Skip to main content

Biochemical and immunological mechanisms by which sickle cell trait protects against malaria

Abstract

Sickle cell trait (HbAS) is the best-characterized genetic polymorphism known to protect against falciparum malaria. Although the protective effect of HbAS against malaria is well known, the mechanism(s) of protection remain unclear. A number of biochemical and immune-mediated mechanisms have been proposed, and it is likely that multiple complex mechanisms are responsible for the observed protection. Increased evidence for an immune component of protection as well as novel mechanisms, such as enhanced tolerance to disease mediated by HO-1 and reduced parasitic growth due to translocation of host micro-RNA into the parasite, have recently been described. A better understanding of relevant mechanisms will provide valuable insight into the host-parasite relationship, including the role of the host immune system in protection against malaria.

Background

Malaria, especially that caused by Plasmodium falciparum, has been a major cause of morbidity and mortality throughout human history. As a result, malaria has exerted extraordinary evolutionary pressure on the human genome and appears to have selected for multiple genetic polymorphisms that provide protection against severe disease [14]. The best-characterized human genetic polymorphism associated with malaria results in sickle haemoglobin (HbS). The high prevalence of HbS in sub-Saharan Africa and some other tropical areas is almost certainly due to the protection against malaria afforded to heterozygotes [13, 5]. Since the protective effect of sickle cell trait on malaria was first described over 60 years ago [68] our understanding of the epidemiology and mechanisms of protection of this genotype have continued to expand, as will be discussed below.

Sickle haemoglobin, sickle cell disease and sickle cell trait

Sickle haemoglobin (HbS) is a structural variant of normal adult haemoglobin. Adult haemoglobin (HbAA) is made up of two alpha and two beta globin chains. HbS is the result of a single point mutation (Glu → Val) on the sixth codon of the beta globin gene [9]. Homozygotes for haemoglobin S (HbSS) with two affected beta chains develop sickle cell disease, in which polymerized haemoglobin causes red blood cells to sickle and occlude blood vessels. Vaso-occlusion affects many organs and tissues, and results in high morbidity and mortality. Heterozygotes for sickle haemoglobin (HbAS) have sickle cell trait and are generally asymptomatic [10].

HbAS and protection against malaria

Despite the obvious deleterious nature of HbSS, it is now widely accepted that the persistence of the sickle mutation in human populations is due to the protection from malaria afforded to heterozygous individuals. Haldane first proposed the concept of a heterozygote advantage against malaria in 1949 [11]. In this seminal paper, Haldane suggested that individuals heterozygous for thalassaemia, another haemoglobinopathy, were protected against malaria.

Contemporaneous to this hypothesis, epidemiologic evidence for protection against malaria in those with HbS was emerging. In 1946, a British medical officer reported that the prevalence of malarial parasitaemia was lower in a cohort of 'sicklers” compared to “non-sicklers” in Northern Rhodesia [6], findings corroborated in 1952 [8]. The first true test of Haldane’s hypothesis came in 1954 through the work of Alison linking HbAS to protection against malaria in Uganda [7]. This study showed a reduced prevalence of parasitaemia (particularly P. falciparum) in Ugandans with HbAS compared to those with HbAA. In addition, after experimental infection Ugandans with HbAS had reduced parasitaemia and less clinical malaria than those with HbAA. Since these observations, strong evidence for the protective effects of HbAS against malaria has been generated in multiple case control and cohort studies [1217]. Recently, an evidence-based map of the global distribution of the sickle cell variant has been created in a bayesian geostatistical framework and compared to the global prevalence of P. falciparum[5]. These data provide comprehensive evidence for a global geographical association between malaria burden and HbS allele frequency, particularly in sub-Saharan Africa. Furthermore, evidence for the protective effects of other red blood cell polymorphisms against malaria, including haemoglobin C, haemoglobin E, thalassaemias, and ovalocytosis have also been described [1728].

In endemic countries, infection with P. falciparum causes a range of outcomes, including asymptomatic parasitaemia, uncomplicated disease and severe malaria, which commonly progresses to death. HbAS provides significant protection against both severe and uncomplicated malaria. Case–control and cohort studies in multiple African countries have consistently found that HbAS is 70-90% protective against severe malaria [1216] and 75% protective against hospitalization for malaria [29]. A recent meta-analysis reviewed 44 studies of children with HbAS and reported significant protection from severe malaria syndromes, including greater than 90% protection from severe malaria, cerebral malaria and severe malarial anaemia [17]. In addition, a cohort study showed a 60% reduction in overall mortality in HbAS children aged two to 16 months, compared to HbAA children, in an area of high malaria transmission [30]. Children with HbAS are also protected from uncomplicated malaria, with cohort studies showing that HbAS is 30-50% protective [15, 17, 27, 3136].

While associations between HbAS and protection against malaria are clear, data from clinical studies aiming to identify mechanism(s) of protection have been less consistent. Older studies found a lower prevalence of parasitaemia in HbAS individuals irrespective of symptoms [7, 37], suggesting HbAS exerts protection against the establishment of parasitaemia. Multiple other reports failed to identify an association between HbAS and the prevalence of asymptomatic parasitaemia [29, 31, 3840], but three recent studies found that HbAS children had significantly less asymptomatic parasitaemia than HbAA children [4143]. Further, HbAS children in Ghana had significantly lower parasite densities and a higher proportion of submicroscopic P. falciparum infection compared to HbAA children [41]. Data on associations between HbAS and the multiplicity of infection, the number of genetically distinct parasites causing an infection, are limited and results have been conflicting [26, 35, 44, 45]. A potential reason for these discrepancies is that, depending on the epidemiological context, high multiplicity of infection may reflect either lack of protection against infection, allowing the establishment of a larger number of patent parasites, or protection against symptomatic disease, allowing parasite clones to “stack up” since patients are less likely to seek care and receive antimalarial therapy. To further investigate the effect of HbAS on parasitaemia, another study followed a cohort of Ugandan children aged one to ten years for asymptomatic parasitaemia and symptomatic malaria, using genotyping to detect and follow individual parasite clones longitudinally [35]. This study found that HbAS protected against the establishment of parasitaemia by decreasing the force of infection, or the average number of parasite strains causing blood stream infections, and the probability of developing clinical symptoms once parasitaemic. HbAS children were also protected against high parasite densities during symptomatic malaria, consistent with prior studies [26, 2931, 33, 35, 41, 46], likely contributing importantly to protection against severe malaria. These discrepancies suggest that the mechanism of protection afforded by HbAS is complex, with impacts on both the development of parasitaemia and the control of parasitaemia once it is established.

Molecular mechanism of protection

Some decades ago, investigators found that P. falciparum parasites induced sickling of HbAS red blood cells in vitro. In the 1970s, two groups showed that parasitized HbAS cells sickled at a two to eight times higher rate than non-parasitized cells [47, 48]. One group also visualized polymerized haemoglobin in parasitized red blood cells and hypothesized that an increase in the polymerized haemoglobin or a reduced intracellular pH might cause increased sickling [48]. Increased sickling of parasitized red blood cells in HbAS individuals may promote enhanced phagocytosis of infected cells and, therefore, result in reduced parasitaemia compared to that in HbAA individuals (Table 1).

Table 1 Hypothesized protective mechanisms of sickle cell trait (HbAS) against malaria

Later in the 1970s, multiple studies found that P. falciparum ring-stage parasites did not grow in HbAS red blood cells under low oxygen tension [4850]. Parasite growth was inhibited in both sickled and non-sickled HbAS red blood cells [50] suggesting that factors in addition to sickling affected parasite growth. It has been hypothesized that specific intra-erythrocytic conditions of HbAS red blood cells, such as low intracellular potassium [49], high concentrations of haemoglobin [51] or osmotic shrinkage of the red blood cell [52] cause an inhospitable environment for parasites. A study also demonstrated that P. falciparum parasites invaded HbAS red blood cells less efficiently than HbAA cells at low oxygen tension [47] but subsequent studies failed to replicate these results [53]. Recent data provide support for the intriguing possibility that human micro RNAs translocated into parasite mRNA reduce intra-erythrocytic growth. This study found two human micro RNAs that were highly enriched in erythrocytes with HbAS, and these micro RNAs inhibited translation of specific parasite mRNA transcripts negatively impacting parasite growth in vitro[54].

Biochemical and mechanical changes in infected HbAS red blood cells have been shown to alter disease progression. Rosette formation, which is the binding of P. falciparum-infected red blood cells to uninfected red blood cells, is thought to lead to microcirculatory obstruction in cerebral malaria [5559]. Rosette formation was found to be impaired in P. falciparum-infected HbAS red blood cells under deoxygenated conditions [57]. Impaired rosette formation with HbAS red blood cells may be due to increased sickling of these cells in deoxygenated conditions [47, 48] or to reduced expression of erythrocyte surface adherence proteins [60]. Decreased rosette formation and the resulting decreased circulatory obstruction might contribute to protection against severe malaria in HbAS individuals.

Reduced cytoadherence has also been implicated as a mechanism of protection in HbAS individuals. Infected red blood cells express one of a family of parasite-encoded P. falciparum erythrocyte membrane protein 1 (PfEMP-1) molecules on the erythrocyte surface, and via this protein adhere to endothelial cells in the microvasculature [6164] .This process, termed cytoadherence, enables parasites to sequester in the vasculature and avoid clearance by the spleen [64]. Cytoadherence also leads to endothelial activation and associated inflammation in the brain and other organs, important in the progression to severe malaria [6568]. Reduced cytoadherence was first seen in infected red blood cells with another haemoglobinopathy, HbC, [69] which is due to a different mutation (Glu → Lys) in the same codon on the beta chain affected in HbS. Altered expression of PfEMP-1 was subsequently found in HbAS red blood cells in vitro[60]. Comparison of binding properties showed reduced adherence to endothelial cells expressing the binding ligand CD36 compared to HbAA red blood cells. PfEMP-1 surface signal was reduced by 14 % in HbAS and HbSS compared to HbAA erythrocytes in flow cytometric assays, suggesting altered surface expression of PfEMP-1, similar to that reported in HbC [60]. In addition, dysfunctional cytoskeletons have been visualized in HbSC erythrocytes [70]. Oxidized haemoglobin present in erythrocytes containing sickled haemoglobin may interfere with actin re-organization in infected HbSC erythrocytes leading to impaired vesicular transport of PfEMP-1 to the erythrocyte surface membrane [70]. These changes may impair parasite-induced remodelling of the red blood cell surface membrane and lead to altered PfEMP-1 surface expression [60, 69]. Reduced cytoadherence of HbAS and HbSS erythrocytes likely leads to increased splenic clearance, and may in part explain lower parasite densities and a lower incidence of severe malaria in HbAS individuals.

Role of the innate immune system

Phagocytosis by monocytes of HbAS red blood cells infected with ring-stage P. falciparum was found to be enhanced compared to that of infected HbAA cells, providing evidence for a role of the innate immune system in protection against P. falciparum in HbAS individuals [53]. Enhanced phagocytosis may be due to increased presentation of opsonins, including membrane bound IgG, C3c, membrane-bound hemichromes, and aggregated band 3 [53]. These opsonins, which are thought to be involved in the removal of senescent red blood cells, were first shown to be increased in G6PD deficiency [71, 72], a red blood cell enzyme deficiency also protective against malaria [1] and were also significantly higher in infected HbAS compared to HbAA red blood cells. Clearance by monocytes of red blood cells with exposed phosphatidylserine, a surface marker of damaged erythrocytes [7377], was also enhanced in infected HbAS compared to HbAA cells [73].

Enhanced opsonization and clearance of parasitized HbAS red blood cells by the spleen may lead to increased antigen presentation and earlier development of acquired immunity compared to that in HbAA individuals. A cross-sectional study found decreased levels of peripheral myeloid dendritic cells and monocytes in individuals with HbAS during healthy periods and malaria [78], suggesting increased monocyte and dendritic cell recruitment to the spleen.

Role of the acquired immune system

Population studies have found that the protective effect of HbAS increases with age, suggesting an acquired component of protection. A cross-sectional study of children with malaria in Nigeria found a significantly lower mean parasite density in HbAS compared with HbAA children in those two to four years old, but not in children less than two years old [476]. In Kenyan children, protection afforded by HbAS against symptomatic malaria increased from 20 % in children less than two years old to a peak of 56 % by age ten years [15]. In a recent study, protection against the establishment of parasitaemia and the development of symptomatic malaria once parasitaemic significantly increased between the ages of two and nine years [35]. Several studies shed light on possible immune bases for acquired protection.

Role of cell mediated immunity

Cell mediated responses to P. falciparum appear to be increased in HbAS compared to HbAA individuals. The mean lymphoproliferative response to affinity-purified P. falciparum soluble antigens [7981] was found to be significantly higher in HbAS children compared to HbAA children [8285] but a significant difference has not consistently been found between HbAA and HbAS adults [82, 83]. Thus, available results suggest a more robust cellular response to P. falciparum in HbAS children. However, it is unclear whether this is a cause or effect of the protective effects of HbAS. The lymphoproliferative response is suppressed during and after acute malarial infection in HbAA individuals [64, 81, 8689]. Therefore, a more robust lymphoproliferative response in HbAS individuals could be secondary to protection against malaria from other mechanisms.

Role of humoral immunity

Investigators have also found evidence for an enhanced humoral response in subjects with HbAS. Increased levels of gamma globulin were found in HbAS compared to HbAA children [90, 91]. However, higher levels of specific antibodies directed at parasite surface antigens believed to play a role in protective responses, including Pf155/ring infected erythrocyte surface antigen (RESA), merozoite surface protein 1 (MSP1), merozoite surface protein 2 (MSP2), erythrocyte binding antigen 175 (EBA175), and glutamate-rich protein (GLURP), have not been seen in HbAS compared to HbAA individuals in most studies [31, 85, 9296]. Recently, a study using a protein microarray representing 491 P. falciparum proteins found no increase in the magnitude or breadth of the P. falciparum-specific IgG response [97]. One study did find increased levels of antibodies toward free parasite antigens apical membrane antigen 1 (AMA1), EBA175, MSP1, MSP2, MSP3, circumsporozoite protein (CSP), and parasite schizont extract (PSE) in HbAS vs HbAA children living in areas of low malarial transmission in Burkina Faso [98], but not in children in an area of higher transmission. Another study found lower levels of IgG1 and IgG3 to MSP2 and RESA in HbAS individuals [99], possibly due to reduced exposure to these antigens in HbAS individuals.

In contrast, higher levels of IgG directed at PfEMP-1 family proteins, which are located on the surface of the red blood cell, have been found in individuals with HbAS in a number of studies. On study found HbAS individuals had a higher IgG response to the infected red blood cell in vitro[100]. In The Gambia [31], Gabon [101], and a low transmission area of Burkina Faso [98], HbAS children had higher levels of IgG antibodies toward PfEMP-1 than did those with HbAA [31]. However, studies in areas of high malaria transmission failed to find increased antibody response to PfEMP-1 in HbAS children [97, 98, 102] perhaps because in these high malaria transmission areas robust responses were seen in the majority of children. Other possibilities for discrepancies between these studies include methods of antigen preparation and sample size limitations. The identification of high levels of IgG toward PfEMP-1 and not toward other parasite antigens suggests that the enhanced humoral immune response in HbAS individuals may be directed at proteins on the surface of the infected red blood cell. This phenomenon may be due to increased splenic uptake of infected red blood cells in HbAS individuals and therefore improved presentation of surface antigens. In addition, protection against high parasite densities seen in younger ages [26, 29, 30, 35, 41, 46] may improve the development of acquired immunity, as parasitaemia may interfere with development of effective immune memory [103]. Higher levels of antibodies to PfEMP-1 may mediate protection in HbAS individuals via enhanced opsonization and phagocytosis of infected red blood cells, or through destabilization of cytoadherence. Accelerated acquisition of antibodies to PfEMP-1 may therefore underly the age-dependent increase in protective effects of HbAS found in three studies [15, 35, 46]. Alternatively, or in conjunction, antibodies to PfEMP-1 may be more effective in HbAS than in HbAA children due to reduced or altered surface expression of PfEMP-1 levels in the former.

Modulation of immunopathogenesis

In addition to direct antiparasitic effects, HbAS may confer protection against severe malaria by limiting pathogenesis independent of effects on pathogen load. A recent study using a well-established mouse model of sickle cell disease implicated increased levels of hemeoxygenase-1 (HO-1), an enzyme that breaks down free haem, in the mechanism of protection against cerebral malaria [104]. The authors concluded that sickle haemoglobin suppresses the pathogenesis of cerebral malaria by inducing the expression of HO-1 and preventing the accumulation of cytotoxic-free haem, limiting subsequent tissue damage after Plasmodium berghei infection. They also reported decreased expansion of cytotoxic CD8 T cells that they hypothesized may also contribute to protection against severe disease. Protection was shown to be present irrespective of parasite density. Although the majority of human studies have shown that HbAS is, in fact, associated with lower parasite densities during symptomatic malaria [26, 2931, 33, 46] data from this mouse model suggest that modulation of pathogenic host inflammatory responses may be an additional mechanism of protection against severe disease. Figure 1 summarizes a number of these potential mechanisms.

Figure 1
figure 1

Model of mechanisms by which sickle cell trait may protect against malaria.

Conclusions

It is likely that both biochemical and immune mechanisms contribute to the protection afforded against falciparum malaria by the HbAS genotype. HbS clearly induces biochemical changes in the red blood cell that may affect parasite metabolism and growth. Plasmodium falciparum also likely inflicts oxidative damage on the HbAS red blood cell. Chronic low levels of oxidized haem in sickled red blood cells may induce HO-1, leading to host tolerance in severe disease. Oxidized haem may also interfere with the formation of an actin cytoskeleton, leading to altered PfEMP-1 expression on the infected red blood cell surface membrane and reduced cytoadherence. Reduced cytoadherence may lead to decreased endothelial activation and decreased inflammation implicated in the pathogenesis of severe malaria. In addition, it could also lead to increased splenic uptake of infected erythrocytes. Within the spleen, increased antigen presentation may lead to accelerated development of protective immune responses, including antibodies to PfEMP-1, potentially augmented by biochemical protection against high parasite densities that might otherwise dampen an effective response. Higher levels of antibodies directed against PfEMP-1 and possibly other surface proteins may enhance opsonization and phagocytosis of infected red blood cells and further destabilize the cytoadherence properties of infected red blood cells. While there are some data to support all of these mechanisms, it is still unclear which ones are relevant in vivo. Together, however, it is clear that relevant mechanisms lead to better control of parasitaemia in HbAS children and protect against both uncomplicated and severe malaria. Improved understanding of the mechanisms of protection derived from this single point mutation will give further insight into the host-parasite relationship including how P. falciparum interacts with the human immune system.

Abbreviations

AMA1:

Apical membrane antigen 1 (AMA1)

CSP:

Circumsporozoite protein

EBA175:

Erythrocyte binding antigen 175

GLURP:

Glutamate-rich protein

HbAA:

Normal adult haemoglobin

HbAS:

Sickle cell trait

HbC:

Haemoglobin C

HbS:

Sickle haemoglobin

HO-1:

Hemeoxygenase-1

MSP1:

Merozoite surface protein 1

MSP1:

Merozoite surface protein 2

PfEMP-1:

Plasmodium falciparum erythrocyte membrane protein 1

PSE:

Parasite schizont extract

RESA:

Pf155/ring infected erythrocyte surface antigen.

References

  1. Williams TN: Human red blood cell polymorphisms and malaria. Curr Opin Microbiol. 2006, 9: 388-394.

    CAS  PubMed  Google Scholar 

  2. Kwiatkowski DP, Luoni G: Host genetic factors in resistance and susceptibility to malaria. Parassitologia. 2006, 48: 450-467.

    PubMed  Google Scholar 

  3. Mackinnon MJ, Mwangi TW, Snow RW, Marsh K, Williams TN: Heritability of malaria in Africa. PLoS Med. 2005, 2: e340-

    PubMed Central  PubMed  Google Scholar 

  4. Taylor SM, Cerami C, Fairhurst RM: Hemoglobinopathies: slicing the Gordian knot of Plasmodium falciparum malaria pathogenesis. PLoS Pathog. 2013, 9: e1003327-

    PubMed Central  CAS  PubMed  Google Scholar 

  5. Piel FB, Patil AP, Howes RE, Nyangiri OA, Gething PW, Williams TN, Weatherall DJ, Hay SI: Global distribution of the sickle cell gene and geographical confirmation of the malaria hypothesis. Nat Commun. 2010, 1: 104-

    PubMed Central  PubMed  Google Scholar 

  6. Beet E: Sickle cell disease in the Balovale District of Northern Rhodesia. East Afr Med J. 1946, 23: 75-86.

    CAS  PubMed  Google Scholar 

  7. Allison A: Protection afforded by sickle-cell trait against subtertian malareal infection. Br Med J. 1954, 1: 290-294.

    PubMed Central  CAS  PubMed  Google Scholar 

  8. Brain P: The sickle cell trait; its clinical significance. S Afr Med J. 1952, 26: 925-928.

    CAS  PubMed  Google Scholar 

  9. Ingram VM: Abnormal human haemoglobins. III. The chemical difference between normal and sickle cell haemoglobins. Biochim Biophys Acta. 1959, 36: 402-411.

    CAS  PubMed  Google Scholar 

  10. Bunn HF: Pathogenesis and treatment of sickle cell disease. N Engl J Med. 1997, 337: 762-769.

    CAS  PubMed  Google Scholar 

  11. Haldane JBS: Disease and evolution. Ric Sci Suppl. 1949, 19: 68-76.

    Google Scholar 

  12. Gilles HM, Fletcher KA, Hendrickse RG, Lindner R, Reddy S, Allan N: Glucose-6-phosphate-dehydrogenase deficiency, sickling, and malaria in African children in South Western Nigeria. Lancet. 1967, 1: 138-140.

    CAS  PubMed  Google Scholar 

  13. Hill AV, Allsopp CE, Kwiatkowski D, Anstey NM, Twumasi P, Rowe PA, Bennett S, Brewster D, McMichael AJ, Greenwood BM: Common west African HLA antigens are associated with protection from severe malaria. Nature. 1991, 352: 595-600.

    CAS  PubMed  Google Scholar 

  14. May J, Evans JA, Timmann C, Ehmen C, Busch W, Thye T, Agbenyega T, Horstmann RD: Hemoglobin variants and disease manifestations in severe falciparum malaria. JAMA. 2007, 297: 2220-2226.

    CAS  PubMed  Google Scholar 

  15. Williams TN, Mwangi TW, Roberts DJ, Alexander ND, Weatherall DJ, Wambua S, Kortok M, Snow RW, Marsh K: An immune basis for malaria protection by the sickle cell trait. PLoS Med. 2005, 2: e128-

    PubMed Central  PubMed  Google Scholar 

  16. Jallow M, Teo YY, Small KS, Rockett KA, Deloukas P, Clark TG, Kivinen K, Bojang KA, Conway DJ, Pinder M, Sirugo G, Sisay-Joof F, Usen S, Auburn S, Bumpstead SJ, Campino S, Coffey A, Dunham A, Fry AE, Green A, Gwilliam R, Hunt SE, Inouye M, Jeffreys AE, Mendy A, Palotie A, Potter S, Ragoussis J, Rogers J, Rowlands K: Genome-wide and fine-resolution association analysis of malaria in West Africa. Nat Genet. 2009, 41: 657-665.

    PubMed Central  CAS  PubMed  Google Scholar 

  17. Taylor SM, Parobek CM, Fairhurst RM: Haemoglobinopathies and the clinical epidemiology of malaria: a systematic review and meta-analysis. Lancet Infect Dis. 2012, 12: 457-468.

    PubMed Central  PubMed  Google Scholar 

  18. Wambua S, Mwangi TW, Kortok M, Uyoga SM, Macharia AW, Mwacharo JK, Weatherall DJ, Snow RW, Marsh K, Williams TN: The effect of alpha + −thalassaemia on the incidence of malaria and other diseases in children living on the coast of Kenya. PLoS Med. 2006, 3: e158-

    PubMed Central  PubMed  Google Scholar 

  19. Williams TN, Wambua S, Uyoga S, Macharia A, Mwacharo JK, Newton CRJC, Maitland K: Both heterozygous and homozygous alpha + thalassemias protect against severe and fatal Plasmodium falciparum malaria on the coast of Kenya. Blood. 2005, 106: 368-371.

    CAS  PubMed  Google Scholar 

  20. Allen SJ, O’Donnell A, Alexander ND, Alpers MP, Peto TE, Clegg JB, Weatherall DJ: alpha + −Thalassemia protects children against disease caused by other infections as well as malaria. Proc Natl Acad Sci USA. 1997, 94: 14736-14741.

    PubMed Central  CAS  PubMed  Google Scholar 

  21. Mockenhaupt FP, Ehrhardt S, Gellert S, Otchwemah RN, Dietz E, Anemana SD, Bienzle U: Alpha(+)-thalassemia protects African children from severe malaria. Blood. 2004, 104: 2003-2006.

    CAS  PubMed  Google Scholar 

  22. Krause MA, Diakite SAS, Lopera-Mesa TM, Amaratunga C, Arie T, Traore K, Doumbia S, Konate D, Keefer JR, Diakite M, Fairhurst RM: α-Thalassemia impairs the cytoadherence of Plasmodium falciparum-infected erythrocytes. PLoS One. 2012, 7: e37214-

    PubMed Central  CAS  PubMed  Google Scholar 

  23. Rosanas-Urgell A, Lin E, Manning L, Rarau P, Laman M, Senn N, Grimberg BT, Tavul L, Stanisic DI, Robinson LJ, Aponte JJ, Dabod E, Reeder JC, Siba P, Zimmerman PA, Davis TME, King CL, Michon P, Mueller I: Reduced risk of Plasmodium vivax malaria in Papua New Guinean children with Southeast Asian ovalocytosis in two cohorts and a case–control study. PLoS Med. 2012, 9: e1001305-

    PubMed Central  CAS  PubMed  Google Scholar 

  24. Hutagalung R, Wilairatana P, Looareesuwan S, Brittenham GM, Aikawa M, Gordeuk VR: Influence of hemoglobin E trait on the severity of Falciparum malaria. J Infect Dis. 1999, 179: 283-286.

    CAS  PubMed  Google Scholar 

  25. Chotivanich K, Udomsangpetch R, Pattanapanyasat K, Chierakul W, Simpson J, Looareesuwan S, White N: Hemoglobin E: a balanced polymorphism protective against high parasitemias and thus severe P falciparum malaria. Blood. 2002, 100: 1172-1176.

    CAS  PubMed  Google Scholar 

  26. Mockenhaupt FP, Ehrhardt S, Cramer JP, Otchwemah RN, Anemana SD, Goltz K, Mylius F, Dietz E, Eggelte TA, Bienzle U: Hemoglobin C and resistance to severe malaria in Ghanaian children. J Infect Dis. 2004, 190: 1006-1009.

    CAS  PubMed  Google Scholar 

  27. Modiano D, Luoni G, Sirima BS, Simporé J, Verra F, Konaté A, Rastrelli E, Olivieri A, Calissano C, Paganotti GM, D’Urbano L, Sanou I, Sawadogo A, Modiano G, Coluzzi M: Haemoglobin C protects against clinical Plasmodium falciparum malaria. Nature. 2001, 414: 305-308.

    CAS  PubMed  Google Scholar 

  28. Agarwal A, Guindo A, Cissoko Y, Taylor JG, Coulibaly D, Koné A, Kayentao K, Djimde A, Plowe CV, Doumbo O, Wellems TE, Diallo D: Hemoglobin C associated with protection from severe malaria in the Dogon of Mali, a West African population with a low prevalence of hemoglobin S. Blood. 2000, 96: 2358-2363.

    CAS  PubMed  Google Scholar 

  29. Williams TN, Mwangi TW, Wambua S, Alexander ND, Kortok M, Snow RW, Marsh K: Sickle cell trait and the risk of Plasmodium falciparum malaria and other childhood diseases. J Infect Dis. 2005, 192: 178-186.

    PubMed Central  PubMed  Google Scholar 

  30. Aidoo M, Terlouw DJ, Kolczak MS, McElroy PD, ter Kuile FO, Kariuki S, Nahlen BL, Lal AA, Udhayakumar V: Protective effects of the sickle cell gene against malaria morbidity and mortality. Lancet. 2002, 359: 1311-1312.

    CAS  PubMed  Google Scholar 

  31. Marsh K, Otoo L, Hayes RJ, Carson DC, Greenwood BM: Antibodies to blood stage antigens of Plasmodium falciparum in rural Gambians and their relation to protection against infection. Trans R Soc Trop Med Hyg. 1989, 83: 293-303.

    CAS  PubMed  Google Scholar 

  32. Clark TD, Greenhouse B, Njama-Meya D, Nzarubara B, Maiteki-Sebuguzi C, Staedke SG, Seto E, Kamya MR, Rosenthal PJ, Dorsey G: Factors determining the heterogeneity of malaria incidence in children in Kampala, Uganda. J Infect Dis. 2008, 198: 393-400.

    PubMed  Google Scholar 

  33. Crompton PD, Traore B, Kayentao K, Doumbo S, Ongoiba A, Diakite SAS, Krause MA, Doumtabe D, Kone Y, Weiss G, Huang C-Y, Doumbia S, Guindo A, Fairhurst RM, Miller LH, Pierce SK, Doumbo OK: Sickle cell trait is associated with a delayed onset of malaria: implications for time-to-event analysis in clinical studies of malaria. J Infect Dis. 2008, 198: 1265-1275.

    PubMed Central  PubMed  Google Scholar 

  34. Kreuels B, Kreuzberg C, Kobbe R, Ayim-Akonor M, Apiah-Thompson P, Thompson B, Ehmen C, Adjei S, Langefeld I, Adjei O, May J: Differing effects of HbS and HbC traits on uncomplicated falciparum malaria, anemia, and child growth. Blood. 2010, 115: 4551-4558.

    CAS  PubMed  Google Scholar 

  35. Gong L, Maiteki-Sebuguzi C, Rosenthal PJ, Hubbard AE, Drakeley CJ, Dorsey G, Greenhouse B: Evidence for both innate and acquired mechanisms of protection from Plasmodium falciparum in children with sickle cell trait. Blood. 2012, 119: 3808-3814.

    PubMed Central  CAS  PubMed  Google Scholar 

  36. Parikh S, Dorsey G, Rosenthal PJ: Host polymorphisms and the incidence of malaria in Ugandan children. Am J Trop Med Hyg. 2004, 71: 750-753.

    CAS  PubMed  Google Scholar 

  37. Motulsky AG, Vandepitte J, Fraser GR: Population genetic studies in the Congo. I. Glucose-6-phosphate dehydrogenase deficiency, hemoglobin S, and malaria. Am J Hum Genet. 1966, 18: 514-537.

    PubMed Central  CAS  PubMed  Google Scholar 

  38. Fleming AF, Storey J, Molineaux L, Iroko EA, Attai ED: Abnormal haemoglobins in the Sudan savanna of Nigeria. I. Prevalence of haemoglobins and relationships between sickle cell trait, malaria and survival. Ann Trop Med Parasitol. 1979, 73: 161-172.

    CAS  PubMed  Google Scholar 

  39. Jakobsen PH, Riley EM, Allen SJ, Larsen SO, Bennett S, Jepsen S, Greenwood BM: Differential antibody response of Gambian donors to soluble Plasmodium falciparum antigens. Trans R Soc Trop Med Hyg. 1991, 85: 26-32.

    CAS  PubMed  Google Scholar 

  40. Migot-Nabias F, Pelleau S, Watier L, Guitard J, Toly C, De Araujo C, Ngom MI, Chevillard C, Gaye O, Garcia A: Red blood cell polymorphisms in relation to Plasmodium falciparum asymptomatic parasite densities and morbidity in Senegal. Microbes Infect. 2006, 8: 2352-2358.

    CAS  PubMed  Google Scholar 

  41. Danquah I, Ziniel P, Eggelte TA, Ehrhardt S, Mockenhaupt FP: Influence of haemoglobins S and C on predominantly asymptomatic Plasmodium infections in northern Ghana. Trans R Soc Trop Med Hyg. 2010, 104: 713-719.

    CAS  PubMed  Google Scholar 

  42. Makani J, Komba AN, Cox SE, Oruo J, Mwamtemi K, Kitundu J, Magesa P, Rwezaula S, Meda E, Mgaya J, Pallangyo K, Okiro E, Muturi D, Newton CR, Fegan G, Marsh K, Williams TN: Malaria in patients with sickle cell anemia: burden, risk factors, and outcome at the outpatient clinic and during hospitalization. Blood. 2010, 115: 215-220.

    PubMed Central  CAS  PubMed  Google Scholar 

  43. Billo MA, Johnson ES, Doumbia SO, Poudiougou B, Sagara I, Diawara SI, Diakité M, Diallo M, Doumbo OK, Tounkara A, Rice J, James MA, Krogstad DJ: Sickle cell trait protects against Plasmodium falciparum infection. Am J Epidemiol. 2012, 176 (Suppl 7): S175-185.

    PubMed Central  PubMed  Google Scholar 

  44. Ntoumi F, Rogier C, Dieye A, Trape JF, Millet P, Mercereau-Puijalon O: Imbalanced distribution of Plasmodium falciparum MSP-1 genotypes related to sickle-cell trait. Mol Med. 1997, 3: 581-592.

    PubMed Central  CAS  PubMed  Google Scholar 

  45. Konaté L, Zwetyenga J, Rogier C, Bischoff E, Fontenille D, Tall A, Spiegel A, Trape JF, Mercereau-Puijalon O: Variation of Plasmodium falciparum msp1 block 2 and msp2 allele prevalence and of infection complexity in two neighbouring Senegalese villages with different transmission conditions. Trans R Soc Trop Med Hyg. 1999, 93 (Suppl 1): 21-28.

    PubMed  Google Scholar 

  46. Guggenmoos-Holzmann I, Bienzle U, Luzzatto L: Plasmodium falciparum malaria and human red cells. II. Red cell genetic traits and resistance against malaria. Int J Epidemiol. 1981, 10: 16-22.

    CAS  PubMed  Google Scholar 

  47. Luzzatto L, Nwachuku-Jarrett ES, Reddy S: Increased sickling of parasitised erythrocytes as mechanism of resistance against malaria in the sickle-cell trait. Lancet. 1970, 1: 319-321.

    CAS  PubMed  Google Scholar 

  48. Roth EF, Friedman M, Ueda Y, Tellez I, Trager W, Nagel RL: Sickling rates of human AS red cells infected in vitro with Plasmodium falciparum malaria. Science. 1978, 202: 650-652.

    PubMed  Google Scholar 

  49. Friedman MJ: Erythrocytic mechanism of sickle cell resistance to malaria. Proc Natl Acad Sci USA. 1978, 75: 1994-1997.

    PubMed Central  CAS  PubMed  Google Scholar 

  50. Pasvol G, Weatherall DJ, Wilson RJ: Cellular mechanism for the protective effect of haemoglobin S against P. falciparum malaria. Nature. 1978, 274: 701-703.

    CAS  PubMed  Google Scholar 

  51. Orjih AU, Chevli R, Fitch CD: Toxic heme in sickle cells: an explanation for death of malaria parasites. Am J Trop Med Hyg. 1985, 34: 223-227.

    CAS  PubMed  Google Scholar 

  52. Ginsburg H, Handeli S, Friedman S, Gorodetsky R, Krugliak M: Effects of red blood cell potassium and hypertonicity on the growth of Plasmodium falciparum in culture. Z Parasitenkd. 1986, 72: 185-199.

    CAS  PubMed  Google Scholar 

  53. Ayi K, Turrini F, Piga A, Arese P: Enhanced phagocytosis of ring-parasitized mutant erythrocytes: a common mechanism that may explain protection against falciparum malaria in sickle trait and beta-thalassemia trait. Blood. 2004, 104: 3364-3371.

    CAS  PubMed  Google Scholar 

  54. LaMonte G, Philip N, Reardon J, Lacsina JR, Majoros W, Chapman L, Thornburg CD, Telen MJ, Ohler U, Nicchitta CV, Haystead T, Chi J-T: Translocation of sickle cell erythrocyte microRNAs into Plasmodium falciparum inhibits parasite translation and contributes to malaria resistance. Cell Host Microbe. 2012, 12: 187-199.

    PubMed Central  CAS  PubMed  Google Scholar 

  55. Trager W, Rudzinska MA, Bradbury PC: The fine structure of Plasmodium falciparum and its host erythrocytes in natural malarial infections in man. Bull World Health Organ. 1966, 35: 883-885.

    PubMed Central  CAS  PubMed  Google Scholar 

  56. Udomsangpetch R, Wåhlin B, Carlson J, Berzins K, Torii M, Aikawa M, Perlmann P, Wahlgren M: Plasmodium falciparum-infected erythrocytes form spontaneous erythrocyte rosettes. J Exp Med. 1989, 169: 1835-1840.

    CAS  PubMed  Google Scholar 

  57. Carlson J, Helmby H, Hill AV, Brewster D, Greenwood BM, Wahlgren M: Human cerebral malaria: association with erythrocyte rosetting and lack of anti-rosetting antibodies. Lancet. 1990, 336: 1457-1460.

    CAS  PubMed  Google Scholar 

  58. Treutiger CJ, Hedlund I, Helmby H, Carlson J, Jepson A, Twumasi P, Kwiatkowski D, Greenwood BM, Wahlgren M: Rosette formation in Plasmodium falciparum isolates and anti-rosette activity of sera from Gambians with cerebral or uncomplicated malaria. Am J Trop Med Hyg. 1992, 46: 503-510.

    CAS  PubMed  Google Scholar 

  59. Aikawa M, Iseki M, Barnwell JW, Taylor D, Oo MM, Howard RJ: The pathology of human cerebral malaria. Am J Trop Med Hyg. 1990, 43: 30-37.

    CAS  PubMed  Google Scholar 

  60. Cholera R, Brittain NJ, Gillrie MR, Lopera-Mesa TM, Diakité SAS, Arie T, Krause MA, Guindo A, Tubman A, Fujioka H, Diallo DA, Doumbo OK, Ho M, Wellems TE, Fairhurst RM: Impaired cytoadherence of Plasmodium falciparum-infected erythrocytes containing sickle hemoglobin. Proc Natl Acad Sci USA. 2008, 105: 991-996.

    PubMed Central  CAS  PubMed  Google Scholar 

  61. Ockenhouse CF, Tandon NN, Magowan C, Jamieson GA, Chulay JD: Identification of a platelet membrane glycoprotein as a falciparum malaria sequestration receptor. Science. 1989, 243: 1469-1471.

    CAS  PubMed  Google Scholar 

  62. Ockenhouse CF, Shear HL: Malaria-induced lymphokines: stimulation of macrophages for enhanced phagocytosis. Infect Immun. 1983, 42: 733-739.

    PubMed Central  CAS  PubMed  Google Scholar 

  63. 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.

    CAS  PubMed  Google Scholar 

  64. Yazdani SS, Mukherjee P, Chauhan VS, Chitnis CE: Immune responses to asexual blood-stages of malaria parasites. Curr Mol Med. 2006, 6: 187-203.

    CAS  PubMed  Google Scholar 

  65. Kaul DK, Roth EF, Nagel RL, Howard RJ, Handunnetti SM: Rosetting of Plasmodium falciparum-infected red blood cells with uninfected red blood cells enhances microvascular obstruction under flow conditions. Blood. 1991, 78: 812-819.

    CAS  PubMed  Google Scholar 

  66. Turner GD, Morrison H, Jones M, Davis TM, Looareesuwan S, Buley ID, Gatter KC, Newbold CI, Pukritayakamee S, Nagachinta B: An immunohistochemical study of the pathology of fatal malaria. Evidence for widespread endothelial activation and a potential role for intercellular adhesion molecule-1 in cerebral sequestration. Am J Pathol. 1994, 145: 1057-1069.

    PubMed Central  CAS  PubMed  Google Scholar 

  67. Miller LH, Baruch DI, Marsh K, Doumbo OK: The pathogenic basis of malaria. Nature. 2002, 415: 673-679.

    CAS  PubMed  Google Scholar 

  68. Dondorp AM, Pongponratn E, White NJ: Reduced microcirculatory flow in severe falciparum malaria: pathophysiology and electron-microscopic pathology. Acta Trop. 2004, 89: 309-317.

    PubMed  Google Scholar 

  69. Fairhurst RM, Baruch DI, Brittain NJ, Ostera GR, Wallach JS, Hoang HL, Hayton K, Guindo A, Makobongo MO, Schwartz OM, Tounkara A, Doumbo OK, Diallo DA, Fujioka H, Ho M, Wellems TE: Abnormal display of PfEMP-1 on erythrocytes carrying haemoglobin C may protect against malaria. Nature. 2005, 435: 1117-1121.

    CAS  PubMed  Google Scholar 

  70. Cyrklaff M, Sanchez CP, Kilian N, Bisseye C, Simpore J, Frischknecht F, Lanzer M: Hemoglobins S and C interfere with actin remodeling in Plasmodium falciparum-infected erythrocytes. Science. 2011, 334: 1283-1286.

    CAS  PubMed  Google Scholar 

  71. Cappadoro M, Giribaldi G, O’Brien E, Turrini F, Mannu F, Ulliers D, Simula G, Luzzatto L, Arese P: Early phagocytosis of glucose-6-phosphate dehydrogenase (G6PD)-deficient erythrocytes parasitized by Plasmodium falciparum may explain malaria protection in G6PD deficiency. Blood. 1998, 92: 2527-2534.

    CAS  PubMed  Google Scholar 

  72. Arese P, Turrini F, Schwarzer E: Band 3/complement-mediated recognition and removal of normally senescent and pathological human erythrocytes. Cell Physiol Biochem. 2005, 16: 133-146.

    CAS  PubMed  Google Scholar 

  73. Lang PA, Kasinathan RS, Brand VB, Duranton C, Lang C, Koka S, Shumilina E, Kempe DS, Tanneur V, Akel A, Lang KS, Foller M, Kun JFJ, Kremsner PG, Wesselborg S, Laufer S, Clemen CS, Herr C, Noegel AA, Wieder T, Gulbins E, Lang F, Huber SM: Accelerated clearance of Plasmodium-infected erythrocytes in sickle cell trait and annexin-A7 deficiency. Cell Physiol Biochem. 2009, 24: 415-428.

    CAS  PubMed  Google Scholar 

  74. Tanaka Y, Schroit AJ: Insertion of fluorescent phosphatidylserine into the plasma membrane of red blood cells. Recognition by autologous macrophages. J Biol Chem. 1983, 258: 11335-11343.

    CAS  PubMed  Google Scholar 

  75. Schroit AJ, Madsen JW, Tanaka Y: In vivo recognition and clearance of red blood cells containing phosphatidylserine in their plasma membranes. J Biol Chem. 1985, 260: 5131-5138.

    CAS  PubMed  Google Scholar 

  76. Boas FE, Forman L, Beutler E: Phosphatidylserine exposure and red cell viability in red cell aging and in hemolytic anemia. Proc Natl Acad Sci USA. 1998, 95: 3077-3081.

    PubMed Central  CAS  PubMed  Google Scholar 

  77. Hoffmann PR, de Cathelineau AM, Ogden CA, Leverrier Y, Bratton DL, Daleke DL, Ridley AJ, Fadok VA, Henson PM: Phosphatidylserine (PS) induces PS receptor-mediated macropinocytosis and promotes clearance of apoptotic cells. J Cell Biol. 2001, 155: 649-659.

    PubMed Central  CAS  PubMed  Google Scholar 

  78. Urban BC, Shafi MJ, Cordery DV, Macharia A, Lowe B, Marsh K, Williams TN: Frequencies of peripheral blood myeloid cells in healthy Kenyan children with alpha + thalassemia and the sickle cell trait. Am J Trop Med Hyg. 2006, 74: 578-584.

    PubMed Central  CAS  PubMed  Google Scholar 

  79. Jepsen S, Andersen BJ: Immunoadsorbent isolation of antigens from the culture medium of in vitro cultivated Plasmodium falciparum. Acta Pathol Microbiol Scand C. 1981, 89: 99-103.

    CAS  PubMed  Google Scholar 

  80. Bygbjerg IC, Jepsen S, Theander TG, Odum N: Specific proliferative response of human lymphocytes to purified soluble antigens from Plasmodium falciparum in vitro cultures and to antigens from malaria patients’ sera. Clin Exp Immunol. 1985, 59: 421-426.

    PubMed Central  CAS  PubMed  Google Scholar 

  81. Riley EM, Andersson G, Otoo LN, Jepsen S, Greenwood BM: Cellular immune responses to Plasmodium falciparum antigens in Gambian children during and after an acute attack of falciparum malaria. Clin Exp Immunol. 1988, 73: 17-22.

    PubMed Central  CAS  PubMed  Google Scholar 

  82. Bayoumi RA, Abu-Zeid YA, Abdulhadi NH, Saeed BO, Theander TG, Hviid L, Ghalib HW, Nugud AH, Jepsen S, Jensen JB: Cell-mediated immune responses to Plasmodium falciparum purified soluble antigens in sickle-cell trait subjects. Immunol Lett. 1990, 25: 243-249.

    CAS  PubMed  Google Scholar 

  83. Abu-Zeid YA, Abdulhadi NH, Hviid L, Theander TG, Saeed BO, Jepsen S, Jensen JB, Bayoumi RA: Lymphoproliferative responses to Plasmodium falciparum antigens in children with and without the sickle cell trait. Scand J Immunol. 1991, 34: 237-242.

    CAS  PubMed  Google Scholar 

  84. Abu-Zeid YA, Theander TG, Abdulhadi NH, Hviid L, Saeed BO, Jepsen S, Jensen JB, Bayoumi RA: Modulation of the cellular immune response during Plasmodium falciparum infections in sickle cell trait individuals. Clin Exp Immunol. 1992, 88: 112-118.

    PubMed Central  CAS  PubMed  Google Scholar 

  85. Le Hesran JY, Personne I, Personne P, Fievet N, Dubois B, Beyemé M, Boudin C, Cot M, Deloron P: Longitudinal study of Plasmodium falciparum infection and immune responses in infants with or without the sickle cell trait. Int J Epidemiol. 1999, 28: 793-798.

    CAS  PubMed  Google Scholar 

  86. Brasseur P, Agrapart M, Ballet JJ, Druilhe P, Warrell MJ, Tharavanij S: Impaired cell-mediated immunity in Plasmodium falciparum-infected patients with high-parasitemia and cerebral malaria. Clin Immunol Immunopathol. 1983, 27: 38-50.

    CAS  PubMed  Google Scholar 

  87. Ho M, Webster HK, Looareesuwan S, Supanaranond W, Phillips RE, Chanthavanich P, Warrell DA: Antigen-specific immunosuppression in human malaria due to Plasmodium falciparum. J Infect Dis. 1986, 153: 763-771.

    CAS  PubMed  Google Scholar 

  88. Theander TG, Bygbjerg IC, Andersen BJ, Jepsen S, Kharazmi A, Odum N: Suppression of parasite-specific response in Plasmodium falciparum malaria. A longitudinal study of blood mononuclear cell proliferation and subset composition. Scand J Immunol. 1986, 24: 73-81.

    CAS  PubMed  Google Scholar 

  89. Angulo I, Fresno M: Cytokines in the pathogenesis of and protection against malaria. Clin Diagn Lab Immunol. 2002, 9: 1145-1152.

    PubMed Central  CAS  PubMed  Google Scholar 

  90. Edozien JC, Boyo AE, Morley DC: The relationship of serum gamma-globulin concentration to malaria and sickling. J Clin Pathol. 1960, 13: 118-123.

    PubMed Central  CAS  PubMed  Google Scholar 

  91. Cornille-Brøgger R, Fleming AF, Kagan I, Matsushima T, Molineaux L: Abnormal haemoglobins in the Sudan savanna of Nigeria. II. Immunological response to malaria in normals and subjects with sickle cell trait. Ann Trop Med Parasitol. 1979, 73: 173-183.

    PubMed  Google Scholar 

  92. Dziegiel M, Rowe P, Bennett S, Allen SJ, Olerup O, Gottschau A, Borre M, Riley EM: Immunoglobulin M and G antibody responses to Plasmodium falciparum glutamate-rich protein: correlation with clinical immunity in Gambian children. Infect Immun. 1993, 61: 103-108.

    PubMed Central  CAS  PubMed  Google Scholar 

  93. Aluoch JR: Higher resistance to Plasmodium falciparum infection in patients with homozygous sickle cell disease in western Kenya. Trop Med Int Health. 1997, 2: 568-571.

    CAS  PubMed  Google Scholar 

  94. Aucan C, Traoré Y, Tall F, Nacro B, Traoré-Leroux T, Fumoux F, Rihet P: High immunoglobulin G2 (IgG2) and low IgG4 levels are associated with human resistance to Plasmodium falciparum malaria. Infect Immun. 2000, 68: 1252-1258.

    PubMed Central  CAS  PubMed  Google Scholar 

  95. Luty AJ, Ulbert S, Lell B, Lehman L, Schmidt-Ott R, Luckner D, Greve B, Matousek P, Schmid D, Herbich K, Dubois B, Deloron P, Kremsner PG: Antibody responses to Plasmodium falciparum: evolution according to the severity of a prior clinical episode and association with subsequent reinfection. Am J Trop Med Hyg. 2000, 62: 566-572.

    CAS  PubMed  Google Scholar 

  96. Miura K, Diakite M, Diouf A, Doumbia S, Konate D, Keita AS, Moretz SE, Tullo G, Zhou H, Lopera-Mesa TM, Anderson JM, Fairhurst RM, Long CA: Relationship between malaria incidence and IgG levels to Plasmodium falciparum merozoite antigens in Malian children: impact of hemoglobins S and C. PLoS ONE. 2013, 8: e60182-

    PubMed Central  CAS  PubMed  Google Scholar 

  97. Tan X, Traore B, Kayentao K, Ongoiba A, Doumbo S, Waisberg M, Doumbo OK, Felgner PL, Fairhurst RM, Crompton PD: Hemoglobin S and C heterozygosity enhances neither the magnitude nor breadth of antibody responses to a diverse array of Plasmodium falciparum antigens. J Infect Dis. 2011, 204: 1750-1761.

    PubMed Central  CAS  PubMed  Google Scholar 

  98. Verra F, Simpore J, Warimwe GM, Tetteh KK, Howard T, Osier FHA, Bancone G, Avellino P, Blot I, Fegan G, Bull PC, Williams TN, Conway DJ, Marsh K, Modiano D: Haemoglobin C and S role in acquired immunity against Plasmodium falciparum malaria. PLoS ONE. 2007, 2: e978-

    PubMed Central  PubMed  Google Scholar 

  99. Sarr JB, Pelleau S, Toly C, Guitard J, Konaté L, Deloron P, Garcia A, Migot-Nabias F: Impact of red blood cell polymorphisms on the antibody response to Plasmodium falciparum in Senegal. Microbes Infect. 2006, 8: 1260-1268.

    CAS  PubMed  Google Scholar 

  100. Diatta A-M, Marrama L, Tall A, Trape J-F, Dieye A, Garraud O, Mercereau-Puijalon O, Perraut R: Relationship of binding of immunoglobulin G to Plasmodium falciparum-infected erythrocytes with parasite endemicity and antibody responses to conserved antigen in immune individuals. Clin Diagn Lab Immuno. 2004, 11: 6-11.

    CAS  Google Scholar 

  101. Cabrera G, Cot M, Migot-Nabias F, Kremsner PG, Deloron P, Luty AJF: The sickle cell trait is associated with enhanced immunoglobulin G antibody responses to Plasmodium falciparum variant surface antigens. J Infect Dis. 2005, 191: 1631-1638.

    CAS  PubMed  Google Scholar 

  102. Allen SJ, Bennett S, Riley EM, Rowe PA, Jakobsen PH, O’Donnell A, Greenwood BM: Morbidity from malaria and immune responses to defined Plasmodium falciparum antigens in children with sickle cell trait in The Gambia. Trans R Soc Trop Med Hyg. 1992, 86: 494-498.

    CAS  PubMed  Google Scholar 

  103. Langhorne J, Ndungu FM, Sponaas A-M, Marsh K: Immunity to malaria: more questions than answers. Nat Immunol. 2008, 9: 725-732.

    CAS  PubMed  Google Scholar 

  104. Ferreira A, Marguti I, Bechmann I, Jeney V, Chora A, Palha NR, Rebelo S, Henri A, Beuzard Y, Soares MP: Sickle hemoglobin confers tolerance to Plasmodium infection. Cell. 2011, 145: 398-409.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was funded, in part, by the National Institutes of Health, National Institute of Allergy and Infectious Diseases (K23 AI076614). BG is a recipient of a Doris Duke Clinical Scientist Development Award (2011043) and supported, in part, by the Doris Duke Charitable Foundation. This study was supported in part by the NIAID program for International Centers of Excellence in Malaria Research (U19 A1089674).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Bryan Greenhouse.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

LG performed a comprehensive review of the primary literature and drafted the manuscript. SP, PJR and BG reviewed and identified additional primary literature, directed the organization of the manuscript and edited the manuscript. All authors read and approved the final version of the manuscript.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Gong, L., Parikh, S., Rosenthal, P.J. et al. Biochemical and immunological mechanisms by which sickle cell trait protects against malaria. Malar J 12, 317 (2013). https://doi.org/10.1186/1475-2875-12-317

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1475-2875-12-317

Keywords