Open access peer-reviewed chapter

Model Human β Thalassemic Erythrocytes: Effect of Unpaired Purified α-Hemoglobin Chains on Normal Erythrocytes

Written By

Mark D. Scott

Submitted: 13 June 2019 Reviewed: 28 October 2019 Published: 25 November 2019

DOI: 10.5772/intechopen.90288

From the Edited Volume

Beta Thalassemia

Edited by Marwa Zakaria and Tamer Hassan

Chapter metrics overview

1,196 Chapter Downloads

View Full Metrics

Abstract

β thalassemias arise from genetic defects that interfere with the synthesis of the β hemoglobin chain and the subsequent production of the normal α2β2 hemoglobin tetramer. As a consequence of this decreased β-chain synthesis, unpaired α-hemoglobin chains are found within the red blood cell (RBC). The unstable α-chains are associated with a number of cellular defects, including: membrane-bound globin; membrane thiol oxidation; altered cytoskeletal proteins; decreased cellular and membrane deformability; and increased susceptibility to both endogenous and exogenous oxidants. Surprisingly, while significant injury to human thalassemic RBC arise from the unpaired α-chains, the underlying intra-RBC mechanisms are not easily studied in patient samples or in mouse models. To better study the fate of excess α-chains in human RBC, the model β Thalassemic cell was developed. Model human β thalassemic RBC is made by entrapping purified human α-chains within normal RBC via osmotic lysis and resealing. This human model allows for the systematic examination of the mechanisms underlying the α-chain mediated damage in the β thalassemic RBC. Studies utilizing the model β thalassemic RBC have demonstrated that the α-chains give rise to an iron and glutathione-dependent, self-amplifying and self-propagating oxidative reaction.

Keywords

  • β thalassemia
  • α-hemoglobin chains
  • iron
  • red blood cell
  • erythrocyte
  • oxidation
  • free radicals
  • glutathione
  • deformability

1. Introduction

The thalassemias are a major cause of morbidity and mortality throughout much of the world [1, 2, 3, 4, 5, 6, 7, 8, 9]. Thalassemias are characterized by the disruption of the synthesis of normal adult hemoglobin (HbA; an α2β2 tetramer; Figure 1) consequent to a diverse array of genetic mutations/deletions to either the β or α-hemoglobin chain genes (Chromosomes 11 and 16, respectively). As a consequence of reduced/absent production of β-chains, β thalassemia is characterized by the presence of highly unstable monomeric α-chains as these chains cannot self-associate and indeed require a chaperone protein to prevent precipitation [10]. In contrast, α thalassemia is characterized by the presence of relatively stable tetrameric β chains. Interestingly, as schematically shown in Figure 1, unlike most genes, there are four copies of the α-globin genes; this is in contrast to the expected two copies of the β-globin genes. The evolutionary duplication of the human α-chain genes may have been favored consequent to the inherent instability of monomeric α-chains. Indeed, the instability of the α-chains is the key factor underlying the pathophysiology of the β thalassemic red blood cell (RBC). Moreover, the pathophysiology of β thalassemia can be further complicated by the geographical prevalence, and high frequency, of a number of mutated β hemoglobin genes (e.g., sickle hemoglobin, hemoglobin E and hemoglobin C). If a mutated β-chain is the only functional β-chain present, the resultant disease will be more severe than that observed in β thalassemia Intermedia (a single normal β-globin gene). Loss of both β-chain genes gives rise to severe β Thalassemia Major which is fatal in the absence of transfusion therapy. The α thalassemias are characterized by a broader range of disease states due to the presence of 4 α genes. The loss of expression from a single gene (α Thalassemia1 Trait) is often asymptomatic and undiagnosed; though the individual is a carrier for α Thalassemia and, in high frequency geographic areas may be at elevated risk for symptomatic disease transmission to an offspring. Deletion of two or three α-genes results in severe disease as a single active α-gene cannot, due to the instability of the chain, produce sufficient mature α-chains to form sufficient HbA. Loss of all four α-genes is fatal (resulting in Hydrops fetalis) due to the crucial role that α-chains play in embryonic and fetal hemoglobin. In contrast to β thalassemia, stable mutated α-chains are rare so typically these do not pose a significant complication in the pathophysiology of α thalassemia.

Figure 1.

Molecular basis and clinical diagnosis of the α and β thalassemias [1, 2, 3, 4, 5, 6, 7, 8, 9]. β* denotes abnormal β hemoglobin gene such as HbS or HbE.

In this chapter we will further explore the pathophysiology of the β thalassemic RBC. Surprisingly, while significant injury to the thalassemic erythrocyte arises from the excess α-chains, the underlying mechanisms by which these chains damage and subsequently destroy the thalassemic RBC in the bone marrow and peripheral blood have not been clearly delineated. Our lack of understanding of the mechanisms of α-chain mediated damage is due, in part, to three major factors: (1) studies of RBC from β thalassemic individuals are difficult to do since these cells, upon collection, already exhibit significant injury and represent a survivorship bias since up to 80% of erythroid precursors are destroyed within the bone; (2) β thalassemic patients are typically transfused to both correct the severe anemia accompanying the disease and to prevent endogenous erythropoiesis of defective RBC; and (3) the lack of a good experimental model by which the pathophysiology of excess globin chains on human RBC can be examined.

While little can be done to change the first two problems, researchers have attempted to tackle the third issue using murine models of thalassemia [11, 12, 13, 14, 15, 16, 17]. Original murine studies examining the knockout of the murine β-chains were not productive as the murine α-chains behave significantly different from their human counterpart. To overcome this problem, human α-chain genes were inserted into the mouse genome in place of the murine genes. Again, these studies failed to give rise to as severe a phenotype as is seen in the human disease. Subsequent studies utilized additional mutations to produce symptomatic disease in the murine context—albeit with still substantial differences from the pathophysiology seen in the human β thalassemic RBC. Hence, an alternative approach for studying the pathophysiology of unpaired α-chains on the human RBC was needed.

Advertisement

2. Model human β thalassemic RBC

To this end, our laboratory developed an in vitro model of the HUMAN β thalassemic erythrocyte [18, 19, 20, 21, 22, 23, 24, 25, 26, 27]. In this model, purified human α-chains are entrapped within normal human RBC (or, if desired, mouse RBC) by osmotic lysis and resealing (Figure 2) [18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34]. As previously shown, osmotic lysis and resealing results in RBC exhibiting normal hemoglobin concentration and volume (Table 1) as well as normal ATP concentration, oxidant sensitivity, morphology and deformability while allowing for the efficient entrapment of exogenous compounds [18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34]. Indeed, studies with resealed human RBC demonstrate that this methodology can be used to correct enzyme deficiencies, enhance antioxidant levels, and be used in vitro to study malarial growth and maturation. Moreover, murine studies demonstrated that osmotically resealed murine RBC exhibited normal in vivo survival. To manufacture the model β thalassemic RBC from normal human donor cells, purified, heme-containing, α-hemoglobin chains were prepared by dissociation of CO-treated HbA in the presence of parahydroxymercuribenzoate, followed by ion exchange chromatography, to isolate the purified CO-α-chains as previously described [1, 18, 20, 21, 22, 35, 36]. Analysis of the purified α-chains by mass spectroscopy demonstrated the expected mass values for the α-globin chain. The purified α-chains can be stored at −80°C as carbon monoxide stabilized chains (CO-α-chains) and then thawed immediately prior to use.

Figure 2.

Generation of model β thalassemic RBC from normal human donor cells via osmotic lysis and resealing [18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34]. Osmotically lysed and resealed RBC have normal morphology and metabolism and exhibit normal in vivo survival.

Parameter N Normal Resealed* Model β thal* Model α thal*,**
Mean cell volume (μm3) 7 88.9 ± 1.5 85.8 ± 4.5 78.6 ± 0.8 76.0 ± 4.0
Mean cell hemoglobin (pg) 7 30.5 ± 0.8 28.5 ± 2.7 23.4 ± 0.6 26.9 ± 2.1
Mean cell Hb conc. (g/dl) 7 34.2 ± 0.7 33.2 ± 1.7 29.6 ± 0.6 35.5 ± 1.3
Red cell distribution width (RDW) 7 12.9 ± 0.2 17.5 ± 4.5 29.2 ± 0.5 21.3 ± 3.8

Table 1.

Cellular characteristics of model β and α thalassemic RBC [18, 19, 20, 21, 22, 23, 24, 25, 26, 27].

Immediately post resealing and washing 3×.


Made via the entrapment of purified human β hemoglobin chains.


Entrapment of the purified α-chains within normal erythrocytes is done by osmotic lysis and resealing as previously described [18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34]. Briefly, washed, packed erythrocytes (80–85% hematocrit) are mixed with the purified CO-α-chains (10 mg/ml packed red cells) and then placed as a thin film within 11.5 mm diameter dialysis tubing (MW cutoff of 3500). The samples are dialyzed against a hypotonic lysis buffer (5 mM potassium phosphate buffer and 2 mM EDTA; pH 7.4) at 4°C for 60 min. The dialysis tubing is then transferred to an isotonic resealing buffer (5 mM potassium phosphate, 160 mM NaCl, and 5 mM glucose; pH 7.4) with gentle agitation for 30 min at 37°C. Following resealing, cells are washed with saline until the supernatant is clear. Using this procedure, approximately 70–80% of the initial packed erythrocyte volume is recovered. Radiolabeled α-chains can be utilized to quantitate the intracellular entrapment [18, 20].

To determine whether the model β thalassemic erythrocytes exhibit the cellular abnormalities characteristic of true β thalassemic cells, a number of cellular parameters have been examined. The results of these studies demonstrate that the α-chain loaded erythrocytes exhibit structural and functional changes very similar to those seen in β thalassemic erythrocytes (Table 2). Consequently, this model allowed for the systematic examination of the mechanisms underlying the α-chain mediated damage within the β thalassemic RBC and to directly determine the ontogeny of the pathologic events underlying the RBC injury and to experimentally test potential therapeutic approaches.

Characteristic β thalassemic RBC Model β thalassemic RBC
Microcytic RBC (MCV <80 fl) + + (progressive)
K+ Loss + +
Unpaired α-chains 1–3% of total hemoglobin in peripheral blood reticulocytes 3–4% of total hemoglobin
Membrane Bound Globin Increased Increased; correlated with α-chain autoxidation & heme release.
Membrane proteins/thiols Loss of spectrin and ankyrin. Oxidation of membrane thiol (-SH) groups. Loss of spectrin and ankyrin correlated with membrane thiol (-SH) oxidation, α-chain autoxidation, and heme release.
Cellular deformability Decreased cellular and membrane deformability Decreased; correlated with iron/heme deposition in membrane.
Intracellular oxidant stress Evidence supporting increased oxidative damage Increased H2O2; correlated with α-chain autoxidation & heme release.
Oxidant susceptibility Increased Increased; correlated with membrane bound iron/heme.
Membrane bound iron/heme Increased membrane associated hemoglobin & Heinz body formation Increased membrane heme; correlated with α-chain autoxidation.
Short in vivo survival + + (murine model β Thal RBC)

Table 2.

Comparison of the pathologic characteristics of β thalassemic and model β thalassemic erythrocytes [18, 19, 20, 21, 22, 23, 24, 25, 26, 27].

Advertisement

3. Iron-glutathione driven progeria of the thalassemic RBC

Containing approximately 20 mM iron, the RBC is the most ferruginous somatic cell in mammals. Under normal conditions, most of this iron is complexed within hemoglobin (as heme) with virtually none present as free metal (i.e., non-heme). This near perfect compartmentalization of iron may, however, break down in certain pathologic states such as β thalassemia and sickle cell disease resulting in the autoxidation of hemoglobin (i.e., formation of methemoglobin and hemichromes). Of physiologic importance, the monomeric α-chains spontaneously autoxidize to methemoglobin, simultaneously generating superoxide (O2 ), at a rate 8-times that of normal hemoglobin (Figure 3) [18, 20, 22, 23, 24, 25, 26, 27, 37, 38, 39]. Moreover, consequent to the formation of methemoglobin, the non-covalently bound heme is more likely to escape the heme pocket of the globin chain giving rise to elevated levels of free, redox-active, intraerythrocytic iron. The O2 produced via the autoxidation of hemoglobin can reduce ferric (Fe+3) to ferrous (Fe+2) iron or form hydrogen peroxide (H2O2; either spontaneously or enzymatically via superoxide dismutase). Importantly, the iron, O2 , and H2O2 can, via the Haber-Weiss Reaction, give rise to the formation of the ‘dreaded’ hydroxyl radical (•OH) which rapidly reacts with virtually all biological constituents converting pristine materials into biological garbage (Figure 3). However, despite the general concept that free iron and the formation of free radicals are bad, the actual iron-dependent pathophysiology of the β thalassemic RBC has been poorly understood.

Figure 3.

Free radical basis of α-hemoglobin autoxidation and free radical injury. Consequent to the formation of methemoglobin, the non-covalently bound heme is 8-times more likely to escape the globin chain. Release of the heme initiates iron-dependent free radical reactions. References: [18, 19, 20, 21, 22, 23, 24, 25, 26, 27].

Mechanistically, studies using the model β thalassemic RBC have demonstrated that the initial autoxidation of the unpaired α-chains initiates an iron and reduced glutathione (GSH) dependent, self-amplifying and self-propagating reaction with the subsequent release of even more heme and, eventually, free iron. Schematically, this self-amplifying, self-propagating injury pathway is shown in Figure 4. As noted, the reaction process is initiated by the autoxidation of the unpaired α-chains which gives rise to the release of free heme and the generation of O2 . Interestingly, at this point, a key component of this pathway is GSH; an ‘anti-oxidant’ present at high intracellular (~2.3 mM) concentrations within the RBC [41, 42]. GSH readily reacts with free heme resulting in the cleavage of the heme ring, the release of free iron, and the formation of a thiol radical (GS•). This reaction leads to the rapid amplification of the oxidative damage to the RBC [18, 20, 22, 23, 24, 25, 26, 27]. .The importance of the released iron and GSH was documented experimentally. As shown in Figure 5A, addition of Fe+3 to hemolysates from normal RBC results in the rapid oxidation of oxyhemoglobin in an iron-dose dependent manner. The oxidation of hemoglobin can be inhibited by the inclusion of an iron chelator (shown is deferoxamine; DFO) or by chemical depletion of GSH (not shown) [23, 27]. Moreover, in the intact model β thalassemic RBC, chemical depletion of GSH inhibited iron-driven hemoglobin oxidation (Figure 5B) [23, 27]. In contrast, as shown in Figure 5C, increasing the amount of intracellular GSH (via osmotic lysis and resealing; see Figure 2) in the model β thalassemic RBC significantly exacerbated injury to the cell. This enhanced injury is readily seen by the significantly reduced deformability (i.e., increased mean cell transit time) of the GSH-loaded model β thalassemic cells relative to the control model β thalassemic RBC. Importantly, as shown, GSH supplementation of normal RBC in the absence of iron had no detrimental effects on cellular deformability.

Figure 4.

Damage to multiple components of the β thalassemic RBC is mediated by an Fe-GSH dependent mechanism. As thalassemic RBC circulate, α-hemoglobin chains autoxidize (A) giving rise to superoxide (O2 ) and methemoglobin [MetHb]. The heme moiety of the α metHb chains are released yielding free heme and globin (B). Free heme reacts with reduced glutathione [GSH; reaction (C)] resulting in cleavage of the heme group and the formation of a glutathione radical (GS•) and the release of free Fe+2 (D). The free iron (E) reacts with hemoglobin derived reactive oxygen species (ROS: O2 and H2O2) to generate •OH – A highly reactive radical capable of damaging all cellular components including cellular metabolism and cation/anion balance. The ferric iron (Fe+3), in the presence of additional GSH or O2 regenerates Fe+2 which then oxidizes additional hemoglobin (F). Reactions (G) and (H) represent subsequent oxidative and non-oxidative injury to the cell. Oxidative pathway generated from references: [18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 40].

Figure 5.

Role of iron and GSH in the destruction of the β thalassemic RBC. (A) Fe+3 is a potent accelerator of hemoglobin oxidation. Shown is the oxyhemoglobin concentration following the addition of 100, 175 and 250 μM Fe+3 to a fresh RBC hemolysate. In contrast to the Fe+3-treated samples, no oxidation was observed in the absence of added Fe+3 or in the presence of an iron chelator [DFO (deferoxamine) or, not shown, the tripeptide Gly-His-Lys]. Also shown are sequential, hemoglobin scans of the hemolysates treated with Fe+3. The time, in minutes, of the individual scan line following addition of Fe+3 are indicated [23, 27]. (B) Depletion of GSH in model β thalassemic RBC inhibits hemoglobin oxidation. Shown is the percent oxyhemoglobin in control-resealed and model β thalassemic cells RBC over 27 h at 37°C. GSH was depleted by treatment with 1-chloro-2.4-dinitrobenzene (CDNB) as previously described [18, 20, 31, 32]. (C) Elevated intracellular GSH levels in model β thalassemic RBC adversely affects cellular deformability as demonstrated by the cell transit analyzer. Less deformable cells take much longer to transit through a pore of known diameter and length. The results shown are the mean ± S.D. of a minimum of four experiments with >2000 RBC measured at each time point. From references: [23, 27].

The enhanced oxidative stress, and consumption of GSH, was further noted by the time dependent decrease in GSH noted in the model β thalassemic RBC (Table 3) [27]. Moreover, a decrease in the NADPH/NADPtotal ratio was noted. NADPH is utilized to regenerate reduced GSH from oxidized (GSSG) glutathione via glutathione reductase. This decrease in the NADPH/NADPtotal ratio is likely reflective of both the high GSH-GSSG-GSH cycling but also of metabolic abnormalities arising consequent to iron and free radical mediated inhibition of normal glucose metabolism (Figure 4). Also of physiological importance, was the finding that the model β thalassemic RBC exhibited significantly (p < 0.001) decreased catalase activity (Table 3). Catalase is the pre-eminent defense against H2O2 within the RBC, making β thalassemic cell particularly increasingly sensitive to H2O2 generated via the pathway described in Figure 4 [31, 32, 33, 34, 43]. Following 20 h incubation at 37°C, only 61.5 ± 2.9% of the initial catalase activity remained in the α-hemoglobin chain loaded cells versus 104.6 ± 4.5% in the control RBC. The loss of catalase arose due to the decrease in the NADPH/NADPtotal ratio as studies have demonstrated that NADPH is essential for maintaining catalase in an enzymatically active state [31, 32, 33, 34, 43, 44, 45, 46, 47]. Indeed, as noted in Table 3, the model β thalassemic erythrocytes exhibit a significant decrease in the NADPH/NADPtotal ratio similar to that seen in severe G6PD deficiency. Hence, consequent to the oxidation of hemoglobin and the formation of free radicals in the thalassemic RBC, significant metabolic and functional changes are noted in the model β thalassemic RBC that mirror those seen in patient derived samples (Figure 4 and Table 2). It is also important to note that non-oxidative driven damage also occurs. As shown in Figure 4, precipitated globin proteins, as well as iron and heme, can alter the intracellular viscosity of the cytoplasm and interact with the cytoskeleton and membrane lipids resulting in mechanical dysfunction. These oxidative and non-oxidative injuries can dramatically affect the function of the RBC.

Population Hours (37°C) GSH (μmol/g Hb) NADPH/NADPtotal (1.0 = Normal) Cat. activity (IU/g Hb)
Control RBC 0 5.6 ± 0.4 0.870 147,300 ± 17,000
20 6.0 ± 0.6 1.063 154,100 ± 14,800
Model β Thal 0 5.2 ± 0.3 0.898 145,600 ± 12,300
20 2.1 ± 0.0* 0.478* 89,540 ± 8200*

Table 3.

Association between NADPH, GSH and catalase.

p < 0.001 from time-matched Control RBC.


Modified from [27].

Advertisement

4. Loss of RBC deformability and vascular survival

From a functional standpoint, perhaps the most important consequence of the oxidative changes to the β thalassemic RBC, as well as other RBC abnormalities, is the loss of cellular deformability [18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 29, 30, 31, 32, 33, 48, 49, 50, 51, 52]. The physiology, fluidics and vascular bed of the circulatory system impart unique rheological stresses on circulating RBC (Figure 6A) [53]. These include extreme variations in shear stress and viscosity as well as biomechanical obstacles (e.g., capillaries and splenic filtration). With an average resting cardiac output of approximately 5 L/min, blood flow varies from approximately 40 cm/s in the aorta to 0.03 cm/s in the smallest capillaries [5354]. Moreover, blood viscosity (affecting shear stress) is also variable. At high RBC counts and high flow rates, blood is highly viscous while at low RBC counts and low flow rates (capillaries), blood viscosity is greatly reduced. Rheological stress is further exacerbated by the biomechanical stresses induced by the extreme disparity in the size of RBC (~8 μm) to the minimum diameter of the vascular capillary beds (4–5 μm) and splenic interendothelial clefts (0.5–1.0 μm) [55, 56]. Hence, consequent to the shear forces, viscosity and biomechanical stresses placed on blood cells, a key biologic/physiologic requirement of the RBC within the vascular space is rheological deformability. Biomechanically, the intracellular viscosity and membrane rigidity of the RBC are the key factors in imparting their vascular rheological deformability; both of which can be dramatically altered consequent to hemoglobin oxidation, heme release and/or redox-damage to cytoplasmic, cytoskeletal, or membrane components (Figure 4) [57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68]. Indeed, as shown in Figure 6B, ektacytometric analysis of the model β thalassemic RBC shows a significant loss of cellular deformability induced by shear stress (e.g., large vessels). Moreover, cell transit analysis of these cells (analogous to capillary deformation) showed a very significant loss of deformability in the model β thalassemic cells as reflected by the very large and significant increase in transit time (Figure 6C).

Figure 6.

Vascular deformability of model β thalassemic RBC. (A) The vascular bed is composed of blood vessels of various sizes which create significant disparity in blood (fluid and cellular) velocity consequent to vessel diameter. The fluid flow induces rheological sheer stress while the vessel size creates biomechanical deformation of cellular elements. Shown is an RBC undergoing deformation in a microfluidic channel. (B) Ektacytometric analysis of α-chain loaded RBC demonstrate that these chains dramatically reduce the sheer-induced shape change of the cell [24]. Ektacytometry bests approximates high flow rates. (C) The mean cell transit time (in ms) of model β thalassemic cells was significantly increased, similarly to that observed in patient samples [20]. This microfluidic flow best approximates capillary flow.

Consequent to the loss of deformability and immune recognition (e.g., Kupffer cells of the liver and, potentially, antibodies), the circulatory survival of β thalassemic RBC is impacted. As demonstrated in Figure 7A, model β thalassemic RBC (Blood group A) exhibited enhanced immune recognition and phagocytosis by autologous monocytes when compared to control cells from the same donor. Indeed, the level of phagocytosis was similar to that of the anti-A opsonized positive control RBC. The loss of deformability and enhanced immune recognition both contribute to decreased in vivo survival. This was demonstrated using mice transfused with model β thalassemic murine cells (mouse RBC + human α-chains) in which the transfused RBC exhibited a dramatic reduction in the circulatory lifespan (Figure 7B). The role of α-chain mediated oxidation was supported by the finding that lightly oxidized (phenazine methosulphate treated; 50 μM) murine cells showed similar circulatory dynamics. These results are comparable to that observed in humans where, consequent to the α-chain driven oxidation, β thalassemic RBC have a very short circulatory lifespan (7–14 days depending on spleen status) compared to the 120 days of a normal RBC.

Figure 7.

Model β thalassemic RBC are immunologically recognized and cleared rapidly from the peripheral circulation in vivo. (A) Blood group A RBC that were loaded with purified α-chains were phagocytosed by human monocytes. Shown for comparison are control cells from the same donor that were either unopsonized or opsonized with an anti-A blood typing antibody. (B) Model β thalassemic mouse RBC were made by the entrapment of purified human α-hemoglobin chains within Normal murine RBC. Oxidatively damaged RBC were made by treating mouse RBC with 50 μM phenazine methosulfate (PMS) for 2.5 h then washing. Both PMS-treatment and the entrapment of α-hemoglobin chains lead to membrane iron deposition and extensive RBC oxidation. The model β thalassemic and PMS-treated RBC have half-lives of 4 and 7 days, respectively in the peripheral circulation versus 26 days for normal murine RBC. Shown is the percent total RBC mass that is PKH-26 labeled. Inset: The peripheral blood lifespan of the human β thalassemic RBC are approximately 6–10 days in a patient with a functioning spleen and 12–16 days in a splenectomized individual. Normal RBC circulates approximately 120 days, data modified from Blendis et al. [69].

Advertisement

5. Immunological dysfunction: Effect on antigen presentation

Interestingly, thalassemias have been clinically associated with an increased risk of recurrent bacterial infections [70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87]. This is most evident in under-developed nations where sanitary and medical facilities are most lacking. Despite the clinical evidence of recurrent bacterial infections in thalassemic patients, the biological events underlying this finding are unclear. This confusion arises as a natural consequence of the heterogeneity of the microbial disease itself, the patients age, the state of splenic function, the frequency of transfusion, the degree of similarity between the patient and the blood donor pool, the nutritional status of the patient (e.g., United States versus Thailand) and whether one is looking at humoral or cell-mediated immunity [70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87].

In general, studies on the humoral (i.e., immunoglobulin-based) immunity of thalassemic patients suggest that this arm of the immune system is ‘relatively’ normal. These studies have indicated normal to elevated levels of IgG, IgA, and IgM but decreased levels of Factor B, C3, and C4 (perhaps due to consumption via oxidatively damaged β thalassemic cells). Reflective of this normality, and consequent to the extensive oxidant injury to the thalassemic cells, circulating immune complexes and an elevated risk of autoimmune hemolytic anemia have been described in β thalassemia intermedia and major patients. Serum fractions from these patients also exhibited increased amounts of C1q-precipitable immune complexes. In contrast, as suggested by the clinically described recurrent bacterial infections, cell-mediated immunity is highly suspect in the thalassemic patient (and sickle cell patients). The few direct studies on cell-mediated immunity in thalassemic patients were, typically, enumeration of the mononuclear cell populations (T cells, B cells, NK cells and monocytes). In general, these studies suggest normal cell numbers but a skewed distribution of the CD4+ to CD8+ T cell ratio. The altered ratio was characterized by a relative depression in CD4+ T cells (i.e., helper T cells) and NK (Natural Killer) cells and a relative rise in CD8+ (cytotoxic and suppressor) T cells that increased linearly with the number of units transfused. However, very few functional studies have been done in thalassemic patients to answer the question: Why are β thalassemic patients at risk of recurrent bacterial infections?

Previous studies have suggested that increased bioavailable iron in transfused patients might facilitate the growth of organisms in which iron is a limiting nutrient (i.e., most bacteria). Other studies have implicated the loss of splenic function. While both of these factors may indeed play important roles in recurrent bacterial infections, they may not offer a complete explanation. In addition to thalassemia, a number of other diseases and trauma scenarios are characterized by recurrent bacterial infections (e.g., malaria and burn injury) suggestive of impaired cell-mediated immunity. Interestingly, a common characteristic of all these conditions is erythrophagocytosis. Previous studies have demonstrated that phagocytic uptake of IgG-coated and oxidatively stressed RBC resulted in a transient depression of further macrophage phagocytosis, decreased respiratory burst (i.e., NADPH-oxidase activity; O2 production), and impaired killing of bacteria [88, 89, 90, 91, 92, 93]. Interestingly, in the case of Plasmodium falciparum-infected RBC, only phagocytosis of mature (trophozoite), but not immature (ring stage), stages had an inhibitory effect on monocyte function. Importantly, a major difference between the mature and immature malarial infected RBC is the presence of malarial pigment (hemozoin), an iron/heme rich degradation product of parasite hemoglobin catabolism. Τhe heme- and iron-rich membranes of the β thalassemic RBC, which we have previously documented [22], may function in a manner analogous to malarial pigment or iron salts and impair cell-mediated immunity—primarily at the level of the APC but potentially extending to the T cell level. Some data from thalassemic patients support the hypothesis for impairment of the T cell response. For example, patients with thalassemia intermedia have been reported to have diminished T cell mitogen responses when their serum iron and ferritin were higher than 200 and 600 μg/dl, respectively [94].

Hence, injury arising from the iron-GSH pathway can result in (any combination of) RBC opsonization by endogenous antibodies, phosphatidylserine (PS) exposure, protein clustering, sublytic levels of complement binding, and/or loss of cellular deformability (Figure 7) that leads to the removal of the damaged β thalassemic cells from the circulatory system by components of the mononuclear phagocytic system (MPS). Erythrophagocytosis can occur within the spleen (if present and functioning), liver (Kupffer cells) or the microvasculature itself when non-deformable RBC are trapped and then cleared by circulating macrophages. Regardless of the location of removal, erythrophagocytosis results in impaired MPS function. As shown in Figure 8A, antigen presentation of purified tetanus toxoid (TT; a peptide) or fixed, intact, S. mutans (SM; an intact bacteria) by normal human antigen presenting cells (APC; blood monocytes) was dramatically, and differentially, affected by the presence of either control (unoxidized) or oxidized (50 μM PMS as per Figure 7) human RBC. As shown, oxidized RBC prevented successful antigen presentation to human T cells while normal RBC showed no detrimental effects. Further experimentation demonstrated that the inhibitory effect was due to heme/iron. As shown in Figure 8B,C, direct addition of hemin to the APC impaired successful antigen presentation of both tetanus toxoid and Strep. mutans in a dose dependent manner.

Figure 8.

Antigen processing and presentation is inhibited by oxidized RBC and hemin. (A) Antigen presentation of TT and SM was inhibited by the erythrophagocytosis of oxidized RBC. Normal RBC had no inhibitory effect. The efficacy of antigen processing and/or presentation was assessed by 3H-thymidine incorporation in proliferating T cells. PBMC were resuspended in Aim V media at a final concentration of 2.5 × 105 PBMC per 200 μl. Final RBC concentration was 8 × 106 per 200 μl. Antigens were diluted in Aim V and added at the indicated concentrations. Results shown are of a representative experiment with quadruplicate samples. (B, C) Heme pretreatment of PBMC (2 h at 37°C) dramatically inhibits the proliferative response to tetanus toxoid (B) and S. mutans (C). Interestingly, the ability of PBMC to respond to intact bacteria is more significantly blunted than is the response to tetanus toxoid at intermediate hemin concentrations (50 μM). Results shown are of a representative experiment with quadruplicate samples.

Advertisement

6. Conclusions

β thalassemias arise from a number of underlying genetic defects that interfere with the synthesis of the β hemoglobin chain and the subsequent production of the normal α2β2 hemoglobin tetramer. As a consequence of this decreased/absent β-chain synthesis, unpaired, monomeric, α-hemoglobin chains are produced. While the presence of the highly unstable α-chains mediate the pathophysiology of the RBC, it has been difficult to fully elucidate the mechanisms underlying their destructive processes in human cells. This lack of understanding of the mechanisms of α-chain mediated damage is due, in large part, to the fact that peripheral RBC isolated from β thalassemic individuals are already severely damaged cells (with most being destroyed within the bone marrow). Moreover, severe β thalassemia patients are typically transfused to both correct the severe anemia accompanying the disease and to prevent endogenous erythropoiesis of defective RBC. Hence, murine models of β thalassemia have been developed and extensively studied. However, problems exist with these models (e.g., mouse vs. human α-chains; interaction of human globins with mouse cytoskeletal proteins) and these mice, as in human patients, still suffer from the heterogeneity of RBC changes arising from the different ages of the peripheral blood RBC [11, 12, 13, 14, 15, 16, 17].

To better study the fate of unpaired α-chains in human RBC, the model β thalassemic cell was developed [18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34]. The entrapment of purified α-hemoglobin chains within normal erythrocytes via osmotic lysis and resealing provides an excellent and reproducible human model for studying the pathologic effects of the unpaired α-chains on the structural and functional characteristics of the RBC. Indeed, as noted in Table 2, the α-chain induced structural and functional RBC changes are very similar to those observed in human donor derived β thalassemic RBC. Schematically the pathophysiology of the β thalassemic RBC, and its downstream consequences, as elucidated by the model human β thalassemic RBC, are summarized in Figure 9. Importantly, these studies have demonstrated that the unpaired α-chains initiate an iron, GSH-dependent, self-amplifying and self-propagating reaction with the subsequent release of even more heme and, eventually, free iron (Figure 4). Membrane proteins and reactive thiol groups (not shown) were rapidly decreased in a pattern similar to that observed in vivo in β thalassemia [18, 20, 21, 22, 23, 25, 26, 27]. These oxidative events also result in membrane vesiculation of the thalassemic RBC. One consequence of membrane vesiculation is the preferential loss of phosphatidylinositol (PI) anchored proteins from the RBC. Among these PI-anchored proteins are decay accelerating factor (DAF; CD55) and the membrane inhibitor of reactive lysis (MIRL; CD59) both of which play important roles in preventing complement-mediated binding and lysis. The effects of the vesiculation-mediated loss of CD55 and CD59 can range from sublytic levels of bound complement enhancing phagocytosis to overt hemolysis. Indeed, a common endpoint for all the α-chain mediated injury is enhanced erythrophagocytosis. As shown, oxidized RBC or the heme from these cells (Figure 8) significantly inhibits antigen processing, presentation and T cell proliferation. The systemic importance of this on cell-mediated immunity has not be fully appreciated and may potentially explain the predisposition of thalassemic patients to recurrent bacterial infections.

Figure 9.

Schematic representation of the pathophysiology of the β thalassemic RBC and its immunological consequences.

In sum, these findings show the utility of the model β thalassemic human RBC for investigating the pathophysiology of the unpaired α-chains. Moreover, these cells are easily ‘manufactured’ from normal donor RBC and may provide an effective means to evaluate therapeutic approaches to ameliorate the damage to the thalassemic cell in β thalassemia intermedia in order to prolong RBC survival and reduce transfusions [23, 25, 26, 27, 40].

Advertisement

Acknowledgments

This work was supported by grants from Canadian Blood Services and Health Canada. The views expressed herein do not necessarily represent the view of the federal government of Canada. We thank the Canada Foundation for Innovation and the Michael Smith Foundation for Health Research for infrastructure funding at the University of British Columbia Centre for Blood Research. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Advertisement

Conflict of interest

There are no conflicts of interest.

References

  1. 1. Robinson AR, Robson M, Harrison AP, Zuelzer WW. A new technique for differentiation of hemoglobin. The Journal of Laboratory and Clinical Medicine. 1957;50:745-752. PMID: 13476032
  2. 2. Weatherall DJ, Clegg JB. Molecular basis of thalassemia. British Journal of Haematology. 1975;31:133-141. DOI: 10.1111/j.1365-2141.1975.tb00906.x
  3. 3. Weatherall DJ. Pathophysiology of thalassaemia. Baillière’s Clinical Haematology. 1998;11:127-146. PMID: 10872475
  4. 4. Higgs DR. The molecular basis of α-thalassemia. Cold Spring Harbor Perspectives in Medicine. 2013;3:a011718. DOI: 10.1101/cshperspect.a011718
  5. 5. Piel FB, Weatherall DJ. The α-thalassemias. The New England Journal of Medicine. 2014;371:1908-1916. DOI: 10.1056/NEJMra1404415
  6. 6. Mettananda S, Gibbons RJ, Higgs DR. α-Globin as a molecular target in the treatment of β-thalassemia. Blood. 2015;125:3694-3701. DOI: 10.1182/blood-2015-03-633594
  7. 7. Mettananda S, Higgs DR. Molecular basis and genetic modifiers of thalassemia. Hematology/Oncology Clinics of North America. 2018;32:177-191. DOI: 10.1016/j.hoc.2017.11.003
  8. 8. Taher AT, Weatherall DJ, Cappellini MD. Thalassaemia. Lancet. 2018;391:155-167. DOI: 10.1016/S0140-6736(17)31822-6
  9. 9. Weatherall DJ. The evolving spectrum of the epidemiology of thalassemia. Hematology/Oncology Clinics of North America. 2018;32:165-175. DOI: 10.1016/j.hoc.2017.11.008
  10. 10. Turbpaiboon C, Wilairat P. Alpha-hemoglobin stabilizing protein: Molecular function and clinical correlation. Frontiers in Bioscience (Landmark Edition). 2010;15:1-11. PMID: 20036801
  11. 11. Rubin EM, Kan YW, Mohandas N. Effect of human beta (s)-globin chains on cellular properties of red cells from beta-thalassemic mice. The Journal of Clinical Investigation. 1988;82:1129-1133. DOI: 10.1172/JCI113670
  12. 12. Sorensen S, Rubin E, Polster H, Mohandas N, Schrier S. The role of membrane skeletal-associated alpha-globin in the pathophysiology of beta-thalassemia. Blood. 1990;75:1333-1336. PMID: 1690033
  13. 13. Advani R, Rubin E, Mohandas N, Schrier SL. Oxidative red blood cell membrane injury in the pathophysiology of severe mouse β-thalassemias. Blood. 1992;79:1064-1067. PMID: 1737090
  14. 14. Pászty C. Transgenic and gene knock-out mouse models of sickle cell anemia and the thalassemias. Current Opinion in Hematology. 1997;4:88-93. PMID: 9107524
  15. 15. Nishino T, Cao H, Stamatoyannopoulos G, Emery DW. Effects of human gamma-globin in murine beta-thalassaemia. British Journal of Haematology. 2006;134:100-108. DOI: 10.1111/j.1365-2141.2006.06102.x
  16. 16. Huo Y, McConnell SC, Liu SR, Yang R, Zhang TT, Sun CW, et al. Humanized mouse model of Cooley’s anemia. The Journal of Biological Chemistry. 2009;284:4889-4896. DOI: 10.1074/jbc.M805681200
  17. 17. McColl B, Vadolas J. Animal models of β-hemoglobinopathies: Utility and limitations. Journal of Blood Medicine. 2016;7:263-274. DOI: 10.2147/JBM.S87955
  18. 18. Scott MD, Rouyer-Fessard P, Lubin BH, Beuzard Y. Entrapment of purified alpha-hemoglobin chains in normal erythrocytes. A model for beta thalassemia. Journal of Biological Chemistry. 1990;265:17953-17959. PMID: 2211672
  19. 19. Rouyer-Fessard P, Scott MD, Leroy-Viard K, Garel MC, Bachir D, Galacteros F, et al. Fate of alpha-hemoglobin chains and erythrocyte defects in beta-thalassemia. Annals of the New York Academy of Sciences. 1990;612:106-117. DOI: 10.1111/j.1749-6632.1990.tb24296.x
  20. 20. Scott MD, Rouyer-Fessard P, Ba MS, Lubin BH, Beuzard Y. Alpha- and beta-haemoglobin chain induced changes in normal erythrocyte deformability: Comparison to beta thalassaemia intermedia and Hb H disease. British Journal of Haematology. 1992;80:519-526. DOI: 10.1111/j.1365-2141.1992.tb04567.x
  21. 21. Scott MD. Entrapment of purified alpha-hemoglobin chains in normal erythrocytes as a model for human beta thalassemia. Advances in Experimental Medicine and Biology. 1992;326:139-148. PMID: 1295299
  22. 22. Scott MD, van den Berg JJ, Repka T, Rouyer-Fessard P, Hebbel RP, Beuzard Y, et al. Effect of excess alpha-hemoglobin chains on cellular and membrane oxidation in model beta-thalassemic erythrocytes. The Journal of Clinical Investigation. 1993;91:1706-1712. DOI: 10.1172/JCI116380
  23. 23. Scott MD, Eaton JW. Thalassaemic erythrocytes: Cellular suicide arising from iron and glutathione-dependent oxidation reactions? British Journal of Haematology. 1995;91:811-819. DOI: 10.1111/j.1365-2141.1995.tb05394.x
  24. 24. Kuypers FA, Schott MA, Scott MD. Phospholipid composition and organization in model beta-thalassemic erythrocytes. American Journal of Hematology. 1996;51:45-54. DOI: 10.1002/(SICI)1096-8652(199601)51:1<45::AID-AJH8>3.0.CO;2-7
  25. 25. Scott MD, Yang L, Ulrich P, Shupe T. Pharmacologic interception of heme: A potential therapeutic strategy for the treatment of ß thalassemia? Redox Report. 1997;3:159-167. DOI: 10.1080/13510002.1997.11747104
  26. 26. Scott MD. Intraerythrocytic iron chelation: A new therapy for thalassemia. Hematology. 2001;6:73-89. DOI: 10.1080/10245332.2001.11746557
  27. 27. Scott MD. H2O2 injury in beta thalassemic erythrocytes: Protective role of catalase and the prooxidant effects of GSH. Free Radical Biology & Medicine. 2006;40:1264-1272. DOI: 10.1016/j.freeradbiomed.2005.11.017
  28. 28. Scott MD, Eaton JW, Kuypers FA, Chiu D-Y, Lubin BH. Enhancement of erythrocyte superoxide dismutase activity: Effects on cellular oxidant defense. Blood. 1989;74:2542-2549. PMID: 2553167
  29. 29. Scott MD, Kuypers FA, Butikofer P, Bookchin RM, Ortiz OE, Lubin BH. Effect of osmotic lysis and resealing on red cell structure and function. The Journal of Laboratory and Clinical Medicine. 1990;115:470-480. PMID: 1691257
  30. 30. Scott MD, Ranz A, Kuypers FA, Lubin BH, Meshnick SR. Parasite uptake of desferroxamine: A prerequisite for antimalarial activity. British Journal of Haematology. 1990;75:598-602. DOI: 10.1111/j.1365-2141.1990.tb07805.x
  31. 31. Scott MD, Lubin BH, Zuo L, Kuypers FA. Erythrocyte defense against hydrogen peroxide: Preeminent importance of catalase. The Journal of Laboratory and Clinical Medicine. 1991;118:7-16. PMID: 2066646
  32. 32. Scott MD, Zuo L, Lubin BH, Chiu DT. NADPH, not glutathione, status modulates oxidant sensitivity in normal and glucose-6-phosphate dehydrogenase-deficient erythrocytes. Blood. 1991;77:2059-2064. PMID: 2018843
  33. 33. Scott MD, Wagner TC, Chiu DT. Decreased catalase activity is the underlying mechanism of oxidant susceptibility in glucose-6-phosphate dehydrogenase-deficient erythrocytes. Biochimica et Biophysica Acta. 1993;1181:163-168. DOI: 10.1016/0925-4439(93)90106-b
  34. 34. Scott MD. Glucose-6-phosphate dehydrogenase deficiency: A new hypothesis for an old disease. Redox Report. 1995;1:235-237. DOI: 10.1080/13510002.1995.11746992
  35. 35. Bucci E, Fronticelli C. A new method for the preparation of alpha and beta subunits of human hemoglobin. The Journal of Biological Chemistry. 1965;240:PC551-PC552. PMID: 14253474
  36. 36. Abraham EC, Reese A, Stallings M, Huisman TH. Separation of human hemoglobins by DEAE-cellulose chromatography using glycine-KCN-NaC1 developers. Hemoglobin. 1976;1:27-44. PMID: 1052169
  37. 37. Bunn HF, Jandl JH. Exchange of heme among hemoglobins and between hemoglobin and albumin. The Journal of Biological Chemistry. 1968;243:465-475. PMID: 4966113
  38. 38. Brunori M, Falcioni G, Fioretti E, Giardina B, Rotilio G. Formation of superoxide in the autoxidation of the isolated α and β chains of human hemoglobin and its involvement in hemichrome precipitation. European Journal of Biochemistry. 1975;53:99-104. DOI: 10.1111/j.1432-1033.1975.tb04046.x
  39. 39. Joshi W, Leb L, Piotrowski J, Fortier N, Snyder LM. Increased sensitivity of isolated alpha subunits of normal human hemoglobin to oxidative damage and crosslinking with spectrin. The Journal of Laboratory and Clinical Medicine. 1983;102:46-52. PMID: 6854133
  40. 40. Rossi NA, Mustafa I, Jackson JK, Burt HM, Horte SA, Scott MD, et al. In vitro chelating, cytotoxicity, and blood compatibility of degradable poly(ethylene glycol)-based macromolecular iron chelators. Biomaterials. 2009;30:638-648. DOI: 10.1016/j.biomaterials.2008.09.057
  41. 41. Lunn G, Dale GL, Beutler E. Transport accounts for glutathione turnover in human erythrocytes. Blood. 1979;54:238-244. PMID: 444668
  42. 42. Raftos JE, Whillier S, Kuchel PW. Glutathione synthesis and turnover in the human erythrocyte: Alignment of a model based on detailed enzyme kinetics with experimental data. The Journal of Biological Chemistry. 2010;285:23557-23567. DOI: 10.1074/jbc.M109.067017
  43. 43. Wagner TC, Scott MD. Single extraction method for the spectrophotometric quantification of oxidized and reduced pyridine nucleotides in erythrocytes. Analytical Biochemistry. 1994;222:417-426. DOI: 10.1006/abio.1994.1511
  44. 44. Eaton JW, Boraas M, Etkin NL. Catalase activity and red cell metabolism. In: Brewer GJ, editor. Hemoglobin and Red Cell Structure and Function. New York: Plenum Publishing Corp; 1972. pp. 121-131. DOI: 10.1007/978-1-4684-3222-0_8
  45. 45. Kirkman HN, Gaetani GF. Catalase: A tetrameric enzyme with four tightly bound molecules of NADPH. Proceedings of the National Academy of Sciences of the United States of America. 1984;81:4343-4347. DOI: 10.1073/pnas.81.14.4343
  46. 46. Kirkman HN, Galiano S, Gaetani GF. The function of catalase-bound NADPH. The Journal of Biological Chemistry. 1987;262:660-666. PMID: 3805001
  47. 47. Gaetani GF, Canepa L, Galiano S, Ferraris AM, Kirkman HK. Catalase and glutathione peroxidase are equally active in the detoxification of hydrogen peroxide in human erythrocytes. Blood. 1989;73:334-339. PMID: 2491951
  48. 48. Scott MD, Meshnick SR, Williams RA, Chiu DT, Pan HC, Lubin BH, et al. Qinghaosu-mediated oxidation in normal and abnormal erythrocytes. The Journal of Laboratory and Clinical Medicine. 1989;114:401-406. PMID: 2794752
  49. 49. Meshnick SR, Scott MD, Lubin B, Ranz A, Eaton JW. Antimalarial activity of diethyldithiocarbamate. Potentiation by copper. Biochemical Pharmacology. 1990;40:213-216. DOI: 10.1016/0006-2952(90)90680-J
  50. 50. Kuypers FA, Scott MD, Schott MA, Lubin B, Chiu DT. Use of ektacytometry to determine red cell susceptibility to oxidative stress. The Journal of Laboratory and Clinical Medicine. 1990;116:535-545. PMID: 2212862
  51. 51. Hong YL, Pan HZ, Scott MD, Meshnick SR. Activated oxygen generation by a primaquine metabolite: Inhibition by antioxidants derived from Chinese herbal remedies. Free Radical Biology & Medicine. 1992;12:213-218. DOI: 10.1016/0891-5849(92)90029-G
  52. 52. Minetti M, Mallozzi C, Scorza G, Scott MD, Kuypers FA, Lubin BH. Role of oxygen and carbon radicals in hemoglobin oxidation. Archives of Biochemistry and Biophysics. 1993;302:233-244. DOI: 10.1006/abbi.1993.1205
  53. 53. Aird WC. Spatial and temporal dynamics of the endothelium. Journal of Thrombosis and Haemostasis. 2005;3:1392-1406. DOI: 10.1111/j.1538-7836.2005.01328.x
  54. 54. Wexler L, Bergel DH, Gabe IT, Makin GS, Mills CJ. Velocity of blood flow in normal human venae cavae. Circulation Research. 1968;23:349-359. DOI: 10.1161/01.RES.23.3.349
  55. 55. Weiss L, Tavassoli M. Anatomical hazards to the passage of erythrocytes through the spleen. Seminars in Hematology. 1970;7:372-380. PMID: 5473419
  56. 56. Chen LT, Weiss L. The role of the sinus wall in the passage of erythrocytes through the spleen. Blood. 1973;41:529-537. PMID: 4688868
  57. 57. LaCelle PL. Alteration of membrane deformability in hemolytic anemias. Seminars in Hematology. 1970;7:355-371. PMID: 5473418
  58. 58. Weed RI. The importance of erythrocyte deformability. The American Journal of Medicine. 1970;49:147-150. DOI: 10.1016/S0002-9343(70)80069-9
  59. 59. Chien S, Usami S, Bertles JF. Abnormal rheology of oxygenated blood in sickle cell anemia. The Journal of Clinical Investigation. 1970;49:623-634. DOI: 10.1172/JCI106273
  60. 60. Chien S, Usami S, Dellenback RJ, Gregersen MI. Shear-dependent deformation of erythrocytes in rheology of human blood. The American Journal of Physiology. 1970;219:136-142. DOI: 10.1152/ajplegacy.1970.219.1.136
  61. 61. Bessis M, Mohandas N. Red cell structure, shapes and deformability. British Journal of Haematology. 1975;31:5-11. DOI: 10.1111/j.1365-2141.1975.tb00893.x
  62. 62. La Celle PL. Pathogenic erythrocytes in the capillary microcirculation. Blood Cells. 1975;1:269-284
  63. 63. Havell TC, Hillman D, Lessin LS. Deformability characteristics of sickle cells by microelastimetry. American Journal of Hematology. 1978;4:9-16. DOI: 10.1002/ajh.2830040103
  64. 64. Mohandas N, Phillips WM, Bessis M. Red blood cell deformability and hemolytic anemias. Seminars in Hematology. 1979;16:95-114. PMID: 384522
  65. 65. Clark MR, Mohandas N, Shohet SB. Deformability of oxygenated irreversibly sickled cells. The Journal of Clinical Investigation. 1980;65:189-195. DOI: 10.1172/JCI109650
  66. 66. Clark MR, Mohandas N, Shohet SB. Osmotic gradient ektacytometry: Comprehensive characterization of red cell volume and surface maintenance. Blood. 1983;61:899-910. PMID: 6831052
  67. 67. Snyder LM, Fortier NL, Trainor J, Jacobs J, Leb L, Lubin B, et al. Effect of hydrogen peroxide exposure on normal human erythrocyte deformability, morphology, surface characteristics, and spectrin-hemoglobin cross-linking. The Journal of Clinical Investigation. 1985;76:1971-1977. DOI: 10.1172/JCI112196
  68. 68. Snyder LM, Fortier NL, Leb L, McKenney J, Trainor J, Sheerin H, et al. The role of membrane protein sulfhydryl groups in hydrogen peroxide-mediated membrane damage in human erythrocytes. Biochimica et Biophysica Acta. 1988;937:229-240. DOI: 10.1016/0005-2736(88)90245-3
  69. 69. Blendis LM, Modell CB, Bowdler AJ, Williams R. Some effects of splenectomy in thalassaemia major. British Journal of Haematology. 1974;28:77-87. DOI: 10.1111/j.1365-2141.1974.tb06641.x
  70. 70. Dickerman JD. Bacterial infection and the asplenic host: A review. The Journal of Trauma. 1976;16:662-668. DOI: 10.1097/00005373-197608000-00012
  71. 71. Dickerman JD. Splenectomy and sepsis: A warning. Pediatrics. 1979;63:938-941. PMID: 377203
  72. 72. Borgna-Pignatti C, De Stefano P, Barone F, Concia E. Penicillin compliance in splenectomized thalassemics. European Journal of Pediatrics. 1984;142:83-85. PMID: 6468437
  73. 73. Fucharoen S, Piankijagum A, Wasi P. Deaths in beta-thalassemia/Hb E patients secondary to infections. Birth Defects Original Article Series. 1987;23:495-500. PMID: 3689937
  74. 74. Aswapokee P, Aswapokee N, Fucharoen S, Sukroongreung S, Wasi P. Severe infection in thalassemia: A prospective study. Birth Defects Original Article Series. 1987;23:521-526. PMID: 3689941
  75. 75. Wasi C, Kuntang R, Louisirirotchanakul S, Siritantikorn S, Fucharoen S, Aswapokee P, et al. Viral infections in beta-thalassemia/hemoglobin E patients. Birth Defects Original Article Series. 1987;23:547-555. PMID: 2825835
  76. 76. Swarup-Mitra S. Immunologic status of Hb E-thalassemia patients. Birth Defects. 1988;23:571-579. PMID: 3689944
  77. 77. Swarup-Mitra S. Morbidity pattern in Hb E-thalassemia disease. Birth Defects Original Article Series. 1988;23:501-504. PMID: 3689938
  78. 78. Sukroongreung S, Nilakul C, Aswapokee N, Aswapokee P, Fucharoen S, Wasi P. Oropharyngeal colonization with aerobic bacteria in β-thalassemia/hemoglobin E disease. Birth Defects Original Article Series. 1988;23:535-541. PMID: 3318952
  79. 79. Sukroongreung S, Wasi P. Serum of thalassemic patients promotes growth of Streptococci. Birth Defects Original Article Series. 1988;23:543-546. PMID: 3689943
  80. 80. Ampel NM, Van Wyck DB, Aguirre ML, Willis DG, Popp RA. Resistance to infection in murine beta-thalassemia. Infection and Immunity. 1989;57:1011-1017. PMCID: PMC313221
  81. 81. Ampel NM, Bejarano GC, Saavedra M. Deferoxamine increases the susceptibility of beta-thalassemic, iron-overloaded mice to infection with Listeria monocytogenes. Life Sciences. 1992;50:1327-1332. DOI: 10.1016/0024-3205(92)90283-u
  82. 82. Cherchi GB, Cossellu S, Pacifico L, Gallisai D, Ranucci A, Zanetti S, et al. Incidence and outcome of Yersinia enterocolitica infection in thalassemic patients. Contributions to Microbiology and Immunology. 1995;13:16-18. PMID: 8833786
  83. 83. Wanachiwanawin W, Phucharoen J, Pattanapanyasat K, Fucharoen S, Webster HK. Lymphocytes in beta-thalassemia/HbE: Subpopulations and mitogen responses. European Journal of Haematology. 1996;56:153-157. PMID: 8598234
  84. 84. Vento S, Cainelli F, Cesario F. Infections and thalassaemia. The Lancet Infectious Diseases. 2006;6:226-233. DOI: 10.1016/S1473-3099(06)70437-6
  85. 85. Rahav G, Volach V, Shapiro M, Rund D, Rachmilewitz EA, Goldfarb A. Severe infections in thalassaemic patients: Prevalence and predisposing factors. British Journal of Haematology. 2006;133:667-674. DOI: 10.1111/j.1365-2141.2006.06082.x
  86. 86. Yapp AR, Lindeman R, Gilroy N, Gao Z, Macintyre CR. Infection outcomes in splenectomized patients with hemoglobinopathies in Australia. International Journal of Infectious Diseases. 2009;13:696-700. DOI: 10.1016/j.ijid.2008.10.011
  87. 87. Kao JK, Wang SC, Ho LW, Huang SW, Chang SH, Yang RC, et al. Chronic Iron overload results in impaired bacterial killing of THP-1 derived macrophage through the inhibition of lysosomal acidification. PLoS One. 2016;11:e0156713. DOI: 10.1371/journal.pone.0156713
  88. 88. Loegering DJ, Grover GJ, Schneidkraut MJ. Effect of red blood cells and red blood cell ghosts on reticuloendothelial system function. Experimental and Molecular Pathology. 1984;41:67-73. DOI: 10.1016/0014-4800(84)90008-X
  89. 89. Loegering DJ, Blumenstock FA. Depressing hepatic macrophage complement receptor function causes increased susceptibility to endotoxemia and infection. Infection and Immunity. 1985;47:659-664. PMCID: PMC261348
  90. 90. Commins LM, Loegering DJ, Gudewicz PW. Effect of phagocytosis of erythrocytes and erythrocyte ghosts on macrophage phagocytic function and hydrogen peroxide production. Inflammation. 1990;14:705-716. DOI: 10.1007/BF00916373
  91. 91. Loegering DJ, Schwacha MG. Macrophage hydrogen peroxide production and phagocytic function and decreased following phagocytosis mediated by Fc receptors but not complement receptors. Biochemical and Biophysical Research Communications. 1991;180:268-272. DOI: 10.1016/S0006-291X(05)81287-2
  92. 92. Schwacha MG, Gudewicz PW, Snyder JA, Loegering DJ. Depression of macrophage respiratory burst capacity and arachidonic acid release after Fc receptor-mediated phagocytosis. Journal of Immunology. 1993;150:236-245. PMID: 8417125
  93. 93. Loegering DJ, Raley MJ, Reho TA, Eaton JW. Macrophage dysfunction following the phagocytosis of IgG coated erythrocytes: Production of lipid peroxidation products. Journal of Leukocyte Biology. 1996;59:357-363. DOI: 10.1002/jlb.59.3.357
  94. 94. de Sousa M. Blood transfusions and allograft survival: Iron-related immunosuppression [Letter]. Lancet. 1983;2(8351):681-682. DOI: 10.1016/s0140-6736(83)92558-8

Written By

Mark D. Scott

Submitted: 13 June 2019 Reviewed: 28 October 2019 Published: 25 November 2019