremic syndrome. J. Mol. Med. (Berl.) 2006, 84, 378–388. [Google Scholar]
  85. Kempe, D.S.; Akel, A.; Lang, P.A.; Hermle, T.; Biswas, R.; Muresanu, J.; Friedrich, B.; Dreischer, P.; Wolz, C.; Schumacher, U.; et al. Suicidal erythrocyte death in sepsis. J. Mol. Med. 2007, 85, 269–277. [Google Scholar]
  86. Foller, M.; Bobbala, D.; Koka, S.; Huber, S.M.; Gulbins, E.; Lang, F. Suicide for survival—Death of infected erythrocytes as a host mechanism to survive malaria. Cell. Physiol. Biochem. 2009, 24, 133–140. [Google Scholar]
  87. Lang, P.A.; Schenck, M.; Nicolay, J.P.; Becker, J.U.; Kempe, D.S.; Lupescu, A.; Koka, S.; Eisele, K.; Klarl, B.A.; Rubben, H.; et al. Liver cell death and anemia in wilson disease involve acid sphingomyelinase and ceramide. Nat. Med. 2007, 13, 164–170. [Google Scholar]
  88. Kempe, D.S.; Lang, P.A.; Duranton, C.; Akel, A.; Lang, K.S.; Huber, S.M.; Wieder, T.; Lang, F. Enhanced programmed cell death of iron-deficient erythrocytes. FASEB J. 2006, 20, 368–370. [Google Scholar]
  89. Qadri, S.M.; 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. [Google Scholar]
  90. Birka, C.; Lang, P.A.; Kempe, D.S.; Hoefling, L.; Tanneur, V.; Duranton, C.; Nammi, S.; Henke, G.; Myssina, S.; Krikov, M.; et al. Enhanced susceptibility to erythrocyte “apoptosis” following phosphate depletion. Pflugers Arch. 2004, 448, 471–477. [Google Scholar]
  91. Foller, M.; Kasinathan, R.S.; Koka, S.; Huber, S.M.; Schuler, B.; Vogel, J.; Gassmann, M.; Lang, F. Enhanced susceptibility to suicidal death of erythrocytes from transgenic mice overexpressing erythropoietin. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2007, 293, R1127–R1134. [Google Scholar] [Green Version]
  92. Foller, M.; Mahmud, H.; Koka, S.; Lang, F. Reduced Ca2+ entry and suicidal death of erythrocytes in pdk1 hypomorphic mice. Pflugers Arch. 2008, 455, 939–949. [Google Scholar]
  93. Kucherenko, Y.V.; Wagner-Britz, L.; Bernhardt, I.; Lang, F. Effect of chloride channel inhibitors on cytosolic Ca2+ levels and Ca2+-activated k+ (gardos) channel activity in human red blood cells. J. Membr. Biol. 2013, 246, 315–326. [Google Scholar]
  94. Shaik, N.; Lupescu, A.; Lang, F. Inhibition of suicidal erythrocyte death by probucol. J. Cardiovasc. Pharmacol. 2013, 61, 120–126. [Google Scholar]
  95. Sinha, A.; Singh, A.; Satchidanandam, V.; Natarajan, K. Impaired generation of reactive oxygen species during differentiation of dendritic cells (dcs) by mycobacterium tuberculosis secretory antigen (mtsa) and subsequent activation of mtsa-dcs by mycobacteria results in increased intracellular survival. J. Immunol. 2006, 177, 468–478. [Google Scholar]

Share and Cite

MDPI and ACS Style

Bissinger, R.; Malik, A.; Warsi, J.; Jilani, K.; Lang, F. Piperlongumine-Induced Phosphatidylserine Translocation in the Erythrocyte Membrane. Toxins 2014, 6, 2975-2988. https://doi.org/10.3390/toxins6102975

AMA Style

Bissinger R, Malik A, Warsi J, Jilani K, Lang F. Piperlongumine-Induced Phosphatidylserine Translocation in the Erythrocyte Membrane. Toxins. 2014; 6(10):2975-2988. https://doi.org/10.3390/toxins6102975

Chicago/Turabian Style

Bissinger, Rosi, Abaid Malik, Jamshed Warsi, Kashif Jilani, and Florian Lang. 2014. "Piperlongumine-Induced Phosphatidylserine Translocation in the Erythrocyte Membrane" Toxins 6, no. 10: 2975-2988. https://doi.org/10.3390/toxins6102975

Article Metrics

Back to TopTop