Skip to main content
Transfusion Medicine and Hemotherapy logoLink to Transfusion Medicine and Hemotherapy
. 2012 Sep 6;39(5):308–314. doi: 10.1159/000342534

Physiology and Pathophysiology of Eryptosis

Florian Lang 1,*, Elisabeth Lang 1, Michael Föller 1
PMCID: PMC3678267  PMID: 23801921

Abstract

Summary

Suicidal erythrocyte death (eryptosis) is characterized by cell shrinkage, cell membrane blebbing, and cell membrane phospholipid scrambling with phosphatidylserine exposure at the cell surface. Eryptotic cells adhere to the vascular wall and are rapidly cleared from circulating blood. Eryptosis is stimulated by an increase in cytosolic Ca2+ activity, ceramide, hyperosmotic shock, oxidative stress, energy depletion, hyperthermia, and a wide variety of xenobiotics and endogenous substances. Inhibitors of eryptosis include erythropoietin and nitric oxide. Enhanced eryptosis is observed in diabetes, renal insufficiency, hemolytic uremic syndrome, sepsis, mycoplasma infection, malaria, iron deficiency, sickle cell anemia, beta-thalassemia, glucose-6-phosphate dehydrogenase-(G6PD) deficiency, hereditary spherocytosis, paroxysmal nocturnal hemoglobinuria, Wilson's disease, myelodysplastic syndrome, and phosphate depletion. Eryptosis is further enhanced in gene-targeted mice with deficient annexin 7, cGMP-dependent protein kinase type I (cGKI), AMP-activated protein kinase (AMPK), anion exchanger 1 (AE1), adenomatous polyposis coli (APC), and Klotho, as well as in mouse models of sickle cell anemia and thalassemia. Decreased eryptosis is observed in mice with deficient phosphoinositide-dependent kinase 1 (PDK1), platelet activating factor (PAF) receptor, transient receptor potential channel 6 (TRPC6), janus kinase 3 (JAK3), and taurine transporter (TAUT). Eryptosis may be a useful mechanism to remove defective erythrocytes prior to hemolysis. Excessive eryptosis may, however, compromise microcirculation and lead to anemia.

KeyWords: Erythrocytes, Apoptosis, Anemia, Malaria, Iron deficiency, HUS, Sepsis, Renal insufficiency, Diabetes

Introduction

In the absence of injury, the erythrocyte lifespan approaches 100–120 days [1]. It is ultimately limited by senescence [2] which results from binding of modified hemoglobin to band 3, followed by modification of band 3, binding of autologous immunoglobulin G (IgG), disruption of the band 3-dependent anchorage of the cytoskeleton to the lipid bilayer, and formation of vesicles that expose cell antigens and phosphatidylserine at their surface [2]. The vesicles are subsequently bound to scavenger receptors, engulfed, and thus removed [2]. Erythrocytes may experience survival-threatening injury prior to senescence. In that case, they may enter programmed cell death or eryptosis [3]. Similar to programmed death of nucleated cells or apoptosis, eryptosis is a coordinated suicidal death eventually leading to disposal of defective cells without rupture of the cell membrane and release of intracellular material [3]. In contrast to nucleated cells, however, erythrocytes lack nuclei and mitochondria [1] which actively participate in the machinery underlying apoptosis [4, 5]. Eryptosis thus lacks several hallmarks of apoptosis, such as mitochondrial depolarization and condensation of nuclei. Moreover, the signaling eventually leading to eryptosis [3] is not identical to that underlying apoptosis [4, 5]. Nevertheless, eryptosis shares several features of apoptosis, such as cell shrinkage, cell membrane blebbing, and cell membrane scrambling with phosphatidylserine exposure at the cell surface [3]. Similar to apoptotic cells and particles, eryptotic cells and particles are engulfed and degraded by macrophages [3, 6]. Also similar to apoptosis, eryptosis allows removal of defective, infected, or otherwise potentially harmful cells. The present review briefly describes mechanisms known to trigger and inhibit eryptosis. Due to limitation of space, a previous review [3] is cited instead of earlier original papers.

Triggers and Signaling of Eryptosis

Eryptosis is stimulated by an increase in cytosolic Ca2+ activity which may be the consequence of increased Ca2+ entry across the erythrocyte cell membrane [3]. Erythrocytes express Ca2+-permeable non-selective cation channels. The molecular identity of those channels has remained ill-defined but somehow involves transient receptor potential channel 6 TRPC6 [3]. Along those lines, Ca2+ entry is blunted in erythrocytes drawn from TRPC6 knockout mice [3]. Stimulators of the non-selective cation channels include oxidative stress, Cl removal, and hyperosmotic shock [3]. Osmotic shock is in part effective by stimulating the release of prostaglandin E2 which in turn activates the cation channels [3]. An increase in cytosolic Ca2+ is followed by activation of Ca2+-sensitive K+ channels [3] with subsequent exit of K+, hyperpolarization of the cell membrane, Cl exit, cellular loss of KCl, and exit of osmotically obliged water resulting in cell shrinkage [3]. In addition, Ca2+ activates calpain, a cysteine endopeptidase degrading cytoskeletal proteins and thus fostering cell membrane blebbing [3]. Increased cytosolic Ca2+ activity further leads to stimulation of cell membrane scrambling with breakdown of phospholipid asymmetry of the cell membrane and phosphatidylserine exposure at the cell surface [3]. Cell shrinkage augments the Ca2+-induced cell membrane scrambling [3].

The Ca2+ sensitivity of erythrocyte cell membrane scrambling is enhanced by ceramide which, similar to cellular Ca2+ accumulation, stimulates eryptosis. Ceramide may be generated by acid sphingomyelinase which produces ceramide from membrane sphingomyelin [3]. Stimulators of acid sphingomyelinase include platelet-activating factor (PAF) which is formed by phospholipase-dependent degradation of cell membrane lipids [3]. A stimulator of phospholipase is hyperosmotic shock [3]. Accordingly, eryptosis following osmotic shock is blunted in gene-targeted mice lacking PAF receptors [3].

Erythrocytes express p38 kinase which is activated by hyperosmotic shock [7]. The p38 kinase inhibitors SB203580 and p38 Inh III blunt the eryptosis following osmotic shock [7]. Erythrocytes further express casein kinase 1 (CK1). The CK1 inhibitors D4476 and (R)-DRF053 blunt eryptosis following energy depletion or oxidative stress [8]. Conversely, eryptosis is stimulated by the CK1α-specific activator pyrvinium pamoate [8]. CK1 is effective by activation of Cl-sensitive Ca2+-permeable cation channels [8]. On the other hand, pyrvinium pamoate inhibits Ca2+-activated K+ channels and thus counteracts eryptotic cell shrinkage [9].

Erythrocytes further express Janus kinase 3 (JAK3) which is phosphorylated and thus activated following glucose depletion [10]. Erythrocytes from JAK3-deficient mice are slightly smaller than erythrocytes from wild-type mice. Genetic knockout of JAK3 or treatment with JAK3 inhibitors WHI-P131/JANEX-1 (4-(4′-hydroxyphenyl)-amino-6.7-dimethoxyquinazoline) and WHI-P154 (4-(3′-bromo-4′-hydroxyphenyl) amino-6.7-dimethoxyquinazoline) do not significantly modify cell membrane scrambling in the absence of triggers of eryptosis, but counteract eryptosis following energy depletion [10].

Eryptosis is further stimulated following activation of protein kinase C (PKC) [3]. Similar to JAK3, PKC is activated during energy depletion, a powerful stimulator of eryptosis [3]. Inhibition of PKC blunts eryptosis following energy depletion [3].

Another stimulator of eryptosis is oxidative stress which is at least in part effective by opening of the unspecific cation channels with subsequent Ca2+ entry, by activation of erythrocyte oxidation-sensitive Cl channels, and by activation of caspases [3]. It is noteworthy, however, that caspases are not required for stimulation of eryptosis following Ca2+ entry. The role of CD95, an important apoptosis-inducing receptor, for erythrocyte phosphatidylserine exposure is controversial [11, 12].

The sensitivity of erythrocytes to suicidal death depends on age. Erythrocytes of different age can be separated according to density which increases with age. Utilizing this method, a positive correlation was found between age and the percentage of erythrocytes undergoing spontaneous cell membrane scrambling [13]. The increase of cell membrane scrambling in aged erythrocytes is reversed in addition of the antioxidant N-acetyl-L-cysteine which increases the in vivo half-life of circulating mouse erythrocytes [13]. Accordingly, the increased susceptibility of aged erythrocytes to eryptosis is at least partially due to increasing sensitivity to oxidative stress.

On the other hand, preferential suicidal death of young erythrocytes or ‘neocytolysis’ has been observed in individuals returning from high altitude or space flight [14]. It has been speculated that the enhanced erythropoietin concentrations as a consequence of high altitude or space flight confer survival of progenitor cells otherwise prone to undergo apoptosis, thus generating erythrocytes particularly sensitive to eryptosis [3]. Those erythrocytes survive at high erythropoietin concentrations but die as soon as erythropoietin concentrations decline. Eryptosis following decreasing erythropoietin concentrations upon return from hypoxic conditions may rapidly readjust the erythrocyte number to the requirements under normoxic conditions. Thus, an otherwise extremely slow negative feedback loop is accelerated. Additional experimental effort is needed to prove or disprove this speculation.

Eryptosis is further stimulated by a myriad of xenobiotics and a wide variety of endogenous substances and challenges (table 1).

Table 1.

Substances stimulating eryptosis

Substance Reference Substance Reference
A23187 (Ca2+ ionophore) [3, 41] Hemin [3]
α-Lipoic acid [42] Hemolysin [3]
Aluminium [43] Immunoglobulin A [55]
Amantadine [3] IPA3 [56]
Amiodarone [3] Lead [3]
Amphotericin B [3] Leukotriene C4 [3]
Amyloid [3] Lipopeptides [3]
Anandamide [3] Listeriolysin [3]
Anti-A IgG [3] Lithium [57]
Apigenin [44] Lysophosphatidic acid [58]
Arsenic [3] Mercury [3]
Azathioprine [3] Methyldopa [3]
Bay 11–7082 [45] Methylglyoxal [3]
Bay-Y5884 [3] Monensin [59]
Beauvericin [46] Oridonin [60]
Benzethonium [47] Paclitaxel [3]
Bismuth chloride [3] PAF [3]
Cadmium [3] Parthenolide [45]
Ceramide (acylsphingosine) [3] Peptidoglycan [3]
Chlorpromazine [3] Phytic acid [61]
Chromium [48] Phorbol-12 myristate-13 acetate [58]
Ciglitazone [3] Polyphyllin [62]
Cisplatin [3] Prostaglandin E2 [3]
Copper [3] Radiocontrast agents [3]
Cordycepin [3] Retinoic acid [3]
Curcumin [3] Selenium (sodium selenite) [3]
Cyclosporine [3] Silver ions [3]
CD95/Fas/ligand [3, 12] Sphingosine [63]
Dicoumarol [49] Thrombospondin-1 receptor CD47 [3]
Dimethylfumarate [50] Thymoquinone [3]
Enniatin A [51] Tin [3]
FTY720 [52] Ursolic acid [64]
Glycation [26] Valinomycin [3]
Glycophorin-C [3] Vanadate [3]
Gold [53] Vitamin K(3) [3]
H2O2 [54] Zinc [3]

Inhibitors of Eryptosis

The unselective Ca2+-permeable cation channels are inhibited by erythropoietin which thus may counteract Ca2+ entry and eryptosis [3]. Administration of erythropoietin to patients on hemodialyis thus decreases the percentage of circulating eryptotic erythrocytes [3]. The effect of erythropoietin is rapid and reversible. Ca2+ entry and subsequent eryptosis following energy depletion are further counteracted by endothelin 1 and the endothelin B (ETB) receptor agonist sarafotoxin, while endothelin 2 or endothelin 3 do not appreciably modify eryptosis [15]. Mice lacking the ETB receptor suffer from increased eryptosis leading to splenomegaly and compensatory stimulation of erythropoiesis with reticulocytosis [15].

Eryptosis is inhibited by the AMP-activated kinase (AMPK) which is activated by energy depletion [3]. Similar to ETB receptor knockout mice, AMPK1α-deficient mice suffer from excessive eryptosis with splenomegaly and reticulocytosis [3]. Activation of AMPK is able to blunt but not abrogate eryptosis upon energy depletion.

Eryptosis is further inhibited by cGMP-dependent protein kinase type I (cGKI) [3]. Gene-targeted mice lacking cGKI thus suffer from excessive eryptosis [3]. Dibuturyl-cGMP blunts the Ca2+-induced, but not the ceramide-induced, stimulation of cell membrane scrambling [16]. cGK1 is activated by nitric oxide (NO), a powerful inhibitor of erythrocyte cell membrane scrambling [3]. The NO donors nitroprusside and papanonoate abrogate the cell membrane scrambling following treatment with the Ca2+ ionophore ionomycin without preventing the increase in cytosolic Ca2+ activity [3]. Accordingly, NO disrupts at least in part the effect of Ca2+ on cell membrane scrambling. Excessive nitroprusside concentrations, however, trigger eryptosis [3]. Ionomycin decreases thioredoxin activity and abundance of nitrosylated protein, effects reversed by the NO donor papanonoate [3].

Eryptosis is also inhibited by adenosine [3], amitriptyline [3], blebbistatin [17], caffeine [3], catecholamines such as isoproterenol [3], chloride [3], EIPA [3], endothelin 1 (see above), erythropoietin [3], flufenamic acid [3], NBQX/CNQX [3], niflumic acid [3], NO (nitroprusside, see above), NPPB [3], phlorhizin [18], resveratrol [3], sarafotoxin 6c (see above), staurosporine [3], thymol [3], urea [3], WHI-P131/JANEX-1 [10], WHI-P154 [10], and xanthohumol [3].

Physiological Significance of Eryptosis

Suicidal erythrocyte death accomplishes the clearance of defective erythrocytes prior to hemolysis. The phosphatidylserine at the surface fosters adherence of eryptotic cells to endothelial cells and macrophages with subsequent engulfment and intracellular degradation. Without removal, the defective erythrocytes may undergo hemolysis. Compromised Na+/K+-ATPase activity (e.g. in energy depletion) or increased cell membrane leakiness (e.g. following exposure to toxins) result in cellular gain of Na+ and Cl together with osmotically obliged water and thus lead to cell swelling [3]. Excessive cell swelling is followed by rupture of the cell membrane with release of cellular hemoglobin which may be filtered in the renal glomerula and occlude renal tubules [19]. Activation of Ca2+-sensitive K+ channels during eryptosis hyperpolarizes the cell membrane, thus enhancing the electrical driving force for Clexit. Cellular loss of KCl counteracts cell swelling and delays hemolysis, thus providing some additional time for the clearance of the defective erythrocytes.

Suicidal erythrocyte death further accomplishes the clearance of infected erythrocytes [3]. A major pathogen entering erythrocytes is the malaria parasite Plasmodium [20]. The intracellular pathogen generates oxidative stress resulting in activation of several channels including the unselective cation channels [20]. Pathogen survival depends on Na+ and Ca2+ entry through those channels. However, opening of the channels triggers eryptosis which may eventually lead to clearance of the infected erythrocyte from circulating blood. Accordingly, pathogen-induced eryptosis decreases parasitemia and thus may be an important defense mechanism against Plasmodium infection. Accordingly, several diseases or genetic traits predisposing to eryptosis confer some protection against a severe course of malaria, such as sickle cell trait, beta-thalassemia trait, glucose-6-phosphate dehydrogenase (G6PD) deficiency, and iron deficiency. Moreover, several eryptosis-inducing drugs delay the development of parasitemia and ameliorate the clinical course of malaria, such as paclitaxel, chlorpromazine, cyclosporine, curcumin, PGE2, and lead.

Pathophysiological Significance of Eryptosis

The clearance of eryptotic cells from circulating blood may result in anemia [3], if accelerated eryptosis cannot be compensated by similarly enhanced formation of new erythrocytes. Prior to anemia accelerated eryptosis may be apparent from the enhanced appearance of reticulocytes [3].

Eryptotic cells adhere to endothelial CXCL16/SR-PSO [21] and presumably further, as yet undefined, molecular binding partners. The phosphatidylserine-binding glycoprotein lactadherin confers adhesion of apoptotic lymphocytes but is apparently not important for the clearance of phosphatidylserine-exposing erythrocytes [22]. The adherence of phosphatidylserine-exposing erythrocytes to the vascular wall may impede microcirculation [3]. Phosphatidylserine-exposing erythrocytes further stimulate blood clotting [23]. Thus, excessive eryptosis may foster thrombosis.

Several clinical conditions are associated with enhanced eryptosis [3]. Accelerated eryptosis in iron deficiency may result from decreased erythrocyte volume which sensitizes erythrocytes to triggers of eryptosis [3]. Enhanced eryptosis is further observed in diabetes [24, 25]. Exposure of erythrocytes to excessive glucose concentrations stimulates cell membrane scrambling [26]. Moreover, in diabetes, eryptosis may be stimulated by methylglyoxal (see above). Eryptosis is further enhanced in renal insufficiency [3]. Enhanced eryptosis in renal insufficiency may similarly be in part due to methylglyoxal (see above). Eryptosis is further enhanced following phosphate depletion [3].

Eryptosis is stimulated by hyperthermia and may thus be enhanced during fever [27]. Eryptosis in hyperthermia is at least in part the result of enhanced leukotriene formation and could thus be inhibited by the leukotriene receptor CysLT1 antagonist cinalukast [27]. Excessive eryptosis is observed in blood drawn from septic patients who harbor a factor in their plasma triggering formation of ceramide and stimulating Ca2+ entry, thus leading to erythrocyte cell membrane scrambling and erythrocyte shrinkage [3]. Some pathogens produce eryptosis-inducing molecules such as bacterial sphingomyelinase which may at least contribute to the eryptosis in septic patients [3]. Excessive eryptosis is further observed in hemolytic uremic syndrome (HUS) [3]. Also, plasma isolated from patients with HUS stimulates erythrocyte cell membrane scrambling. The eryptosis in HUS is probably in part the consequence of complement activation [3]. Eryptosis is further stimulated by bacterial hemolysin [3]. The eryptosis in hemolytic disorders presumably serves to clear defective erythrocytes prior to hemolysis (see above). Possibly, hemolysis is only experienced by those erythrocytes that are not cleared away fast enough by a phagocytosing cell.

Eryptosis is triggered following infection with mycoplasma [28] or, as pointed out above, by the malaria pathogen Plasmodium [20, 29, 30, 31, 32, 33, 34]. Intraerythrocytic plasmodia induce oxidative stress which in turn activates the Ca2+-permeable cation channels leading to Ca2+ entry and eryptosis. The phosphatidylserine-exposing erythrocytes are phagocytosed, which leads to the removal not only of the erythrocytes but also of the pathogen (see above). Accordingly, eryptosis counteracts parasitemia. Thus, at least in mice, several eryptosis-inducing substances favorably influence the clinical course of the disease [3, 29, 30, 32]. Moreover, genetic defects in humans and mice resulting in accelerated eryptosis may confer partial protection against a severe course of malaria [20].

Eryptosis is accelerated in sickle cell anemia [3, 35], thalassemia [36], G6PD deficiency [3, 37], defective anion exchanger 1 (AE1) [3], in a GLUT1 mutation turning this carrier into a Ca2+ channel [3], and hereditary spherocytosis [36]. Enhanced eryptosis is further observed in Wilson's disease [3], paroxysmal nocturnal hemoglobinuria [37], and myelodysplastic syndrome [38]. Paroxysmal nocturnal hemoglobinuria and myelodysplastic syndrome affect preferably lighter and thus presumably younger erythrocytes, pointing to enhanced neocytolysis in those disorders [38]. Accelerated eryptosis has been observed in several gene-targeted mice, such as mice mimicking the defect of sickle cell anemia or thalassemia [3]. Enhanced eryptosis is further observed in annexin 7-deficient mice with enhanced formation of PGE2 [39] which activates the cation channels and thus triggers eryptosis (see above). Eryptosis is further enhanced in mice lacking cGKI (see above), AMPK (see above), and ETB receptor (see above). Gene-targeted mice lacking the AE1 similarly suffer from excessive eryptosis leading to anemia despite enhanced formation of new erythrocytes [3]. Erythropoietin-overexpressing mice generate erythrocytes with enhanced sensitivity to eryptotic stimuli [3]. Eryptosis is further enhanced in Klotho-deficient mice [3]. In those mice, the excessive eryptosis could be reversed with a vitamin D-deficient diet, and thus directly or indirectly results from excessive 1.25(OH)2D3 formation [3]. In adenomatous polyposis coli (APC)-deficient mice, eryptosis appears in parallel to the development of intestinal tumors [40].

Decreased eryptosis has been observed in gene-targeted mice lacking phosphoinositide-dependent kinase (PDK1) [3], PAF receptor [3], TRPC6 [3], JAK3 [10], and taurine transporter (TAUT) [3].

Conclusion

Much has been learned in the past few years about cellular mechanisms triggering and counteracting suicidal erythrocyte death or eryptosis. It is obvious that suicidal erythrocyte death is triggered by a myriad of xenobiotics and endogenous challenges and that it participates in the pathophysiology of several clinical conditions. Nevertheless, our knowledge about mechanisms underlying eryptosis is still incomplete. Moreover, a wide variety of further xenobiotics and endogenous small molecules presumably stimulate or inhibit eryptosis. Excessive eryptosis most likely also plays a role in other, yet unrecognized, clinical conditions. Clearly, extensive additional experimental effort is required to fully understand this physiologically and pathophysiologically important fundamental cellular mechanism.

Disclosure Statement

The authors have nothing to disclose.

References

  • 1.Jelkmann W. Functional significance of erythrocytes. In: Lang F, Föller M, editors. Erythrocytes. London: Imperial College Press; 2012. in press. [Google Scholar]
  • 2.Bosman GJ, Willekens FL, Werre JM. Erythrocyte aging: a more than superficial resemblance to apoptosis? Cell Physiol Biochem. 2005;16:1–8. doi: 10.1159/000087725. [DOI] [PubMed] [Google Scholar]
  • 3.Lang F, Gulbins E, Lang PA, Zappulla D, Foller M. Ceramide in suicidal death of erythrocytes. Cell Physiol Biochem. 2010;26:21–28. doi: 10.1159/000315102. [DOI] [PubMed] [Google Scholar]
  • 4.Lanza IR, Nair KS. Mitochondrial function as a determinant of life span. Pflugers Arch. 2010;459:277–289. doi: 10.1007/s00424-009-0724-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Wlodkowic D, Telford W, Skommer J, Darzynkiewicz Z. Apoptosis and beyond: cytometry in studies of programmed cell death. Methods Cell Biol. 2011;103:55–98. doi: 10.1016/B978-0-12-385493-3.00004-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Lang F, Qadri SM. Mechanisms and significance of eryptosis, the suicidal death of erythrocytes. Blood Purif. 2012;33:125–130. doi: 10.1159/000334163. [DOI] [PubMed] [Google Scholar]
  • 7.Gatidis S, Zelenak C, Fajol A, Lang E, Jilani K, Michael D, Qadri SM, Lang F. p38 MAPK activation and function following osmotic shock of erythrocytes. Cell Physiol Biochem. 2011;28:1279–1286. doi: 10.1159/000335859. [DOI] [PubMed] [Google Scholar]
  • 8.Zelenak C, Eberhard M, Jilani K, Qadri SM, Macek B, Lang F. Protein kinase CK1alpha regulates erythrocyte survival. Cell Physiol Biochem. 2012;29:171–180. doi: 10.1159/000337598. [DOI] [PubMed] [Google Scholar]
  • 9.Kucherenko Y, Zelenak C, Eberhard M, Qadri SM, Lang F. Effect of casein kinase 1alpha activator pyrvinium pamoate on erythrocyte ion channels. Cell Physiol Biochem. 2012;30:407–417. doi: 10.1159/000339034. [DOI] [PubMed] [Google Scholar]
  • 10.Bhavsar SK, 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. doi: 10.1159/000329956. [DOI] [PubMed] [Google Scholar]
  • 11.Sagan D, Jermnim N, Tangvarasittichai O. CD95 is not functional in human erythrocytes. Int J Lab Hematol. 2010;32:e244–e247. doi: 10.1111/j.1751-553X.2010.01245.x. [DOI] [PubMed] [Google Scholar]
  • 12.Mandal D, Mazumder A, Das P, Kundu M, Basu J. Fas-, caspase 8-, and caspase 3-dependent signaling regulates the activity of the aminophospholipid translocase and phosphatidylserine externalization in human erythrocytes. J Biol Chem. 2005;280:39460–39467. doi: 10.1074/jbc.M506928200. [DOI] [PubMed] [Google Scholar]
  • 13.Ghashghaeinia M, Cluitmans JC, Akel A, Dreischer P, Toulany M, Koberle M, Skabytska Y, Saki M, Biedermann T, Duszenko M, Lang F, Wieder T, Bosman GJ. The impact of erythrocyte age on eryptosis. Br J Haematol. 2012;157:606–614. doi: 10.1111/j.1365-2141.2012.09100.x. [DOI] [PubMed] [Google Scholar]
  • 14.Rice L, Alfrey CP. The negative regulation of red cell mass by neocytolysis: physiologic and pathophysiologic manifestations. Cell Physiol Biochem. 2005;15:245–250. doi: 10.1159/000087234. [DOI] [PubMed] [Google Scholar]
  • 15.Foller M, Mahmud H, Qadri SM, Gu S, Braun M, Bobbala D, Hocher B, Lang F. Endothelin B receptor stimulation inhibits suicidal erythrocyte death. FASEB J. 2010;24:3351–3359. doi: 10.1096/fj.10-159483. [DOI] [PubMed] [Google Scholar]
  • 16.Nicolay JP, Liebig G, Niemoeller OM, Koka S, Ghashghaeinia M, Wieder T, Haendeler J, Busse R, Lang F. Inhibition of suicidal erythrocyte death by nitric oxide. Pflugers Arch. 2008;456:293–305. doi: 10.1007/s00424-007-0393-1. [DOI] [PubMed] [Google Scholar]
  • 17.Lang E, Qadri SM, Zelenak C, Gu S, Rotte A, Draeger A, Lang F. Inhibition of suicidal erythrocyte death by blebbistatin. Am J Physiol Cell Physiol. 2011;301:C490–C498. doi: 10.1152/ajpcell.00043.2011. [DOI] [PubMed] [Google Scholar]
  • 18.Gatidis S, Meier A, Jilani K, Lang E, Zelenak C, Qadri SM, Lang F. Phlorhizin protects against erythrocyte cell membrane scrambling. J Agric Food Chem. 2011;59:8524–8530. doi: 10.1021/jf201938d. [DOI] [PubMed] [Google Scholar]
  • 19.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. doi: 10.1084/jem.86.4.339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Foller M, Bobbala D, Koka S, Huber SM, 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. doi: 10.1159/000233238. [DOI] [PubMed] [Google Scholar]
  • 21.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:C644–651. doi: 10.1152/ajpcell.00340.2011. [DOI] [PubMed] [Google Scholar]
  • 22.Dasgupta SK, Abdel-Monem H, Guchhait P, Nagata S, Thiagarajan P. Role of lactadherin in the clearance of phosphatidylserine-expressing red blood cells. Transfusion. 2008;48:2370–2376. doi: 10.1111/j.1537-2995.2008.01841.x. [DOI] [PubMed] [Google Scholar]
  • 23.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. doi: 10.1161/01.ATV.0000252898.48084.6a. [DOI] [PubMed] [Google Scholar]
  • 24.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. doi: 10.1007/s11010-011-0887-1. [DOI] [PubMed] [Google Scholar]
  • 25.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 2011; Epub ahead of print. [DOI] [PubMed]
  • 26.Kucherenko YV, Bhavsar SK, Grischenko VI, Fischer UR, Huber SM, Lang F. Increased cation conductance in human erythrocytes artificially aged by glycation. J Membr Biol. 2010;235:177–189. doi: 10.1007/s00232-010-9265-2. [DOI] [PubMed] [Google Scholar]
  • 27.Foller M, Braun M, Qadri SM, Lang E, Mahmud H, Lang F. Temperature sensitivity of suicidal erythrocyte death. Eur J Clin Invest. 2010;40:534–540. doi: 10.1111/j.1365-2362.2010.02296.x. [DOI] [PubMed] [Google Scholar]
  • 28.Felder KM, Hoelzle K, Ritzmann M, Kilchling T, Schiele D, Heinritzi K, Groebel K, Hoelzle LE. Hemotrophic mycoplasmas induce programmed cell death in red blood cells. Cell Physiol Biochem. 2011;27:557–564. doi: 10.1159/000329957. [DOI] [PubMed] [Google Scholar]
  • 29.Totino PR, Daniel-Ribeiro CT, Ferreira-da-Cruz MF. Refractoriness of eryptotic red blood cells to plasmodium falciparum infection: a putative host defense mechanism limiting parasitaemia. PLoS One. 2011;6:e26575. doi: 10.1371/journal.pone.0026575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Siraskar B, Ballal A, Bobbala D, Foller M, Lang F. Effect of amphotericin B on parasitemia and survival of plasmodium berghei-infected mice. Cell Physiol Biochem. 2010;26:347–354. doi: 10.1159/000320558. [DOI] [PubMed] [Google Scholar]
  • 31.Bobbala D, Alesutan I, Foller M, Huber SM, Lang F. Effect of anandamide in plasmodium berghei-infected mice. Cell Physiol Biochem. 2010;26:355–362. doi: 10.1159/000320559. [DOI] [PubMed] [Google Scholar]
  • 32.Bobbala D, Alesutan I, Foller M, Tschan S, Huber SM, Lang F. Protective effect of amiodarone in malaria. Acta Trop. 2010;116:39–44. doi: 10.1016/j.actatropica.2010.05.005. [DOI] [PubMed] [Google Scholar]
  • 33.Bottger E, Multhoff G, Kun JF, Esen M. Plasmodium falciparum-infected erythrocytes induce granzyme B by NK cells through expression of host-Hsp70. PLoS One. 2012;7:e33774. doi: 10.1371/journal.pone.0033774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Delic D, Warskulat U, Borsch E, Al Qahtani S, Al Quraishi S, Haussinger D, Wunderlich F. Loss of ability to self-heal malaria upon taurine transporter deletion. Infect Immun. 2010;78:1642–1649. doi: 10.1128/IAI.01159-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Weiss E, Cytlak UM, Rees DC, Osei A, Gibson JS. Deoxygenation-induced and Ca(2+) dependent phosphatidylserine externalisation in red blood cells from normal individuals and sickle cell patients. Cell Calcium. 2012;51:51–6. doi: 10.1016/j.ceca.2011.10.005. [DOI] [PubMed] [Google Scholar]
  • 36.Basu S, Banerjee D, Chandra S, Chakrabarti A. Eryptosis in hereditary spherocytosis and thalassemia: role of glycoconjugates. Glycoconj J. 2010;27:717–722. doi: 10.1007/s10719-009-9257-6. [DOI] [PubMed] [Google Scholar]
  • 37.Ganesan S, Chaurasiya ND, Sahu R, Walker LA, Tekwani BL. 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. doi: 10.1016/j.tox.2012.01.015. [DOI] [PubMed] [Google Scholar]
  • 38.Basu S, Banerjee D, Ghosh M, Chakrabarti A. Erythrocyte membrane defects and asymmetry in paroxysmal nocturnal hemoglobinuria and myelodysplastic syndrome. Hematology. 2010;15:236–239. doi: 10.1179/102453309X12583347114095. [DOI] [PubMed] [Google Scholar]
  • 39.Lang E, Lang PA, Shumilina E, Qadri SM, Kucherenko Y, Kempe DS, Foller M, Capasso A, Wieder T, Gulbins E, Clemen CS, Herr C, Noegel AA, Huber SM, Lang F. Enhanced eryptosis of erythrocytes from gene-targeted mice lacking annexin A7. Pflugers Arch. 2010;460:667–676. doi: 10.1007/s00424-010-0829-x. [DOI] [PubMed] [Google Scholar]
  • 40.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. doi: 10.1111/j.1582-4934.2011.01387.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Nguyen TT, Foller M, Lang F. Tin triggers suicidal death of erythrocytes. J Appl Toxicol. 2009;29:79–83. doi: 10.1002/jat.1390. [DOI] [PubMed] [Google Scholar]
  • 42.Bhavsar SK, Bobbala D, Xuan NT, Foller M, Lang F. Stimulation of suicidal erythrocyte death by alpha-lipoic acid. Cell Physiol Biochem. 2010;26:859–868. doi: 10.1159/000323995. [DOI] [PubMed] [Google Scholar]
  • 43.Vota DM, Crisp RL, Nesse AB, Vittori DC. Oxidative stress due to aluminium exposure induces eryptosis which is prevented by erythropoietin. J Cell Biochem. 2012;113:1581–1589. doi: 10.1002/jcb.24026. [DOI] [PubMed] [Google Scholar]
  • 44.Zbidah M, Lupescu A, Jilani K, Fajol A, Michael D, Qadri SM, Lang F. Apigenin-induced suicidal erythrocyte death. J Agric Food Chem. 2012;60:533–538. doi: 10.1021/jf204107f. [DOI] [PubMed] [Google Scholar]
  • 45.Ghashghaeinia M, Toulany M, Saki M, Bobbala D, Fehrenbacher B, Rupec R, Rodemann HP, Ghoreschi K, Rocken M, Schaller M, Lang F, Wieder T. The NFkB pathway inhibitors Bay 11–7082 and parthenolide induce programmed cell death in anucleated erythrocytes. Cell Physiol Biochem. 2011;27:45–54. doi: 10.1159/000325204. [DOI] [PubMed] [Google Scholar]
  • 46.Qadri SM, Kucherenko Y, Lang F. Beauvericin induced erythrocyte cell membrane scrambling. Toxicology. 2011;283:24–31. doi: 10.1016/j.tox.2011.01.023. [DOI] [PubMed] [Google Scholar]
  • 47.Lang E, Jilani K, Zelenak C, Pasham V, Bobbala D, Qadri SM, Lang F. Stimulation of suicidal erythrocyte death by bezethonium. Cell Physiol Biochem. 2011;28:347–354. doi: 10.1159/000331751. [DOI] [PubMed] [Google Scholar]
  • 48.Lupescu A, Jilani K, Zelenak C, Zbidah M, Qadri SM, Lang F. Hexavalent chromium-induced erythrocyte membrane phospholipid asymmetry. Biometals. 2012;25:309–318. doi: 10.1007/s10534-011-9507-5. [DOI] [PubMed] [Google Scholar]
  • 49.Qadri SM, Kucherenko Y, Zelenak C, Jilani K, Lang E, Lang F. Dicoumarol activates Ca-permeable cation channels triggering erythrocyte cell membrane scrambling. Cell Physiol Biochem. 2011;28:857–864. doi: 10.1159/000335800. [DOI] [PubMed] [Google Scholar]
  • 50.Ghashghaeinia M, Bobbala D, Wieder T, Koka S, Bruck J, Fehrenbacher B, Rocken M, Schaller M, Lang F, Ghoreschi K. Targeting glutathione by dimethylfumarate protects against experimental malaria by enhancing erythrocyte cell membrane scrambling. Am J Physiol Cell Physiol. 2010;299:C791–C804. doi: 10.1152/ajpcell.00014.2010. [DOI] [PubMed] [Google Scholar]
  • 51.Jilani K, Qadri SM, Lang E, Zelenak C, Rotte A, Bobbala D, Lang F. Stimulation of erythrocyte phospholipid scrambling by enniatin A. Mol Nutr Food Res. 2011;55(suppl 2):S294–302. doi: 10.1002/mnfr.201100156. [DOI] [PubMed] [Google Scholar]
  • 52.Eberhard M, Ferlinz K, Alizzi K, Cacciato PM, Faggio C, Foller M, Lang F. FTY720-induced suicidal erythrocyte death. Cell Physiol Biochem. 2010;26:761–766. doi: 10.1159/000322343. [DOI] [PubMed] [Google Scholar]
  • 53.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 2011; Epub ahead of print. [DOI] [PubMed]
  • 54.Qian EW, Ge DT, Kong SK. Salidroside protects human erythrocytes against hydrogen peroxide-induced apoptosis. J Nat Prod. 2012;75:531–537. doi: 10.1021/np200555s. [DOI] [PubMed] [Google Scholar]
  • 55.Chadebech P, Michel M, Janvier D, Yamada K, Copie-Bergman C, Bodivit G, Bensussan A, Fournie JJ, Godeau B, Bierling P, Izui S, Noizat-Pirenne F. IgA-mediated human autoimmune hemolytic anemia as a result of hemagglutination in the spleen, but independent of complement activation and FcalphaRI. Blood. 2010;116:4141–4147. doi: 10.1182/blood-2010-03-276162. [DOI] [PubMed] [Google Scholar]
  • 56.Zelenak C, Foller M, Velic A, Krug K, Qadri SM, 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. doi: 10.1021/pr101004j. [DOI] [PubMed] [Google Scholar]
  • 57.Nicolay JP, Gatz S, Lang F, Lang UE. Lithium-induced suicidal erythrocyte death. J Psychopharmacol. 2010;24:1533–1539. doi: 10.1177/0269881109102631. [DOI] [PubMed] [Google Scholar]
  • 58.Nguyen DB, Wagner-Britz L, Maia S, Steffen P, Wagner C, Kaestner L, Bernhardt I. Regulation of phosphatidylserine exposure in red blood cells. Cell Physiol Biochem. 2011;28:847–856. doi: 10.1159/000335798. [DOI] [PubMed] [Google Scholar]
  • 59.Bhavsar SK, Eberhard M, Bobbala D, Lang F. Monensin induced suicidal erythrocyte death. Cell Physiol Biochem. 2010;25:745–752. doi: 10.1159/000315094. [DOI] [PubMed] [Google Scholar]
  • 60.Jilani K, Qadri SM, Zelenak C, Lang F. Stimulation of suicidal erythrocyte death by oridonin. Arch Biochem Biophys. 2011;511:14–20. doi: 10.1016/j.abb.2011.05.001. [DOI] [PubMed] [Google Scholar]
  • 61.Eberhard M, Foller M, Lang F. Effect of phytic acid on suicidal erythrocyte death. J Agric Food Chem. 2010;58:2028–2033. doi: 10.1021/jf903666b. [DOI] [PubMed] [Google Scholar]
  • 62.Gao M, Cheung KL, Lau IP, Yu WS, Fung KP, Yu B, Loo JF, Kong SK. Polyphyllin D induces apoptosis in human erythrocytes through Ca(2+) rise and membrane permeabilization. Arch Toxicol. 2012;86:741–752. doi: 10.1007/s00204-012-0808-4. [DOI] [PubMed] [Google Scholar]
  • 63.Qadri SM, Bauer J, Zelenak C, Mahmud H, Kucherenko Y, Lee SH, Ferlinz K, Lang F. Sphingosine but not sphingosine-1-phosphate stimulates suicidal erythrocyte death. Cell Physiol Biochem. 2011;28:339–346. doi: 10.1159/000331750. [DOI] [PubMed] [Google Scholar]
  • 64.Jilani K, Abed M, Zelenak C, Lang E, Qadri SM, Lang F. Triggering of erythrocyte cell membrane scrambling by ursolic acid. J Nat Prod. 2011;74:2181–2186. doi: 10.1021/np2005133. [DOI] [PubMed] [Google Scholar]

Articles from Transfusion Medicine and Hemotherapy are provided here courtesy of Karger Publishers

RESOURCES