Next Article in Journal
Effects of Vitamin D3, Calcipotriol and FTY720 on the Expression of Surface Molecules and Cytolytic Activities of Human Natural Killer Cells and Dendritic Cells
Previous Article in Journal
Cyanobacteria and Cyanotoxins: From Impacts on Aquatic Ecosystems and Human Health to Anticarcinogenic Effects
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Effect of Thioridazine on Erythrocytes

1
Department of Physiology, Eberhard-Karls-University of Tuebingen, Gmelinstr.5, Tuebingen 72076, Germany
2
Department of Biological and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres, 31, S. Agata-Messina 98166, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Toxins 2013, 5(10), 1918-1931; https://doi.org/10.3390/toxins5101918
Submission received: 4 September 2013 / Revised: 16 October 2013 / Accepted: 18 October 2013 / Published: 23 October 2013

Abstract

:
Background: Thioridazine, a neuroleptic phenothiazine with antimicrobial efficacy is known to trigger anemia. At least in theory, the anemia could result from stimulation of suicidal erythrocyte death or eryptosis, which is characterized by cell shrinkage and by phospholipid scrambling of the cell membrane with phosphatidylserine exposure at the erythrocyte surface. Triggers of eryptosis include increase of cytosolic Ca2+-concentration ([Ca2+]i) and activation of p38 kinase. The present study explored, whether thioridazine elicits eryptosis. Methods: [Ca2+]i has been estimated from Fluo3-fluorescence, cell volume from forward scatter, phosphatidylserine exposure from annexin-V-binding, and hemolysis from hemoglobin release. Results: A 48 hours exposure to thioridazine was followed by a significant increase of [Ca2+]i (30 µM), decrease of forward scatter (30 µM), and increase of annexin-V-binding (≥12 µM). Nominal absence of extracellular Ca2+ and p38 kinase inhibitor SB203580 (2 µM) significantly blunted but did not abolish annexin-V-binding following thioridazine exposure. Conclusions: Thioridazine stimulates eryptosis, an effect in part due to entry of extracellular Ca2+ and activation of p38 kinase.

1. Introduction

Thioridazine, a phenothiazine drug, has both antipsychotic efficacy [1,2] and anti-microbial activity [2]. It is particularly useful for the treatment of multidrug resistant tuberculosis [3,4,5,6].
Side effects of thioridazine include anemia [7,8]. Drug induced anemia could, at least in theory, result from stimulation of suicidal erythrocyte death or eryptosis [9]. Eryptotic erythrocytes are rapidly cleared from circulating blood and are, thus, removed prior to hemolysis [9]. The most important hallmark of eryptosis is breakdown of phosphatidylserine asymmetry of the erythrocyte cell membrane with translocation of phosphatidylserine to the erythrocyte surface [9]. Eryptosis is further typically paralleled by erythrocyte shrinkage [10]. Eryptosis may be triggered by increased cytosolic Ca2+ concentration ([Ca2+]i) due to Ca2+ entry through Ca2+-permeable cation channels [11,12], or due to permeabilization of the erythrocyte membrane [13]. Increased [Ca2+]i leads to cell shrinkage due to activation of Ca2+-sensitive K+ channels [14], K+ exit, hyperpolarization, Cl exit and thus cellular loss of KCl and osmotically obliged water [10]. Increased [Ca2+]i further leads to translocation of phosphatidylserine from the inner leaflet of the cell membrane to the erythrocyte surface [15]. The sensitivity of cell membrane scrambling to cytosolic Ca2+ is enhanced by ceramide [16]. Eryptosis is further triggered by energy depletion [17] and caspase activation [18,19,20,21,22]. The cytosolic machinery governing eryptosis further involves AMP activated kinase AMPK [12], cGMP-dependent protein kinase [23], Janus-activated kinase JAK3 [24], casein kinase [25,26], p38 kinase [27], PAK2 kinase [28], as well as sorafenib [29] and sunifinib [30] sensitive kinases.
Eryptosis may be triggered by a wide variety of xenobiotics [30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61]. Moreover, excessive eryptosis contributes to the pathophysiology of several clinical disorders [9], such as diabetes [22,62,63], renal insufficiency [64], hemolytic uremic syndrome [65], sepsis [66], malaria [67,68,69,70,71], sickle cell disease [72], Wilson’s disease [70], iron deficiency [73], malignancy [74], phosphate depletion [75], and metabolic syndrome [57].
The present study explored, whether thioridazine triggers eryptosis. To this end, the effect of thioridazine on [Ca2+]i, cell volume and phosphatidylserine abundance at the erythrocyte surface has been determined.

2. Results and Discussion

The present study explored whether thioridazine triggers eryptosis, the suicidal death of erythrocytes. The key hallmark of eryptosis is the triggering of cell membrane scrambling with increase of phosphatidylserine abundance at the cell surface. Accordingly, phosphatidylserine exposing erythrocytes were identified by annexin-V-binding in flow cytometry. As shown in Figure 1, treatment of human erythrocytes from healthy individuals with thioridazine increased the percentage of annexin-V-binding erythrocytes, an effect reaching statistical significance at 12 µM thioridazine concentration (Figure 1A,B). In order to explore whether thioridazine treatment may trigger hemolysis, the percentage of hemolysed erythrocytes was estimated from the hemoglobin concentration in the supernatant. As illustrated in Figure 1B, thioridazine treatment significantly increased the hemoglobin concentration in the supernatant. The percentage of hemolytic erythrocytes was, however, clearly smaller than the percentage of annexin V binding erythrocytes (Figure 1B).
Figure 1. Effect of thioridazine on phosphatidylserine exposure and hemolysis.(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 thioridazine; (B) Arithmetic means ± SEM (n = 6) of erythrocyte annexin-V-binding following incubation for 48 h to Ringer solution without (white bar) or with (black bars) presence of thioridazine (6–30 µM). For comparison, arithmetic means ± SEM (n = 5) of the percentage of hemolysis is shown as grey bars. *** (p < 0.001) indicate significant differences from the absence of thioridazine (ANOVA).
Figure 1. Effect of thioridazine on phosphatidylserine exposure and hemolysis.(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 thioridazine; (B) Arithmetic means ± SEM (n = 6) of erythrocyte annexin-V-binding following incubation for 48 h to Ringer solution without (white bar) or with (black bars) presence of thioridazine (6–30 µM). For comparison, arithmetic means ± SEM (n = 5) of the percentage of hemolysis is shown as grey bars. *** (p < 0.001) indicate significant differences from the absence of thioridazine (ANOVA).
Toxins 05 01918 g001
Another hallmark of eryptosis is cell shrinkage. Thus, cell volume was estimated utilizing forward scatter in flow cytometry. As illustrated in Figure 2A,B, thioridazine treatment resulted in a decrease of forward scatter, an effect reaching statistical significance at a 30 µM thioridazine concentration.
Both, cell membrane scrambling and cell shrinkage could have resulted from increase of cytosolic Ca2+ concentration ([Ca2+]i). Accordingly, [Ca2+]i was determined utilizing Fluo3 fluorescence. To this end, erythrocytes were loaded with Fluo3-AM and Fluo3 fluorescence determined in flow cytometry following prior incubation of the erythrocytes in Ringer solution without or with thioridazine. As illustrated in Figure 3A,B, treatment of human erythrocytes with thioridazine increased Fluo3 fluorescence, an effect reaching statistical significance at a 30 µM thioridazine concentration.
In order to explore whether extracellular Ca2+ entry was required for the effect of thioridazine on cell membrane scrambling, erythrocytes were exposed to 30 µM of thioridazine for 48 hours, either in the presence of 1 mM Ca2+ or in the absence of Ca2+ and presence of Ca2+ chelator EGTA (1 mM). As shown in Figure 4, the effect of thioridazine on annexin-V-binding was significantly blunted in the nominal absence of Ca2+. However, even in the absence of extracellular Ca2+, thioridazine still significantly increased the percentage of annexin-V-bindiung erythrocytes pointing to additional mechanisms involved.
Figure 2. Effect of thioridazine 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 thioridazine; (B) Arithmetic means ± SEM (n = 6) of the normalized erythrocyte forward scatter (FSC) following incubation for 48 h to Ringer solution without (white bar) or with (black bars) thioridazine (6–30 µM); *** (p < 0.001) indicates significant difference from the absence of thioridazine (ANOVA).
Figure 2. Effect of thioridazine 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 thioridazine; (B) Arithmetic means ± SEM (n = 6) of the normalized erythrocyte forward scatter (FSC) following incubation for 48 h to Ringer solution without (white bar) or with (black bars) thioridazine (6–30 µM); *** (p < 0.001) indicates significant difference from the absence of thioridazine (ANOVA).
Toxins 05 01918 g002
Figure 3. Effect of thioridazine 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 thioridazine; (B) Arithmetic means ± SEM (n = 6) of the Fluo3 fluorescence (arbitrary units) in erythrocytes exposed for 48 h to Ringer solution without (white bar) or with (black bars) thioridazine (6–30 µM); *** (p < 0.001) indicates significant difference from the absence of thioridazine (ANOVA).
Figure 3. Effect of thioridazine 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 thioridazine; (B) Arithmetic means ± SEM (n = 6) of the Fluo3 fluorescence (arbitrary units) in erythrocytes exposed for 48 h to Ringer solution without (white bar) or with (black bars) thioridazine (6–30 µM); *** (p < 0.001) indicates significant difference from the absence of thioridazine (ANOVA).
Toxins 05 01918 g003
Figure 4. Effect of Ca2+ withdrawal on thioridazine induced annexin-V-binding.Arithmetic means ± SEM (n = 7) 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 thioridazine in the presence (left bars, +Ca) and absence (right bars, −Ca) of calcium. *** (p < 0.001) indicates significant difference from the absence of thioridazine (ANOVA); # (p < 0.05) indicates significant difference from the respective values in the presence of Ca2+ (ANOVA).
Figure 4. Effect of Ca2+ withdrawal on thioridazine induced annexin-V-binding.Arithmetic means ± SEM (n = 7) 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 thioridazine in the presence (left bars, +Ca) and absence (right bars, −Ca) of calcium. *** (p < 0.001) indicates significant difference from the absence of thioridazine (ANOVA); # (p < 0.05) indicates significant difference from the respective values in the presence of Ca2+ (ANOVA).
Toxins 05 01918 g004
In order to explore whether the additional mechanisms could include p38 kinase, erythrocytes were exposed in further experiments to 6–30 µM of thioridazine for 48 h in either the presence or absence of the p38 kinase inhibitor SB203580 (2 µM). As shown in Figure 5, the effect of thioridazine on annexin-V-binding was significantly blunted in the presence of SB203580. However, even in the presence of SB203580, thioridazine still significantly increased the percentage of annexin-V-binding erythrocytes.
Additional experiments were performed in the absence and presence of pancaspase inhibitor zVAD (10 µM). As illustrated in Figure 6, a 48 h exposure to thioridazine (30 µM) increased the percentage of annexin V binding erythrocytes to a similar value in the presence and absence of zVAD. Further experiments were performed in the absence and presence of antioxidant N-acetyl-cysteine (1 mM). As a result, a 48 h exposure to thioridazine (30 µM) increased the percentage of annexin V binding erythrocytes again to a similar value in the presence and absence of N-acetyl-cysteine.
The present study demonstrates a novel effect of thioridazine, i.e., the stimulation of eryptosis, the suicidal erythrocyte death. Exposure of human erythrocytes to thioridazine decreased cell volume and triggered erythrocyte of eryptosis. The concentrations required to trigger eryptosis were within the range of concentrations (6 µg/mL ≈ 15 µM) encountered in vivo [76].
Thioridazine influenced erythrocyte cell volume most likely by stimulating entry and/or impairing extrusion of Ca2+ with subsequent increase of cytosolic Ca2+ concentration ([Ca2+]i), activation of Ca2+ sensitive K+ channels [14,77], K+ exit, cell membrane hyperpolarisation, Cl exit and cellular loss of KCl with osmotically obliged water [10]. The Ca2+ entry may have been accomplished by activation of the endogenous Ca2+ permeable non-selective cation channels [11], which are known to be activated by oxidative stress [78]. Ca2+ is extruded by the Ca2+ ATPase, which has previously been shown to be inhibited by thioridazine [79]. Stimulation of extracellular Ca2+ entry with subsequent increase of [Ca2+]i further contributed to the triggering of cell membrane scrambling by thioridazine. However, even in the absence of extracellular Ca2+ thioridazine still significantly triggered cell membrane scrambling, an observation pointing to the involvement of additional mechanisms. Mechanisms mediating the stimulation of eryptosis following osmotic shock include p38 kinase [27], which is expressed in human erythrocytes and is activated by hyperosmotic shock, a known trigger of eryptosis [27]. As shown earlier [27], p38 kinase is phosphorylated upon osmotic shock and inhibitors of the kinase significantly blunt the decrease of forward scatter and the increase of annexin-V-binding following osmotic shock. Inhibition of p38 kinase significantly blunted but did not abrogate thioridazine induced cell membrane scrambling again indicating that p38 kinase contributes to but does not fully account for the stimulation of eryptosis. Thus, the present results do not rule out the involvement of further cellular mechanisms paticipating in the triggering of eryptosis by thioridazine.
Whatever mechanisms involved in the triggering of phosphatidylserine translocation by thioridazine, phosphatidylserine exposed at the surface of eryptotic cells fosters the binding of the affected erythrocytes to phagocytosing cells with subsequent engulfment and thus clearance of those cells [16]. The clearance of eryptotic erythrocytes from circulating blood may lead to anemia [9].
Figure 5. Effect of thioridazine on phosphatidylserine exposure in the presence or absence of p38 kinase inhibitor SB203580. Arithmetic means ± SEM (n = 6) of erythrocyte annexin-V-binding following incubation for 48 h to Ringer solution without or with presence of thioridazine (6–30 µM) in the absence (white bars) or presence (black bars) of 2 µM SB203580. ** (p < 0.01); *** (p < 0.001) indicate significant differences from the absence of thioridazine (ANOVA); # (p < 0.05); ### (p < 0.001) indicate significant differences from the absence of SB203580 (ANOVA).
Figure 5. Effect of thioridazine on phosphatidylserine exposure in the presence or absence of p38 kinase inhibitor SB203580. Arithmetic means ± SEM (n = 6) of erythrocyte annexin-V-binding following incubation for 48 h to Ringer solution without or with presence of thioridazine (6–30 µM) in the absence (white bars) or presence (black bars) of 2 µM SB203580. ** (p < 0.01); *** (p < 0.001) indicate significant differences from the absence of thioridazine (ANOVA); # (p < 0.05); ### (p < 0.001) indicate significant differences from the absence of SB203580 (ANOVA).
Toxins 05 01918 g005
Phosphatidylserine exposing erythrocytes may further adhere to endothelial CXCL16/SR-PSO of the vascular wall [80] and, thus, interfere with blood flow [80,81,82,83,84,85]. Phosphatidylserine exposing erythrocytes could in addition foster blood clotting and, thus, cause thrombosis [81,86,87].
Besides causing anemia [7,8], thioridazine intoxication leads to impairment of consciousness, cardiac arrhythmia and subsequent cardiac failure with pulmonary edema, severe hypotension, and renal failure [88,89,90]. At least in theory, similar mechanisms may be effective in the derangement of cardiac excitation and suicidal erythrocyte death. Notably, pathogenesis of cardiac arrhythmia may involve activation of p38 kinase [91,92]. However, whether or not thioridazine activates p38 kinase in nucleated cells, remains to be shown.
Figure 6. Effect of thioridazine on phosphatidylserine exposure in the presence or absence of pancapsase inhibitor zVAD or antioxidant N-acetylcysteine. Arithmetic means ± SEM (n = 4 each) of erythrocyte annexin-V-binding following incubation for 48 h to Ringer solution without (white bars) or with (black bars) presence of thioridazine (30 µM) in the absence (control, left bars) or presence of 10 µM zVAD (middle bars) or 1 mM N-acetylcysteine (right bars). ** (p < 0.01); *** (p < 0.001) indicate significant differences from the absence of thioridazine (ANOVA); # (p < 0.05); ### (p < 0.001) indicate significant differences from the absence of SB203580 (ANOVA).
Figure 6. Effect of thioridazine on phosphatidylserine exposure in the presence or absence of pancapsase inhibitor zVAD or antioxidant N-acetylcysteine. Arithmetic means ± SEM (n = 4 each) of erythrocyte annexin-V-binding following incubation for 48 h to Ringer solution without (white bars) or with (black bars) presence of thioridazine (30 µM) in the absence (control, left bars) or presence of 10 µM zVAD (middle bars) or 1 mM N-acetylcysteine (right bars). ** (p < 0.01); *** (p < 0.001) indicate significant differences from the absence of thioridazine (ANOVA); # (p < 0.05); ### (p < 0.001) indicate significant differences from the absence of SB203580 (ANOVA).
Toxins 05 01918 g006
On the other hand, triggering of eryptosis may result in the clearance of defective erythrocytes prior to rupture of the cell membrane and thus prevent release of cellular hemoglobin, which is filtered in renal glomerula and subsequently occludes renal tubules [93].

3. Experimental Section

3.1. Erythrocytes, Solutions and Chemicals

Leukocyte-depleted erythrocytes were kindly provided by the blood bank of the University of Tuebingen. The study is approved by the ethics committee of the University of Tuebingen (184/2003V). 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), 5 glucose, 1 CaCl2; pH 7.4 at 37 °C for 48 h. Where indicated, erythrocytes were exposed to thioridazine (Sigma, Aldrich, Germany) at the indicated concentrations (dissolved in 0.001% DMSO). In Ca2+-free Ringer solution, 1 mM CaCl2 was substituted by 1 mM glycol-bis(2-aminoethylether)-N,N,N',N'-tetraacetic acid (EGTA) and/or p38 kinase inhibited by addition of SB203580 (2 μM; Tocris, Bristol, UK).

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

After incubation under the respective experimental condition, 50 µL cell suspension were 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 utilizing 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 150 µL Ringer. Following this, Ca2+-dependent fluorescence intensity was measured with an excitation wavelength of 488 nm and an emission wavelength of 530 nm utilizing a FACS Calibur (BD, Heidelberg, Germany).

3.4. Measurement of Hemolysis

For the determination of hemolysis the samples were centrifuged (3 min at 400 g, 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.

3.5. Statistics

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

4. Conclusions

In conclusion, thioridazine is shown to stimulate eryptosis, which is characterized by cell membrane scrambling and cell shrinkage. The substance is partially effective by activation of p38 kinase and by increase of cytosolic Ca2+ concentration.

Acknowledgments

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

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Purhonen, M.; Koponen, H.; Tiihonen, J.; Tanskanen, A. Outcome of patients after market withdrawal of thioridazine: A retrospective analysis in a nationwide cohort. Pharmacoepidemiol. Drug Saf. 2012, 21, 1227–1231. [Google Scholar] [CrossRef]
  2. Thanacoody, R.H. Thioridazine: The good and the bad. Recent Pat. Antiinfect. Drug Discov. 2011, 6, 92–98. [Google Scholar] [CrossRef]
  3. Amaral, L. Thioridazine: An old neuroleptic effective against totally drug resistant tuberculosis. Acta Med. Port. 2012, 25, 118–121. [Google Scholar]
  4. Amaral, L.; Molnar, J. Potential therapy of multidrug-resistant and extremely drug-resistant tuberculosis with thioridazine. In Vivo 2012, 26, 231–236. [Google Scholar]
  5. Amaral, L.; Viveiros, M. Why thioridazine in combination with antibiotics cures extensively drug-resistant mycobacterium tuberculosis infections. Int. J. Antimicrob. Agents. 2012, 39, 376–380. [Google Scholar] [CrossRef]
  6. Sohaskey, C. Latent tuberculosis: Is there a role for thioridazine? Recent Pat. Antiinfect. Drug Discov. 2011, 6, 139–146. [Google Scholar] [CrossRef]
  7. Cooper, J.W., Jr.; Pesnell, L.H. Thioridazine-associated immune hemolytic anemia. South Med. J. 1978, 71, 1443–1444. [Google Scholar] [CrossRef]
  8. King, D.J.; Wager, E. Haematological safety of antipsychotic drugs. J. Psychopharmacol. 1998, 12, 283–288. [Google Scholar] [CrossRef]
  9. Lang, F.; Gulbins, E.; Lerche, H.; Huber, S.M.; Kempe, D.S.; Föller, M. Eryptosis, a window to systemic disease. Cell. Physiol. Biochem. 2008, 22, 373–380. [Google Scholar] [CrossRef]
  10. 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] [CrossRef]
  11. Föller, M.; Kasinathan, R.S.; Koka, S.; Lang, C.; Shumilina, E.; Birnbaumer, L.; Lang, F.; Huber, S.M. Trpc6 contributes to the Ca2+ leak of human erythrocytes. Cell. Physiol. Biochem. 2008, 21, 183–192. [Google Scholar] [CrossRef]
  12. Föller, 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] [CrossRef]
  13. Lang, P.A.; Kaiser, S.; Myssina, S.; Birka, C.; Weinstock, C.; Northoff, H.; Wieder, T.; Lang, F.; Huber, S.M. Effect of vibrio parahaemolyticus haemolysin on human erythrocytes. Cell. Microbiol. 2004, 6, 391–400. [Google Scholar] [CrossRef]
  14. Brugnara, C.; de Franceschi, L.; Alper, S.L. Inhibition of Ca2+-dependent K+ transport and cell dehydration in sickle erythrocytes by clotrimazole and other imidazole derivatives. J. Clin. Invest. 1993, 92, 520–526. [Google Scholar] [CrossRef]
  15. Berg, C.P.; Engels, I.H.; Rothbart, A.; Lauber, K.; Renz, A.; Schlosser, S.F.; Schulze-Osthoff, K.; Wesselborg, S. Human mature red blood cells express caspase-3 and caspase-8, but are devoid of mitochondrial regulators of apoptosis. Cell. Death. Differ. 2001, 8, 1197–1206. [Google Scholar] [CrossRef]
  16. Lang, F.; Gulbins, E.; Lang, P.A.; Zappulla, D.; Föller, M. Ceramide in suicidal death of erythrocytes. Cell. Physiol. Biochem. 2010, 26, 21–28. [Google Scholar] [CrossRef]
  17. 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]
  18. Bhavsar, S.K.; Bobbala, D.; Xuan, N.T.; Föller, M.; Lang, F. Stimulation of suicidal erythrocyte death by alpha-lipoic acid. Cell. Physiol. Biochem. 2010, 26, 859–868. [Google Scholar] [CrossRef]
  19. Föller, M.; Huber, S.M.; Lang, F. Erythrocyte programmed cell death. IUBMB Life 2008, 60, 661–668. [Google Scholar] [CrossRef]
  20. Föller, M.; Mahmud, H.; Gu, S.; Wang, K.; Floride, E.; Kucherenko, Y.; Luik, S.; Laufer, S.; Lang, F. Participation of leukotriene C4 in the regulation of suicidal erythrocyte death. J. Physiol. Pharmacol. 2009, 60, 135–143. [Google Scholar]
  21. 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] [CrossRef]
  22. 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] [CrossRef]
  23. Föller, 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] [CrossRef] [Green Version]
  24. 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] [CrossRef]
  25. Kucherenko, Y.; Zelenak, C.; Eberhard, M.; Qadri, S.M.; Lang, F. Effect of casein kinase 1 α activator pyrvinium pamoate on erythrocyte ion channels. Cell. Physiol. Biochem. 2012, 30, 407–417. [Google Scholar] [CrossRef]
  26. Zelenak, C.; Eberhard, M.; Jilani, K.; Qadri, S.M.; Macek, B.; Lang, F. Protein kinase CK1α regulates erythrocyte survival. Cell. Physiol. Biochem. 2012, 29, 171–180. [Google Scholar] [CrossRef]
  27. 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] [CrossRef]
  28. Zelenak, C.; Föller, 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] [CrossRef]
  29. Lupescu, A.; Shaik, N.; Jilani, K.; Zelenak, C.; Lang, E.; Pasham, V.; Zbidah, M.; Plate, A.; Bitzer, M.; Föller, 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] [CrossRef]
  30. Shaik, N.; Lupescu, A.; Lang, F. Sunitinib-sensitive suicidal erythrocyte death. Cell. Physiol. Biochem. 2012, 30, 512–522. [Google Scholar] [CrossRef]
  31. 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] [CrossRef]
  32. Abed, M.; Towhid, S.T.; Shaik, N.; Lang, F. Stimulation of suicidal death of erythrocytes by rifampicin. Toxicology 2012, 302, 123–128. [Google Scholar] [CrossRef]
  33. 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] [CrossRef]
  34. Felder, K.M.; Hoelzle, K.; Ritzmann, M.; Kilchling, T.; Schiele, D.; Heinritzi, K.; Groebel, K.; Hoelzle, L.E. Hemotrophic mycoplasmas induce programmed cell death in red blood cells. Cell. Physiol. Biochem. 2011, 27, 557–564. [Google Scholar] [CrossRef] [Green Version]
  35. 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, 316–384. [Google Scholar]
  36. 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] [CrossRef]
  37. 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 Ca2+ rise and membrane permeabilization. Arch. Toxicol. 2012, 86, 741–752. [Google Scholar] [CrossRef]
  38. 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] [CrossRef]
  39. Ghashghaeinia, M.; Toulany, M.; Saki, M.; Bobbala, D.; Fehrenbacher, B.; Rupec, R.; Rodemann, H.P.; Ghoreschi, K.; Rocken, M.; Schaller, M.; et al. The nfkb pathway inhibitors bay 11-7082 and parthenolide induce programmed cell death in anucleated erythrocytes. Cell. Physiol. Biochem. 2011, 27, 45–54. [Google Scholar] [CrossRef]
  40. 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] [CrossRef]
  41. 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 2012, 27, 52–58. [Google Scholar]
  42. 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] [CrossRef]
  43. 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] [CrossRef]
  44. 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]
  45. Lang, F.; Qadri, S.M. Mechanisms and significance of eryptosis, the suicidal death of erythrocytes. Blood Purif. 2012, 33, 125–130. [Google Scholar] [CrossRef]
  46. Lupescu, A.; Jilani, K.; Zbidah, M.; Lang, E.; Lang, F. Enhanced Ca2+ entry, ceramide formation, and apoptotic death of erythrocytes triggered by plumbagin. J. Nat. Prod. 2012, 75, 1956–1961. [Google Scholar] [CrossRef]
  47. Lupescu, A.; Jilani, K.; Zbidah, M.; Lang, F. Induction of apoptotic erythrocyte death by rotenone. Toxicology 2012, 300, 132–137. [Google Scholar] [CrossRef]
  48. Lupescu, A.; Jilani, K.; Zelenak, C.; Zbidah, M.; Qadri, S.M.; Lang, F. Hexavalent chromium-induced erythrocyte membrane phospholipid asymmetry. Biometals 2012, 25, 309–318. [Google Scholar] [CrossRef]
  49. Polak-Jonkisz, D.; Purzyc, L. Ca influx versus efflux during eryptosis in uremic erythrocytes. Blood Purif. 2012, 34, 209–210. [Google Scholar] [CrossRef]
  50. Qadri, S.M.; Bauer, J.; Zelenak, C.; Mahmud, H.; Kucherenko, Y.; Lee, S.H.; Ferlinz, K.; Lang, F. Sphingosine but not sphingosine-1-phosphate stimulates suicidal erythrocyte death. Cell. Physiol. Biochem. 2011, 28, 339–346. [Google Scholar] [CrossRef]
  51. Qadri, S.M.; Kucherenko, Y.; Lang, F. Beauvericin induced erythrocyte cell membrane scrambling. Toxicology 2011, 283, 24–31. [Google Scholar] [CrossRef]
  52. Qadri, S.M.; Kucherenko, Y.; Zelenak, C.; Jilani, K.; Lang, E.; Lang, F. Dicoumarol activates Ca2+-permeable cation channels triggering erythrocyte cell membrane scrambling. Cell. Physiol. Biochem. 2011, 28, 857–864. [Google Scholar] [CrossRef]
  53. 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] [CrossRef]
  54. Shaik, N.; Zbidah, M.; Lang, F. Inhibition of Ca2+ entry and suicidal erythrocyte death by naringin. Cell. Physiol. Biochem. 2012, 30, 678–686. [Google Scholar] [CrossRef]
  55. 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. 2012, 65, 145–157. [Google Scholar]
  56. Weiss, E.; Cytlak, U.M.; Rees, D.C.; Osei, A.; Gibson, J.S. Deoxygenation-induced and Ca2+ dependent phosphatidylserine externalisation in red blood cells from normal individuals and sickle cell patients. Cell. Calcium. 2012, 51, 51–56. [Google Scholar] [CrossRef]
  57. Zappulla, D. Environmental stress, erythrocyte dysfunctions, inflammation, and the metabolic syndrome: Adaptations to CO2 increases? Cardiometab. Syndr. 2008, 3, 30–34. [Google Scholar] [CrossRef]
  58. Zbidah, M.; Lupescu, A.; Jilani, K.; Lang, F. Stimulation of suicidal erythrocyte death by fumagillin. Basic Clin. Pharmacol. Toxicol. 2012, 112, 346–351. [Google Scholar]
  59. Zbidah, M.; Lupescu, A.; Shaik, N.; Lang, F. Gossypol-induced suicidal erythrocyte death. Toxicology 2012, 302, 101–105. [Google Scholar] [CrossRef]
  60. 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] [CrossRef]
  61. Lang, E.; Qadri, S.M.; Lang, F. Killing me softly—Suicidal erythrocyte death. Int. J. Biochem. Cell. Biol. 2012, 44, 1236–1243. [Google Scholar] [CrossRef]
  62. 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] [CrossRef]
  63. 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] [CrossRef]
  64. 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] [CrossRef]
  65. 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. 2006, 84, 378–388. [Google Scholar] [CrossRef]
  66. 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]
  67. Bobbala, D.; Alesutan, I.; Föller, M.; Huber, S.M.; Lang, F. Effect of anandamide in plasmodium berghei-infected mice. Cell. Physiol. Biochem. 2010, 26, 355–362. [Google Scholar] [CrossRef]
  68. Föller, 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] [CrossRef]
  69. Koka, S.; Bobbala, D.; Lang, C.; Boini, K.M.; Huber, S.M.; Lang, F. Influence of paclitaxel on parasitemia and survival of plasmodium berghei infected mice. Cell. Physiol. Biochem. 2009, 23, 191–198. [Google Scholar] [CrossRef]
  70. 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] [CrossRef]
  71. Siraskar, B.; Ballal, A.; Bobbala, D.; Föller, M.; Lang, F. Effect of amphotericin B on parasitemia and survival of plasmodium berghei-infected mice. Cell. Physiol. Biochem. 2010, 26, 347–354. [Google Scholar] [CrossRef]
  72. Lang, P.A.; Kasinathan, R.S.; Brand, V.B.; Duranton, C.; Lang, C.; Koka, S.; Shumilina, E.; Kempe, D.S.; Tanneur, V.; Akel, A.; et al. Accelerated clearance of plasmodium-infected erythrocytes in sickle cell trait and annexin-a7 deficiency. Cell. Physiol. Biochem. 2009, 24, 415–428. [Google Scholar] [CrossRef]
  73. 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]
  74. Qadri, S.M.; Mahmud, H.; Lang, E.; Gu, S.; Bobbala, D.; Zelenak, C.; Jilani, K.; Siegfried, A.; Föller, 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] [CrossRef]
  75. 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. Pflüg. Arch. 2004, 448, 471–477. [Google Scholar]
  76. Gottschalk, L.A.; Biener, R.; Noble, E.P.; Birch, H.; Wilbert, D.E.; Heiser, J.F. Thioridazine plasma levels and clinical response. Compr. Psychiatry 1975, 16, 323–337. [Google Scholar] [CrossRef]
  77. Bookchin, R.M.; Ortiz, O.E.; Lew, V.L. Activation of calcium-dependent potassium channels in deoxygenated sickled red cells. Prog. Clin. Biol. Res. 1987, 240, 193–200. [Google Scholar]
  78. Brand, V.B.; Sandu, C.D.; Duranton, C.; Tanneur, V.; Lang, K.S.; Huber, S.M.; Lang, F. Dependence of plasmodium falciparum in vitro growth on the cation permeability of the human host erythrocyte. Cell. Physiol. Biochem. 2003, 13, 347–356. [Google Scholar] [CrossRef]
  79. Palacios, J.; Sepulveda, M.R.; Lee, A.G.; Mata, A.M. Ca2+ transport by the synaptosomal plasma membrane Ca2+-atpase and the effect of thioridazine. Biochemistry 2004, 43, 2353–2358. [Google Scholar] [CrossRef]
  80. Borst, O.; Abed, M.; Alesutan, I.; Towhid, S.T.; Qadri, S.M.; Föller, 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] [CrossRef]
  81. Andrews, D.A.; Low, P.S. Role of red blood cells in thrombosis. Curr. Opin. Hematol. 1999, 6, 76–82. [Google Scholar] [CrossRef]
  82. 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] [CrossRef]
  83. 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] [CrossRef]
  84. 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]
  85. 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]
  86. 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]
  87. 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] [CrossRef]
  88. Le Blaye, I.; Donatini, B.; Hall, M.; Krupp, P. Acute overdosage with thioridazine: A review of the available clinical exposure. Vet. Hum. Toxicol. 1993, 35, 147–150. [Google Scholar]
  89. Isbister, G.K.; Balit, C.R.; Kilham, H.A. Antipsychotic poisoning in young children: A systematic review. Drug Saf. 2005, 28, 1029–1044. [Google Scholar] [CrossRef]
  90. Schmidt, W.; Lang, K. Life-threatening dysrhythmias in severe thioridazine poisoning treated with physostigmine and transient atrial pacing. Crit. Care Med. 1997, 25, 1925–1930. [Google Scholar] [CrossRef]
  91. Lee, Y.M.; Cheng, P.Y.; Chen, S.Y.; Chung, M.T.; Sheu, J.R. Wogonin suppresses arrhythmias, inflammatory responses, and apoptosis induced by myocardial ischemia/reperfusion in rats. J. Cardiovasc. Pharmacol. 2011, 58, 133–142. [Google Scholar] [CrossRef]
  92. Surinkaew, S.; Kumphune, S.; Chattipakorn, S.; Chattipakorn, N. Inhibition of p38 mapk during ischemia, but not reperfusion, effectively attenuates fatal arrhythmia in ischemia/reperfusion heart. J. Cardiovasc. Pharmacol. 2013, 61, 133–141. [Google Scholar] [CrossRef]
  93. 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] [CrossRef]

Share and Cite

MDPI and ACS Style

Lang, E.; Modicano, P.; Arnold, M.; Bissinger, R.; Faggio, C.; Abed, M.; Lang, F. Effect of Thioridazine on Erythrocytes. Toxins 2013, 5, 1918-1931. https://doi.org/10.3390/toxins5101918

AMA Style

Lang E, Modicano P, Arnold M, Bissinger R, Faggio C, Abed M, Lang F. Effect of Thioridazine on Erythrocytes. Toxins. 2013; 5(10):1918-1931. https://doi.org/10.3390/toxins5101918

Chicago/Turabian Style

Lang, Elisabeth, Paola Modicano, Markus Arnold, Rosi Bissinger, Caterina Faggio, Majed Abed, and Florian Lang. 2013. "Effect of Thioridazine on Erythrocytes" Toxins 5, no. 10: 1918-1931. https://doi.org/10.3390/toxins5101918

Article Metrics

Back to TopTop