Skip to main content
Transfusion Medicine and Hemotherapy logoLink to Transfusion Medicine and Hemotherapy
. 2012 Sep 6;39(5):353–361. doi: 10.1159/000342229

Red Blood Cell Clearance in Inflammation

Marleen Straat a,*, Robin van Bruggen b, Dirk de Korte c, Nicole P Juffermans a
PMCID: PMC3678279  PMID: 23801928

Abstract

Summary

Anemia is a frequently encountered problem in the critically ill patient. The inability to compensate for anemia includes several mechanisms, collectively referred to as anemia of inflammation: reduced production of erythropoietin, impaired bone marrow response to erythropoietin, reduced iron availability, and increased red blood cell (RBC) clearance. This review focuses on mechanisms of RBC clearance during inflammation. We state that phosphatidylserine (PS) expression in inflammation is mainly enhanced due to an increase in ceramide, caused by an increase in sphingomyelinase activity due to either platelet activating factor, tumor necrosis factor-α, or direct production by bacteria. Phagocytosis of RBCs during inflammation is mediated via RBC membrane protein band 3. Reduced deformability of RBCs seems an important feature in inflammation, also mediated by band 3 as well as by nitric oxide, reactive oxygen species, and sialic acid residues. Also, adherence of RBCs to the endothelium is increased during inflammation, most likely due to increased expression of endothelial adhesion molecules as well as PS on the RBC membrane, in combination with decreased capillary blood flow. Thereby, clearance of RBCs during inflammation shows similarities to clearance of senescent RBCs, but also has distinct entities, including increased adhesion to the endothelium.

KeyWords: Erythrocyte clearance, Senescence, Inflammation

Introduction

It is commonly known that anemia is a frequently encountered problem in the critically ill patient. Almost 95% of patients admitted to the intensive care unit (ICU) have a hemoglobin level below normal after 3 days of ICU admission [1]. As a consequence, critically ill patients are frequently transfused. The CRIT study showed that 44% of the patients received at least 1 or more units of red blood cells (RBCs). In sepsis patients, transfusion rates even reach 73% [2]. The association between transfusion and adverse outcome found in a number of observational studies in several critically ill patient populations [3, 4, 5, 6] calls for a thorough understanding of the causes of anemia. In this review, we discuss the causes of anemia in sepsis patients, focusing on anemia of inflammation. We discuss mechanisms of anemia of inflammation, with a special emphasis on increased RBC clearance.

Causes of Anemia in Sepsis

In sepsis, pre-existent factors which contribute to chronic anemia before admission to the ICU are often present, including poor nutritional status, co-morbidities such as renal failure, or intensive treatment for malignancies. These factors not only contribute to anemia, but at the same time put these patients at increased risk of sepsis. Causes of anemia in sepsis are multifactorial and are summarized in table 1. One cause of anemia which stands somewhat separate is blood loss. Increased blood loss can occur via the gastrointestinal tract, from surgical procedures, or through repeated phlebotomy [7]. In total, a median blood loss of 128 ml/day has been calculated in critically ill patients [8]. Healthy individuals donating blood can compensate a loss of about 10 ml of RBCs per day [9]. Critically ill patients, however, are impaired in their ability to regenerate these losses. Causes of this inability to correct for deficiencies are the consequence of inflammatory processes, collectively referred to as anemia of inflammation. This type of anemia was once known as anemia of chronic disease. However, it turned out not to be restricted to chronic disease but also occur in patients with acute inflammation [7]. Moreover, some of the chronic disorders that were covered by the term led to anemia through a different mechanism, for example chronic kidney failure. Therefore, by 1983 it was advocated to change the term to anemia of inflammation [10].

Table 1.

Causes of anemia in sepsis

Pre-existing conditions
 Renal failure
 Poor nutritional status
 Chemotherapy
Nutritional deficiency
 Iron deficiency
 Folate deficiency
Blood loss
 Gastrointestinal tract
 Surgical procedures
 Repeated phlebotomy
Anemia of inflammation
 Reduced production of erythropoietin
 Impaired bone marrow response to
 erytropoietin
 Reduced iron availability
 Increased RBC clearance
  Phosphatidylserine exposure
  Erythrocyte phagocytosis
  Reduced deformability
  Adherence to endothelium
Hemolysis
Diffuse intravascular coagulation

Pathogenesis of Anemia of Inflammation

The pathogenesis of anemia of inflammation comprises 4 different mechanisms: reduced production of erythropoietin (EPO), impaired bone marrow response to EPO, reduced iron availability, and increased RBC clearance.

Decreased Production of Erythropoietin

Reduced erythropoiesis is a result of reduced maturation of erythroid precursors. When hemoglobin levels drop, the normal response is an increase in EPO production. In sepsis, however, the rise in EPO levels in response to anemia is blunted, corresponding with low hematocrit [11]. The blunted endogenous EPO response to anemia in critically ill patients is irrespective of the presence of renal failure [12]. As low levels of EPO are found in several subsets of patients, including trauma and sepsis [13, 14], the common denominator most likely is the presence of an inflammatory condition.

Impaired Bone Marrow Response to Erythropoietin

Critically ill patients are able to respond to EPO administered exogenously in terms of an increase in reticulocyte count [12] and decreased need for blood transfusion [15]. However, the erythropoid response to exogenously administered EPO is also blunted, as doses of EPO given to generate a response are higher when compared to doses administered to patients with renal failure. At present, a recent meta-analysis of the efficacy of EPO has not led to the recommendation to supply EPO to the critically ill [16]. The cause of bone marrow hyporesponsiveness to EPO is not known but is likely to include inflammatory pathways. EPO binds to a specific receptor, resulting in proliferation and differentiation of erythroid progenitors. This process may be counterbalanced by death signals, resulting in impaired RBC production. In line with this, increased apoptosis of bone marrow erythroid precursors was demonstrated in sepsis patients. Also, erythroid progenitors can be inhibited by incubation with serum of septic patients [17]. This suggests that inflammation directly suppresses erythroid precursors. Alternatively, as the amplification of erythropoiesis that results from the administration of EPO increases the need for iron, a lack of iron may underlie the blunted response to EPO. In anemia of chronic kidney disease, it is well recognized that response to EPO is greatly enhanced by giving intravenous iron. It should be noted that only 1 study in the meta-analysis administered intravenous iron in association with EPO. In that study, the effect of EPO treatment was greater in terms of a reduced need for blood transfusion and increase in hemoglobin concentrations [18].

Reduced Iron Availability

Iron for erythropoiesis comes from dietary sources through absorption in the gut. However, this accounts for only a small part of our daily iron need. The majority of iron used for erythropoiesis comes from lysis of aged RBCs, which generates free heme which is then degraded to iron and recycled towards the circulation or stored in ferritin molecules. Hepcidin is a major regulator of the iron metabolism, which acts by binding to the iron exporter ferroportin, causing internalization of iron and inhibiting the release of iron from tissue macrophages. Thus, hepcidin reduces the concentration of iron in the blood. The production of hepcidin is upregulated in response to elevated serum iron levels. Inflammation also induces hepcidin, which is a fast response resulting in a drop in iron levels within hours [19]. The induction of hepcidin synthesis by inflammation is not fully understood but depends on IL-6, as infusion of IL-6 into human volunteers induces increased hepcidin synthesis with a decrease in plasma iron levels [20]. Indeed, hepcidin levels have been found to be elevated in critically ill trauma patients, correlating with the duration of the anemia [21], as well as in critically ill patients not suspected for iron deficiency [22]. Thereby, it is reasonable to assume that hepcidin is elevated in inflammatory conditions, contributing to anemia. However, given the complexity of separating iron deficiency from anemia of inflammation, more research is needed in this area.

Increased Red Blood Cell Clearance

Although some microbial agents can elicit severe hemolytic reactions in the course of sepsis, increased hemolysis in general is thought to play a minor role in anemia of inflammation, as parameters of hemolysis are usually not disturbed. The decreased RBC lifespan in inflammation is rather thought to be due to an altered morphology of the RBCs, resulting in increased adherence to the endothelium and clearance from the circulation. The following chapters describe these changes in morphology in detail. As changes occurring during inflammation to some extend are similar to those observed during RBC ageing, a short description of changes during ageing is given first.

Senescence of Red Blood Cells

Phosphatidylserine Exposure

Apoptosis is the term used for the suicidal cell death of nucleated cells and is characterized by loss of cellular K+ with subsequent cell shrinkage, nuclear condensation, DNA fragmentation, mitochondrial depolarization, cell membrane blebbing, and phosphatidylserine (PS) exposure at the cell surface [23]. However, since RBCs are devoid of nuclei and mitochondria, the term eryptosis was introduced to describe the process of apoptosis in these cells [24]. Eryptosis is triggered by an increased cytosolic Ca2+ concentration due to activation of cation channels by a number of different causes [23, 25]. In an in vivo experiment, an increase in PS expression with increasing age of the RBCs has been found, which correlated with the rate of RBC removal from the circulation [26]. It was long believed that after increased PS expression, macrophages eliminate these apoptotic RBCs by recognizing them through their specific PS receptor [27, 28]. However, various receptors have been identified recently, such as Tim1, Tim4 and Stabilin-2, that can mediate binding and phagocytosis of apoptotic cells by the recognition of PS on these cells [29, 30]. In addition, several plasma proteins, such as lactadherin, GAS6 and protein S, have been described to bind to PS and act as bridging molecules to direct PS to receptors on phagocytes, αvβ3/5 integrins, and receptors of the Axl family and mediate clearance of PS-positive cells [31].

Erythrocyte Phagocytosis – Band 3 and CD47

In contrast to eryptosis, RBCs can also be phagocytosed directly without being apoptotic first. There are 2 distinct mechanisms of erythrophagocytosis due to ageing. Naturally occurring antibodies (NAbs) that bind to band 3 are implicated in the clearance of senescent RBCs [32]. Band 3 is an abundant RBC integral membrane protein which has 2 different domains: the membrane-spanning domain catalyzes anion exchange and is recognized by NAbs, whereas the cytoplasmic domain binds different proteins and thereby regulates the structure and function of the RBCs [33, 34]. Band 3 undergoes a conformational change in senescent RBCs, although no consensus exists over the exact mechanism leading to this change. It is thought that oxidative damage to hemoglobin, which occurs during senescence, and the subsequent formation of hemichromes which bind to band 3, can eventually lead to clustering of band 3 into large aggregates. These clusters show enhanced affinity for NAbs [35, 36, 37, 38, 39]. Indeed, a mutual correlation has been shown to exist between the amount of membrane-bound hemichromes, percentage of aggregated band 3, and phagocytosis intensity [40, 41]. Another hypothesis is that proteolytic degradation of band 3 is required to form the band 3 epitope(s) recognized by NAbs [42, 43]. NAbs are not efficient opsonins, due to their low affinity and low circulating numbers. Their efficiency is increased by the activation of the classical complement pathway, which significantly lowers the amount of NAbs needed for induction of phagocytosis [44, 45, 46]. For example, an in vitro experiment showed that phagocytosis of sheep RBCs was at least 10-fold more effective when opsonized with C3b immunoglobulin G (C3b-IgG) compared to opsonization with IgG alone [47]. NAbs form complexes with C3b, which are more resistant to inactivation by factors H and I than free C3b. Furthermore, the activation of C3 convertase by these complexes is more potent than by immobilized C3b [45]. However, the exact mechanism how opsonization with C3B-IgG increases phagocytosis compared to opsonization with IgG or C3B alone remains unclear. An alternative mechanism of erytrophagocytosis involves the regulation of expression of ‘eat me’ and ‘don't eat me’ signals. CD47, one of the membrane proteins expressed by RBCs, has an inhibitory effect on erythrocyte phagocytosis by macrophages [48, 49, 50, 51, 52]. CD47 binds to SIRPα on the macrophage, eventually leading to inhibition of phagocytosis [53, 54, 55, 56]. However, CD47-SIRPα can, through an unknown mechanism, also promote phagocytosis of apoptotic cells [57, 58]. Recently, it was shown that SIRPα plays a role in the removal of aged RBCs through CD47 binding [53]. CD47 undergoes a conformational change in response to oxidative damage due to ageing, after which CD47 binds thrombospondin-1 [59, 60] and is subsequently recognized as an ‘eat me’ signal by SIRPα [118].

Reduced Deformability

To be able to pass through capillaries with a diameter 2–3 μm while their own diameter is 8 μm, RBCs are highly deformable. The ability to deform relies on different characteristics of the RBC, including membrane composition, cellular geometry, and cytoplasmic viscosity [61, 62]. The filtering of senescent RBCs from the circulation is performed by the spleen as a consequence of its unique structure. Arterial blood passes the red pulp that contains many macrophages, and then on to the venous sinuses which are eventually drained into the vena lienalis. To reach these sinuses, the blood from the cords is forced through very small slits that are formed by stress fibers running parallel to the endothelial cells. This passage is more difficult for senescent RBCs which have stiffening membranes such that they stick in the cords and are phagocytosed by the earlier mentioned red pulp macrophages [63, 64]. Decreased RBC deformability is not only observed in senescence, but also occurs in a wide variety of pathological conditions, such as malaria [65], hereditary spherocytosis [66], sickle cell disease [67], and sepsis [68, 69, 70, 71, 72, 73]. In the following chapter, the effects of inflammation on RBC clearance are discussed by comparing mechanisms occurring in ageing with inflammatory pathways (table 2).

Table 2.

Identified mechanisms of erythrocyte clearance in physiologic senescence and inflammation

Physiologic senescence Inflammation
Phosphatidylserine exposure +/− +
Erythrocyte phagocytosis + +/−
Reduced deformability + +
Adherence to endothelium +

Effects of Inflammation on Clearance of Red Blood Cells

Phosphatidylserine Exposure in Inflammation

PS exposure has been claimed to be involved in accelerated RBC clearance during inflammation. This is thought to be due to an increase in the plasma concentrations of sphingomyelinase, an enzyme that converts sphingomyelin into ceramide [74]. Ceramide enhances the sensitivity of RBCs to an already increased intracellular Ca2+ concentration, and thereby enhances PS exposure [75]. Sphingomyelinase normally resides in the lysosomes of macrophages, but can be secreted into plasma [76]. Many factors can lead to an increase in plasma levels of sphingomyelinase, and some of these are also implicated in sepsis, such as tumor necrosis factor-alpha (TNF-α) [77] and platelet activating factor (PAF) [78]. Furthermore, bacteria such as Staphylococcus aureus can also produce sphingomyelinase [79]. In vitro experiments show that PS exposure is induced by incubation of RBCs with plasma of sepsis patients but not by incubation with plasma of healthy volunteers [79]. Furthermore, PS exposure is induced after treatment with supernatant from cultured S. aureus with sphingomyelinase activity, but not after exposure to supernatant from mutated S. aureus lacking sphingomyelinase activity [79].

Erythrocyte Phagocytosis in Inflammation – Band 3 and CD47

As in ageing, band 3 may be involved in mediating erythrocyte phagocytosis in inflammation. It is shown that the RBC band 3/α-spectrin ratio increased in septic mice compared to non-septic mice in a cecal ligation and puncture (CLP) model [80]. In a different experiment, but using the same in vivo model, the same group found an increase in band 3 phosphorylation in septic mice compared to non-septic mice [81]. However, no evidence exists linking these changes in band 3 directly to an increased RBC clearance in a sepsis model. No evidence has been published concerning the CD47-thrombospondin-1 combination that is recognized as an ‘eat me’ signal by SIRPα. However, since this is a recently elucidated mechanism, additional research is needed.

Reduced Deformability in Inflammation

In vitro, lipopolysaccharide (LPS) induces a change in the deformability in RBCs after whole blood stimulation, but not in isolated RBCs [68]. Also, in patients with sepsis due to both Gram-negative and Gram-positive bacteria, reduced RBC deformability has been shown when compared to healthy controls [69, 70, 71, 72, 73]. This may implicate that other factors besides the erythrocyte itself may be needed to induce reduced deformability. Several factors are involved in the decrease of RBC deformability during sepsis, which we will discuss here. As in aging, band 3 may be involved in reduced deformability in inflammation. It was found in an in vivo CLP mouse model that a higher RBC band 3/α-spectrin ratio was associated with decreased RBC deformability [80]. Also, in vitro, RBC deformability was found to depend on the band 3 phosphorylation state [82, 83]. Reactive oxygen species (ROS) influence RBC deformability in inflammation. ROS can lead to protein degradation in RBCs in vitro [84], in particular membrane proteins such as band 3 and spectrin [85]. A clear link between ROS and loss of deformability was established in an in vitro experiment showing that human RBCs undergo loss of deformability after exposure to H2O2 [86]. Also, in an in vivo CLP rat model, decreased RBC deformability was found in septic rats when compared to rats that underwent sham surgery. Decreased RBC deformability was prevented by pretreating the rats with the ROS scavenger α-tocopherol [73]. Another mechanism that is implicated in the reduced deformability encountered in inflammation is nitric oxide (NO). NO is a mediator that is released by vascular endothelial cells and acts mainly as a vasodilator [87, 88, 89]. Small amounts of NO are present in the blood under physiological conditions, but during inflammation and infection its concentration may increase by 10-fold [90]. Several in vitro experiments showed that NO causes a decrease in RBC deformability [91, 92], and this was also shown in an in vitro sepsis model in which this loss of RBC deformability was attenuated by the NO inhibitor Nmonomethyl arginine [93]. Furthermore, a selective inhibitor of NO synthase prevented overproduction of NOS, accumulation of NO within the RBC, as well as a decrease in RBC deformability in murine sepsis [94]. Another factor of importance in clearance of RBCs during inflammation are sialic acid residues (SA) which are bound to glycophorin and account for the negative force of the RBC membrane [95]. Due to this negative force, RBCs have repellent properties. When the SA content is cleaved from glycophorin after treatment with neuraminidase, RBCs have a reduced mean curvature [96]. A reduction in SA content was found in RBCs from critically ill patients when compared to RBCs from healthy volunteers, associated with a decrease in RBC deformability [97].

Increased Adherence of Red Blood Cells to the Endothelium in Inflammation

In sepsis, there are profound disturbances in microcirculation, already occurring in the early phase [98, 99, 100] and clearly contributing to adverse outcome [99, 100, 101]. The resistance of microcirculatory disorders to vasodilators [102] and the apparent independence of the mean arterial blood pressure and cardiac output [98, 99, 103] raises the hypothesis that these disorders may be caused by an obstruction due to adherence of cells to the endothelium and may not be solely due to a low flow state. Indeed, in diffuse intravascular coagulation which occurs in 25% of patients with sepsis [104], increased aggregation of cells with formation of microthrombi is apparent. However, RBC adherence to the endothelium may also play a role. Although not extensively investigated, there is some evidence for this phenomenon. Incubation of both endothelial cells and RBCs with endotoxin increased adherence of RBCs to endothelial monolayers [105]. This effect was also observed after stimulation with TNF-α [106]. The mechanisms mediating RBC adhesiveness to the endothelium during inflammation are not well characterized. Most knowledge comes from specific diseases characterized by the presence of vascular pathology, including sickle cell disease, diabetes mellitus, and malaria. Specific ligand-receptor interactions have been identified in enhanced RBC adherence to the endothelium in sickle cell disease [107]. In diabetes, advanced glycation end products (AGE) expressed on diabetic RBCs ligate with the receptor for AGE (RAGE) expressed on endothelial cells. Also, AGE-RAGE interactions have been found in other inflammatory states, including trauma [108]. Of interest, AGE-RAGE interactions may play a role in adverse effects of blood transfusion, as AGE formed in stored RBCs was found to ligate to endothelial bound RAGE, resulting in endothelial damage [109]. Whether specific ligand-receptor interactions play a role in other inflammatory states is not known. However, there is evidence that non-receptor cytoadhesion is mediated by exposure of PS on the RBC membrane. RBCs expressing PS on their outer membrane are more prone to adhere to endothelial cells, irrespective of the cause of the band 3/α-spectrin ratio PS exposure [110, 111, 112, 113, 114, 115, 116]. A definite role for PS in adherence of RBCs to endothelium has been shown by a reversal of adhesion following blocking of PS by PS liposomes [110]. As discussed before, inflammatory conditions are able to induce PS exposure on RBCs [79]. Besides specific RBC-endothelial interactions, low flow may contribute to increased RBC adhesiveness. Flow in the microcirculation in sepsis is diminished, with a decrease in the number of perfused capillaries which display intermittent flow and differences in RBC velocities. This led to the hypothesis that flow may have an effect on adhesion of RBCs to vascular endothelium. Indeed, it was found that higher flow rates reduced and lower flow rates increased RBC adherence to the endothelium [117]. Taken together, similar to the Virchow's triad in thrombosis, both alterations in the microcirculatory flow as well as activation of endothelial adhesion markers may contribute to increased RBC adherence to the endothelium in sepsis.

Conclusion

Anemia is a common feature in sepsis due to several inflammatory pathways collectively referred to as anemia of inflammation. RBC clearance is likely to contribute significantly to anemia of inflammation. We postulate that besides mechanisms that also play a role in RBC clearance during senescence, RBC-endothelial interactions are important features underlying the clearance of RBCs from the circulation during inflammation.

Disclosure Statement

The authors declare that they have no conflict of interest relating to the material covered in this manuscript.

Erratum

Transfus Med Hemother 2012;39:

In the article

Straat M, van Bruggen R, de Korte D, Juffermans NP: Red blood cell clearance in inflammation. Transfus Med Hemother 2012;39:353–360

there was a mistake.

Correction Statement

We are very sorry to note an error that has slipped into our above mentioned review article, which was pointed out to us by Dr. H.U. Lutz. In the last sentence on page 355, the ‘classical complement pathway’ should be replaced by the ‘alternative complement pathway’.

Marleen Straat, Nicole Juffermans

References

  • 1.Rodriguez RM, Corwin HL, Gettinger A, Corwin MJ, Gubler D, Pearl RG. Nutritional deficiencies and blunted erythropoietin response as causes of the anemia of critical illness. J Crit Care. 2001;16:36–41. doi: 10.1053/jcrc.2001.21795. [DOI] [PubMed] [Google Scholar]
  • 2.Corwin HL, Gettinger A, Pearl RG, Fink MP, Levy MM, Abraham E, MacIntyre NR, Shabot MM, Duh MS, Shapiro MJ. The CRIT Study: anemia and blood transfusion in the critically ill – current clinical practice in the United States. Crit Care Med. 2004;32:39–52. doi: 10.1097/01.CCM.0000104112.34142.79. [DOI] [PubMed] [Google Scholar]
  • 3.Croce MA, Tolley EA, Claridge JA, Fabian TC. Transfusions result in pulmonary morbidity and death after a moderate degree of injury. J Trauma. 2005;59:19–23. doi: 10.1097/01.ta.0000171459.21450.dc. discussion 23–14. [DOI] [PubMed] [Google Scholar]
  • 4.Gajic O, Rana R, Mendez JL, Rickman OB, Lymp JF, Hubmayr RD, Moore SB. Acute lung injury after blood transfusion in mechanically ventilated patients. Transfusion. 2004;44:1468–1474. doi: 10.1111/j.1537-2995.2004.04053.x. [DOI] [PubMed] [Google Scholar]
  • 5.Gong MN, Thompson BT, Williams P, Pothier L, Boyce PD, Christiani DC. Clinical predictors of and mortality in acute respiratory distress syndrome: potential role of red cell transfusion. Crit Care Med. 2005;33:1191–1198. doi: 10.1097/01.ccm.0000165566.82925.14. [DOI] [PubMed] [Google Scholar]
  • 6.Vamvakas EC. Pneumonia as a complication of blood product transfusion in the critically ill: transfusion-related immunomodulation (TRIM) Crit Care Med. 2006;34:S151–159. doi: 10.1097/01.CCM.0000214310.70642.8C. [DOI] [PubMed] [Google Scholar]
  • 7.Aird WC. The hematologic system as a marker of organ dysfunction in sepsis. Mayo Clin Proc. 2003;78:869–881. doi: 10.4065/78.7.869. [DOI] [PubMed] [Google Scholar]
  • 8.Von Ahsen N, Muller C, Serke S, Frei U, Eckardt KU. Important role of nondiagnostic blood loss and blunted erythropoietic response in the anemia of medical intensive care patients. Crit Care Med. 1999;27:2630–2639. doi: 10.1097/00003246-199912000-00005. [DOI] [PubMed] [Google Scholar]
  • 9.Weisbach V, Corbiere C, Strasser E, Zingsem J, Zimmermann R, Eckstein R. The variability of compensatory erythropoiesis in repeated autologous blood donation. Transfusion. 2001;41:179–183. doi: 10.1046/j.1537-2995.2001.41020179.x. [DOI] [PubMed] [Google Scholar]
  • 10.Hansen NE. The anaemia of chronic disorders. A bag of unsolved questions. Scand J Haematol. 1983;31:397–402. doi: 10.1111/j.1600-0609.1983.tb01532.x. [DOI] [PubMed] [Google Scholar]
  • 11.Rogiers P, Zhang H, Leeman M, Nagler J, Neels H, Melot C, Vincent JL. Erythropoietin response is blunted in critically ill patients. Intensive Care Med. 1997;23:159–162. doi: 10.1007/s001340050310. [DOI] [PubMed] [Google Scholar]
  • 12.Van Iperen CE, Gaillard CA, Kraaijenhagen RJ, Braam BG, Marx JJ, van de Wiel A. Response of erythropoiesis and iron metabolism to recombinant human erythropoietin in intensive care unit patients. Crit Care Med. 2000;28:2773–2778. doi: 10.1097/00003246-200008000-00015. [DOI] [PubMed] [Google Scholar]
  • 13.Corwin HL, Gettinger A, Fabian TC, May A, Pearl RG, Heard S, An R, Bowers PJ, Burton P, Klausner MA, Corwin MJ. Efficacy and safety of epoetin alfa in critically ill patients. N Engl J Med. 2007;357:965–976. doi: 10.1056/NEJMoa071533. [DOI] [PubMed] [Google Scholar]
  • 14.DeAngelo AJ, Bell DG, Quinn MW, Long DE, Ouellette DR. Erythropoietin response in critically ill mechanically ventilated patients: a prospective observational study. Crit Care. 2005;9:R172–176. doi: 10.1186/cc3480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Corwin HL, Gettinger A, Pearl RG, Fink MP, Levy MM, Shapiro MJ, Corwin MJ, Colton T. Efficacy of recombinant human erythropoietin in critically ill patients: a randomized controlled trial. JAMA. 2002;288:2827–2835. doi: 10.1001/jama.288.22.2827. [DOI] [PubMed] [Google Scholar]
  • 16.Zarychanski R, Turgeon AF, McIntyre L, Fergus-son DA. Erythropoietin-receptor agonists in critically ill patients: a meta-analysis of randomized controlled trials. CMAJ. 2007;177:725–734. doi: 10.1503/cmaj.071055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Claessens YE, Fontenay M, Pene F, Chiche JD, Guesnu M, Hababou C, Casadevall N, Dhainaut JF, Mira JP, Cariou A. Erythropoiesis abnormalities contribute to early-onset anemia in patients with septic shock. Am J Respir Crit Care Med. 2006;174:51–57. doi: 10.1164/rccm.200504-561OC. [DOI] [PubMed] [Google Scholar]
  • 18.Georgopoulos D, Matamis D, Routsi C, Michalopoulos A, Maggina N, Dimopoulos G, Zakynthinos E, Nakos G, Thomopoulos G, Mandragos K, Maniatis A. Recombinant human erythropoietin therapy in critically ill patients: a dose-response study (ISRCTN48523317) Crit Care. 2005;9:R508–515. doi: 10.1186/cc3786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Kemna E, Pickkers P, Nemeth E, van der Hoeven H, Swinkels D. Time-course analysis of hepcidin, serum iron, and plasma cytokine levels in humans injected with LPS. Blood. 2005;106:1864–1866. doi: 10.1182/blood-2005-03-1159. [DOI] [PubMed] [Google Scholar]
  • 20.Nemeth E, Rivera S, Gabayan V, Keller C, Taudorf S, Pedersen BK, Ganz T. Il-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin. J Clin Invest. 2004;113:1271–1276. doi: 10.1172/JCI20945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Sihler KC, Raghavendran K, Westerman M, Ye W, Napolitano LM. Hepcidin in trauma: linking injury, inflammation, and anemia. J Trauma. 2010;69:831–837. doi: 10.1097/TA.0b013e3181f066d5. [DOI] [PubMed] [Google Scholar]
  • 22.Lasocki S, Baron G, Driss F, Westerman M, Puy H, Boutron I, Beaumont C, Montravers P. Diagnostic accuracy of serum hepcidin for iron deficiency in critically ill patients with anemia. Intensive Care Med. 2010;36:1044–1048. doi: 10.1007/s00134-010-1794-8. [DOI] [PubMed] [Google Scholar]
  • 23.Foller M, Huber SM, Lang F. Erythrocyte programmed cell death. IUBMB Life. 2008;60:661–668. doi: 10.1002/iub.106. [DOI] [PubMed] [Google Scholar]
  • 24.Lang KS, Lang PA, Bauer C, Duranton C, Wieder T, Huber SM, Lang F. Mechanisms of suicidal erythrocyte death. Cell Physiol Biochem. 2005;15:195–202. doi: 10.1159/000086406. [DOI] [PubMed] [Google Scholar]
  • 25.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]
  • 26.Boas FE, Forman L, Beutler E. Phosphatidylserine exposure and red cell viability in red cell aging and in hemolytic anemia. Proc Natl Acad Sci U S A. 1998;95:3077–3081. doi: 10.1073/pnas.95.6.3077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Fadok VA, Bratton DL, Rose DM, Pearson A, Ezekewitz RA, Henson PM. A receptor for phosphatidylserine-specific clearance of apoptotic cells. Nature. 2000;405:85–90. doi: 10.1038/35011084. [DOI] [PubMed] [Google Scholar]
  • 28.Li MO, Sarkisian MR, Mehal WZ, Rakic P, Flavell RA. Phosphatidylserine receptor is required for clearance of apoptotic cells. Science. 2003;302:1560–1563. doi: 10.1126/science.1087621. [DOI] [PubMed] [Google Scholar]
  • 29.Park SY, Jung MY, Kim HJ, Lee SJ, Kim SY, Lee BH, Kwon TH, Park RW, Kim IS. Rapid cell corpse clearance by stabilin-2, a membrane phosphatidylserine receptor. Cell Death Differ. 2008;15:192–201. doi: 10.1038/sj.cdd.4402242. [DOI] [PubMed] [Google Scholar]
  • 30.Kobayashi N, Karisola P, Pena-Cruz V, Dorfman DM, Jinushi M, Umetsu SE, Butte MJ, Nagumo H, Chernova I, Zhu B, Sharpe AH, Ito S, Dranoff G, Kaplan GG, Casasnovas JM, Umetsu DT, Dekruyff RH, Freeman GJ. TIM-1 and TIM-4 glycoproteins bind phosphatidylserine and mediate uptake of apoptotic cells. Immunity. 2007;27:927–940. doi: 10.1016/j.immuni.2007.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Raymond A, Ensslin MA, Shur BD. SED1/MFG-E8: A bi-motif protein that orchestrates diverse cellular interactions. J Cell Biochem. 2009;106:957–966. doi: 10.1002/jcb.22076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kay MM, Goodman SR, Sorensen K, Whitfield CF, Wong P, Zaki L, Rudloff V. Senescent cell antigen is immunologically related to band 3. Proc Natl Acad Sci U S A. 1983;80:1631–1635. doi: 10.1073/pnas.80.6.1631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Zhang D, Kiyatkin A, Bolin JT, Low PS. Crystal-lographic structure and functional interpretation of the cytoplasmic domain of erythrocyte membrane band 3. Blood. 2000;96:2925–2933. [PubMed] [Google Scholar]
  • 34.Pantaleo A, Giribaldi G, Mannu F, Arese P, Turrini F. Naturally occurring anti-band 3 antibodies and red blood cell removal under physiological and pathological conditions. Autoimmun Rev. 2008;7:457–462. doi: 10.1016/j.autrev.2008.03.017. [DOI] [PubMed] [Google Scholar]
  • 35.Low PS. Structure and function of the cytoplasmic domain of band 3: center of erythrocyte membrane-peripheral protein interactions. Biochim Biophys Acta. 1986;864:145–167. doi: 10.1016/0304-4157(86)90009-2. [DOI] [PubMed] [Google Scholar]
  • 36.Hornig R, Lutz HU. Band 3 protein clustering on human erythrocytes promotes binding of naturally occurring anti-band 3 and anti-spectrin antibodies. Exp Gerontol. 2000;35:1025–1044. doi: 10.1016/s0531-5565(00)00126-1. [DOI] [PubMed] [Google Scholar]
  • 37.Arese P, Turrini F, Schwarzer E. Band 3/complement-mediated recognition and removal of normally senescent and pathological human erythrocytes. Cell Physiol Biochem. 2005;16:133–146. doi: 10.1159/000089839. [DOI] [PubMed] [Google Scholar]
  • 38.Mannu F, Arese P, Cappellini MD, Fiorelli G, Cappadoro M, Giribaldi G, Turrini F. Role of hemichrome binding to erythrocyte membrane in the generation of band-3 alterations in beta-thalassemia intermedia erythrocytes. Blood. 1995;86:2014–2020. [PubMed] [Google Scholar]
  • 39.Waugh SM, Willardson BM, Kannan R, Labotka RJ, Low PS. Heinz bodies induce clustering of band 3, glycophorin, and ankyrin in sickle cell erythrocytes. J Clin Invest. 1986;78:1155–1160. doi: 10.1172/JCI112696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ayi K, Turrini F, Piga A, Arese P. Enhanced phagocytosis of ring-parasitized mutant erythrocytes: a common mechanism that may explain protection against falciparum malaria in sickle trait and beta-thalassemia trait. Blood. 2004;104:3364–3371. doi: 10.1182/blood-2003-11-3820. [DOI] [PubMed] [Google Scholar]
  • 41.Cappellini MD, Tavazzi D, Duca L, Graziadei G, Mannu F, Turrini F, Arese P, Fiorelli G. Metabolic indicators of oxidative stress correlate with haemichrome attachment to membrane, band 3 aggregation and erythrophagocytosis in beta-thalassaemia intermedia. Br J Haematol. 1999;104:504–512. doi: 10.1046/j.1365-2141.1999.01217.x. [DOI] [PubMed] [Google Scholar]
  • 42.Kay MM. Posttranslational changes in band 3 in aging and Alzheimer's disease: do anion and glucose transport changes reflect or foreshadow the eventual metabolic abnormalities? Cell Mol Biol (Noisy-le-grand) 1998;44:271–272. [PubMed] [Google Scholar]
  • 43.Kay MM. Band 3 and its alterations in health and disease. Cell Mol Biol (Noisy-le-grand) 2004;50:117–138. [PubMed] [Google Scholar]
  • 44.Lutz HU, Bussolino F, Flepp R, Fasler S, Stammler P, Kazatchkine MD, Arese P. Naturally occurring anti-band-3 antibodies and complement together mediate phagocytosis of oxidatively stressed human erythrocytes. Proc Natl Acad Sci U S A. 1987;84:7368–7372. doi: 10.1073/pnas.84.21.7368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Lutz HU, Nater M, Stammler P. Naturally occurring anti-band 3 antibodies have a unique affinity for C3. Immunology. 1993;80:191–196. [PMC free article] [PubMed] [Google Scholar]
  • 46.Lutz HU, Jelezarova E. Complement amplification revisited. Mol Immunol. 2006;43:2–12. doi: 10.1016/j.molimm.2005.06.020. [DOI] [PubMed] [Google Scholar]
  • 47.Fries LF, Siwik SA, Malbran A, Frank MM. Phagocytosis of target particles bearing C3b-IgG covalent complexes by human monocytes and polymorphonuclear leucocytes. Immunology. 1987;62:45–51. [PMC free article] [PubMed] [Google Scholar]
  • 48.Oldenborg PA, Zheleznyak A, Fang YF, Lagenaur CF, Gresham HD, Lindberg FP. Role of CD47 as a marker of self on red blood cells. Science. 2000;288:2051–2054. doi: 10.1126/science.288.5473.2051. [DOI] [PubMed] [Google Scholar]
  • 49.Oldenborg PA, Gresham HD, Lindberg FP. CD47-signal regulatory protein alpha (SIRPαlpha) regulates Fcgamma and complement receptor-mediated phagocytosis. J Exp Med. 2001;193:855–862. doi: 10.1084/jem.193.7.855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Olsson M, Nilsson A, Oldenborg PA. Dose-dependent inhibitory effect of CD47 in macrophage uptake of IgG-opsonized murine erythrocytes. Biochem Biophys Res Commun. 2007;352:193–197. doi: 10.1016/j.bbrc.2006.11.002. [DOI] [PubMed] [Google Scholar]
  • 51.Olsson M, Oldenborg PA. CD47 on experimentally senescent murine RBCs inhibits phagocytosis following Fcgamma receptor-mediated but not scavenger receptor-mediated recognition by macrophages. Blood. 2008;112:4259–4267. doi: 10.1182/blood-2008-03-143008. [DOI] [PubMed] [Google Scholar]
  • 52.Tsai RK, Rodriguez PL, Discher DE. Self inhibition of phagocytosis: The affinity of ‘marker of self’ CD47 for SIRPαlpha dictates potency of inhibition but only at low expression levels. Blood Cells Mol Dis. 2010;45:67–74. doi: 10.1016/j.bcmd.2010.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Burger P, Hilarius-Stokman P, de Korte D, van den Berg TK, van Bruggen R: CD47 functions as a molecular switch for erythrocyte phagocytosis. Blood 2012 [DOI] [PubMed]
  • 54.Matozaki T, Murata Y, Okazawa H, Ohnishi H. Functions and molecular mechanisms of the CD47-SIRPαlpha signalling pathway. Trends Cell Biol. 2009;19:72–80. doi: 10.1016/j.tcb.2008.12.001. [DOI] [PubMed] [Google Scholar]
  • 55.Sato R, Ohnishi H, Kobayashi H, Kiuchi D, Hayashi A, Kaneko Y, Honma N, Okazawa H, Hirata Y, Matozaki T. Regulation of multiple functions of SHPS-1, a transmembrane glycoprotein, by its cytoplasmic region. Biochem Biophys Res Commun. 2003;309:584–590. doi: 10.1016/j.bbrc.2003.08.031. [DOI] [PubMed] [Google Scholar]
  • 56.Ishikawa-Sekigami T, Kaneko Y, Okazawa H, Tomizawa T, Okajo J, Saito Y, Okuzawa C, Suga-wara-Yokoo M, Nishiyama U, Ohnishi H, Matozaki T, Nojima Y. SHPS-1 promotes the survival of circulating erythrocytes through inhibition of phagocytosis by splenic macrophages. Blood. 2006;107:341–348. doi: 10.1182/blood-2005-05-1896. [DOI] [PubMed] [Google Scholar]
  • 57.Babic I, Schallhorn A, Lindberg FP, Jirik FR. SHPS-1 induces aggregation of Ba/F3 pro-B cells via an interaction with CD47. J Immunol. 2000;164:3652–3658. doi: 10.4049/jimmunol.164.7.3652. [DOI] [PubMed] [Google Scholar]
  • 58.Tada K, Tanaka M, Hanayama R, Miwa K, Shinohara A, Iwamatsu A, Nagata S. Tethering of apoptotic cells to phagocytes through binding of CD47 to SRC homology 2 domain-bearing protein tyrosine phosphatase substrate-1. J Immunol. 2003;171:5718–5726. doi: 10.4049/jimmunol.171.11.5718. [DOI] [PubMed] [Google Scholar]
  • 59.Bornstein P. Thrombospondins as matricellular modulators of cell function. J Clin Invest. 2001;107:929–934. doi: 10.1172/JCI12749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Carlson CB, Lawler J, Mosher DF. Structures of thrombospondins. Cell Mol Life Sci. 2008;65:672–686. doi: 10.1007/s00018-007-7484-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Piagnerelli M, Boudjeltia KZ, Vanhaeverbeek M, Vincent JL. Red blood cell rheology in sepsis. Intensive Care Med. 2003;29:1052–1061. doi: 10.1007/s00134-003-1783-2. [DOI] [PubMed] [Google Scholar]
  • 62.Piagnerelli M, Cotton F, Van Nuffelen M, Vincent JL, Gulbis B. Modifications in erythrocyte membrane protein content are not responsible for the alterations in rheology seen in sepsis. Shock. 2012;37:17–21. doi: 10.1097/SHK.0b013e318237d55a. [DOI] [PubMed] [Google Scholar]
  • 63.Mebius RE, Kraal G. Structure and function of the spleen. Nat Rev Immunol. 2005;5:606–616. doi: 10.1038/nri1669. [DOI] [PubMed] [Google Scholar]
  • 64.Bratosin D, Mazurier J, Tissier JP, Estaquier J, Huart JJ, Ameisen JC, Aminoff D, Montreuil J. Cellular and molecular mechanisms of senescent erythrocyte phagocytosis by macrophages. A review. Biochimie. 1998;80:173–195. doi: 10.1016/s0300-9084(98)80024-2. [DOI] [PubMed] [Google Scholar]
  • 65.Dondorp AM, Chotivanich KT, Fucharoen S, Silamut K, Vreeken J, Kager PA, White NJ. Red cell deformability, splenic function and anaemia in thalassaemia. Br J Haematol. 1999;105:505–508. doi: 10.1111/j.1365-2141.1999.01367.x. [DOI] [PubMed] [Google Scholar]
  • 66.Deplaine G, Safeukui I, Jeddi F, Lacoste F, Brousse V, Perrot S, Biligui S, Guillotte M, Guitton C, Dokmak S, Aussilhou B, Sauvanet A, Cazals Hatem D, Paye F, Thellier M, Mazier D, Milon G, Mohandas N, Mercereau-Puijalon O, David PH, Buffet PA. The sensing of poorly deformable red blood cells by the human spleen can be mimicked in vitro. Blood. 2011;117:e88–95. doi: 10.1182/blood-2010-10-312801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Barabino GA, Platt MO, Kaul DK. Sickle cell biomechanics. Annu Rev Biomed Eng. 2010;12:345–367. doi: 10.1146/annurev-bioeng-070909-105339. [DOI] [PubMed] [Google Scholar]
  • 68.Betticher DC, Keller H, Maly FE, Reinhart WH. The effect of endotoxin and tumour necrosis factor on erythrocyte and leucocyte deformability in vitro. Br J Haematol. 1993;83:130–137. doi: 10.1111/j.1365-2141.1993.tb04643.x. [DOI] [PubMed] [Google Scholar]
  • 69.Astiz ME, DeGent GE, Lin RY, Rackow EC. Microvascular function and rheologic changes in hyperdynamic sepsis. Crit Care Med. 1995;23:265–271. doi: 10.1097/00003246-199502000-00011. [DOI] [PubMed] [Google Scholar]
  • 70.Baskurt OK, Gelmont D, Meiselman HJ. Red blood cell deformability in sepsis. Am J Respir Crit Care Med. 1998;157:421–427. doi: 10.1164/ajrccm.157.2.9611103. [DOI] [PubMed] [Google Scholar]
  • 71.Kirschenbaum LA, Aziz M, Astiz ME, Saha DC, Rackow EC. Influence of rheologic changes and platelet-neutrophil interactions on cell filtration in sepsis. Am J Respir Crit Care Med. 2000;161:1602–1607. doi: 10.1164/ajrccm.161.5.9902105. [DOI] [PubMed] [Google Scholar]
  • 72.Poschl JM, Ruef P, Schnauffer M, Pohl S, Sonntag HG, Linderkamp O. Group B streptococcus impairs erythrocyte deformability in neonates more than in adults. Arch Dis Child Fetal Neonatal Ed. 1996;74:F187–190. doi: 10.1136/fn.74.3.f187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Powell RJ, Machiedo GW, Rush BF, Jr, Dikdan G. Oxygen free radicals: effect on red cell deformability in sepsis. Crit Care Med. 1991;19:732–735. [PubMed] [Google Scholar]
  • 74.Huwiler A, Kolter T, Pfeilschifter J, Sandhoff K. Physiology and pathophysiology of sphingolipid metabolism and signaling. Biochim Biophys Acta. 2000;1485:63–99. doi: 10.1016/s1388-1981(00)00042-1. [DOI] [PubMed] [Google Scholar]
  • 75.Lang KS, Myssina S, Brand V, Sandu C, Lang PA, Berchtold S, Huber SM, Lang F, Wieder T. Involvement of ceramide in hyperosmotic shock-induced death of erythrocytes. Cell Death Differ. 2004;11:231–243. doi: 10.1038/sj.cdd.4401311. [DOI] [PubMed] [Google Scholar]
  • 76.Jenkins RW, Canals D, Hannun YA. Roles and regulation of secretory and lysosomal acid sphingomyelinase. Cell Signal. 2009;21:836–846. doi: 10.1016/j.cellsig.2009.01.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Chatterjee S. Neutral sphingomyelinase: past, present and future. Chem Phys Lipids. 1999;102:79–96. doi: 10.1016/s0009-3084(99)00077-8. [DOI] [PubMed] [Google Scholar]
  • 78.Lang PA, Kempe DS, Tanneur V, Eisele K, Klarl BA, Myssina S, Jendrossek V, Ishii S, Shimizu T, Waidmann M, Hessler G, Huber SM, Lang F, Wieder T. Stimulation of erythrocyte ceramide formation by platelet-activating factor. J Cell Sci. 2005;118:1233–1243. doi: 10.1242/jcs.01730. [DOI] [PubMed] [Google Scholar]
  • 79.Kempe DS, Akel A, Lang PA, Hermle T, Biswas R, Muresanu J, Friedrich B, Dreischer P, Wolz C, Schumacher U, Peschel A, Gotz F, Doring G, Wieder T, Gulbins E, Lang F. Suicidal erythrocyte death in sepsis. J Mol Med. 2007;85:273–281. doi: 10.1007/s00109-006-0123-8. [DOI] [PubMed] [Google Scholar]
  • 80.Spolarics Z, Condon MR, Siddiqi M, Machiedo GW, Deitch EA. Red blood cell dysfunction in septic glucose-6-phosphate dehydrogenase-deficient mice. Am J Physiol Heart Circ Physiol. 2004;286:H2118–2126. doi: 10.1152/ajpheart.01085.2003. [DOI] [PubMed] [Google Scholar]
  • 81.Condon MR, Feketova E, Machiedo GW, Deitch EA, Spolarics Z. Augmented erythrocyte band-3 phosphorylation in septic mice. Biochim Biophys Acta. 2007;1772:580–586. doi: 10.1016/j.bbadis.2007.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Saldanha C, Silva AS, Goncalves S, Martins-Silva J. Modulation of erythrocyte hemorheological properties by band 3 phosphorylation and dephosphorylation. Clin Hemorheol Microcirc. 2007;36:183–194. [PubMed] [Google Scholar]
  • 83.de Almeida JP, Freitas-Santos T, Saldanha C: Erythrocyte deformability dependence on band 3 protein in an in-vitro model of hyperfibrinogenemia. Clin Hemorheol Microcirc 2011 [DOI] [PubMed]
  • 84.Davies KJ, Goldberg AL. Oxygen radicals stimulate intracellular proteolysis and lipid peroxidation by independent mechanisms in erythrocytes. J Biol Chem. 1987;262:8220–8226. [PubMed] [Google Scholar]
  • 85.Uyesaka N, Hasegawa S, Ishioka N, Ishioka R, Shio H, Schechter AN. Effects of superoxide anions on red cell deformability and membrane proteins. Biorheology. 1992;29:217–229. doi: 10.3233/bir-1992-292-303. [DOI] [PubMed] [Google Scholar]
  • 86.Srour, Bilto YY, Juma M, Irhimeh MR. Exposure of human erythrocytes to oxygen radicals causes loss of deformability, increased osmotic fragility, lipid peroxidation and protein degradation. Clin Hemorheol Microcirc. 2000;23:13–21. [PubMed] [Google Scholar]
  • 87.Bhagat K, Vallance P. Nitric oxide 9 years on. J R Soc Med. 1996;89:667–673. doi: 10.1177/014107689608901204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Lloyd-Jones DM, Bloch KD. The vascular biology of nitric oxide and its role in atherogenesis. Annu Rev Med. 1996;47:365–375. doi: 10.1146/annurev.med.47.1.365. [DOI] [PubMed] [Google Scholar]
  • 89.Marin J, Rodriguez-Martinez MA. Role of vascular nitric oxide in physiological and pathological conditions. Pharmacol Ther. 1997;75:111–134. doi: 10.1016/s0163-7258(97)00051-x. [DOI] [PubMed] [Google Scholar]
  • 90.Eiserich JP, Patel RP, O'Donnell VB. Pathophysiology of nitric oxide and related species: free radical reactions and modification of biomolecules. Mol Aspects Med. 1998;19:221–357. doi: 10.1016/s0098-2997(99)00002-3. [DOI] [PubMed] [Google Scholar]
  • 91.Korbut R, Gryglewski RJ. Nitric oxide from polymorphonuclear leukocytes modulates red blood cell deformability in vitro. Eur J Pharmacol. 1993;234:17–22. doi: 10.1016/0014-2999(93)90700-r. [DOI] [PubMed] [Google Scholar]
  • 92.Mesquita R, Picarra B, Saldanha C, Martins e Silva J. Nitric oxide effects on human erythrocytes structural and functional properties – an in vitro study. Clin Hemorheol Microcirc. 2002;27:137–147. [PubMed] [Google Scholar]
  • 93.Ismail NH, Cohn EJ, Jr, Mollitt DL. Nitric oxide synthase inhibition negates septic-induced alterations in cytoplasmic calcium homeostasis and membrane dynamics. Am Surg. 1997;63:20–23. [PubMed] [Google Scholar]
  • 94.Bateman RM, Jagger JE, Sharpe MD, Ellsworth ML, Mehta S, Ellis CG. Erythrocyte deformability is a nitric oxide-mediated factor in decreased capillary density during sepsis. Am J Physiol Heart Circ Physiol. 2001;280:H2848–2856. doi: 10.1152/ajpheart.2001.280.6.H2848. [DOI] [PubMed] [Google Scholar]
  • 95.Mohandas N, Gallagher PG. Red cell membrane: past, present, and future. Blood. 2008;112:3939–3948. doi: 10.1182/blood-2008-07-161166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Grebe R, Wolff H, Schmid-Schonbein H. Influence of red cell surface charge on red cell membrane curvature. Pflugers Arch. 1988;413:77–82. doi: 10.1007/BF00581231. [DOI] [PubMed] [Google Scholar]
  • 97.Piagnerelli M, Boudjeltia KZ, Brohee D, Piro P, Carlier E, Vincent JL, Lejeune P, Vanhaeverbeek M. Alterations of red blood cell shape and sialic acid membrane content in septic patients. Crit Care Med. 2003;31:2156–2162. doi: 10.1097/01.CCM.0000079608.00875.14. [DOI] [PubMed] [Google Scholar]
  • 98.Kanoore Edul VS, Enrico C, Laviolle B, Risso Vazquez A, Ince C, Dubin A. Quantitative assessment of the microcirculation in healthy volunteers and in patients with septic shock. Crit Care Med. 2012;40:1443–1448. doi: 10.1097/CCM.0b013e31823dae59. [DOI] [PubMed] [Google Scholar]
  • 99.De Backer D, Creteur J, Preiser JC, Dubois MJ, Vincent JL. Microvascular blood flow is altered in patients with sepsis. Am J Respir Crit Care Med. 2002;166:98–104. doi: 10.1164/rccm.200109-016oc. [DOI] [PubMed] [Google Scholar]
  • 100.Trzeciak S, Dellinger RP, Parrillo JE, Guglielmi M, Bajaj J, Abate NL, Arnold RC, Colilla S, Zanotti S, Hollenberg SM. Early microcirculatory perfusion derangements in patients with severe sepsis and septic shock: relationship to hemodynamics, oxygen transport, and survival. Ann Emerg Med. 2007;49:88–98. doi: 10.1016/j.annemergmed.2006.08.021. 98.e81–82. [DOI] [PubMed] [Google Scholar]
  • 101.Sakr Y, Dubois MJ, De Backer D, Creteur J, Vincent JL. Persistent microcirculatory alterations are associated with organ failure and death in patients with septic shock. Crit Care Med. 2004;32:1825–1831. doi: 10.1097/01.ccm.0000138558.16257.3f. [DOI] [PubMed] [Google Scholar]
  • 102.Boerma EC, Koopmans M, Konijn A, Kaiferova K, Bakker AJ, van Roon EN, Buter H, Bruins N, Egbers PH, Gerritsen RT, Koetsier PM, Kingma WP, Kuiper MA, Ince C. Effects of nitroglycerin on sublingual microcirculatory blood flow in patients with severe sepsis/septic shock after a strict resuscitation protocol: a double-blind randomized placebo controlled trial. Crit Care Med. 2010;38:93–100. doi: 10.1097/CCM.0b013e3181b02fc1. [DOI] [PubMed] [Google Scholar]
  • 103.De Backer D, Creteur J, Dubois MJ, Sakr Y, Koch M, Verdant C, Vincent JL. The effects of dobutamine on microcirculatory alterations in patients with septic shock are independent of its systemic effects. Crit Care Med. 2006;34:403–408. doi: 10.1097/01.ccm.0000198107.61493.5a. [DOI] [PubMed] [Google Scholar]
  • 104.Zeerleder S, Hack CE, Wuillemin WA. Disseminated intravascular coagulation in sepsis. Chest. 2005;128:2864–2875. doi: 10.1378/chest.128.4.2864. [DOI] [PubMed] [Google Scholar]
  • 105.Eichelbronner O, Sielenkamper A, Cepinskas G, Sibbald WJ, Chin-Yee IH. Endotoxin promotes adhesion of human erythrocytes to human vascular endothelial cells under conditions of flow. Crit Care Med. 2000;28:1865–1870. doi: 10.1097/00003246-200006000-00030. [DOI] [PubMed] [Google Scholar]
  • 106.Anniss AM, Sparrow RL. Variable adhesion of different red blood cell products to activated vascular endothelium under flow conditions. Am J Hematol. 2007;82:439–445. doi: 10.1002/ajh.20837. [DOI] [PubMed] [Google Scholar]
  • 107.Shiu YT, McIntire LV. In vitro studies of erythrocyte-vascular endothelium interactions. Ann Biomed Eng. 2003;31:1299–1313. doi: 10.1114/1.1630320. [DOI] [PubMed] [Google Scholar]
  • 108.Cohen MJ, Brohi K, Calfee CS, Rahn P, Chesebro BB, Christiaans SC, Carles M, Howard M, Pittet JF. Early release of high mobility group box nuclear protein 1 after severe trauma in humans: role of injury severity and tissue hypoperfusion. Crit Care. 2009;13:R174. doi: 10.1186/cc8152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Mangalmurti NS, Chatterjee S, Cheng G, Andersen E, Mohammed A, Siegel DL, Schmidt AM, Albelda SM, Lee JS. Advanced glycation end products on stored red blood cells increase endothelial reactive oxygen species generation through interaction with receptor for advanced glycation end products. Transfusion. 2010;50:2353–2361. doi: 10.1111/j.1537-2995.2010.02689.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Closse C, Dachary-Prigent J, Boisseau MR. Phosphatidylserine-related adhesion of human erythrocytes to vascular endothelium. Br J Haematol. 1999;107:300–302. doi: 10.1046/j.1365-2141.1999.01718.x. [DOI] [PubMed] [Google Scholar]
  • 111.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]
  • 112.Andrews DA, Low PS. Role of red blood cells in thrombosis. Curr Opin Hematol. 1999;6:76–82. doi: 10.1097/00062752-199903000-00004. [DOI] [PubMed] [Google Scholar]
  • 113.Gallagher PG, Chang SH, Rettig MP, Neely JE, Hillery CA, Smith BD, Low PS. Altered erythrocyte endothelial adherence and membrane phospholipid asymmetry in hereditary hydrocytosis. Blood. 2003;101:4625–4627. doi: 10.1182/blood-2001-12-0329. [DOI] [PubMed] [Google Scholar]
  • 114.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. doi: 10.1002/jcp.21138. [DOI] [PubMed] [Google Scholar]
  • 115.Wood BL, Gibson DF, Tait JF. Increased erythrocyte phosphatidylserine exposure in sickle cell disease: Flow-cytometric measurement and clinical associations. Blood. 1996;88:1873–1880. [PubMed] [Google Scholar]
  • 116.Zwaal RF, Comfurius P, Bevers EM. Surface exposure of phosphatidylserine in pathological cells. Cell Mol Life Sci. 2005;62:971–988. doi: 10.1007/s00018-005-4527-3. [DOI] [PubMed] [Google Scholar]
  • 117.Eichelbronner O, Sibbald WJ, Chin-Yee IH. Intermittent flow increases endotoxin-induced adhesion of human erythrocytes to vascular endothelial cells. Intensive Care Med. 2003;29:709–714. doi: 10.1007/s00134-003-1698-y. [DOI] [PubMed] [Google Scholar]
  • 118.Burger P, de Korte D, van den Berg TK, van Bruggen R. CD47 in Erythrocyte Ageing and Clearance – the Dutch point of view. Transfus Med Hemother. 2012;39(5) doi: 10.1159/000342231. DOI: 10.1159/000342231. [DOI] [PMC free article] [PubMed] [Google Scholar]

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

RESOURCES