Severe malaria is manifest by a variety of clinical syndromes dependent on properties of both the host and the parasite. In young infants, severe malarial anemia (SMA) is the most common syndrome of severe disease and contributes substantially to the considerable mortality and morbidity from malaria. There is now growing evidence, from both human and mouse studies of malaria, to show that anemia is due not only to increased hemolysis of infected and clearance of uninfected red blood cells (RBCs) but also to an inability of the infected host to produce an adequate erythroid response. In this review, we will summarize the recent clinical and experimental studies of malaria to highlight similarities and differences in human and mouse pathology that result in anemia and so inform the use of mouse models in the study of severe malarial anemia in humans.

The vast majority of morbidity and mortality from malaria is caused by infection with Plasmodium falciparum, although P vivax, P ovale, and P malariae also are responsible for human infections. The total burden of disease has recently been estimated to 515 million episodes annually and malaria is responsible for 18% of all childhood deaths in sub-Saharan Africa, equivalent to 800 000 deaths each year.1,2 

The study of malarial anemia has somewhat belatedly excited academic and professional interest. Severe malarial anemia (SMA) certainly merits concern as a major public health problem because of the very large numbers of children affected, and it is likely that these numbers may increase as drug resistance spreads. Concerns have also been raised by data from recent vaccine studies, which suggest that monkeys immunized with erythrocytic-stage antigens, and that have acquired protection from acute infection, may succumb to severe anemia during a subacute or chronic phase of infection.3,4  Moreover, there is increasing awareness of the difficulty of satisfactory treatment by blood transfusion outside specialist centers in many endemic areas as a result of the limited availability of a rapid and safe supply of blood.1,5 

SMA is seen most frequently in areas of very high malaria transmission and most commonly in young children and pregnant women.6  The prevalence of anemia, defined as a hematocrit (Hct) level higher than 0.33, in malaria-endemic areas of Africa, varies between 31% and 91% in children, and between 60% and 80% in pregnant women.7,8  Given the high degree of morbidity that is associated with SMA in children, this term will be used to refer to severe anemia in this group, unless stated otherwise.

It is very difficult to determine the number of cases of severe anemia attributable to malaria as the WHO definition of SMA (a hemoglobin [Hb] concentration of < 50 g/L [5 g/dL], or a Hct < 0.15, in the presence of a parasitemia > 10 000 parasites per microliter [μL], and a normocytic blood film) may exclude the considerable proportion of children admitted with severe anemia that has a blood smear negative for malaria parasites but that responds to antimalarial treatment.9,10  It may be difficult to attribute anemia to a single cause since the background to malarial anemia is often complex in endemic areas and hematinic deficiencies, genetic traits, and intercurrent infection all may contribute to anemia (see Roberts et al11  for review). Nevertheless, a randomized placebo-controlled trial of malaria chemoprophylaxis and iron supplementation in infants, from an endemic area, has shown that malaria infection was the main etiologic factor underlying anemia.9,12 

The pathology of malaria is associated with the blood stage of infection (for overview of life cycle see Figure 1).13 P falciparum infections have high multiplication rates while also expressing clonally variant antigens at the surface of infected erythrocytes (Pf-EMP-1). Pf-EMP-1 binds to ligands on the surface of endothelial cells and mediates sequestration of infected erythrocytes in postcapillary venules. Both of these characteristics allow the P falciparum parasite to evade the host immune system, which results in the occurrence of high parasitemias with repeated infections that contribute to the chronic nature of this disease.14  In P vivax and P ovale malaria, high parasitemias are rare as invasion of erythrocytes is limited to reticulocytes. However, P vivax can, on occasion, cause severe disease including anemia with severe hemolysis.15,17 

Figure 1

Plasmodia life cycle. (A) The asexual life cycle begins when sporozoites from a female mosquito taking a blood meal enter the circulation and invade hepatocytes. (B) Up to 10 000 merozoites are formed. Following rupture of the hepatocyte, infective merozoites are released and invade erythrocytes (RBCs). (C) Within RBCs, the parasite develops through the stages of rings, trophozoites, and schizonts. Mature schizonts burst to release erythrocytic merozoites that invade new RBCs. (D) A small proportion of merozoites in RBCs transform into male and female gametocytes that are ingested by the mosquito. (E) The male and female gametes fuse and transform into an öocyst that divides asexually into many sporozoites that migrate to the salivary glands from where they are released during the next blood meal. Reproduced from Ocana-Morgher et al13  with permission.

Figure 1

Plasmodia life cycle. (A) The asexual life cycle begins when sporozoites from a female mosquito taking a blood meal enter the circulation and invade hepatocytes. (B) Up to 10 000 merozoites are formed. Following rupture of the hepatocyte, infective merozoites are released and invade erythrocytes (RBCs). (C) Within RBCs, the parasite develops through the stages of rings, trophozoites, and schizonts. Mature schizonts burst to release erythrocytic merozoites that invade new RBCs. (D) A small proportion of merozoites in RBCs transform into male and female gametocytes that are ingested by the mosquito. (E) The male and female gametes fuse and transform into an öocyst that divides asexually into many sporozoites that migrate to the salivary glands from where they are released during the next blood meal. Reproduced from Ocana-Morgher et al13  with permission.

Close modal

The spectrum of the clinical presentation and severity of P falciparum infection is broad. In endemic areas, many infections in semi-immune and immune children and adults present as uncomplicated febrile illness. In more severe disease, nonimmune individuals may exhibit a number of syndrome(s) including anemia, coma, respiratory distress, and hypoglycemia, and have a high frequency of concurrent bacteremia.18,20  Many children present with mild, moderate, and even severe anemia without other syndromes of severe disease. However, severe anemia may be accompanied by other syndromes of severe disease.18  For example, children with anemia may also present with malaise, fatigue, dyspnoea, or respiratory distress as metabolic acidosis supervenes18,21,22  (for review see English et al23 ). The age distribution of the syndromes of severe disease is striking, but poorly understood. Children born in endemic areas are largely protected from severe malaria in the first 6 months of life by the passive transfer of maternal immunoglobulins and by fetal hemoglobin. The presentation of disease changes from severe anemia in children aged between 1 and 3 years in areas of high transmission to cerebral malaria in older children in areas of lower transmission.24  As transmission intensity declines, severe malaria is most frequently found in older age groups.

The anemia of P falciparum malaria is typically normocytic and normochromic, with a notable absence of reticulocytes, although microcytosis and hypochromia may be present due to the very high frequency of alpha and beta thalassemia traits and/or iron deficiency in many endemic areas25,26  (for a general review of the hematologic features of malaria infection see Abdalla et al27  and Roberts et al28 ). The exact differences in the pathophysiology of anemia in the various clinical settings, ages, and geographic areas are poorly defined and surely represent a fruitful field for further study. A less common form of anemia in malaria is “blackwater fever” characterized by the sudden appearance of hemoglobin in the urine associated with irregular use of quinine.29,30 

The clinical setting of severe anemia is therefore varied and complex: acute infection may lead to anemia and/or cerebral malaria, respiratory distress, and hypoglycemia; and chronic, repeated infection may lead to severe anemia. In addition, there may be a background of normal or low Hb. As such, any understanding of the pathophysiological processes has ultimately to be linked to the diverse clinical contexts.

In searching for tractable model systems, we are fortunate that laboratory mice and rats can be infected with natural species of rodent malaria. Strictly speaking the rodent host-parasite systems are not “models” (“representation on a smaller scale” or “an example”) since the progression of disease may be quite different than that seen in P falciparum malaria. A detailed comparative description of the features of the rodent “models” of malaria is therefore a prerequisite to improving the use of these systems in understanding severe anemia in human disease.

The scope of mouse models of malaria is enhanced by the availability of differentially susceptible inbred mouse strains in which infections can be lethal or nonlethal. The course of infection and immunologic response of infection is variable but specific for each host-parasite combination, and always results in anemia (Table 1; and see Lamb et al31  for review). There are several strains of Plasmodia that cause infection in mice, such as P chabaudi, P yoelii, P berghei, and P vinckei. As for P falciparum infections, P chabaudi invades RBCs of all ages,32,34  while P yoelii has a preference for reticulocytes35  and therefore may serve better as a model for P vivax. However, the majority of these parasite-mouse models demonstrate an acute malaria infection with parasitemias often exceeding 20%, which is in contrast to severe anemia in humans where acute malaria frequently occurs with a lower parasitemia (Table 2).

In attempting to develop an in vivo model of SMA, it may not be necessary to demonstrate all features of severe falciparum anemia but to represent a component of this syndrome that may help to identify factors that contribute to its severity. These should then be considered in the context of concurrent infections that may exacerbate the anemia.36  The examples of possible causes of SMA given in this article will be from clinical studies in which participants with coinfections, hemoglobinopathies, or folate or G6PD deficiencies were excluded from the study.

The underlying causes of SMA in humans may include one or more of the following mechanisms: (1) the clearance and/or destruction of infected RBCs, (2) the clearance of uninfected RBCs, (3) erythropoietic suppression and dyserythropoiesis. Each of these mechanisms has been implicated in both human and mouse malarial anemias (Table 3), but their relative contribution between the species is likely to differ. In the remainder of this review, the pathophysiological mechanisms of human and mouse malarial anemia will be discussed, highlighting similarities and differences.

Loss of infected erythrocytes

During infection there is obvious loss of infected erythrocytes through parasite maturation as well as through recognition by macrophages. The pathways of phagocytic clearance for both humans and mice are summarized in Table 3 (see Casals-Pascual and Roberts37  for further discussion).

It is clear that similar mechanisms exist for the clearance of infected erythrocytes in humans and mice. However, the removal of infected erythrocytes in humans with parasitemias of less than 1% is unlikely to have a significant impact on the degree of anemia. This removal, therefore, may prove more pertinent for the onset of anemia in individuals suffering from an acute infection, in particular children where parasitemias are frequently greater than 10%. Since malaria infections in mice often attain high parasitemias with intravascular hemolysis of infected erythrocytes,38  they may provide suitable models to study the factors that contribute to the pathology of anemia during acute falciparum infection.

Loss of uninfected erythrocytes

During human malaria infection, many uninfected red cells are destroyed, in the spleen and quite possibly the liver, and their destruction has been identified as the major contributor to malarial anemia.39,41  Both mathematical modeling and clinical observations suggest that 10 times as many uninfected RBCs are removed from the circulation for each infected erythrocyte.40  Although few direct measurements of red cell survival have been made in human infections, reduced half-life of normal erythrocyte and increased clearance of heat-treated erythrocytes have been demonstrated in patients with malaria, consistent with these observations.39,42 

The activity and the number of macrophages are also increased during human malarial infection, and may therefore contribute to the increased removal of uninfected cells.43,,46  The increased clearance of uninfected erythrocytes is due not only to the activation of splenic macrophages but also to extrinsic and intrinsic changes to the red blood cells that enhance their recognition and phagocytosis. First, uninfected RBCs have a reduced deformability leading to enhanced clearance in the spleen. The mechanism responsible for the loss of deformability is not completely understood. Increased oxidation of membranes in uninfected erythrocytes has been shown in children with severe P falciparum malaria, and the ongoing inflammatory insults associated with acute malaria (proinflammatory cytokines), or direct effect of parasite products have been shown to cause loss of RBC deformability.44,47,48  Intriguingly, a severe reduction in red cell deformability is also a strong predictor for mortality measured on admission, both in adults and children with severe malaria.49,50  Second, the deposition of immunoglobulin and complement on uninfected RBCs may enhance receptor-mediated uptake by macrophages (Table 3).

Parasite products that may be part of the immunoglobulin-antigen complexes deposited on uninfected RBCs include the P falciparum ring surface protein 2 (RSP-2). This protein, expressed shortly after merozoite invasion of RBCs, mediates adhesion of iRBCs to endothelial cells.51,52  RSP-2 is also deposited on uninfected RBCs and the opsonization of these RSP-2–bearing uninfected RBCs provides a mechanism of removing uninfected RBCs. Indeed high levels of these antibodies that facilitate complement-mediated phagocytosis of cells expressing RSP-2 are found in sera from immune adults and children with severe anemia.53  This antigen is also present on the surface of erythroblasts in bone marrows of P falciparum–infected patients, indicating that clearance or damage to circulating or developing erythroid cells by RSP-2 and anti–RSP-2 could contribute to the development of SMA.

Due to the high level of parasitemia, it may appear that the clearance of uninfected RBCs for the development of anemia is less significant in mice than in the majority of human malaria infections. However, in P chabaudi and P berghei infections the degree of anemia is not always related to peak parasitemia,54,55  and premature removal of uninfected RBCs has been shown in P yoelii infection. Interestingly, immunoglobulin-mediated removal of erythrocytes in this infection was not implicated, but rather it was thought that abnormalities in the erythrocytes themselves was responsible.56  It remains to be determined whether removal of RBCs via immune complex deposition takes place in mouse infection, and the parasite product RSP-2 has not yet been identified in rodent parasites. So far it seems that rodent models in this respect may be representative of acute human infections only where removal of uninfected RBCs is independent of opsonization. However, this is clearly an area where further research is needed. More recently, a model of chronic anemia has been developed where P berghei ANKA–infected semi-immune rodents show that, although parasitemia remains less than 1%, there is a 5- to 10-fold increase in the clearance of uninfected RBCs.57  In this study, clearance of uninfected RBCs was delayed in mice depleted of macrophages supporting the view that removal of uninfected RBCs is an important factor in anemia. Moreover, macrophage activation was dependent on CD4+ T cells. This model could therefore represent one feature of chronic human infection where removal of uninfected erythrocytes contributes to the severity of anemia.

Erythropoietic suppression and dyserythropoiesis

Normal erythropoiesis (as described in Figure 2) is perturbed during malaria infection. The earliest observations of reduced erythropoiesis in acute human malaria were made more than 60 years ago where reticulocytopenia was observed in P vivax and P falciparum malaria infection followed by reticulocytosis after parasite clearance.58  Later, it was demonstrated that low reticulocyte counts in Thai patients with malaria was accompanied by suppression of erythropoiesis (reviewed in Casals-Pascual and Roberts37 ).

Figure 2

Human and murine erythropoiesis. At birth, erythropoiesis occurs throughout the human skeleton, although over time hematopoietic activity is confined to the sternum and pelvic region of most adults. Likewise, in adult mice the bone marrow is the major organ, with the majority of hematopoietically active cells typically present in the femur and tibia, though this is dependent on the age and strain of the mouse. Erythropoiesis from the multipotent hematopoietic stem cell (HSC) through to the mature red blood cell is shown to allow comparison of human and mouse cell surface phenotypes. The time at which these markers are expressed and lost at the cell surface is very similar between species indicating that as a first approximation the mechanisms used to control cell expansion, differentiation, and removal of precursors may not differ greatly. Developing erythroid cells respond to signals from stromal cells of the bone marrow or spleen. Erythroblasts are organized into erythroblastic islands that consist of macrophages surrounded by developing erythroid precursors.144,145  Macrophages provide many of the cellular mediators that control erythropoietic activity: GM-CSF, IL-3, and stem cell factor (SCF) generate colony-forming unit erythroid macrophage-granulocyte megakaryocyte (CFU-GEMM) and burst-forming unit erythroid (BFU-E), whereas TGF-β, TNF-α, and MIP1-α inhibit cell cycle activity146  and BFU-E development. Other factors that negatively regulate numbers include proapoptotic activity initiated by interaction of the Fas receptor (CD95) with Fas ligand (CD95L).147,148  Expression of caspases also results in cleavage of GATA-1 and loss of its antiapoptotic activity.148  Secretion of Epo induces the expansion of colony-forming unit erythroid (CFU-E) and initiates differentiation through a number of erythroid-specific events. SCF and Epo synergize to drive the proliferation of human erythroid progenitors and precursors150,151  and induce anti-apoptotic activity.152,153  In human and mouse erythropoiesis, levels of the transferrin receptor (CD71) peak when the highest transport of transferrin-bound iron is required for synthesis of heme from protoporphyrin. During erythroid precursor maturation an increase in expression of glycophorin A (gpA) in humans or Ter119 in mice is coupled with a drop in CD71 expressed at the cell surface. This loss of CD71 indicates reduced proliferation and heme synthesis with continued differentiation into reticulocytes.

Figure 2

Human and murine erythropoiesis. At birth, erythropoiesis occurs throughout the human skeleton, although over time hematopoietic activity is confined to the sternum and pelvic region of most adults. Likewise, in adult mice the bone marrow is the major organ, with the majority of hematopoietically active cells typically present in the femur and tibia, though this is dependent on the age and strain of the mouse. Erythropoiesis from the multipotent hematopoietic stem cell (HSC) through to the mature red blood cell is shown to allow comparison of human and mouse cell surface phenotypes. The time at which these markers are expressed and lost at the cell surface is very similar between species indicating that as a first approximation the mechanisms used to control cell expansion, differentiation, and removal of precursors may not differ greatly. Developing erythroid cells respond to signals from stromal cells of the bone marrow or spleen. Erythroblasts are organized into erythroblastic islands that consist of macrophages surrounded by developing erythroid precursors.144,145  Macrophages provide many of the cellular mediators that control erythropoietic activity: GM-CSF, IL-3, and stem cell factor (SCF) generate colony-forming unit erythroid macrophage-granulocyte megakaryocyte (CFU-GEMM) and burst-forming unit erythroid (BFU-E), whereas TGF-β, TNF-α, and MIP1-α inhibit cell cycle activity146  and BFU-E development. Other factors that negatively regulate numbers include proapoptotic activity initiated by interaction of the Fas receptor (CD95) with Fas ligand (CD95L).147,148  Expression of caspases also results in cleavage of GATA-1 and loss of its antiapoptotic activity.148  Secretion of Epo induces the expansion of colony-forming unit erythroid (CFU-E) and initiates differentiation through a number of erythroid-specific events. SCF and Epo synergize to drive the proliferation of human erythroid progenitors and precursors150,151  and induce anti-apoptotic activity.152,153  In human and mouse erythropoiesis, levels of the transferrin receptor (CD71) peak when the highest transport of transferrin-bound iron is required for synthesis of heme from protoporphyrin. During erythroid precursor maturation an increase in expression of glycophorin A (gpA) in humans or Ter119 in mice is coupled with a drop in CD71 expressed at the cell surface. This loss of CD71 indicates reduced proliferation and heme synthesis with continued differentiation into reticulocytes.

Close modal

Bone marrow aspirates taken from Gambian children with acute anemia revealed that despite an increase in cellularity no significant difference in the total number of erythroblasts was observed when compared with uninfected patients, providing evidence for a suppressed erythroid response. Children who presented with chronic anemia (parasitemia < 1%) had higher levels of erythroid hyperplasia and dyserythropoiesis59,60  Dyserythropoiesis or morphologically and/or functionally abnormal production of RBCs was demonstrated by cytoplasmic vacuolation, stippling, fragmentation, intercytoplasmic bridges, nuclear fragmentation, and multinuclearity. This coincided with reduced reticulocytosis indicating functional disruption of RBC output from the bone marrow59,60  (Figure 3). In a smaller study of 6 children with chronic disease, an increased proportion of polychromatic erythroblasts in the G2 phase of division was observed.61  Treatment of these patients with antimalarial drugs increased reticulocyte numbers, which pointed to P falciparum as the cause of dyserythropoiesis and ineffective erythropoiesis.

Figure 3

Direct and indirect effects of parasite on the development of malarial anemia. Severe malarial anemia is characterized by destruction of infected red blood cell (iRBC) following schizogony and clearance of both iRBCs and uninfected RBCs. During malarial infection, changes in membrane protein composition occur and the resultant immune complexes of RBCs, Ag, and immunoglobulin (Ig) (eg, RBC:RSP2:Ig) are cleared by macrophages to the spleen where they become activated (see Table 3 for more examples). Pigment-containing macrophages may release inflammatory cytokines and other biologically active mediators such as hydroxy-nonenal (HNE). It is possible that malarial pigment or other parasite products may have a direct inhibitory effect on erythropoiesis. Inhibition of erythropoiesis may be at one or more sites in the growth and differentiation of hematopoietic progenitors. Both indirect and direct effects may cause suppression of the bone marrow and spleen resulting in inadequate reticulocyte counts for the degree of anemia. The mechanisms of insufficient erythropoiesis in murine malaria have been summarized in Chang and Stevenson.154  Blue box indicates demonstrated in human infection; pink box, demonstrated in mouse infection; yellow box, demonstrated in both human and mouse infections. Hz indicates hemozoin; GPI, glycophosphatidylinositol anchors of merozoite proteins; Epo, erythropoietin; Epo-R, erythropoietin receptor; MΦ, macrophage; RSP-2, ring surface protein-2; and Ig, immunoglobulin. Figure modified with permission from Springer Science Business Media, Heidelberg, Germany.

Figure 3

Direct and indirect effects of parasite on the development of malarial anemia. Severe malarial anemia is characterized by destruction of infected red blood cell (iRBC) following schizogony and clearance of both iRBCs and uninfected RBCs. During malarial infection, changes in membrane protein composition occur and the resultant immune complexes of RBCs, Ag, and immunoglobulin (Ig) (eg, RBC:RSP2:Ig) are cleared by macrophages to the spleen where they become activated (see Table 3 for more examples). Pigment-containing macrophages may release inflammatory cytokines and other biologically active mediators such as hydroxy-nonenal (HNE). It is possible that malarial pigment or other parasite products may have a direct inhibitory effect on erythropoiesis. Inhibition of erythropoiesis may be at one or more sites in the growth and differentiation of hematopoietic progenitors. Both indirect and direct effects may cause suppression of the bone marrow and spleen resulting in inadequate reticulocyte counts for the degree of anemia. The mechanisms of insufficient erythropoiesis in murine malaria have been summarized in Chang and Stevenson.154  Blue box indicates demonstrated in human infection; pink box, demonstrated in mouse infection; yellow box, demonstrated in both human and mouse infections. Hz indicates hemozoin; GPI, glycophosphatidylinositol anchors of merozoite proteins; Epo, erythropoietin; Epo-R, erythropoietin receptor; MΦ, macrophage; RSP-2, ring surface protein-2; and Ig, immunoglobulin. Figure modified with permission from Springer Science Business Media, Heidelberg, Germany.

Close modal

In the mouse, both lethal and nonlethal malaria infections induce ineffective erythropoiesis, with alterations in erythropoietic progenitor and precursor populations, as well as in the sites of erythropoiesis (Figure 3). In contrast to the human studies cited, a decrease in mouse bone marrow cellularity of between 40% to 75% of normal levels is seen at peak parasitemia or death, whichever is reached first,62,,65  with reductions in cellularity proportional to the severity of infection.54,63,66  However, despite these discrepancies with the reported pathology in human infection, reports of only a minimal reduction in the total number of BFU-Es, and at best only a modest increase in CFU-Es in mice,63,,66  suggest that the bone marrow is unable to compensate for RBC loss in both humans and mice through increased erythropoiesis during the acute phase of infection. At present there are no studies reporting the production of dyserythropoietic erythroblasts during mouse malaria infection, most probably because there are few chronic infection models. Dyserythropoiesis has been observed only in chronic human infections, thus future studies on the production of abnormal erythroblasts in the newly described chronic model of P berghei57  may help elucidate the mechanism by which this occurs.

A parasite by-product of hemoglobin digestion, hemozoin, may have a role in the impaired erythroid development through its effects on human monocyte function. Hemozoin reduces human macrophage oxidative burst activity, prevents up-regulation of activation markers,67,68  and also stimulates the secretion of biologically active endoperoxides from monocytes, such as 15(S)-hydroxyeicosatetraenoic (HETE) and 4-hydroxy-nonenal (4-HNE) through oxidation of membrane lipids,69,70  which may effect erythroid growth.71  Macrophage dysfunction could also disrupt the function of erythroblastic islands in which macrophages support terminal differentiation of erythroblasts in the bone marrow.

Hemozoin and TNF-α also have additive effects on erythropoiesis in vitro, and in a clinical study hemozoin-containing macrophages and plasma hemozoin were associated with anemia and reticulocyte suppression.72  Moreover, bone marrow sections from children who died from severe malaria show a significant association between the quantity of hemozoin (located in erythroid precursors and macrophages) and the proportion of erythroid cells that was abnormal. These findings are consistent with a direct inhibitory effect of hemozoin on erythropoiesis and therefore warrant further investigation. Disappointingly, however, there has been little focus on the role of hemozoin on erythropoiesis during mouse malaria infection, where effects of hemozoin-induced suppression of erythropoiesis could be dissected in an in vivo setting. Clearly hemozoin present in bone marrow may have a role in mice, as the efficient reticulocyte response is not observed until there is parasite clearance.55,66 

Cytokine suppression of erythropoiesis.

During the acute phase of both human and mouse infections there is a strong inflammatory response, which results in increases in TNF-α and IFN-γ.73,75  TNF-α inhibits all stages of erythropoiesis,76  and IFN-γ works with TNFα to inhibit erythroid growth and differentiation by up-regulating expression of TRAIL, TWEAK, and CD95L in developing erythroblasts.77  However, in a nonlethal P chabaudi (AS) infection of C57BL/6 mice, neutralization of TNF-α or IFN-γ has no effect on in vitro erythropoiesis.74  While severe disease in children is associated with elevated levels of proinflammatory and anti-inflammatory cytokines, the severity of anemia seems to be dependent on levels of TNF-α relative to its regulator, the potent anti-inflammatory cytokine IL-10. Several clinical studies have demonstrated that a low ratio of plasma IL-10/TNF-α is associated with SMA in young children.78,79  Furthermore, a number of polymorphisms in the human TNF-α promoter show greater association with anemia than with cerebral malaria.80  The IL-10/TNF-α ratio is also important in mice where IL-10 knock-out mice infected with P chabaudi display increased anemia,81  which is reversed following TNF-α neutralization in vivo.82  It is therefore conceivable that in both humans and mice, IL-10 may protect against bone marrow suppression and erythrophagocytic activity induced by TNF-α and/or mitigate other proinflammatory stimuli.

Many other proinflammatory cytokines such as IL-12, IL-18, and migration inhibitory factor (MIF) have also been implicated in the pathogenesis of anemia in malaria. In humans, the secretion of IL-12 and IL-18 from macrophages induces production of IFN-γ from natural killer (NK), B cells, and T cells,83  while MIF is produced by activated T cells and macrophages and inhibits the anti-inflammatory activity of glucocorticoids (for review see Clark and Cowden84 ).

IL-12 is present at higher levels in nonlethal, compared with lethal, infections of P chabaudi, suggesting that this cytokine may be a stimulator of erythropoiesis.85,86  Conversely, with elevated levels found during infection, MIF has been shown to suppress hematopoiesis,87  and more recently P chabaudi infection of MIF knock-out mice resulted in a less severe anemia with improved development of erythroid progenitors in the bone marrow.88  In this latter study, a hyperparasitemic model of malaria (P chabaudi AS in Balb/c) was used. However the levels of parasitemia, TNF-α, and IFN-γ in wild-type and knock-out mice were equally raised, therefore indicating a direct effect of MIF on erythroid development during the acute phase of disease.

The association of IL-12 with severe falciparum malaria is less clear. While some studies observe moderate increases in IL-12 and IL-18 in patients with severe anemia,83,89  others report decreases in IL-12 in patients with severe malaria (Hb of < 75 g/L [7.5 g/dL]) compared with uncomplicated controls (Hb of > 100 g/L [10 g/dL]), or no significant increases in patients with severe disease compared with uncomplicated malaria.90,91  In these last 2 examples, anti-inflammatory cytokines such as TGF-β or IL-10 were also reduced in patients with severe disease. In contrast, patients with acute disease and elevated levels of IL-12 had marked increases in IL-10.83  Since the majority of patients with anemia in this last study had average Hb levels of 90 g/L (9 g/dL) it is possible that, as with mice, increases in IL-12 are associated with reduced severity of SMA. The data on serum levels of MIF in patients with malaria are, however, consistent with its role as a hematopoietic suppressor in mice: MIF concentrations in those with moderate anemia are decreased,89  and MIF is elevated in patients with more severe anemia.90 

These observations, in both human and mouse infections, show the complexity of cytokine responses, and also highlight the importance of a balance between proinflammatory and anti-inflammatory cytokines, which can either be protective or detrimental to the host. Understanding the role of these cytokines will require more data from adequately powered studies to enable use of more sophisticated multivariate analyses that may allow for intricate interactions between each factor. In addition, significant similarities do indeed exist between humans and mice, and the ready availability of gene knock-out mouse models, for example, will allow for more in-depth analysis of proinflammatory and anti-inflammatory mechanisms without the confusion of human genetic variability.

A parasite product found in plasma during human and mouse infections that may be implicated in the proinflammatory cytokine effects on SMA is the glycophosphatidylinositol (GPI) anchor of the merozoite proteins, MSP-1, MSP-2, and MSP-4.92  GPIs are likely to contribute to malarial anemia since they can induce the release of TNF-α from human macrophages,93  which could contribute to the pathology of SMA. More specifically, it has recently been demonstrated that the proinflammatory response from human monocytes is through interaction of GPIs with TLR2, and to a lesser extent TLR4.94  The role of GPI-induced anemia in mice has not been studied directly. The injection of crude infected RBC lysate into mice does result in a transient decrease in the number of circulating RBCs,95  which is probably through the induction of host inflammatory responses,96  however this could be the effect of a range of other parasite products as well as GPI. Purified GPI immunization of mice in one study did result in reduced cerebral pathology and fatality, however effects on SMA were not reported.97  Further analysis of GPI-induced effects in mouse models of malaria to investigate a similar mechanism in humans would be of both academic and therapeutic interest because antibodies specific to GPIs are found in sera of adults from endemic regions in Kenya, but at reduced levels in children who, in general, have more severe disease and malarial anemia.98 

A product discussed earlier, hemozoin, may also be more intimately linked to an innate immune response, and thus proinflammatory cytokine release, than previously thought. In humans, synthetic pigment induces expression of TNF-α, which has been linked to the ability of hemozoin to induce the metalloproteinase MMP-9,99,101  and relatively recent studies in mice have suggested that hemozoin stimulates an innate proinflammatory response102  via a MyD88-dependent TLR9 pathway.103 

Erythropoietin.

A fall in Hb and subsequent reduction in oxygen tension should stimulate elevated levels of erythropoietin (Epo) in patients with SMA. The clinical evidence for appropriately raised levels of Epo in malaria is somewhat contradictory. Studies in adults from Thailand and Sudan have suggested that the Epo concentration, although raised, was inappropriate for the degree of anemia.104,105  However, several studies of malaria in African children suffering from malarial anemia have shown appropriately raised Epo concentrations.25,106,108  In fact, the Epo levels in malarial anemia are more than 3-fold higher when compared with anemic children without malaria.72  It is possible that ineffective or inadequate Epo synthesis does contribute to malarial anemia in some settings, possibly related to age, ethnic origin, or presentation of the patient. However, in African children with malaria, Epo synthesis is indeed elevated more than expected and it is more likely that a reduced response to Epo, not an inappropriately low level of Epo, is the more significant contribution to pathology.

Similarly, in mice, P chabaudi AS infection of resistant C57BL/6 and susceptible A/J strains results in anemia despite increases in serum Epo.109  However, the increase in Epo, albeit driving a somewhat blunted reticulocyte response, is a crucial determinant of the outcome of infection. Neutralization of Epo transforms P chabaudi AS infection in normally resistant C57BL/6 mice into a fatal infection. Intriguingly, induction of reticulocytosis by exogenous Epo prior to infection enhanced parasite multiplication and resulted in lethal infection. However, timely induction of reticulocytosis shortly after infection could alleviate malarial anemia and increase survival of A/J (susceptible) mice.55 

Thus, in both human and mouse malaria infections, increased serum Epo appears essential for recovery, despite the possibility of an attenuated bone marrow response to this growth factor. Clearly, more extensive human studies are required to investigate the kinetics of erythrocyte production in response to elevated Epo during infection. However, a detailed phenotypic analysis of the abnormalities in erythropoiesis during the rodent P chabaudi infection may provide an insight to this mechanism, which has revealed that not only is Epo-induced proliferation of early erythroblasts suppressed, but also terminal differentiation and maturation of TER119+ erythroblasts is impaired.110  Perhaps surprisingly, these abnormalities of erythropoiesis were accompanied by neither an increase in apoptosis nor dysregulation of the cell cycle, suggesting that serum Epo is increased appropriately but that a reduced response to Epo is responsible for an insufficient erythrocyte output.

Hematinic deficiency.

Although dietary deficiencies are widespread in malaria-endemic regions, the influence of reduced folate and iron levels is not thought to be a major contributor to dyserythropoiesis seen during SMA.60  However dysregulation of iron metabolism may contribute to the severity of disease in children that present with SMA. The peptide hormone hepcidin has been implicated in mediating anemia of chronic disease or inflammation by reducing the availability of iron stores for erythropoiesis.111  Hepcidin is regulated by proinflammatory mediators such as IL-6, which is elevated in both murine infections and in patients who present with severe falciparum malaria.38,91  To date, there are no reports that describe the association of elevated hepcidin with SMA.

Extramedullary hematopoiesis.

One substantial difference in human and mouse responses to malarial anemia is that in mice the marked decrease in bone marrow cellularity appears to be compensated by increased erythropoietic activity in the spleen (Figure 3). However, to date there are no studies that have investigated extramedullary erythropoiesis during P falciparum infection. In mice, massive splenomegaly is observed with cellularity increasing 20-fold at peak parasitemia compared with that of naive animals.64,81  This rise is reflected in greater erythroid progenitor populations with up to an 8-fold increase of BFU-E and 100-fold increase of CFU-E total numbers in the spleen.65,66  Interestingly, there is a strong correlation between the severity of disease and the observed increases in splenic erythropoiesis. Lower rises in BFU-E and CFU-E populations are seen in fatal infections, when compared with nonfatal infections, which strongly suggests that a marked splenic erythropoietic response is required for survival in mice.54,112  It is unknown whether this is relevant to human studies because although splenomegaly is also observed in human patients with severe anemia, this has been primarily attributed to increased sequestration and erythrophagocytosis of infected and uninfected RBCs113,114  (Figure 3). However, extramedullary hematopoiesis commonly occurs in other conditions such as thalassemia in both humans and mice, and it would therefore be important to explore the erythropoietic role of the spleen in future human malaria studies, since if this is the case then the mouse model gains in relevance.

Finally, it should be noted that microarray analysis of peripheral responses to human and mouse malaria infection could prove extremely useful. One human study has allowed for some validation of clinical observations through the description of the gene expression profiles in children with severe malaria and during their convalescence.115  In this case, a positive correlation between neutrophil counts and increased expression of genes activated in the innate immune response as well as an inverse relationship between reticulocyte count and expression of genes associated with hemoglobin synthesis was observed. The advantage of a rodent model for SMA would be that a more complete study of bone marrow and splenic erythrocytic responses to malaria infection is possible, and several studies analyzing different organs from infected mice have described some promising results, such as a reduction in the level of gene transcripts involved in erythropoiesis and erythroid cell survival, during the early stages of infection.116,118  These preliminary observations reveal that this technology is capable of extensive analyses in both humans and mice. Future studies should be considered because, as well as being able to identify the similarities and differences of the human and mouse peripheral response, microarrays may also facilitate the development of therapeutics.

In summary, it is possible to identify specific similarities and differences between disease pathology during P falciparum infection in humans, and infection with specific strains of Plasmodia in mice. The mouse is particularly useful for studying anemia associated with the acute phase of infection. One of the key similarities at this stage is that the erythropoietic response to Epo does not correct the deficit in hematocrit caused by hemolysis and sequestration of RBCs and that this is associated with abnormalities in the bone marrow. In acute disease the imbalance between proinflammatory and anti-inflammatory mediators may be the main cause of dyserythropoiesis. Mouse models that may represent this etiology of disease include those that use P chabaudi and P yoelii. However, the causative factors of SMA in chronic infection in humans are less clear and have not been extensively investigated in mice. In addition to the chronic P bergei model of rodent malaria mentioned previously, more chronic models of malaria infection with persistent but low parasitemias need to be developed. During the course of a chronic infection, there would be impaired clearance of parasite products (eg, RSP2, Hz, and GPI), and their accumulation may together or individually contribute to the chronic nature of SMA. Future studies of the longer term effects of parasite products on cytokine regulation and hematopoiesis in mice may be valuable in elucidating the mechanism involved in suppression of RBC production. Such studies coupled with in vitro investigations of the effects of parasite products on human hematopoietic cells will allow us to determine the stages of erythropoiesis involved and will increase our understanding of the etiology of SMA. However, it will be essential to link these animal and experimental data with studies in patients with malaria aimed at understanding not only the mechanisms leading to severe anemia but also their relative importance in different clinical settings. Already, from the studies summarized in this review on both mouse and human SMA, several interesting hypotheses await further clinical investigation and may lay the foundation for novel interventions to prevent or treat severe malarial anemia.

This work was supported by the National Blood Service, the Howard Hughes Medical Institute (A.A.L. and D.J.R.), the MRC (D.B., A.P., and J. L.), and the EU BioMalPar NoE (A.A.L., D.J.R., D.B., A.P., and J. L.).

We would like to thank Kevin Marsh, Paulo Arese, Suzanne Watt, and Bill Wood for helpful discussions and critical thoughts. The research was carried out in part at the NBS-Oxford Center and benefits from NHS R&D funding.

Contribution: A.A.L. and D.B. contributed to design and content of paper, to review of literature, and to writing the draft; A.P. and C.C.-P. contributed to content of the review; J.L. and D.J.R. critically reviewed text and edited tables and figures.

A.A.L. and D.B. contributed equally to the review.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Jean Langhorne, National Institute for Medical Research, The Ridgeway, London, NW7 1AA; e-mail: jlangho@nimr.mrc.ac.uk; and David J. Roberts, Nuffield Department of Clinical Laboratory Sciences and National Blood Service Oxford Centre, Oxford OX3 9BQ; e-mail: david.roberts@ndcls.ox.ac.uk.

1
Rowe
AK
Rowe
SY
Snow
RW
et al
The burden of malaria mortality among African children in the year 2000.
Int J Epidemiol
2006
35
691
704
2
Snow
RW
Guerra
CA
Noor
AM
Myint
HY
Hay
SI
The global distribution of clinical episodes of Plasmodium falciparum malaria.
Nature
2005
434
214
217
3
Egan
AF
Fabucci
ME
Saul
A
Kaslow
DC
Miller
LH
Aotus New World monkeys: model for studying malaria-induced anemia.
Blood
2002
99
3863
3866
4
Jones
TR
Stroncek
DF
Gozalo
AS
et al
Anemia in parasite- and recombinant protein-immunized aotus monkeys infected with Plasmodium falciparum.
Am J Trop Med Hyg
2002
66
672
679
5
Fleming
AF
HIV and blood transfusion in sub-Saharan Africa.
Transfus Sci
1997
18
167
179
6
Greenwood
BM
The epidemiology of malaria.
Ann Trop Med Parasitol
1997
91
763
769
7
Menendez
C
Fleming
AF
Alonso
PL
Malaria-related anaemia.
Parasitol Today
2000
16
469
476
8
Schellenberg
D
Schellenberg
JR
Mushi
A
et al
The silent burden of anaemia in Tanzanian children: a community-based study.
Bull World Health Organ
2003
81
581
590
9
Menendez
C
Kahigwa
E
Hirt
R
et al
Randomised placebo-controlled trial of iron supplementation and malaria chemoprophylaxis for prevention of severe anaemia and malaria in Tanzanian infants.
Lancet
1997
350
844
850
10
Warrell
DA
Molyneux
ME
Beales
PF
Severe and Complicated Malaria.
World Health Organization Division of Control of Tropical Diseases
2nd ed.
London, United Kingdom
Royal Society of Tropical Medicine and Hygiene
1990
11
Roberts
DJ
Casals-Pascual
C
Weatherall
DJ
The clinical and pathophysiological features of malarial anaemia.
Curr Top Microbiol Immunol
2005
295
137
167
12
Schellenberg
D
Menendez
C
Kahigwa
E
et al
Intermittent treatment for malaria and anaemia control at time of routine vaccinations in Tanzanian infants: a randomised, placebo-controlled trial.
Lancet
2001
357
1471
1477
13
Ocaňa-Morgner
C
Mota
MM
Rodriguez
A
Malaria blood stage suppression of liver stage immunity by dendritic cells.
J Exp Med
2003
197
143
151
14
Hviid
L
Naturally acquired immunity to Plasmodium falciparum malaria in Africa.
Acta Tropica
2005
95
270
275
15
Tjitra
E
Warikar
N
Ebsworth
EP
et al
Major burden of P. vivax infection in Papua Indonesia: a region with high levels of chloroquine resistance.
XVIe International Congress for Tropical Medicine and Malaria
September 11-15 2005
France
Marseille
Paper presented at
16
Rodriguez-Morales
AJ
Sanchez
E
Vargas
M
Piccolo
C
Colina
R
Arria
M
Anemia and thrombocytopenia in children with Plasmodium vivax malaria.
J Trop Pediatr
2006
52
49
51
17
Nosten
F
McGready
R
Simpson
JA
et al
Effects of Plasmodium vivax malaria in pregnancy.
Lancet
1999
354
546
549
18
Marsh
K
Forster
D
Waruiru
C
et al
Indicators of life-threatening malaria in African children.
N Engl J Med
1995
332
1399
1404
19
Berkley
J
Mwarumba
S
Bramham
K
Lowe
B
Marsh
K
Bacteraemia complicating severe malaria in children.
Trans R Soc Trop Med Hyg
1999
93
283
286
20
Berkley
JA
Maitland
K
Mwangi
I
et al
Use of clinical syndromes to target antibiotic prescribing in seriously ill children in malaria endemic area: observational study.
BMJ
2005
330
995
1000
21
Krishna
S
Waller
DW
ter Kuile
F
et al
Lactic acidosis and hypoglycaemia in children with severe malaria: pathophysiological and prognostic significance.
Trans R Soc Trop Med Hyg
1994
88
67
73
22
English
M
Muambi
B
Mithwani
S
Marsh
K
Lactic acidosis and oxygen debt in African children with severe anaemia.
Qjm
1997
90
563
569
23
English
M
Waruiru
C
Marsh
K
Transfusion for respiratory distress in life-threatening childhood malaria.
Am J Trop Med Hyg
1996
55
525
530
24
Snow
RW
Omumbo
JA
Lowe
B
et al
Relation between severe malaria morbidity in children and level of Plasmodium falciparum transmission in Africa [see comments].
Lancet
1997
349
1650
1654
25
Newton
CR
Warn
PA
Winstanley
PA
et al
Severe anaemia in children living in a malaria endemic area of Kenya.
Trop Med Int Health
1997
2
165
178
26
Yeats
J
Daley
H
Hardie
D
Parvovirus B19 infection does not contribute significantly to severe anaemia in children with malaria in Malawi [letter] [In Process Citation].
Eur J Haematol
1999
63
276
277
27
Abdalla
SH
Pasvol
G
Malaria: A Hematological Perspective.
2004
Vol 4.
London, United Kingdom
Imperial College Press
28
Roberts
DJ
Casals-Pascual
C
Weatherall
DJ
Sullivan
D
Krishna
S
The Clinical and Pathophysiological Features of Malarial Anaemia.
Malaria: Drugs, Disease and Post-genomic Biology
2005
Vol 295.
Heidelberg, Germany
Springer
29
Stephens
JWW
Blackwater Fever.
1937
Liverpool, United Kingdom
University of Liverpool Press
30
Price
R
van Vugt
M
Phaipun
L
et al
Adverse effects in patients with acute falciparum malaria treated with artemisinin derivatives.
Am J Trop Med Hyg
1999
60
547
555
31
Lamb
TJ
Brown
DE
Potocnik
AJ
Langhorne
J
Insights into the immunopathogenesis of malaria using mouse models.
Expert Rev Mol Med
2006
8
1
22
32
Carter
R
Walliker
D
New observations on the malaria parasites of rodents of the Central African Republic: Plasmodium vinckei petteri subsp. nov and Plasmodium chabaudi Landau, 1965.
Ann Trop Med Parasitol
1975
69
187
196
33
Jarra
W
Brown
KN
Protective immunity to malaria: studies with cloned lines of rodent malaria in CBA/Ca mice IV: the specificity of mechanisms resulting in crisis and resolution of the primary acute phase parasitaemia of Plasmodium chabaudi chabaudi and P. yoelii.
Parasite Immunol
1989
11
1
13
34
Chotivanich
K
Udomsangpetch
R
Simpson
JA
et al
Parasite multiplication potential and the severity of Falciparum malaria.
J Infect Dis
2000
181
1206
1209
35
Garnham
PC
Bird
RG
Baker
JR
Desser
SS
el-Nahal
HM
Electron microscope studies on motile stages of malaria parasites, VI: the ookinete of Plasmodium berghei yoelii and its transformation into the early oocyst.
Trans R Soc Trop Med Hyg
1969
63
187
194
36
Wildig
J
Michon
P
Siba
P
Mellombo
M
et al
Parvovirus B19 infection contributes to severe anemia in young children in Papua New Guinea.
J Infect Dis
2006
194
146
153
37
Casals-Pascual
C
Roberts
DJ
Severe malarial anaemia.
Curr Mol Med
2006
6
155
168
38
Langhorne
J
Quin
SJ
Sanni
LA
Mouse models of blood-stage malaria infections: immune responses and cytokines involved in protection and pathology.
Chem Immunol
2002
80
204
228
39
Looareesuwan
S
Ho
M
Wattanagoon
Y
et al
Dynamic alteration in splenic function during acute falciparum malaria.
N Engl J Med
1987
317
675
679
40
Jakeman
GN
Saul
A
Hogarth
WL
Collins
WE
Anaemia of acute malaria infections in non-immune patients primarily results from destruction of uninfected erythrocytes [In Process Citation].
Parasitology
1999
119
127
133
41
Price
RN
Simpson
JA
Nosten
F
et al
Factors contributing to anemia after uncomplicated falciparum malaria.
Am J Trop Med Hyg
2001
65
614
622
42
Looareesuwan
S
Merry
AH
Phillips
RE
et al
Reduced erythrocyte survival following clearance of malarial parasitaemia in Thai patients.
Br J Haematol
1987
67
473
478
43
Brown
AE
Webster
HK
Teja-Isavadharm
P
Keeratithakul
D
Macrophage activation in falciparum malaria as measured by neopterin and interferon-gamma.
Clin Exp Immunol
1990
82
97
101
44
Mohan
K
Dubey
ML
Ganguly
NK
Mahajan
RC
Plasmodium falciparum: role of activated blood monocytes in erythrocyte membrane damage and red cell loss during malaria.
Exp Parasitol
1995
80
54
63
45
Ladhani
S
Lowe
B
Cole
AO
Kowuondo
K
Newton
CR
Changes in white blood cells and platelets in children with falciparum malaria: relationship to disease outcome.
Br J Haematol
2002
119
839
847
46
Jenkins
NE
Chakravorty
SJ
Urban
BC
Kai
OK
Marsh
K
Craig
AG
The effect of Plasmodium falciparum infection on expression of monocyte surface molecules.
Trans R Soc Trop Med Hyg
2006
100
1007
1012
47
Dondorp
AM
Omodeo-Sale
F
Chotivanich
K
Taramelli
D
White
NJ
Oxidative stress and rheology in severe malaria.
Redox Rep
2003
8
292
294
48
Omodeo-Sale
F
Motti
A
Dondorp
A
White
NJ
Taramelli
D
Destabilisation and subsequent lysis of human erythrocytes induced by Plasmodium falciparum haem products.
Eur J Haematol
2005
74
324
332
49
Dondorp
AM
Angus
BJ
Hardeman
MR
et al
Prognostic significance of reduced red blood cell deformability in severe falciparum malaria.
Am J Trop Med Hyg
1997
57
507
511
50
Dondorp
AM
Nyanoti
M
Kager
PA
Mithwani
S
Vreeken
J
Marsh
K
The role of reduced red cell deformability in the pathogenesis of severe falciparum malaria and its restoration by blood transfusion.
Trans R Soc Trop Med Hyg
2002
96
282
286
51
Pouvelle
B
Buffet
PA
Lepolard
C
Scherf
A
Gysin
J
Cytoadhesion of Plasmodium falciparum ring-stage-infected erythrocytes.
Nat Med
2000
6
1264
1268
52
Douki
JB
Sterkers
Y
Lepolard
C
et al
Adhesion of normal and Plasmodium falciparum ring-infected erythrocytes to endothelial cells and the placenta involves the rhoptry-derived ring surface protein-2.
Blood
2003
101
5025
5032
53
Layez
C
Nogueira
P
Combes
V
et al
Plasmodium falciparum rhoptry protein RSP2 triggers destruction of the erythroid lineage.
Blood
2005
106
3632
3638
54
Villeval
JL
Lew
A
Metcalf
D
Changes in hemopoietic and regulator levels in mice during fatal or nonfatal malarial infections, I: erythropoietic populations.
Exp Parasitol
1990
71
364
374
55
Chang
KH
Tam
M
Stevenson
MM
Modulation of the course and outcome of blood-stage malaria by erythropoietin-induced reticulocytosis.
J Infect Dis
2004
189
735
743
56
Salmon
MG
De Souza
JB
Butcher
GA
Playfair
JH
Premature removal of uninfected erythrocytes during malarial infection of normal and immunodeficient mice.
Clin Exp Immunol
1997
108
471
476
57
Evans
KJ
Hansen
DS
van Rooijen
N
Buckingham
LA
Schofield
L
Severe malarial anemia of low parasite burden in rodent models results from accelerated clearance of uninfected erythrocytes.
Blood
2006
107
1192
1199
58
Vryonis
G
Observations in the parasitizaton of erythrocytes by Plasmodium vivax, with special reference to reticulocytes.
Am J Hyg
1939
30
41
59
Abdalla
S
Weatherall
DJ
Wickramasinghe
SN
Hughes
M
The anaemia of P. falciparum malaria.
Br J Haematol
1980
46
171
183
60
Abdalla
SH
Hematopoiesis in human malaria.
Blood Cells
1990
16
401
416
discussion 417–419
61
Wickramasinghe
SN
Abdalla
S
Weatherall
DJ
Cell cycle distribution of erythroblasts in P. falciparum malaria.
Scand J Haematol
1982
29
83
88
62
Rencricca
NJ
Coleman
RM
Altered erythropoiesis during the course of virulent murine malaria.
Proc Soc Exp Biol Med
1979
162
424
428
63
Maggio-Price
L
Brookoff
D
Weiss
L
Changes in hematopoietic stem cells in bone marrow of mice with Plasmodium berghei malaria.
Blood
1985
66
1080
1085
64
Weiss
L
Johnson
J
Weidanz
W
Mechanisms of splenic control of murine malaria: tissue culture studies of the erythropoietic interplay of spleen, bone marrow, and blood in lethal (strain 17XL) Plasmodium yoelii malaria in BALB/c mice.
Am J Trop Med Hyg
1989
41
135
143
65
Asami
M
Owhashi
M
Abe
T
Nawa
Y
A comparative study of the kinetic changes of hemopoietic stem cells in mice infected with lethal and non-lethal malaria.
Int J Parasitol
1992
22
43
47
66
Yap
GS
Stevenson
MM
Plasmodium chabaudi AS: erythropoietic responses during infection in resistant and susceptible mice.
Exp Parasitol
1992
75
340
352
67
Schwarzer
E
Turrini
F
Ulliers
D
Giribaldi
G
Ginsburg
H
Arese
P
Impairment of macrophage functions after ingestion of Plasmodium falciparum-infected erythrocytes or isolated malarial pigment.
J Exp Med
1992
176
1033
1041
68
Schwarzer
E
Alessio
M
Ulliers
D
Arese
P
Phagocytosis of the malarial pigment, hemozoin, impairs expression of major histocompatibility complex class II antigen, CD54, and CD11c in human monocytes.
Infect Immun
1998
66
1601
1606
69
Schwarzer
E
Ludwig
P
Valente
E
Arese
P
15(S)-hydroxyeicosatetraenoic acid (15-HETE), a product of arachidonic acid peroxidation, is an active component of hemozoin toxicity to monocytes.
Parassitologia
1999
41
199
202
70
Schwarzer
E
Kuhn
H
Valente
E
Arese
P
Malaria-parasitized erythrocytes and hemozoin nonenzymatically generate large amounts of hydroxy fatty acids that inhibit monocyte functions.
Blood
2003
101
722
728
71
Giribaldi
G
Ulliers
D
Schwarzer
E
Roberts
I
Piacibello
W
Arese
P
Hemozoin- and 4-hydroxynonenal-mediated inhibition of erythropoiesis: possible role in malarial dyserythropoiesis and anemia.
Haematologica
2004
89
492
493
72
Casals-Pascual
C
Kai
O
Cheung
JO
et al
Suppression of erythropoiesis in malarial anemia is associated with hemozoin in vitro and in vivo.
Blood
2006
108
2569
2577
73
Kwiatkowski
D
Hill
AV
Sambou
I
et al
TNF concentration in fatal cerebral, non-fatal cerebral, and uncomplicated Plasmodium falciparum malaria.
Lancet
1990
336
1201
1204
74
Yap
GS
Stevenson
MM
Inhibition of in vitro erythropoiesis by soluble mediators in Plasmodium chabaudi AS malaria: lack of a major role for interleukin 1, tumor necrosis factor alpha, and gamma interferon.
Infect Immun
1994
62
357
362
75
Miller
KL
Silverman
PH
Kullgren
B
Mahlmann
LJ
Tumor necrosis factor alpha and the anemia associated with murine malaria.
Infect Immun
1989
57
1542
1546
76
Dufour
C
Corcione
A
Svahn
J
et al
TNF-alpha and IFN-gamma are overexpressed in the bone marrow of Fanconi anemia patients and TNF-alpha suppresses erythropoiesis in vitro.
Blood
2003
102
2053
2059
77
Felli
N
Pedini
F
Zeuner
A
et al
Multiple members of the TNF superfamily contribute to IFN-gamma-mediated inhibition of erythropoiesis.
J Immunol
2005
175
1464
1472
78
Kurtzhals
JA
Adabayeri
V
Goka
BQ
et al
Low plasma concentrations of interleukin 10 in severe malarial anaemia compared with cerebral and uncomplicated malaria.
Lancet
1998
351
1768
1772
79
Othoro
C
Lal
AA
Nahlen
B
Koech
D
Orago
AS
Udhayakumar
V
A low interleukin-10 tumor necrosis factor-alpha ratio is associated with malaria anemia in children residing in a holoendemic malaria region in western Kenya.
J Infect Dis
1999
179
279
282
80
McGuire
W
Knight
JC
Hill
AV
Allsopp
CE
Greenwood
BM
Kwiatkowski
D
Severe malarial anemia and cerebral malaria are associated with different tumor necrosis factor promoter alleles.
J Infect Dis
1999
179
287
290
81
Linke
A
Kuhn
R
Muller
W
Honarvar
N
Li
C
Langhorne
J
Plasmodium chabaudi chabaudi: differential susceptibility of gene-targeted mice deficient in IL-10 to an erythrocytic-stage infection.
Exp Parasitol
1996
84
253
263
82
Li
C
Sanni
LA
Omer
F
Riley
E
Langhorne
J
Pathology of Plasmodium chabaudi chabaudi infection and mortality in interleukin-10-deficient mice are ameliorated by anti-tumor necrosis factor alpha and exacerbated by anti-transforming growth factor beta antibodies.
Infect Immun
2003
71
4850
4856
83
Malaguarnera
L
Pignatelli
S
Musumeci
M
Simpore
J
Musumeci
S
Plasma levels of interleukin-18 and interleukin-12 in Plasmodium falciparum malaria.
Parasite Immunol
2002
24
489
492
84
Clark
IA
Cowden
WB
The pathophysiology of falciparum malaria.
Pharmacol Ther
2003
99
221
260
85
Mohan
K
Stevenson
MM
Dyserythropoiesis and severe anaemia associated with malaria correlate with deficient interleukin-12 production.
Br J Haematol
1998
103
942
949
86
Mohan
K
Stevenson
MM
Interleukin-12 corrects severe anemia during blood-stage Plasmodium chabaudi AS in susceptible A/J mice.
Exp Hematol
1998
26
45
52
87
Martiney
JA
Sherry
B
Metz
CN
et al
Macrophage migration inhibitory factor release by macrophages after ingestion of Plasmodium chabaudi-infected erythrocytes: possible role in the pathogenesis of malarial anemia.
Infect Immun
2000
68
2259
2267
88
McDevitt
MA
Xie
J
Shanmugasundaram
G
et al
A critical role for the host mediator macrophage migration inhibitory factor in the pathogenesis of malarial anemia.
J Exp Med
2006
203
1185
1196
89
Awandare
GA
Hittner
JB
Kremsner
PG
et al
Decreased circulating macrophage migration inhibitory factor (MIF) protein and blood mononuclear cell MIF transcripts in children with Plasmodium falciparum malaria.
Clin Immunol
2006
119
219
225
90
Chaiyaroj
SC
Rutta
AS
Muenthaisong
K
Watkins
P
Na Ubol
M
Looareesuwan
S
Reduced levels of transforming growth factor-beta1, interleukin-12 and increased migration inhibitory factor are associated with severe malaria.
Acta Trop
2004
89
319
327
91
Lyke
KE
Burges
R
Cissoko
Y
et al
Serum levels of the proinflammatory cytokines interleukin-1 beta (IL-1beta), IL-6, IL-8, IL-10, tumor necrosis factor alpha, and IL-12(p70) in Malian children with severe Plasmodium falciparum malaria and matched uncomplicated malaria or healthy controls.
Infect Immun
2004
72
5630
5637
92
Miller
LH
Roberts
T
Shahabuddin
M
McCutchan
TF
Analysis of sequence diversity in the Plasmodium falciparum merozoite surface protein-1 (MSP-1).
Mol Biochem Parasitol
1993
59
1
14
93
Schofield
L
Hackett
F
Signal transduction in host cells by a glycosylphosphatidylinositol toxin of malaria parasites.
J Exp Med
1993
177
145
153
94
Krishnegowda
G
Hajjar
AM
Zhu
J
et al
Induction of proinflammatory responses in macrophages by the glycosylphosphatidylinositols of Plasmodium falciparum: cell signaling receptors, glycosylphosphatidylinositol (GPI) structural requirement, and regulation of GPI activity.
J Biol Chem
2005
280
8606
8616
95
Rudin
W
Quesniaux
V
Favre
N
Bordmann
G
Malaria toxins from P. chabaudi chabaudi AS and P. berghei ANKA cause dyserythropoiesis in C57BL/6 mice.
Parasitology
1997
115
pt 5
467
474
96
Bordmann
G
Favre
N
Rudin
W
Malaria toxins: effects on murine spleen and bone marrow cell proliferation and cytokine production in vitro.
Parasitology
1997
115
5
475
483
97
Schofield
L
Hewitt
MC
Evans
K
Siomos
MA
Seeberger
PH
Synthetic GPI as a candidate anti-toxic vaccine in a model of malaria.
Nature
2002
418
785
789
98
Naik
RS
Branch
OH
Woods
AS
et al
Glycosylphosphatidylinositol anchors of Plasmodium falciparum: molecular characterization and naturally elicited antibody response that may provide immunity to malaria pathogenesis.
J Exp Med
2000
192
1563
1576
99
Pichyangkul
S
Saengkrai
P
Webster
HK
Plasmodium falciparum pigment induces monocytes to release high levels of tumor necrosis factor-alpha and interleukin-1 beta.
Am J Trop Med Hyg
1994
51
430
435
100
Sherry
BA
Alava
G
Tracey
KJ
Martiney
J
Cerami
A
Slater
AF
Malaria-specific metabolite hemozoin mediates the release of several potent endogenous pyrogens (TNF, MIP-1 alpha, and MIP-1 beta) in vitro, and altered thermoregulation in vivo.
J Inflamm
1995
45
85
96
101
Prato
M
Giribaldi
G
Polimeni
M
Gallo
V
Arese
P
Phagocytosis of hemozoin enhances matrix metalloproteinase-9 activity and TNF-alpha production in human monocytes: role of matrix metalloproteinases in the pathogenesis of falciparum malaria.
J Immunol
2005
175
6436
6442
102
Jaramillo
M
Plante
I
Ouellet
N
Vandal
K
Tessier
PA
Olivier
M
Hemozoin-inducible proinflammatory events in vivo: potential role in malaria infection.
J Immunol
2004
172
3101
3110
103
Coban
C
Ishii
KJ
Kawai
T
et al
Toll-like receptor 9 mediates innate immune activation by the malaria pigment hemozoin.
J Exp Med
2005
201
19
25
104
Burgmann
H
Looareesuwan
S
Kapiotis
S
et al
Serum levels of erythropoietin in acute Plasmodium falciparum malaria.
Am J Trop Med Hyg
1996
54
280
283
105
el Hassan
AM
Saeed
AM
Fandrey
J
Jelkmann
W
Decreased erythropoietin response in Plasmodium falciparum malaria-associated anaemia.
Eur J Haematol
1997
59
299
304
106
Kurtzhals
JA
Rodrigues
O
Addae
M
Commey
OO
et al
Reversible Suppression of bone marrow response to erythropoietin in Plasmodium falciparum malaria.
Brit J Haematol
1997
97
169
174
107
Nussenblatt
V
Mukasa
G
Metzger
A
Ndeezi
G
Garrett
E
Semba
RD
Anemia and interleukin-10, tumor necrosis factor alpha, and erythropoietin levels among children with acute, uncomplicated Plasmodium falciparum malaria.
Clin Diagn Lab Immunol
2001
8
1164
1170
108
Verhoef
H
West
CE
Kraaijenhagen
R
et al
Malarial anemia leads to adequately increased erythropoiesis in asymptomatic Kenyan children.
Blood
2002
100
3489
3494
109
Chang
KH
Stevenson
MM
Effect of anemia and renal cytokine production on erythropoietin production during blood-stage malaria.
Kidney Intl
2004
65
1640
1646
110
Chang
KH
Tam
M
Stevenson
MM
Inappropriately low reticulocytosis in severe malarial anemia correlates with suppression in the development of late erythroid precursors.
Blood
2004
103
3727
3735
111
Nemeth
E
Valore
E
Territo
M
Schiller
G
Lichtenstein
A
Ganz
T
Hepcidin, a putative mediator of anemia of inflammation, is a type II acute-phase protein.
Blood
2003
101
2461
2463
112
Weiss
L
Mechanisms of splenic control of murine malaria: cellular reactions of the spleen in lethal (strain 17XL) Plasmodium yoelii malaria in BALB/c mice, and the consequences of pre-infective splenectomy.
Am J Trop Med Hyg
1989
41
144
160
113
Zingman
BS
Viner
BL
Splenic complications in malaria: case report and review.
Clin Infect Dis
1993
16
223
232
114
Urban
BC
Hien
TT
Day
NP
et al
Fatal Plasmodium falciparum malaria causes specific patterns of splenic architectural disorganization.
Infect Immun
2005
73
1986
1994
115
Griffiths
MJ
Shafi
MJ
Popper
SJ
et al
Genomewide analysis of the host response to malaria in Kenyan children.
J Infect Dis
2005
191
1599
1611
116
Sexton
AC
Good
RT
Hansen
DS
et al
Transcriptional profiling reveals suppressed erythropoiesis, up-regulated glycolysis, and interferon-associated responses in murine malaria.
J Infect Dis
2004
189
1245
1256
117
Sarfo
BY
Armah
HB
Irune
I
et al
Plasmodium yoelii 17XL infection up-regulates RANTES, CCR1, CCR3 and CCR5 expression, and induces ultrastructural changes in the cerebellum.
Malar J
2005
4
63
75
118
Delahaye
NF
Coltel
N
Puthier
D
et al
Gene-expression profiling discriminates between cerebral malaria (CM)-susceptible mice and CM-resistant mice.
J Infect Dis
2006
193
312
321
119
Spira
D
Zuckerman
A
Blood loss and replacement in plasmodial infections. VI. Plasmodium berghei in splenectomized rats.
J Infect Dis
1965
115
337
344
120
Kaul
DK
Liu
XD
Nagel
RL
Shear
HL
Microvascular hemodynamics and in vivo evidence for the role of intercellular adhesion molecule-1 in the sequestration of infected red blood cells in a mouse model of lethal malaria.
Am J Trop Med Hyg
1998
58
240
247
121
Miller
KL
Schooley
JC
Smith
KL
Kullgren
B
Mahlmann
LJ
Silverman
PH
Inhibition of erythropoiesis by a soluble factor in murine malaria.
Exp Hematol
1989
17
379
385
122
Glenister
FK
Coppel
RL
Cowman
AF
Mohandas
N
Cooke
BM
Contribution of parasite proteins to altered mechanical properties of malaria-infected red blood cells.
Blood
2002
99
1060
1063
123
Groux
H
Gysin
J
Opsonization as an effector mechanism in human protection against asexual blood stages of Plasmodium falciparum: functional role of IgG subclasses.
Res Immunol
1990
141
529
542
124
Scholander
C
Carlson
J
Kremsner
PG
Wahlgren
M
Extensive immunoglobulin binding of Plasmodium falciparum-infected erythrocytes in a group of children with moderate anemia.
Infect Immun
1998
66
361
363
125
Winograd
E
Sherman
IW
Naturally occurring anti-band 3 autoantibodies recognize a high molecular weight protein on the surface of Plasmodium falciparum infected erythrocytes.
Biochem Biophys Res Commun
1989
160
1357
1363
126
McGilvray
ID
Serghides
L
Kapus
A
Rotstein
OD
Kain
KC
Nonopsonic monocyte/macrophage phagocytosis of Plasmodium falciparum-parasitized erythrocytes: a role for CD36 in malarial clearance.
Blood
2000
96
3231
3240
127
Angus
BJ
Chotivanich
K
Udomsangpetch
R
White
NJ
In vivo removal of malaria parasites from red blood cells without their destruction in acute falciparum malaria.
Blood
1997
90
2037
2040
128
Mota
MM
Brown
KN
Holder
AA
Jarra
W
Acute Plasmodium chabaudi chabaudi malaria infection induces antibodies which bind to the surfaces of parasitized erythrocytes and promote their phagocytosis by macrophages in vitro.
Infect Immun
1998
66
4080
4086
129
Ternynck
T
Falanga
PB
Unterkirscher
C
Gregoire
J
da Silva
LP
Avrameas
S
Induction of high levels of IgG autoantibodies in mice infected with Plasmodium chabaudi.
Int Immunol
1991
3
29
37
130
Ockenhouse
CF
Shear
HL
Oxidative killing of the intraerythrocytic malaria parasite Plasmodium yoelii by activated macrophages.
J Immunol
1984
132
424
431
131
Shear
HL
Nussenzweig
RS
Bianco
C
Immune phagocytosis in murine malaria.
J Exp Med
1979
149
1288
1298
132
Landau
I
Chabaud
AG
Mora-Silvera
E
et al
Survival of rodent malaria merozoites in the lymphatic network: potential role in chronicity of the infection.
Parasite
1999
6
311
322
133
Facer
CA
Bray
RS
Brown
J
Direct Coombs antiglobulin reactions in Gambian children with Plasmodium falciparum malaria, I: incidence and class specificity.
Clin Exp Immunol
1979
35
119
127
134
Facer
CA
Direct Coombs antiglobulin reactions in Gambian children with Plasmodium falciparum malaria, II: specificity of erythrocyte-bound IgG.
Clin Exp Immunol
1980
39
279
288
135
Waitumbi
JN
Opollo
MO
Muga
RO
Misore
AO
Stoute
JA
Red cell surface changes and erythrophagocytosis in children with severe plasmodium falciparum anemia.
Blood
2000
95
1481
1486
136
Stoute
JA
Odindo
AO
Owuor
BO
Mibei
EK
Opollo
MO
Waitumbi
JN
Loss of red blood cell-complement regulatory proteins and increased levels of circulating immune complexes are associated with severe malarial anemia.
J Infect Dis
2003
187
522
525
137
Das
BS
Nanda
NK
Evidence for erythrocyte lipid peroxidation in acute falciparum malaria.
Trans R Soc Trop Med Hyg
1999
93
58
62
138
Griffiths
MJ
Ndungu
F
Baird
KL
Muller
DP
Marsh
K
Newton
CR
Oxidative stress and erythrocyte damage in Kenyan children with severe Plasmodium falciparum malaria.
Br J Haematol
2001
113
486
491
139
Atkinson
SH
Rockett
K
Sirugo
G
et al
Seasonal childhood anaemia in West Africa is associated with the haptoglobin 2-2 genotype.
PLoS Med
2006
5
3
5
e172
140
Hunt
NH
Driussi
C
Sai-Kiang
L
Haptoglobin and malaria.
Redox Rep
2001
6
389
392
141
Abdalla
SH
Wickramasinghe
SN
A study of erythroid progenitor cells in the bone marrow of Gambian children with falciparum malaria.
Clin Lab Haematol
1988
10
33
40
142
Silverman
PH
Schooley
JC
Mahlmann
LJ
Murine malaria decreases hemopoietic stem cells.
Blood
1987
69
408
413
143
Prunescu
P
Hepatic hematopoiesis in adult mouse during experimental rodent malaria.
Roum Arch Microbiol Immunol
1999
58
79
94
144
Hanspal
M
Hanspal
JS
The association of erythroblasts with macrophages promotes erythroid proliferation and maturation: a 30-kD heparin-binding protein is involved in this contact.
Blood
1994
84
3494
3504
145
Lee
G
Spring
FA
Parsons
SF
et al
Novel secreted isoform of adhesion molecule ICAM-4: potential regulator of membrane-associated ICAM-4 interactions.
Blood
2003
101
1790
1797
146
Zermati
Y
Fichelson
S
Valensi
F
et al
Transforming growth factor inhibits erythropoiesis by blocking proliferation and accelerating differentiation of erythroid progenitors.
Exp Hematol
2000
28
885
894
147
De Maria
R
Testa
U
Luchetti
L
et al
Apoptotic role of Fas/Fas ligand system in the regulation of erythropoiesis.
Blood
1999
93
796
803
148
Liu
Y
Pop
R
Sadegh
C
Brugnara
C
Haase
VH
Socolovsky
M
Suppression of Fas-FasL coexpression by erythropoietin mediates erythroblast expansion during the erythropoietic stress response in vivo.
Blood
2006
108
123
133
149
De Maria
R
Zeuner
A
Eramo
A
et al
Negative regulation of erythropoiesis by caspase-mediated cleavage of GATA-1.
Nature
1999
401
489
493
150
McNiece
IK
Langley
KE
Zsebo
KM
Recombinant human stem cell factor synergises with GM-CSF, G-CSF, IL-3 and epo to stimulate human progenitor cells of the myeloid and erythroid lineages.
Exp Hematol
1991
19
226
231
151
Arcasoy
MO
Jiang
X
Co-operative signalling mechanisms required for erythroid precursor expansion in response to erythropoietin and stem cell factor.
Br J Haematol
2005
130
121
129
152
Sui
X
Krantz
SB
Zhao
ZJ
Stem cell factor and erythropoietin inhibit apoptosis of human erythroid progenitor cells through different signalling pathways.
Br J Haematol
2000
110
63
70
153
Munugalavadla
V
Dore
LC
Tan
BL
et al
Repression of c-kit and its downstream substrates by GATA-1 inhibits cell proliferation during erythroid maturation.
Mol Cell Biol
2005
25
6747
6759
154
Chang
KH
Stevenson
MM
Malarial anaemia: mechanisms and implications of insufficient erythropoiesis during blood-stage malaria.
Int J Parasitol
2004
34
1501
1516
Sign in via your Institution