Hostname: page-component-8448b6f56d-qsmjn Total loading time: 0 Render date: 2024-04-19T02:03:19.155Z Has data issue: false hasContentIssue false

Rosetting revisited: a critical look at the evidence for host erythrocyte receptors in Plasmodium falciparum rosetting

Published online by Cambridge University Press:  16 September 2019

Fiona McQuaid
Affiliation:
Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, School of Biological Sciences, Ashworth Laboratories, Kings Buildings, Charlotte Auerbach Rd, Edinburgh, EH9 3FL, UK
J. Alexandra Rowe*
Affiliation:
Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, School of Biological Sciences, Ashworth Laboratories, Kings Buildings, Charlotte Auerbach Rd, Edinburgh, EH9 3FL, UK
*
Author for correspondence: J. Alexandra Rowe, E-mail: alex.rowe@ed.ac.uk

Abstract

Malaria remains a major cause of mortality in African children, with no adjunctive treatments currently available to ameliorate the severe clinical forms of the disease. Rosetting, the adhesion of infected erythrocytes (IEs) to uninfected erythrocytes, is a parasite phenotype strongly associated with severe malaria, and hence is a potential therapeutic target. However, the molecular mechanisms of rosetting are complex and involve multiple distinct receptor–ligand interactions, with some similarities to the diverse pathways involved in P. falciparum erythrocyte invasion. This review summarizes the current understanding of the molecular interactions that lead to rosette formation, with a particular focus on host uninfected erythrocyte receptors including the A and B blood group trisaccharides, complement receptor one, heparan sulphate, glycophorin A and glycophorin C. There is strong evidence supporting blood group A trisaccharides as rosetting receptors, but evidence for other molecules is incomplete and requires further study. It is likely that additional host erythrocyte rosetting receptors remain to be discovered. A rosette-disrupting low anti-coagulant heparin derivative is being investigated as an adjunctive therapy for severe malaria, and further research into the receptor–ligand interactions underlying rosetting may reveal additional therapeutic approaches to reduce the unacceptably high mortality rate of severe malaria.

Type
Review Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © Cambridge University Press 2019

Introduction

Rosetting is a Plasmodium falciparum infected erythrocyte (IE) adhesion phenotype that is associated with severe malaria in sub-Saharan Africa (summarized in Doumbo et al., Reference Doumbo, Thera, Koné, Raza, Tempest, Lyke, Plowe and Rowe2009). It is a form of cell adhesion in which erythrocytes infected with mature, asexual parasites bind to uninfected erythrocytes to form clusters of cells (Fig. 1). Rosetting is a phenotypically variable property, which is common in parasite isolates collected from severe malaria patients, but infrequent in parasites from uncomplicated malaria cases. For culture-adapted P. falciparum isolates, only a subset of parasite lines can be selected in vitro for the rosetting phenotype, and many of the commonly used laboratory strains such as 3D7, rosette poorly or not at all. The relative rarity of rosetting in culture-adapted parasite lines may explain why rosetting is studied infrequently, despite being a virulence-associated phenotype in clinical isolates.

Fig. 1. Plasmodium falciparum rosetting in an in vitro culture. Rosettes consisting of clusters of infected and uninfected erythrocytes are shown. Inset image shows a single infected erythrocyte (centre) and three adherent uninfected erythrocytes. Images were taken using a Yenway microscope camera on a Leica DM LB2 fluorescent microscope using the ×40 and ×100 (inset) objectives.

Rosetting can contribute to IE sequestration and microvascular congestion, leading to obstruction to blood flow (Kaul et al., Reference Kaul, Roth, Nagel, Howard and Handunnetti1991), one of the major pathological events in severe falciparum malaria contributing to inflammation, tissue damage and organ failure (Miller et al., Reference Miller, Baruch, Marsh and Doumbo2002; White et al., Reference White, Turner, Day and Dondorp2013). Rosetting also causes membrane changes in uninfected erythrocytes that may contribute to phagocytic removal and anaemia (Uyoga et al., Reference Uyoga, Skorokhod, Opiyo, Orori, Williams, Arese and Schwarzer2012). In Africa, high levels of rosetting occur in parasites sampled from severe malaria patients with all clinical types of disease including cerebral malaria (Carlson et al., Reference Carlson, Helmby, Hill, Brewster, Greenwood and Wahlgren1990; Treutiger et al., Reference Treutiger, Hedlund, Helmby, Carlson, Jepson, Twumasi, Kwiatkowski, Greenwood and Wahlgren1992; Ringwald et al., Reference Ringwald, Peyron, Lepers, Rabarison, Rakotomalala, Razanamparany, Rabodonirina, Roux and Le Bras1993; Rowe et al., Reference Rowe, Obeiro, Newbold and Marsh1995; Kun et al., Reference Kun, Schmidt-Ott, Lehman, Lell, Luckner, Greve, Matousek and Kremsner1998; Doumbo et al., Reference Doumbo, Thera, Koné, Raza, Tempest, Lyke, Plowe and Rowe2009), severe malarial anaemia (Newbold et al., Reference Newbold, Warn, Black, Berendt, Craig, Snow, Msobo, Peshu and Marsh1997; Doumbo et al., Reference Doumbo, Thera, Koné, Raza, Tempest, Lyke, Plowe and Rowe2009) and respiratory distress (Warimwe et al., Reference Warimwe, Fegan, Musyoki, Newton, Opiyo, Githinji, Andisi, Menza, Kitsao, Marsh and Bull2012). Rosette-like clusters of cells have been seen in the microvasculature in histological studies of fatal malaria cases (Dondorp et al., Reference Dondorp, Pongponratn and White2004; Barrera et al., Reference Barrera, MacCormick, Czanner, Hiscott, White, Craig, Beare, Culshaw, Zheng, Biddolph, Milner, Kamiza, Molyneux, Taylor and Harding2018). The major Plasmodium species that infect humans are all able to form rosettes (Udomsanpetch et al., Reference Udomsanpetch, Thanikkul, Pukrittayakamee and White1995; Angus et al., Reference Angus, Thanikkul, Silamut, White and Udomsangpetch1996; Chotivanich et al., Reference Chotivanich, Pukrittayakamee, Simpson, White and Udomsangpetch1998; Lowe et al., Reference Lowe, Mosobo and Bull1998). However, the link between severity of disease and rosetting is confined to P. falciparum, possibly due to the unique ability to bind both endothelial cells and uninfected erythrocytes simultaneously (Udomsangpetch et al., Reference Udomsangpetch, Webster, Pattanapanyasat, Pitchayangkul and Thaithong1992; Adams et al., Reference Adams, Kuhnrae, Higgins, Ghumra and Rowe2014), such that P. falciparum rosetting IEs are sequestered and are not seen in peripheral blood. Recently it has been suggested that rosetting may contribute to anaemia in Plasmodium vivax infections (Marín-Menéndez et al., Reference Marín-Menéndez, Bardají, Martínez-Espinosa, Bôtto-Menezes, Lacerda, Ortiz, Cisteró, Piqueras, Felger, Müeller, Ordi, del Portillo, Menéndez, Wahlgren and Mayor2013).

The biological function of rosetting in vivo remains unknown. Rosettes may shield IEs from host immune attack, or close contact with uninfected erythrocytes in rosettes might enhance merozoite invasion (Wahlgren et al., Reference Wahlgren, Carlson, Udomsangpetch and Perlmann1989; Deans and Rowe, Reference Deans and Rowe2006). However, firm evidence to support either of these hypotheses is lacking. Most rosetting parasite isolates form larger, stronger rosettes with blood group A erythrocytes compared to other blood groups (Carlson and Wahlgren, Reference Carlson and Wahlgren1992), and these group A rosettes may shield IEs to reduce antibody binding to parasite variant surface antigens (VSAs) (Moll et al., Reference Moll, Palmkvist, Ch'ng, Kiwuwa and Wahlgren2015). Whether this translates into the reduced clearance of IEs and subsequent higher parasite burdens in vivo is unclear, although some studies have noted a positive correlation between rosetting and parasitaemia (Rowe et al., Reference Rowe, Obiero, Marsh and Raza2002). Another study showed that rosetting does not prevent IgG-mediated phagocytosis of IEs (Stevenson et al., Reference Stevenson, Huda, Jeppesen, Laursen, Rowe, Craig, Streicher, Barfod and Hviid2015a), although experiments were only performed in group O cells. Parasite invasion of erythrocytes is not increased in vitro in rosetting compared to isogenic non-rosetting parasites (Clough et al., Reference Clough, Atilola and Pasvol1998; Deans and Rowe, Reference Deans and Rowe2006; Ribacke et al., Reference Ribacke, Moll, Albrecht, Ahmed Ismail, Normark, Flaberg, Szekely, Hultenby, Persson, Egwang and Wahlgren2013), nor in the presence of larger rosettes (Moll et al., Reference Moll, Palmkvist, Ch'ng, Kiwuwa and Wahlgren2015). However, in vivo studies using splenectomized Saimiri sciureus monkeys demonstrated a 1.5 times higher parasite multiplication rate with rosetting compared to isogenic non-rosetting parasites (Le Scanf et al., Reference Le Scanf, Vigan-Womas, Contamin, Guillotte, Bischoff and Mercereau-Puijalon2008). This suggests either increased invasion or decreased clearance of rosetting parasites in vivo, which requires further investigation.

This review will discuss the molecular mechanisms of rosetting and describe recent advances exploring the potential of rosetting as a therapeutic target in severe P. falciparum malaria. Rosetting is a complex cell adhesion phenotype involving parasite adhesion molecules on the IE surface and host receptors on uninfected erythrocytes (Fig. 2). Current evidence suggests that there are multiple distinct pathways of rosette formation, similar to the diverse pathways involved in merozoite invasion of erythrocytes (Cowman et al., Reference Cowman, Tonkin, Tham and Duraisingh2017). Interestingly, although the parasite molecules that mediate rosetting are different from those involved in merozoite invasion, both sets of proteins have ‘Duffy-Binding-Like’ adhesion domains and many of the same host erythrocyte receptors are used (e.g. glycophorin A, glycophorin C and complement receptor one). The diversity in P. falciparum merozoite invasion pathways is thought to have evolved to allow parasites to successfully establish infections despite host genetic variation and/or development of host antibodies blocking single pathways. The same arguments can be applied to rosetting, and the existence of multiple rosetting pathways suggests that there has been significant selection pressure in favour of the phenotype, and that rosetting somehow improves parasite fitness.

Fig. 2. Parasite-derived adhesion ligands and host receptors that interact to form rosettes. UE, uninfected erythrocyte; IE, infected erythrocyte; GAGs, glycosaminoglycans; HS, heparan sulphate; CS, chondroitin sulphate; CR1, complement receptor 1; GYPA, glycophorin A; GYPC, glycophorin C. Dotted lines represent proposed host receptors for each parasite ligand.

Several recent reviews have discussed the parasite adhesion molecules involved in rosetting (Hviid and Jensen, Reference Hviid and Jensen2015; Wang and Hviid, Reference Wang and Hviid2015; Yam et al., Reference Yam, Niang, Madnani and Preiser2017), so these will not be described in detail here. Briefly, multiple studies have identified members of the VSA family P. falciparum erythrocyte membrane protein one (PfEMP1) as rosette-mediating adhesion molecules (Rowe et al., Reference Rowe, Moulds, Newbold and Miller1997; Vigan-Womas et al., Reference Vigan-Womas, Guillotte, Le Scanf, Igonet, Petres, Juillerat, Badaut, Nato, Schneider, Lavergne, Contamin, Tall, Baril, Bentley and Mercereau-Puijalon2008, Reference Vigan-Womas, Guillotte, Juillerat, Vallieres, Lewit-Bentley, Tall, Baril, Bentley and Mercereau-Puijalon2011; Albrecht et al., Reference Albrecht, Moll, Blomqvist, Normark, Chen and Wahlgren2011; Ghumra et al., Reference Ghumra, Semblat, Ataide, Kifude, Adams, Claessens, Anong, Bull, Fennell, Arman, Amambua-Ngwa, Walther, Conway, Kassambara, Doumbo, Raza and Rowe2012), and recent reports suggest that other VSAs such as RIFIN (Goel et al., Reference Goel, Palmkvist, Moll, Joannin, Lara, Akhouri, Moradi, Öjemalm, Westman, Angeletti, Kjellin, Lehtiö, Blixt, Ideström, Gahmberg, Storry, Hult, Olsson, von Heijne, Nilsson and Wahlgren2015) and STEVOR (Niang et al., Reference Niang, Bei, Madnani, Pelly, Dankwa, Kanjee, Gunalan, Amaladoss, Yeo, Bob, Malleret, Duraisingh and Preiser2014) may also contribute to rosette formation. Further work is needed to determine the relative contributions of the different VSAs to rosetting, especially in clinical isolates.

Host serum proteins such as IgM, α2macroglobulin, albumin and fibrinogen also contribute to rosetting, either by binding directly to parasite adhesion molecules or by non-specific erythrocyte aggregating effects (Scholander et al., Reference Scholander, Treutiger, Hultenby and Wahlgren1996; Treutiger et al., Reference Treutiger, Scholander, Carlson, McAdam, Raynes, Falksveden and Wahlgren1999; Luginbuhl et al., Reference Luginbuhl, Nikolic, Beck, Wahlgren and Lutz2007; Ghumra et al., Reference Ghumra, Semblat, McIntosh, Raza, Rasmussen, Braathen, Johansen, Sandlie, Mongini, Rowe and Pleass2008, Reference Ghumra, Semblat, Ataide, Kifude, Adams, Claessens, Anong, Bull, Fennell, Arman, Amambua-Ngwa, Walther, Conway, Kassambara, Doumbo, Raza and Rowe2012; Semblat et al., Reference Semblat, Ghumra, Czajkowsky, Wallis, Mitchell, Raza and Rowe2015; Stevenson et al., Reference Stevenson, Huda, Jeppesen, Laursen, Rowe, Craig, Streicher, Barfod and Hviid2015a, Reference Stevenson, Laursen, Cowan, Bandoh, Barfod, Cavanagh, Andersen and Hviid2015b). The extent to which host serum proteins influence rosetting, sequestration and microvascular obstruction in vivo is unknown, and would be a valuable area of future study.

Rosetting receptors on host erythrocytes

A number of different molecules on uninfected erythrocytes have been proposed as receptors for P. falciparum rosetting (Fig. 2 and Table 1), and multiple receptor–ligand interactions may contribute to rosetting in any given parasite isolate. Some of the proposed rosetting receptor molecules, including blood group A and B sugars, heparan sulphate (HS)-like molecules and complement receptor one (CR1) are widely accepted as having a role in rosetting, whereas other recent candidates such glycophorin A (GYPA) and glycophorin C (GYPC) are less well-authenticated. However, a close examination of the underlying data shows that in most cases, the evidence is incomplete, as discussed in detail below.

Table 1. Summary of host erythrocyte receptors for Plasmodium falciparum rosetting

a Parasite strains used are not consistent between studies with a wide range of culture-adapted and clinical isolates in use. Results are therefore not necessarily generalizable from single studies.

b Many studies included here use heparin instead of/in addition to heparan sulphate.

Evidence needed to establish a role for a specific host receptor in rosetting

In order to prove that a particular molecule acts as a host receptor for P. falciparum rosetting, a variety of different types of evidence have been provided. Essential data include proof that the molecule in question is found on normal human erythrocytes and that erythrocytes lacking the molecule show reduced/absent rosetting. Direct binding between IEs and/or recombinant parasite adhesion proteins and the receptor molecule should be demonstrated. Ideally, a crystal structure of the parasite adhesion molecule–host receptor complex should show the precise binding interaction site. Supportive evidence includes the ability of antibodies against the receptor or soluble receptor proteins to inhibit rosetting, and biochemical approaches to remove or alter the receptor on erythrocytes. Human genetic evidence can also provide indirect supportive evidence that particular molecules are important in life-threatening malaria. Several putative rosetting receptors have high-frequency polymorphisms in populations from malaria endemic regions that reduce rosetting and are associated with protection against severe malaria and death [reviewed in Rowe et al. (Reference Rowe, Claessens, Corrigan and Arman2009a, Reference Rowe, Opi and Williams2009b)]. These various lines of evidence are summarized below for each potential host rosetting receptor.

Blood group A and B trisaccharides

The most well-validated rosetting receptors are the blood group A and B trisaccharides (Fig. 3). In vitro experiments have shown that rosetting parasites have a ‘preference’ for blood groups A, B or AB rather than O (Carlson and Wahlgren, Reference Carlson and Wahlgren1992; Udomsangpetch et al., Reference Udomsangpetch, Todd, Carlson and Greenwood1993; Barragan et al., Reference Barragan, Kremsner, Wahlgren and Carlson2000b; Pipitaporn et al., Reference Pipitaporn, Sueblinvong, Dharmkrong-at and Udomsangpetch2000; Vigan-Womas et al., Reference Vigan-Womas, Guillotte, Juillerat, Hessel, Raynal, England, Cohen, Bertrand, Peyrard, Bentley, Lewit-Bentley and Mercereau-Puijalon2012; Moll et al., Reference Moll, Palmkvist, Ch'ng, Kiwuwa and Wahlgren2015). This varies by parasite genotype, with A-preference being the commonest. Clinical isolates from non-O (i.e. A, B or AB) patients show higher levels of rosetting than isolates from group O patients in studies from sub-Saharan Africa (Rowe et al., Reference Rowe, Obeiro, Newbold and Marsh1995, Reference Rowe, Handel, Thera, Deans, Lyke, Koné, Diallo, Raza, Kai, Marsh, Plowe, Doumbo and Moulds2007) and India (Rout et al., Reference Rout, Dhangadamajhi, Ghadei, Mohapatra, Kar and Ranjit2012), although the same result was not seen in one Thai study (Lee et al., Reference Lee, Malleret, Lau, Mauduit, Fong, Cho, Suwanarusk, Zhang, Albrecht, Costa, Preiser, McGready, Renia, Nosten and Russell2014). When parasites are cultured in their ‘preferred’ blood group, they form larger, stronger rosettes that are more resistant to disruption by antibodies or chemical agents than in group O cells (Carlson and Wahlgren, Reference Carlson and Wahlgren1992; Barragan et al., Reference Barragan, Kremsner, Wahlgren and Carlson2000b; Ch'ng et al., Reference Ch'ng, Moll, Quintana, Chan, Masters, Moles, Liu, Eriksson and Wahlgren2016). Enzymatic removal of the terminal sugars (N-acetyl-D-galactosamine for A and D-galactose for B) results in smaller, weaker rosettes, equivalent to those seen in group O erythrocytes (Barragan et al., Reference Barragan, Kremsner, Wahlgren and Carlson2000b). Rosettes do, however, still occur with blood group O erythrocytes (that express the H antigen), and also in Bombay phenotype red cells that lack the ABO blood group core fucose residue (Fig. 3) (Carlson and Wahlgren, Reference Carlson and Wahlgren1992; Rowe et al., Reference Rowe, Moulds, Newbold and Miller1997). This indicates that other red cell surface molecules in addition to the A and B antigens can act as host receptors for rosette formation.

Fig. 3. Diagram of the ABO blood group sugars. Schematic representation of the terminal structure of the A (blue square), B (purple) H (green; H is the antigen carried on blood group O erythrocytes) and Bombay (orange) antigens. Yellow circle: D-Galactose (Gal), yellow square: N-acetyl-D-galactosamine (GalNac), red triangle: L-Fucose (Fuc). The symbols α and β indicate the position of the hydroxyl group and the numbers indicate the specific carbon atoms that are linked between the sugars. The H, A and B antigens are synthesized by a series of glycosyltransferase enzymes that add monosaccharides to create oligosaccharide chains attached to lipids and proteins in the erythrocyte membrane.

For the blood group A-preferring parasite line, Palo Alto 89F5, direct binding between the VarO PfEMP1 adhesion molecule and the blood group A trisaccharide was shown by Surface Plasmon Resonance (Vigan-Womas et al., Reference Vigan-Womas, Guillotte, Juillerat, Hessel, Raynal, England, Cohen, Bertrand, Peyrard, Bentley, Lewit-Bentley and Mercereau-Puijalon2012). The VarO PfEMP1 variant also binds to the B trisaccharide, but with lower affinity (Vigan-Womas et al., Reference Vigan-Womas, Guillotte, Juillerat, Hessel, Raynal, England, Cohen, Bertrand, Peyrard, Bentley, Lewit-Bentley and Mercereau-Puijalon2012). A crystal structure of the PfEMP1 N-terminal region was obtained and the A-trisaccharide binding site mapped (Vigan-Womas et al., Reference Vigan-Womas, Guillotte, Juillerat, Hessel, Raynal, England, Cohen, Bertrand, Peyrard, Bentley, Lewit-Bentley and Mercereau-Puijalon2012). A recent study suggests that P. falciparum RIFIN molecules may also be able to interact with blood group A sugars to contribute to rosette formation (Goel et al., Reference Goel, Palmkvist, Moll, Joannin, Lara, Akhouri, Moradi, Öjemalm, Westman, Angeletti, Kjellin, Lehtiö, Blixt, Ideström, Gahmberg, Storry, Hult, Olsson, von Heijne, Nilsson and Wahlgren2015), although direct RIFIN-A trisaccharide interaction was not shown.

The importance of the A and B antigens in rosetting is emphasized by the fact that the non-O blood groups are associated with increased risk of severe malaria and death compared to O (Rowe et al., Reference Rowe, Handel, Thera, Deans, Lyke, Koné, Diallo, Raza, Kai, Marsh, Plowe, Doumbo and Moulds2007; Fry et al., Reference Fry, Griffiths, Auburn, Diakite, Forton, Green, Richardson, Wilson, Jallow, Sisay-Joof, Pinder, Peshu, Williams, Marsh, Molyneux, Taylor, Rockett and Kwiatkowski2008; Tekeste and Petros, Reference Tekeste and Petros2010; Rout et al., Reference Rout, Dhangadamajhi, Ghadei, Mohapatra, Kar and Ranjit2012; Malaria Genomic Epidemiology Network, 2014; Ndila et al., Reference Ndila, Uyoga, Macharia, Nyutu, Peshu, Ojal, Shebe, Awuondo, Mturi, Tsofa, Sepulveda, Clark, Band, Clarke, Rowlands, Hubbart, Jeffreys, Kariuki, Marsh, Mackinnon, Maitland, Kwiatkowski, Rockett, Williams and Malaria2018; Degarege et al., Reference Degarege, Gebrezgi, Ibanez, Wahlgren and Madhivanan2019). Reduced rosetting in blood group O, and therefore reduced microvascular obstruction and reduced downstream pathological effects, is the proposed mechanism for the protective association with group O (Udomsangpetch et al., Reference Udomsangpetch, Todd, Carlson and Greenwood1993; Rowe et al., Reference Rowe, Handel, Thera, Deans, Lyke, Koné, Diallo, Raza, Kai, Marsh, Plowe, Doumbo and Moulds2007). ABO blood group does not influence parasite burden (Rowe et al., Reference Rowe, Handel, Thera, Deans, Lyke, Koné, Diallo, Raza, Kai, Marsh, Plowe, Doumbo and Moulds2007; Degarege et al., Reference Degarege, Gebrezgi, Ibanez, Wahlgren and Madhivanan2019), and evidence for an effect of ABO on P. falciparum invasion or other host–parasite interactions is conflicting and requires further study (Chung et al., Reference Chung, Gardiner, Hyland, Gatton, Kemp and Trenholme2005; Wolofsky et al., Reference Wolofsky, Ayi, Branch, Hult, Olsson, Liles, Cserti-Gazdewich and Kain2012; Pathak et al., Reference Pathak, Colah and Ghosh2016; Theron et al., Reference Theron, Cross, Cawkill, Bustamante and Rayner2018). The ABH antigens are known to be present on endothelial cells (Ito et al., Reference Ito, Nishi, Kawahara, Okamura, Hirota, Rand, Fechner and Brinkmann1990) and it is likely, but has not been shown experimentally, that cytoadhesion and overall levels of sequestration of rosetting parasites are enhanced in group A/B/AB patients compared to O.

Despite the progress in identifying the A and B trisaccharides as rosetting receptors and key genetic determinants of host susceptibility to severe malaria, there have been no attempts to develop specific therapies to block P. falciparum interaction with A/B antigens. The PfEMP1-blood group A trisaccharide binding pair described above (Vigan-Womas et al., Reference Vigan-Womas, Guillotte, Juillerat, Hessel, Raynal, England, Cohen, Bertrand, Peyrard, Bentley, Lewit-Bentley and Mercereau-Puijalon2012) remains the most clearly defined molecular interaction between parasite ligand and host receptor in rosetting, and could be used as a starting point to develop rosette-blocking therapeutics. Vigan-Womas et al. did report that the interaction between PfEMP1 and the A and B trisaccharides is indirectly inhibited by heparin (Vigan-Womas et al., Reference Vigan-Womas, Guillotte, Juillerat, Hessel, Raynal, England, Cohen, Bertrand, Peyrard, Bentley, Lewit-Bentley and Mercereau-Puijalon2012), and the development of a heparin-derivative as a potential adjunctive therapy for severe malaria is described below (Leitgeb et al., Reference Leitgeb, Charunwatthana, Rueangveerayut, Uthaisin, Silamut, Chotivanich, Sila, Moll, Lee, Lindgren, Holmer, Färnert, Kiwuwa, Kristensen, Herder, Tarning, Wahlgren and Dondorp2017).

Complement receptor one (CR1, CD35)

CR1 is a red cell membrane glycoprotein that regulates complement activation on cell surfaces (Thielen et al., Reference Thielen, Zeerleder and Wouters2018) and carries the Knops Blood Group antigens (Moulds, Reference Moulds2010). In malaria, CR1 plays a role in both rosetting and parasite invasion of erythrocytes (Schmidt et al., Reference Schmidt, Kennedy and Tham2015). CR1 was first identified as a rosetting receptor from a screen of 23 naturally occurring erythrocyte null mutants, each missing a particular blood group molecule or membrane glycoprotein (Rowe et al., Reference Rowe, Moulds, Newbold and Miller1997). The only variant to show substantially reduced rosetting with five P. falciparum parasite lines was Knops null cells, which are deficient in CR1. Normally, erythrocytes have between 100 and 1000 molecules of CR1 per cell (Wilson et al., Reference Wilson, Murphy, Wong, Klickstein, Weis and Fearon1986), whereas Knops null cells have fewer than 100 molecules per cell (Moulds et al., Reference Moulds, Moulds, Brown and Atkinson1992). Erythrocytes with fewer than 50 CR1 molecules per cell form rosettes poorly (Rowe et al., Reference Rowe, Moulds, Newbold and Miller1997), with normal rosetting occurring above a threshold of around 100 molecules per cell (JA Rowe, unpublished data).

Soluble CR1 and CR1 antibodies were shown to inhibit rosetting in some but, not all P. falciparum rosetting laboratory strains and clinical isolates, with only monoclonal antibodies (mAbs) that map to the C3b binding site on CR1 being effective inhibitors (Rowe et al., Reference Rowe, Moulds, Newbold and Miller1997, Reference Rowe, Rogerson, Raza, Moulds, Kazatchkine, Marsh, Newbold, Atkinson and Miller2000; Vigan-Womas et al., Reference Vigan-Womas, Guillotte, Juillerat, Hessel, Raynal, England, Cohen, Bertrand, Peyrard, Bentley, Lewit-Bentley and Mercereau-Puijalon2012). A recent paper suggested that the commercially available CR1 mAb E11 that recognizes epitopes outside the C3b binding site (Nickells et al., Reference Nickells, Hauhart, Krych, Subramanian, Geoghegan-Barek, Marsh and Atkinson1998) may inhibit P. falciparum rosetting (Lee et al., Reference Lee, Malleret, Lau, Mauduit, Fong, Cho, Suwanarusk, Zhang, Albrecht, Costa, Preiser, McGready, Renia, Nosten and Russell2014), but this was not seen in our hands (Rowe et al., Reference Rowe, Rogerson, Raza, Moulds, Kazatchkine, Marsh, Newbold, Atkinson and Miller2000). Further evidence of a role for CR1 in rosetting came from the expression of recombinant PfEMP1 domains in COS-7 cells, which bound to normal erythrocytes but not to CR1-deficient cells (Rowe et al., Reference Rowe, Moulds, Newbold and Miller1997).

Despite these supportive data, direct binding of IEs to CR1 protein has not been demonstrated, and recombinant rosette-mediating PfEMP1 proteins produced in E. coli (Ghumra et al., Reference Ghumra, Semblat, Ataide, Kifude, Adams, Claessens, Anong, Bull, Fennell, Arman, Amambua-Ngwa, Walther, Conway, Kassambara, Doumbo, Raza and Rowe2012) do not bind to CR1 in Surface Plasmon Resonance experiments (Tetteh-Quarcoo et al., Reference Tetteh-Quarcoo, Schmidt, Tham, Hauhart, Mertens, Rowe, Atkinson, Cowman, Rowe and Barlow2012). This could reflect a genuine lack of interaction between the two molecules, or could be due to technical reasons (e.g. the recombinant CR1 used in experiments was produced in mouse rather than human cells, whereas CR1 glycosylation, which may affect function, is cell-type specific) (Lublin et al., Reference Lublin, Griffith and Atkinson1986). It is also possible that a serum protein mediates the interaction between PfEMP1 on IEs and CR1 on uninfected erythrocytes, as the original experiments were all performed in the presence of serum (Rowe et al., Reference Rowe, Moulds, Newbold and Miller1997).

Human genetic studies provide additional support for the importance of CR1 in malaria host–parasite interactions. Erythrocyte CR1 deficiency is common in some malaria-endemic countries such as Papua New Guinea (Cockburn et al., Reference Cockburn, Mackinnon, O'Donnell, Allen, Moulds, Baisor, Bockarie, Reeder and Rowe2004) and India (Sinha et al., Reference Sinha, Jha, Anand, Qidwai, Pati, Mohanty, Mishra, Tyagi, Sharma, Venkatesh and Habib2009), and is associated with protection against severe malaria in medium to high transmission areas (Cockburn et al., Reference Cockburn, Mackinnon, O'Donnell, Allen, Moulds, Baisor, Bockarie, Reeder and Rowe2004; Sinha et al., Reference Sinha, Jha, Anand, Qidwai, Pati, Mohanty, Mishra, Tyagi, Sharma, Venkatesh and Habib2009; Rout et al., Reference Rout, Dhangadamajhi, Mohapatra, Kar and Ranjit2011; Panda et al., Reference Panda, Panda, Tripathy, Pattanaik, Ravindran and Das2012). However, erythrocyte CR1 deficiency may be detrimental in areas such as Thailand, where malaria transmission is low (Nagayasu et al., Reference Nagayasu, Ito, Akaki, Nakano, Kimura, Looareesuwan and Aikawa2001; Teeranaipong et al., Reference Teeranaipong, Ohashi, Patarapotikul, Kimura, Nuchnoi, Hananantachai, Naka, Putaporntip, Jongwutiwes and Tokunaga2008). There is also evidence that the CR1 Swain Langley 2 (Sl2) Knops blood group polymorphism that is common in African populations (Moulds, Reference Moulds2010) is associated with protection against severe malaria (Thathy et al., Reference Thathy, Moulds, Guyah, Otieno and Stoute2005; Opi et al., Reference Opi, Swann, Macharia, Uyoga, Band, Ndila, Harrison, Thera, Kone, Diallo, Doumbo, Lyke, Plowe, Moulds, Shebbe, Mturi, Peshu, Maitland, Raza, Kwiatkowski, Rockett, Williams and Rowe2018). Red cells carrying the Sl2 antigen on CR1 show reduced rosetting (Rowe et al., Reference Rowe, Moulds, Newbold and Miller1997; Opi et al., Reference Opi, Swann, Macharia, Uyoga, Band, Ndila, Harrison, Thera, Kone, Diallo, Doumbo, Lyke, Plowe, Moulds, Shebbe, Mturi, Peshu, Maitland, Raza, Kwiatkowski, Rockett, Williams and Rowe2018), and Sl2 may have additional effects on complement activation and regulation (Opi et al., Reference Opi, Swann, Macharia, Uyoga, Band, Ndila, Harrison, Thera, Kone, Diallo, Doumbo, Lyke, Plowe, Moulds, Shebbe, Mturi, Peshu, Maitland, Raza, Kwiatkowski, Rockett, Williams and Rowe2018).

Overall, the ability of CR1 mAbs and soluble protein to reverse rosettes suggests that CR1 plays a role in rosetting for some P. falciparum isolates. However, further work is needed to fully investigate the molecular interactions between parasite adhesion molecules and CR1, and to explore the potential for CR1 reagents (Li et al., Reference Li, Jaggers and Anderson2006; Reddy et al., Reference Reddy, Siedlecki and Francis2017) as therapeutic disruptors of rosetting.

Heparan sulphate and chondroitin sulphate

The glycosaminoglycans HS and chondroitin sulphate (CS) are found on cell surfaces and in the extracellular matrix of many tissues, and have a role in multiple aspects of the P. falciparum life cycle including hepatocyte invasion (Frevert et al., Reference Frevert, Sinnis, Cerami, Shreffler, Takacs and Nussenzweig1993), endothelial cell cytoadherence (Vogt et al., Reference Vogt, Barragan, Chen, Kironde, Spillmann and Wahlgren2003; Adams et al., Reference Adams, Kuhnrae, Higgins, Ghumra and Rowe2014) and, for CS, placental sequestration (Fried and Duffy, Reference Fried and Duffy1996). A number of papers have showed that heparin (which is a highly-sulphated form of HS found only in mast cells) can partially disrupt rosettes in about one-third to one-half of P. falciparum clinical isolates in vitro (Udomsangpetch et al., Reference Udomsangpetch, Wåhlin, Carlson, Berzins, Torii, Aikawa, Perlmann and Wahlgren1989; Carlson et al., Reference Carlson, Ekre, Helmby, Gysin, Greenwood and Wahlgren1992; Rogerson et al., Reference Rogerson, Reeder, al-Yaman and Brown1994; Rowe et al., Reference Rowe, Berendt, Marsh and Newbold1994; Barragan et al., Reference Barragan, Spillmann, Kremsner, Wahlgren and Carlson1999). It was shown that treating erythrocytes with heparinase III, which selectively cleaves HS chains, reduces rosetting in two P. falciparum lines (Barragan et al., Reference Barragan, Spillmann, Kremsner, Wahlgren and Carlson1999), and therefore suggested that ‘HS-like’ molecules on red cells are receptors for rosetting (Chen et al., Reference Chen, Heddini, Barragan, Fernandez, Pearce and Wahlgren2000). However, there has been only one paper reporting the existence of HS on normal human erythrocytes (Vogt et al., Reference Vogt, Winter, Wahlgren and Spillmann2004) and we have been unable to confirm this, and unable to detect any rosette-reducing effect of heparinase III in a range of parasite lines (McQuaid and Rowe, unpublished data).

Fluorescently-labelled heparin does bind to the surface of erythrocytes infected with rosetting parasites more than non-rosetting lines (Barragan et al., Reference Barragan, Fernandez, Chen, von Euler, Wahlgren and Spillmann2000a; Heddini et al., Reference Heddini, Pettersson, Kai, Shafi, Obiero, Chen, Barragan, Wahlgren and Marsh2001), and some rosette-mediating PfEMP1 variants bind directly to heparin (Barragan et al., Reference Barragan, Fernandez, Chen, von Euler, Wahlgren and Spillmann2000a; Vogt et al., Reference Vogt, Barragan, Chen, Kironde, Spillmann and Wahlgren2003; Juillerat et al., Reference Juillerat, Igonet, Vigan-Womas, Guillotte, Gangnard, Faure, Baron, Raynal, Mercereau-Puijalon and Bentley2010, Reference Juillerat, Lewit-Bentley, Guillotte, Gangnard, Hessel, Baron, Vigan-Womas, England, Mercereau-Puijalon and Bentley2011; Adams et al., Reference Adams, Kuhnrae, Higgins, Ghumra and Rowe2014). The heparin binding site in the N-terminal region of the varO PfEMP1 variant was mapped onto a crystal structure (Juillerat et al., Reference Juillerat, Lewit-Bentley, Guillotte, Gangnard, Hessel, Baron, Vigan-Womas, England, Mercereau-Puijalon and Bentley2011), and shown to be on the opposite side of the molecule from the erythrocyte binding site (Vigan-Womas et al., Reference Vigan-Womas, Guillotte, Juillerat, Hessel, Raynal, England, Cohen, Bertrand, Peyrard, Bentley, Lewit-Bentley and Mercereau-Puijalon2012). Hence, the rosette-disrupting effect of heparin is not due to direct blocking of receptor binding, but may result from aggregating PfEMP1 monomers and preventing their interaction with erythrocyte receptors (Vigan-Womas et al., Reference Vigan-Womas, Guillotte, Juillerat, Hessel, Raynal, England, Cohen, Bertrand, Peyrard, Bentley, Lewit-Bentley and Mercereau-Puijalon2012). Similarly, for another rosette-mediating PfEMP1 variant IT4var60, site-directed mutagenesis studies of recombinant proteins showed that mutations that disrupt heparin binding are distinct from mutations that disrupt erythrocyte binding, indicating that heparin-like molecules are not the main host rosetting receptor in this case (Angeletti et al., Reference Angeletti, Sandalova, Wahlgren and Achour2015).

Overall, whether HS is present on normal erythrocytes and is a host receptor for rosetting requires further confirmation. HS in present on the luminal surface of microvascular endothelial cells (albeit at a much lower density than on basolateral surfaces) (de Agostini et al., Reference de Agostini, Watkins, Slayter, Youssoufian and Rosenberg1990; Stoler-Barak et al., Reference Stoler-Barak, Moussion, Shezen, Hatzav, Sixt and Alon2014), therefore interactions between IE and endothelial HS (Vogt et al., Reference Vogt, Barragan, Chen, Kironde, Spillmann and Wahlgren2003; Adams et al., Reference Adams, Kuhnrae, Higgins, Ghumra and Rowe2014) are physiologically relevant and are likely to contribute to cytoadherence and sequestration in vivo.

Despite the uncertainty on the precise role of HS as an erythrocyte rosetting receptor, heparin and other sulphated glycoconjugate compounds have clear potential as adjunctive therapies for severe malaria due to their rosette-disrupting effects (Udomsangpetch et al., Reference Udomsangpetch, Wåhlin, Carlson, Berzins, Torii, Aikawa, Perlmann and Wahlgren1989; Carlson et al., Reference Carlson, Ekre, Helmby, Gysin, Greenwood and Wahlgren1992; Rogerson et al., Reference Rogerson, Reeder, al-Yaman and Brown1994; Rowe et al., Reference Rowe, Berendt, Marsh and Newbold1994; Kyriacou et al., Reference Kyriacou, Steen, Raza, Arman, Warimwe, Bull, Havlik and Rowe2007). There are reports of successful heparin treatment in severe malaria (Rampengan, Reference Rampengan1991) but its use is not recommended due to a high incidence of bleeding complications (World Health Organisation, 1986). As an alternative, Wahlgren and coworkers have developed a low anti-coagulant heparin derivative, Sevuparin, that reverses rosetting and cytoadherence in some P. falciparum isolates (Leitgeb et al., Reference Leitgeb, Blomqvist, Cho-Ngwa, Samje, Nde, Titanji and Wahlgren2011; Saiwaew et al., Reference Saiwaew, Sritabal, Piaraksa, Keayarsa, Ruengweerayut, Utaisin, Sila, Niramis, Udomsangpetch, Charunwatthana, Pongponratn, Pukrittayakamee, Leitgeb, Wahlgren, Lee, Day, White, Dondorp and Chotivanich2017) and also blocks merozoite invasion (Leitgeb et al., Reference Leitgeb, Charunwatthana, Rueangveerayut, Uthaisin, Silamut, Chotivanich, Sila, Moll, Lee, Lindgren, Holmer, Färnert, Kiwuwa, Kristensen, Herder, Tarning, Wahlgren and Dondorp2017). Sevuparin has been shown to be safe in adults with uncomplicated malaria (Leitgeb et al., Reference Leitgeb, Charunwatthana, Rueangveerayut, Uthaisin, Silamut, Chotivanich, Sila, Moll, Lee, Lindgren, Holmer, Färnert, Kiwuwa, Kristensen, Herder, Tarning, Wahlgren and Dondorp2017), but has not yet been tested in severe malaria patients.

The evidence for CS as a rosetting receptor is minimal. One report shows that rosetting in the P. falciparum line TM284 was partially inhibited by soluble CS and by chondroitinase enzyme treatment of erythrocytes, and that several clinical isolates showed reduced rosetting in the presence of CS (Barragan et al., Reference Barragan, Spillmann, Kremsner, Wahlgren and Carlson1999). However, other studies have found no effect of CS on rosetting in a variety of culture-adapted lines and clinical isolates (Rogerson et al., Reference Rogerson, Reeder, al-Yaman and Brown1994; Rowe et al., Reference Rowe, Berendt, Marsh and Newbold1994). There is also no convincing evidence that CS is found on the surface of normal human erythrocytes. Overall, current data do not support a role for CS in rosetting.

CD36

The membrane glycoprotein CD36 is a scavenger receptor for oxidized lipoproteins and a fatty acid translocase (Silverstein and Febbraio, Reference Silverstein and Febbraio2009). It is expressed on a variety of cell types including monocytes, macrophages, platelets, microvascular endothelial cells and adipocytes (Silverstein and Febbraio, Reference Silverstein and Febbraio2009), and at low levels on erythrocytes (van Schravendijk et al., Reference van Schravendijk, Handunnetti, Barnwell and Howard1992). The binding of PfEMP1 (group B and C variants) to CD36 on microvascular endothelial cells plays a major role in P. falciparum sequestration (Baruch et al., Reference Baruch, Gormely, Ma, Howard and Pasloske1996; Robinson et al., Reference Robinson, Welch and Smith2003). Almost all P. falciparum isolates bind to CD36, and increased CD36 binding (Newbold et al., Reference Newbold, Warn, Black, Berendt, Craig, Snow, Msobo, Peshu and Marsh1997; Ochola et al., Reference Ochola, Siddondo, Ocholla, Nkya, Kimani, Williams, Makale, Liljander, Urban, Bull, Szestak, Marsh and Craig2011) and predominant expression of group B and C PfEMP1 (Kraemer and Smith, Reference Kraemer and Smith2006; Kyriacou et al., Reference Kyriacou, Stone, Challis, Raza, Lyke, Thera, Koné, Doumbo, Plowe and Rowe2006) are associated with uncomplicated malaria.

The role of CD36 in rosetting is less clear. Anti-CD36 mAbs are capable of disrupting rosettes in a single culture-adapted parasite line, Malayan Camp (Handunnetti et al., Reference Handunnetti, van Schravendijk, Hasler, Barnwell, Greenwalt and Howard1992), but not in a wide range of other laboratory lines or clinical isolates (Udomsangpetch et al., Reference Udomsangpetch, Wåhlin, Carlson, Berzins, Torii, Aikawa, Perlmann and Wahlgren1989; Wahlgren et al., Reference Treutiger, Hedlund, Helmby, Carlson, Jepson, Twumasi, Kwiatkowski, Greenwood and Wahlgren1992; Rowe et al., Reference Rowe, Rogerson, Raza, Moulds, Kazatchkine, Marsh, Newbold, Atkinson and Miller2000; Niang et al., Reference Niang, Bei, Madnani, Pelly, Dankwa, Kanjee, Gunalan, Amaladoss, Yeo, Bob, Malleret, Duraisingh and Preiser2014). The PfEMP1 variants identified as parasite rosetting ligands (Rowe et al., Reference Rowe, Moulds, Newbold and Miller1997; Vigan-Womas et al., Reference Vigan-Womas, Guillotte, Juillerat, Vallieres, Lewit-Bentley, Tall, Baril, Bentley and Mercereau-Puijalon2011; Ghumra et al., Reference Ghumra, Semblat, Ataide, Kifude, Adams, Claessens, Anong, Bull, Fennell, Arman, Amambua-Ngwa, Walther, Conway, Kassambara, Doumbo, Raza and Rowe2012) are mostly of the group A type, which do not bind to CD36 (Robinson et al., Reference Robinson, Welch and Smith2003).

Intriguingly, while CD36 deficiency is fairly common in African populations, large-scale genetic studies have shown that CD36 polymorphisms do not influence severe malaria risk (Fry et al., Reference Fry, Ghansa, Small, Palma, Auburn, Diakite, Green, Campino, Teo, Clark, Jeffreys, Wilson, Jallow, Sisay-Joof, Pinder, Griffiths, Peshu, Williams, Newton, Marsh, Molyneux, Taylor, Koram, Oduro, Rogers, Rockett, Sabeti and Kwiatkowski2009). There is some evidence that interaction between IEs and CD36 may benefit the host, as CD36 may contribute to innate immune clearance of IEs and platelet-mediated parasite death (McGilvray et al., Reference McGilvray, Serghides, Kapus, Rotstein and Kain2000; McMorran et al., Reference McMorran, Wieczorski, Drysdale, Chan, Huang, Smith, Mitiku, Beeson, Burgio and Foote2012; Cabrera et al., Reference Cabrera, Neculai and Kain2014). Overall, it is unlikely that CD36 is a clinically significant rosetting receptor or a useful therapeutic target in severe malaria (Cabrera et al., Reference Cabrera, Neculai and Kain2014).

Glycophorin C (GYPC; GPC; CD236)

GYPC is a red cell membrane glycoprotein that carries the Gerbich blood group antigens (Jaskiewicz et al., Reference Jaskiewicz, Peyrard, Kaczmarek, Zerka, Jodlowska and Czerwinski2018). It is a P. falciparum invasion receptor bound by the merozoite protein EBA-140/BAEBL (Maier et al., Reference Maier, Duraisingh, Reeder, Patel, Kazura, Zimmerman and Cowman2003; Mayer et al., Reference Mayer, Jiang, Achur, Kakizaki, Gowda and Miller2006). Recently, two studies have suggested that GYPC is a rosetting receptor for both P. falciparum (Niang et al., Reference Niang, Bei, Madnani, Pelly, Dankwa, Kanjee, Gunalan, Amaladoss, Yeo, Bob, Malleret, Duraisingh and Preiser2014) and P. vivax (Lee et al., Reference Lee, Malleret, Lau, Mauduit, Fong, Cho, Suwanarusk, Zhang, Albrecht, Costa, Preiser, McGready, Renia, Nosten and Russell2014; Niang et al., Reference Niang, Bei, Madnani, Pelly, Dankwa, Kanjee, Gunalan, Amaladoss, Yeo, Bob, Malleret, Duraisingh and Preiser2014). Niang et al. showed that the rosetting of a 3D7-derived P. falciparum laboratory strain (5A-R+) was partially inhibited by a GYPC mAb (clone Ret40f) and by soluble recombinant GYPC (Niang et al., Reference Niang, Bei, Madnani, Pelly, Dankwa, Kanjee, Gunalan, Amaladoss, Yeo, Bob, Malleret, Duraisingh and Preiser2014). Furthermore, cultured GYPC knockdown erythrocytes failed to rosette, providing strong evidence that GYPC is an essential rosetting receptor for 5A-R+ parasites (Niang et al., Reference Niang, Bei, Madnani, Pelly, Dankwa, Kanjee, Gunalan, Amaladoss, Yeo, Bob, Malleret, Duraisingh and Preiser2014). Other parasite lines or clinical isolates were not tested, therefore the wider role of GYPC in P. falciparum rosetting was not determined.

Lee et al. (Reference Lee, Malleret, Lau, Mauduit, Fong, Cho, Suwanarusk, Zhang, Albrecht, Costa, Preiser, McGready, Renia, Nosten and Russell2014) focussed mainly on P. vivax, but also assessed the ability of GYPC mAb fragments to inhibit rosette formation in ten P. falciparum clinical isolates from Thailand. A significant decrease in rosetting was reported with GYPC mAb BRIC 4, although the reduction in the median rosette frequency was small (from 11.5 to 5.5%), and was based on a single count for each isolate with no replication (Lee et al., Reference Lee, Malleret, Lau, Mauduit, Fong, Cho, Suwanarusk, Zhang, Albrecht, Costa, Preiser, McGready, Renia, Nosten and Russell2014). The biological significance of these results is difficult to assess, given the low starting rosette frequencies and inherent variation in the rosetting assay. Lee et al. also used a different definition of rosetting to all previous studies, defining a rosette as an IE binding one or more uninfected erythrocytes. The usual definition requires the binding of two or more uninfected erythrocytes, which helps to identify genuine cell–cell interactions and avoid spurious identification of rosettes due to close packing of cells under the coverslip during microscopy.

For P. vivax, Lee et al. showed that the GYPC mAb reduced the median rosette frequency from 30 to 22% when tested on 11 Thai isolates, and that GYPC knockdown cultured erythrocytes formed rosettes poorly compared to GYPC-positive control cells (median rosette frequency 6.2 vs. 35.4% in controls, tested on three isolates). Plasmodium falciparum isolates were not tested with the GYPC knockdown erythrocytes.

If GYPC is a rosetting receptor, it is possible that the ‘Gerbich-negative’ blood group type, which is common in Melanesian populations (Patel et al., Reference Patel, Mehlotra, Kastens, Mgone, Kazura and Zimmerman2001), might influence rosetting. As part of a screen of null blood group erythrocytes with five high-rosetting P. falciparum culture-adapted parasite lines, Rowe et al. (Reference Rowe, Moulds, Newbold and Miller1997) tested two donors with the Gerbich-negative blood group (formed by deletion of exon 3 of the GYPC gene on chromosome 2, giving a truncated protein with altered glycosylation). Gerbich-negative erythrocytes formed rosettes normally with the five parasite lines tested. Goel et al. also report normal rosetting of Gerbich-negative erythrocytes from two donors (Goel et al., Reference Goel, Palmkvist, Moll, Joannin, Lara, Akhouri, Moradi, Öjemalm, Westman, Angeletti, Kjellin, Lehtiö, Blixt, Ideström, Gahmberg, Storry, Hult, Olsson, von Heijne, Nilsson and Wahlgren2015). The true null phenotype for GYPC, called the Leach phenotype (which arises due to the deletion of exon 3 and exon 4, encoding the transmembrane and cytoplasmic domains, respectively) is rare and has not been tested in rosetting assays to our knowledge.

Taking into account all existing evidence, further investigation of a wider range of parasite lines is needed to determine whether GYPC is an important host receptor for both P. falciparum and P. vivax rosetting.

Glycophorin A (GYPA, GPA, CD235a)

GYPA is a highly-expressed erythrocyte surface glycoprotein that carries the MNS blood group antigens. It is known to be a receptor for P. falciparum erythrocyte invasion (Sim et al., Reference Sim, Chitnis, Wasniowska, Hadley and Miller1994), and polymorphisms in GYPA are associated with resistance to severe malaria (Band et al., Reference Band, Rockett, Spencer, Kwiatkowski and Network2015; Leffler et al., Reference Leffler, Band, Busby, Kivinen, Le, Clarke, Bojang, Conway, Jallow, Sisay-Joof, Bougouma, Mangano, Modiano, Sirima, Achidi, Apinjoh, Marsh, Ndila, Peshu, Williams, Drakeley, Manjurano, Reyburn, Riley, Kachala, Molyneux, Nyirongo, Taylor, Thornton, Tilley, Grimsley, Drury, Stalker, Cornelius, Hubbart, Jeffreys, Rowlands, Rockett, Spencer and Kwiatkowski2017).

There is some limited evidence to suggest that GYPA may have a role in rosetting. Parasites of the strain FCR3S1.2 transfected with a specific RIFIN gene formed rosettes that were largely dependent on blood group A (Goel et al., Reference Goel, Palmkvist, Moll, Joannin, Lara, Akhouri, Moradi, Öjemalm, Westman, Angeletti, Kjellin, Lehtiö, Blixt, Ideström, Gahmberg, Storry, Hult, Olsson, von Heijne, Nilsson and Wahlgren2015). However, rosetting of the RIFIN-transfected parasites was significantly reduced with GYPA null cells from blood group O and B donors, whereas blood group A GYPA null erythrocytes formed rosettes normally. These data suggest that GYPA may have an accessory role for RIFIN-mediated rosetting in the absence of the A antigen (Goel et al., Reference Goel, Palmkvist, Moll, Joannin, Lara, Akhouri, Moradi, Öjemalm, Westman, Angeletti, Kjellin, Lehtiö, Blixt, Ideström, Gahmberg, Storry, Hult, Olsson, von Heijne, Nilsson and Wahlgren2015), although whether this applies to rosetting in non-genetically manipulated parasites in unknown.

Despite the above positive evidence, there are no other data supporting a role for GYPA in rosetting. GYPA mAb fragments had no inhibitory effect on rosetting in ten P. falciparum and 11 P. vivax clinical isolates (Lee et al., Reference Lee, Malleret, Lau, Mauduit, Fong, Cho, Suwanarusk, Zhang, Albrecht, Costa, Preiser, McGready, Renia, Nosten and Russell2014), and a GYPA mAb did not inhibit 3D7 5A-R+ rosettes (Niang et al., Reference Niang, Bei, Madnani, Pelly, Dankwa, Kanjee, Gunalan, Amaladoss, Yeo, Bob, Malleret, Duraisingh and Preiser2014). Furthermore, GYPA null erythrocytes (MkMk cells, lacking both GYPA and glycophorin B) formed rosettes with five culture-adapted P. falciparum lines (Rowe et al., Reference Rowe, Moulds, Newbold and Miller1997). Overall, existing evidence does not support a major role for GYPA in rosetting, but as with GYPC, further investigation is needed.

New receptors and new approaches

None of the receptors described above fully account for the adhesion interactions between infected and uninfected erythrocytes, and it is likely that other host rosetting receptors remain to be identified. There is evidence to suggest that these unknown host receptors are carbohydrates or protease-resistant proteins, because uninfected group O erythrocytes treated with trypsin and other proteases are still able to form rosettes (Udomsangpetch et al., Reference Udomsangpetch, Wåhlin, Carlson, Berzins, Torii, Aikawa, Perlmann and Wahlgren1989; Rowe et al., Reference Rowe, Berendt, Marsh and Newbold1994).

In order to progress rosetting research, alternative methods are needed. Rosetting experiments with GYPC and CR1 knockdown cultured human red cells derived from CD34+ haematopoetic stem cells have been performed (Lee et al., Reference Lee, Malleret, Lau, Mauduit, Fong, Cho, Suwanarusk, Zhang, Albrecht, Costa, Preiser, McGready, Renia, Nosten and Russell2014; Niang et al., Reference Niang, Bei, Madnani, Pelly, Dankwa, Kanjee, Gunalan, Amaladoss, Yeo, Bob, Malleret, Duraisingh and Preiser2014), using lentiviral transduction of short hairpin RNA (Bei et al., Reference Bei, Brugnara and Duraisingh2010). However, these cultured erythrocytes have a short life-span, limiting their usefulness. The development of immortalized erythroid lines (Kurita et al., Reference Kurita, Suda, Sudo, Miharada, Hiroyama, Miyoshi, Tani and Nakamura2013; Kanjee et al., Reference Kanjee, Grüring, Chaand, Lin, Egan, Manzo, Jones, Yu, Barker, Weekes and Duraisingh2017; Trakarnsanga et al., Reference Trakarnsanga, Griffiths, Wilson, Blair, Satchwell, Meinders, Cogan, Kupzig, Kurita, Nakamura, Toye, Anstee and Frayne2017; Scully et al., Reference Scully, Shabani, Rangel, Gruring, Kanjee, Clark, Chaand, Kurita, Nakamura, Ferreira and Duraisingh2019) may overcome this limitation. Nevertheless, attention must be paid to the subtle but real differences between mature erythrocytes and these, still relatively immature, immortalized CD34+ derived cells (Wilson et al., Reference Wilson, Trakarnsanga, Heesom, Cogan, Green, Toye, Parsons, Anstee and Frayne2016; Dankwa et al., Reference Dankwa, Chaand, Kanjee, Jiang, Nobre, Goldberg, Bei, Moechtar, Grüring, Ahouidi, Ndiaye, Dieye, Mboup, Weekes and Duraisingh2017; Trakarnsanga et al., Reference Trakarnsanga, Griffiths, Wilson, Blair, Satchwell, Meinders, Cogan, Kupzig, Kurita, Nakamura, Toye, Anstee and Frayne2017). CRISPR-Cas9 technology (Doudna and Charpentier, Reference Doudna and Charpentier2014) has led to an explosion in the ability to genetically manipulate multiple cell types, including erythrocyte precursors and immortalized haematopoietic lines (Song et al., Reference Song, Fan, He, Zhu, Niu, Wang, Ou, Luo and Sun2015; Kanjee et al., Reference Kanjee, Grüring, Chaand, Lin, Egan, Manzo, Jones, Yu, Barker, Weekes and Duraisingh2017; Hawksworth et al., Reference Hawksworth, Satchwell, Meinders, Daniels, Regan, Thornton, Wilson, Dobbe, Streekstra, Trakarnsanga, Heesom, Anstee, Frayne and Toye2018; Chung et al., Reference Chung, Magis, Vu, Heo, Wartiovaara, Walters, Kurita, Nakamura, Boffelli, Martin, Corn and DeWitt2019; Scully et al., Reference Scully, Shabani, Rangel, Gruring, Kanjee, Clark, Chaand, Kurita, Nakamura, Ferreira and Duraisingh2019), potentially giving the opportunity to generate multiple knockout lines for rosetting research. A consistent supply of knockout erythrocytes would allow large-scale screens for new rosetting receptors using cells as close to their normal physiological form as possible, raising exciting prospects for future work.

Conclusions

Of the rosetting receptors described over the past 30 years, only the blood group A trisaccharide has been authenticated by a variety of methodological approaches from a range of different investigators. For all other potential rosetting receptors, the evidence remains fragmentary (Table 1) and further research is needed (Table 2). Recent technical advances in genetic manipulation of red cell precursors and immortalised lines should enable reverse genetic studies to bring further clarity to this biologically important topic.

Table 2. Key areas for future research on rosetting receptors

Financial support

This work was supported by the Wellcome Trust (PhD studentship grant number 108685/Z/15/Z).

Conflict of interest

None.

Ethical standards

Not applicable.

References

Adams, Y, Kuhnrae, P, Higgins, MK, Ghumra, A and Rowe, JA (2014) Rosetting Plasmodium falciparum-infected erythrocytes bind to human brain microvascular endothelial cells in vitro, demonstrating a dual adhesion phenotype mediated by distinct P. falciparum erythrocyte membrane protein 1 domains. Infection and Immunity 82, 949959.CrossRefGoogle ScholarPubMed
Albrecht, L, Moll, K, Blomqvist, K, Normark, J, Chen, Q and Wahlgren, M (2011) Var gene transcription and PfEMP1 expression in the rosetting and cytoadhesive Plasmodium falciparum clone FCR3S1.2. Malaria Journal 10, 17.CrossRefGoogle ScholarPubMed
Angeletti, D, Sandalova, T, Wahlgren, M and Achour, A (2015) Binding of subdomains 1/2 of PfEMP1-DBL1alpha to heparan sulfate or heparin mediates Plasmodium falciparum rosetting. PLoS ONE 10, e0118898.CrossRefGoogle ScholarPubMed
Angus, BJ, Thanikkul, K, Silamut, K, White, NJ and Udomsangpetch, R (1996) Short report: rosette formation in Plasmodium ovale infection. American Journal of Tropical Medicine and Hygiene 55, 560561.CrossRefGoogle ScholarPubMed
Band, G, Rockett, KA, Spencer, CC, Kwiatkowski, DP and Network, MGE (2015) A novel locus of resistance to severe malaria in a region of ancient balancing selection. Nature 526, 253257.Google Scholar
Barragan, A, Spillmann, D, Kremsner, PG, Wahlgren, M and Carlson, J (1999) Plasmodium falciparum: molecular background to strain-specific rosette disruption by glycosaminoglycans and sulfated glycoconjugates. Experimental Parasitology 91, 133143.CrossRefGoogle ScholarPubMed
Barragan, A, Fernandez, V, Chen, Q, von Euler, A, Wahlgren, M and Spillmann, D (2000 a) The duffy-binding-like domain 1 of Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) is a heparan sulfate ligand that requires 12 mers for binding. Blood 95, 35943599.CrossRefGoogle ScholarPubMed
Barragan, A, Kremsner, PG, Wahlgren, M and Carlson, J (2000 b) Blood group A antigen is a coreceptor in Plasmodium falciparum rosetting. Infection and Immunity 68, 29712975.CrossRefGoogle ScholarPubMed
Barrera, V, MacCormick, IJC, Czanner, G, Hiscott, PS, White, VA, Craig, AG, Beare, NAV, Culshaw, LH, Zheng, Y, Biddolph, SC, Milner, DA, Kamiza, S, Molyneux, ME, Taylor, TE and Harding, SP (2018) Neurovascular sequestration in paediatric P. falciparum malaria is visible clinically in the retina. Elife 7, e32208. doi: 10.7554/eLife.32208.CrossRefGoogle ScholarPubMed
Baruch, DI, Gormely, JA, Ma, C, Howard, RJ and Pasloske, BL (1996) Plasmodium falciparum erythrocyte membrane protein 1 is a parasitized erythrocyte receptor for adherence to CD36, thrombospondin, and intercellular adhesion molecule 1. Proceedings of the National Academy of Sciences USA 93, 34973502.CrossRefGoogle ScholarPubMed
Bei, AK, Brugnara, C and Duraisingh, MT (2010) In vitro genetic analysis of an erythrocyte determinant of malaria infection. Journal of Infectious Diseases 202, 17221727.CrossRefGoogle ScholarPubMed
Cabrera, A, Neculai, D and Kain, KC (2014) CD36 and malaria: friends or foes? A decade of data provides some answers. Trends in Parasitology 30, 436444.CrossRefGoogle ScholarPubMed
Carlson, J and Wahlgren, M (1992) Plasmodium falciparum erythrocyte rosetting is mediated by promiscuous lectin-like interactions. Journal of Experimental Medicine 176, 13111317.CrossRefGoogle ScholarPubMed
Carlson, J, Helmby, H, Hill, AV, Brewster, D, Greenwood, BM and Wahlgren, M (1990) Human cerebral malaria: association with erythrocyte rosetting and lack of anti-rosetting antibodies. Lancet 336, 14571460.CrossRefGoogle ScholarPubMed
Carlson, J, Ekre, HP, Helmby, H, Gysin, J, Greenwood, BM and Wahlgren, M (1992) Disruption of Plasmodium falciparum erythrocyte rosettes by standard heparin and heparin devoid of anticoagulant activity. American Journal of Tropical Medicine and Hygiene 46, 595602.CrossRefGoogle ScholarPubMed
Chen, Q, Heddini, A, Barragan, A, Fernandez, V, Pearce, SF and Wahlgren, M (2000) The semiconserved head structure of Plasmodium falciparum erythrocyte membrane protein 1 mediates binding to multiple independent host receptors. Journal of Experimental Medicine 192, 110.CrossRefGoogle ScholarPubMed
Ch'ng, JH, Moll, K, Quintana, MEP, Chan, SC, Masters, E, Moles, E, Liu, J, Eriksson, AB and Wahlgren, M (2016) Rosette-disrupting effect of an anti-plasmodial compound for the potential treatment of Plasmodium falciparum malaria complications. Scientific Reports 6, 29317.CrossRefGoogle ScholarPubMed
Chotivanich, KT, Pukrittayakamee, S, Simpson, JA, White, NJ and Udomsangpetch, R (1998) Characteristics of Plasmodium vivax-infected erythrocyte rosettes. American Journal of Tropical Medicine and Hygiene 59, 7376.CrossRefGoogle ScholarPubMed
Chung, WY, Gardiner, DL, Hyland, C, Gatton, M, Kemp, DJ and Trenholme, KR (2005) Enhanced invasion of blood group A1 erythrocytes by Plasmodium falciparum. Molecular and Biochemical Parasitology 144, 128130.CrossRefGoogle ScholarPubMed
Chung, JE, Magis, W, Vu, J, Heo, SJ, Wartiovaara, K, Walters, MC, Kurita, R, Nakamura, Y, Boffelli, D, Martin, DIK, Corn, JE and DeWitt, MA (2019) CRISPR-Cas9 interrogation of a putative fetal globin repressor in human erythroid cells. PLoS ONE 14, e0208237.CrossRefGoogle ScholarPubMed
Clough, B, Atilola, FA and Pasvol, G (1998) The role of rosetting in the multiplication of Plasmodium falciparum: rosette formation neither enhances nor targets parasite invasion into uninfected red cells. British Journal of Haematology 100, 99104.CrossRefGoogle ScholarPubMed
Cockburn, IA, Mackinnon, MJ, O'Donnell, A, Allen, SJ, Moulds, JM, Baisor, M, Bockarie, M, Reeder, JC and Rowe, JA (2004) A human complement receptor 1 polymorphism that reduces Plasmodium falciparum rosetting confers protection against severe malaria. Proceedings of the National Academy of Sciences USA 101, 272277.CrossRefGoogle ScholarPubMed
Cowman, AF, Tonkin, CJ, Tham, WH and Duraisingh, MT (2017) The molecular basis of erythrocyte invasion by malaria parasites. Cell Host and Microbe 22, 232245.CrossRefGoogle ScholarPubMed
Dankwa, S, Chaand, M, Kanjee, U, Jiang, RHY, Nobre, LV, Goldberg, JM, Bei, AK, Moechtar, MA, Grüring, C, Ahouidi, AD, Ndiaye, D, Dieye, TN, Mboup, S, Weekes, MP and Duraisingh, MT (2017) Genetic evidence for erythrocyte receptor glycophorin B expression levels defining a dominant Plasmodium falciparum invasion pathway into human erythrocytes. Infection and Immunity 85, e00074-17.CrossRefGoogle ScholarPubMed
de Agostini, AI, Watkins, SC, Slayter, HS, Youssoufian, H and Rosenberg, RD (1990) Localization of anticoagulantly active heparan sulfate proteoglycans in vascular endothelium: antithrombin binding on cultured endothelial cells and perfused rat aorta. Journal of Cell Biology 111, 12931304.CrossRefGoogle ScholarPubMed
Deans, AM and Rowe, JA (2006) Plasmodium falciparum: rosettes do not protect merozoites from invasion-inhibitory antibodies. Experimental Parasitology 112, 269273.CrossRefGoogle Scholar
Degarege, A, Gebrezgi, MT, Ibanez, G, Wahlgren, M and Madhivanan, P (2019) Effect of the ABO blood group on susceptibility to severe malaria: a systematic review and meta-analysis. Blood Reviews 33, 5362.CrossRefGoogle ScholarPubMed
Dondorp, AM, Pongponratn, E and White, NJ (2004) Reduced microcirculatory flow in severe falciparum malaria: pathophysiology and electron-microscopic pathology. Acta Tropica 89, 309317.CrossRefGoogle ScholarPubMed
Doudna, JA and Charpentier, E (2014) Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 346, 1258096.CrossRefGoogle ScholarPubMed
Doumbo, OK, Thera, MA, Koné, AK, Raza, A, Tempest, LJ, Lyke, KE, Plowe, CV and Rowe, JA (2009) High levels of Plasmodium falciparum rosetting in all clinical forms of severe malaria in African children. American Journal of Tropical Medicine and Hygiene 81, 987993.CrossRefGoogle ScholarPubMed
Frevert, U, Sinnis, P, Cerami, C, Shreffler, W, Takacs, B and Nussenzweig, V (1993) Malaria circumsporozoite protein binds to heparan sulfate proteoglycans associated with the surface membrane of hepatocytes. Journal of Experimental Medicine 177, 12871298.CrossRefGoogle ScholarPubMed
Fried, M and Duffy, PE (1996) Adherence of Plasmodium falciparum to chondroitin sulfate A in the human placenta. Science 272, 15021504.CrossRefGoogle ScholarPubMed
Fry, AE, Griffiths, MJ, Auburn, S, Diakite, M, Forton, JT, Green, A, Richardson, A, Wilson, J, Jallow, M, Sisay-Joof, F, Pinder, M, Peshu, N, Williams, TN, Marsh, K, Molyneux, ME, Taylor, TE, Rockett, KA and Kwiatkowski, DP (2008) Common variation in the ABO glycosyltransferase is associated with susceptibility to severe Plasmodium falciparum malaria. Human Molecular Genetics 17, 567576.CrossRefGoogle ScholarPubMed
Fry, AE, Ghansa, A, Small, KS, Palma, A, Auburn, S, Diakite, M, Green, A, Campino, S, Teo, YY, Clark, TG, Jeffreys, AE, Wilson, J, Jallow, M, Sisay-Joof, F, Pinder, M, Griffiths, MJ, Peshu, N, Williams, TN, Newton, CR, Marsh, K, Molyneux, ME, Taylor, TE, Koram, KA, Oduro, AR, Rogers, WO, Rockett, KA, Sabeti, PC and Kwiatkowski, DP (2009) Positive selection of a CD36 nonsense variant in sub-Saharan Africa, but no association with severe malaria phenotypes. Human Molecular Genetics 18, 26832692.CrossRefGoogle ScholarPubMed
Ghumra, A, Semblat, JP, McIntosh, RS, Raza, A, Rasmussen, IB, Braathen, R, Johansen, FE, Sandlie, I, Mongini, PK, Rowe, JA and Pleass, RJ (2008) Identification of residues in the Cmu4 domain of polymeric IgM essential for interaction with Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1). Journal of Immunology 181, 19882000.CrossRefGoogle Scholar
Ghumra, A, Semblat, JP, Ataide, R, Kifude, C, Adams, Y, Claessens, A, Anong, DN, Bull, PC, Fennell, C, Arman, M, Amambua-Ngwa, A, Walther, M, Conway, DJ, Kassambara, L, Doumbo, OK, Raza, A and Rowe, JA (2012) Induction of strain-transcending antibodies against Group A PfEMP1 surface antigens from virulent malaria parasites. PLoS Pathogens 8, e1002665.CrossRefGoogle Scholar
Goel, S, Palmkvist, M, Moll, K, Joannin, N, Lara, P, Akhouri, RR, Moradi, N, Öjemalm, K, Westman, M, Angeletti, D, Kjellin, H, Lehtiö, J, Blixt, O, Ideström, L, Gahmberg, CG, Storry, JR, Hult, AK, Olsson, ML, von Heijne, G, Nilsson, I and Wahlgren, M (2015) RIFINs are adhesins implicated in severe Plasmodium falciparum malaria. Nature Medicine 21, 314317.CrossRefGoogle ScholarPubMed
Handunnetti, SM, van Schravendijk, MR, Hasler, T, Barnwell, JW, Greenwalt, DE and Howard, RJ (1992) Involvement of CD36 on erythrocytes as a rosetting receptor for Plasmodium falciparum-infected erythrocytes. Blood 80, 20972104.CrossRefGoogle ScholarPubMed
Hawksworth, J, Satchwell, TJ, Meinders, M, Daniels, DE, Regan, F, Thornton, NM, Wilson, MC, Dobbe, JG, Streekstra, GJ, Trakarnsanga, K, Heesom, KJ, Anstee, DJ, Frayne, J and Toye, AM (2018) Enhancement of red blood cell transfusion compatibility using CRISPR-mediated erythroblast gene editing. EMBO Molecular Medicine 10, e8454. doi: 10.15252/emmm.201708454.CrossRefGoogle ScholarPubMed
Heddini, A, Pettersson, F, Kai, O, Shafi, J, Obiero, J, Chen, Q, Barragan, A, Wahlgren, M and Marsh, K (2001) Fresh isolates from children with severe Plasmodium falciparum malaria bind to multiple receptors. Infection and Immunity 69, 58495856.CrossRefGoogle ScholarPubMed
Hviid, L and Jensen, AT (2015) Pfemp1 – a parasite protein family of key importance in Plasmodium falciparum malaria immunity and pathogenesis. Advances in Parasitology 88, 5184.CrossRefGoogle ScholarPubMed
Ito, N, Nishi, K, Kawahara, S, Okamura, Y, Hirota, T, Rand, S, Fechner, G and Brinkmann, B (1990) Difference in the ability of blood group-specific lectins and monoclonal antibodies to recognize the ABH antigens in human tissues. Histochemical Journal 22, 604614.CrossRefGoogle ScholarPubMed
Jaskiewicz, E, Peyrard, T, Kaczmarek, R, Zerka, A, Jodlowska, M and Czerwinski, M (2018) The Gerbich blood group system: old knowledge, new importance. Transfusion Medicine Reviews 32, 111116.CrossRefGoogle Scholar
Juillerat, A, Igonet, S, Vigan-Womas, I, Guillotte, M, Gangnard, S, Faure, G, Baron, B, Raynal, B, Mercereau-Puijalon, O and Bentley, GA (2010) Biochemical and biophysical characterisation of DBL1alpha1-varO, the rosetting domain of PfEMP1 from the VarO line of Plasmodium falciparum. Molecular and Biochemical Parasitology 170, 8492.CrossRefGoogle ScholarPubMed
Juillerat, A, Lewit-Bentley, A, Guillotte, M, Gangnard, S, Hessel, A, Baron, B, Vigan-Womas, I, England, P, Mercereau-Puijalon, O and Bentley, GA (2011) Structure of a Plasmodium falciparum PfEMP1 rosetting domain reveals a role for the N-terminal segment in heparin-mediated rosette inhibition. Proceedings of the National Academy of Sciences USA 108, 52435248.CrossRefGoogle ScholarPubMed
Kanjee, U, Grüring, C, Chaand, M, Lin, KM, Egan, E, Manzo, J, Jones, PL, Yu, T, Barker, R, Weekes, MP and Duraisingh, MT (2017) CRISPR/cas9 knockouts reveal genetic interaction between strain-transcendent erythrocyte determinants of. Proceedings of the National Academy of Sciences USA 114, E9356E9365.CrossRefGoogle Scholar
Kaul, DK, Roth, EF, Nagel, RL, Howard, RJ and Handunnetti, SM (1991) Rosetting of Plasmodium falciparum-infected red blood cells with uninfected red blood cells enhances microvascular obstruction under flow conditions. Blood 78, 812819.CrossRefGoogle ScholarPubMed
Kraemer, SM and Smith, JD (2006) A family affair: var genes, PfEMP1 binding, and malaria disease. Current Opinion in Microbiology 9, 374380.CrossRefGoogle ScholarPubMed
Kun, JF, Schmidt-Ott, RJ, Lehman, LG, Lell, B, Luckner, D, Greve, B, Matousek, P and Kremsner, PG (1998) Merozoite surface antigen 1 and 2 genotypes and rosetting of Plasmodium falciparum in severe and mild malaria in Lambaréné, Gabon. Transactions of the Royal Society of Tropical Medicine and Hygiene 92, 110114.CrossRefGoogle ScholarPubMed
Kurita, R, Suda, N, Sudo, K, Miharada, K, Hiroyama, T, Miyoshi, H, Tani, K and Nakamura, Y (2013) Establishment of immortalized human erythroid progenitor cell lines able to produce enucleated red blood cells. PLoS ONE 8, e59890.CrossRefGoogle ScholarPubMed
Kyriacou, HM, Stone, GN, Challis, RJ, Raza, A, Lyke, KE, Thera, MA, Koné, AK, Doumbo, OK, Plowe, CV and Rowe, JA (2006) Differential var gene transcription in Plasmodium falciparum isolates from patients with cerebral malaria compared to hyperparasitaemia. Molecular and Biochemical Parasitology 150, 211218.CrossRefGoogle ScholarPubMed
Kyriacou, HM, Steen, KE, Raza, A, Arman, M, Warimwe, G, Bull, PC, Havlik, I and Rowe, JA (2007) In vitro inhibition of Plasmodium falciparum rosette formation by Curdlan sulfate. Antimicrobial Agents and Chemotherapy 51, 13211326.CrossRefGoogle ScholarPubMed
Le Scanf, C, Vigan-Womas, I, Contamin, H, Guillotte, M, Bischoff, E and Mercereau-Puijalon, O (2008) Rosetting is associated with increased Plasmodium falciparum in vivo multiplication rate in the Saimiri sciureus monkey. Microbes and Infection 10, 447451.CrossRefGoogle ScholarPubMed
Lee, WC, Malleret, B, Lau, YL, Mauduit, M, Fong, MY, Cho, JS, Suwanarusk, R, Zhang, R, Albrecht, L, Costa, FT, Preiser, P, McGready, R, Renia, L, Nosten, F and Russell, B (2014) Glycophorin C (CD236R) mediates vivax malaria parasite rosetting to normocytes. Blood 123, e100e109.CrossRefGoogle ScholarPubMed
Leffler, EM, Band, G, Busby, GBJ, Kivinen, K, Le, QS, Clarke, GM, Bojang, KA, Conway, DJ, Jallow, M, Sisay-Joof, F, Bougouma, EC, Mangano, VD, Modiano, D, Sirima, SB, Achidi, E, Apinjoh, TO, Marsh, K, Ndila, CM, Peshu, N, Williams, TN, Drakeley, C, Manjurano, A, Reyburn, H, Riley, E, Kachala, D, Molyneux, M, Nyirongo, V, Taylor, T, Thornton, N, Tilley, L, Grimsley, S, Drury, E, Stalker, J, Cornelius, V, Hubbart, C, Jeffreys, AE, Rowlands, K, Rockett, KA, Spencer, CCA, Kwiatkowski, DP and Malaria Genomic Epidemiology Network (2017) Resistance to malaria through structural variation of red blood cell invasion receptors. Science 356, eaam6393. doi: 10.1126/science.aam6393.CrossRefGoogle ScholarPubMed
Leitgeb, AM, Blomqvist, K, Cho-Ngwa, F, Samje, M, Nde, P, Titanji, V and Wahlgren, M (2011) Low anticoagulant heparin disrupts Plasmodium falciparum rosettes in fresh clinical isolates. American Journal of Tropical Medicine and Hygiene 84, 390396.CrossRefGoogle ScholarPubMed
Leitgeb, AM, Charunwatthana, P, Rueangveerayut, R, Uthaisin, C, Silamut, K, Chotivanich, K, Sila, P, Moll, K, Lee, SJ, Lindgren, M, Holmer, E, Färnert, A, Kiwuwa, MS, Kristensen, J, Herder, C, Tarning, J, Wahlgren, M and Dondorp, AM (2017) Inhibition of merozoite invasion and transient de-sequestration by sevuparin in humans with Plasmodium falciparum malaria. PLoS ONE 12, e0188754.CrossRefGoogle ScholarPubMed
Li, JS, Jaggers, J and Anderson, PA (2006) The use of TP10, soluble complement receptor 1, in cardiopulmonary bypass. Expert Reviews in Cardiovascular Therapy 4, 649654.CrossRefGoogle ScholarPubMed
Lowe, BS, Mosobo, M and Bull, PC (1998) All four species of human malaria parasites form rosettes. Transactions of the Royal Society of Tropical Medicine and Hygiene 92, 526.CrossRefGoogle ScholarPubMed
Lublin, DM, Griffith, RC and Atkinson, JP (1986) Influence of glycosylation on allelic and cell-specific Mr variation, receptor processing, and ligand binding of the human complement C3b/C4b receptor. Journal of Biological Chemistry 261, 57365744.Google ScholarPubMed
Luginbuhl, A, Nikolic, M, Beck, HP, Wahlgren, M and Lutz, HU (2007) Complement factor D, albumin, and immunoglobulin G anti-band 3 protein antibodies mimic serum in promoting rosetting of malaria-infected red blood cells. Infection and Immunity 75, 17711777.CrossRefGoogle ScholarPubMed
Maier, AG, Duraisingh, MT, Reeder, JC, Patel, SS, Kazura, JW, Zimmerman, PA and Cowman, AF (2003) Plasmodium falciparum erythrocyte invasion through glycophorin C and selection for Gerbich negativity in human populations. Nature Medicine 9, 8792.CrossRefGoogle ScholarPubMed
Malaria Genomic Epidemiology Network (2014) Reappraisal of known malaria resistance loci in a large multicenter study. Nature Genetics 46, 11971204.CrossRefGoogle Scholar
Marín-Menéndez, A, Bardají, A, Martínez-Espinosa, FE, Bôtto-Menezes, C, Lacerda, MV, Ortiz, J, Cisteró, P, Piqueras, M, Felger, I, Müeller, I, Ordi, J, del Portillo, H, Menéndez, C, Wahlgren, M and Mayor, A (2013) Rosetting in Plasmodium vivax: a cytoadhesion phenotype associated with anaemia. PLoS Neglected Tropical Diseases 7, e2155.CrossRefGoogle ScholarPubMed
Mayer, DC, Jiang, L, Achur, RN, Kakizaki, I, Gowda, DC and Miller, LH (2006) The glycophorin C N-linked glycan is a critical component of the ligand for the Plasmodium falciparum erythrocyte receptor BAEBL. Proceedings of the National Academy of Sciences USA 103, 23582362.CrossRefGoogle ScholarPubMed
McGilvray, ID, Serghides, L, Kapus, A, Rotstein, OD and Kain, KC (2000) Nonopsonic monocyte/macrophage phagocytosis of Plasmodium falciparum-parasitized erythrocytes: a role for CD36 in malarial clearance. Blood 96, 32313240.CrossRefGoogle ScholarPubMed
McMorran, BJ, Wieczorski, L, Drysdale, KE, Chan, JA, Huang, HM, Smith, C, Mitiku, C, Beeson, JG, Burgio, G and Foote, SJ (2012) Platelet factor 4 and Duffy antigen required for platelet killing of Plasmodium falciparum. Science 338, 13481351.CrossRefGoogle ScholarPubMed
Miller, LH, Baruch, DI, Marsh, K and Doumbo, OK (2002) The pathogenic basis of malaria. Nature 415, 673679.CrossRefGoogle ScholarPubMed
Moll, K, Palmkvist, M, Ch'ng, J, Kiwuwa, MS and Wahlgren, M (2015) Evasion of immunity to Plasmodium falciparum: rosettes of blood group A impair recognition of PfEMP1. PLoS ONE 10, e0145120.CrossRefGoogle ScholarPubMed
Moulds, JM (2010) The Knops blood-group system: a review. Immunohematology 26, 27.Google ScholarPubMed
Moulds, JM, Moulds, JJ, Brown, M and Atkinson, JP (1992) Antiglobulin testing for CR1-related (Knops/McCoy/Swain-Langley/York) blood group antigens: negative and weak reactions are caused by variable expression of CR1. Vox Sanguinis 62, 230235.Google ScholarPubMed
Nagayasu, E, Ito, M, Akaki, M, Nakano, Y, Kimura, M, Looareesuwan, S and Aikawa, M (2001) CR1 density polymorphism on erythrocytes of falciparum malaria patients in Thailand. American Journal of Tropical Medicine and Hygiene 64, 15.CrossRefGoogle ScholarPubMed
Ndila, CM, Uyoga, S, Macharia, AW, Nyutu, G, Peshu, N, Ojal, J, Shebe, M, Awuondo, KO, Mturi, N, Tsofa, B, Sepulveda, N, Clark, TG, Band, G, Clarke, G, Rowlands, K, Hubbart, C, Jeffreys, A, Kariuki, S, Marsh, K, Mackinnon, M, Maitland, K, Kwiatkowski, DP, Rockett, KA, Williams, TN and Malaria, GENC (2018) Human candidate gene polymorphisms and risk of severe malaria in children in Kilifi, Kenya: a case-control association study. Lancet Haematology 5, e333e345.CrossRefGoogle ScholarPubMed
Newbold, C, Warn, P, Black, G, Berendt, A, Craig, A, Snow, B, Msobo, M, Peshu, N and Marsh, K (1997) Receptor-specific adhesion and clinical disease in Plasmodium falciparum. American Journal of Tropical Medicine and Hygiene 57, 389398.CrossRefGoogle ScholarPubMed
Niang, M, Bei, AK, Madnani, KG, Pelly, S, Dankwa, S, Kanjee, U, Gunalan, K, Amaladoss, A, Yeo, KP, Bob, NS, Malleret, B, Duraisingh, MT and Preiser, PR (2014) STEVOR is a Plasmodium falciparum erythrocyte binding protein that mediates merozoite invasion and rosetting. Cell Host & Microbe 16, 8193.CrossRefGoogle ScholarPubMed
Nickells, M, Hauhart, R, Krych, M, Subramanian, VB, Geoghegan-Barek, K, Marsh, HC Jr. and Atkinson, JP (1998) Mapping epitopes for 20 monoclonal antibodies to CR1. Clinical and Experimental Immunology 112, 2733.CrossRefGoogle ScholarPubMed
Ochola, LB, Siddondo, BR, Ocholla, H, Nkya, S, Kimani, EN, Williams, TN, Makale, JO, Liljander, A, Urban, BC, Bull, PC, Szestak, T, Marsh, K and Craig, AG (2011) Specific receptor usage in Plasmodium falciparum cytoadherence is associated with disease outcome. PLoS ONE 6, e14741.CrossRefGoogle ScholarPubMed
Opi, DH, Swann, O, Macharia, A, Uyoga, S, Band, G, Ndila, CM, Harrison, E, Thera, MA, Kone, AK, Diallo, DA, Doumbo, OK, Lyke, KE, Plowe, C, Moulds, JM, Shebbe, M, Mturi, N, Peshu, N, Maitland, K, Raza, A, Kwiatkowski, DP, Rockett, KA, Williams, T and Rowe, JA (2018) Two complement receptor one alleles have opposing associations with cerebral malaria and interact with α + thalassaemia. Elife 7, e31579. doi: 10.7554/eLife.31579.CrossRefGoogle ScholarPubMed
Panda, AK, Panda, M, Tripathy, R, Pattanaik, SS, Ravindran, B and Das, BK (2012) Complement receptor 1 variants confer protection from severe malaria in Odisha, India. PLoS ONE 7, e49420.CrossRefGoogle ScholarPubMed
Patel, SS, Mehlotra, RK, Kastens, W, Mgone, CS, Kazura, JW and Zimmerman, PA (2001) The association of the glycophorin C exon 3 deletion with ovalocytosis and malaria susceptibility in the Wosera, Papua New Guinea. Blood 98, 34893491.CrossRefGoogle ScholarPubMed
Pathak, V, Colah, R and Ghosh, K (2016) Correlation between ‘H’ blood group antigen and Plasmodium falciparum invasion. Annals of Hematology 95, 10671075.CrossRefGoogle ScholarPubMed
Pipitaporn, B, Sueblinvong, T, Dharmkrong-at, A and Udomsangpetch, R (2000) Rosetting of Plasmodium falciparum required multiple components of the uninfected erythrocytes. Asian Pacific Journal of Allergy and Immunology 18, 2935.Google ScholarPubMed
Rampengan, TH (1991) Cerebral malaria in children. Comparative study between heparin, dexamethasone and placebo. Paediatrica Indonesiana 31, 5966.Google ScholarPubMed
Reddy, YN, Siedlecki, AM and Francis, JM (2017) Breaking down the complement system: a review and update on novel therapies. Current Opinion in Nephrology and Hypertension 26, 123128.Google ScholarPubMed
Ribacke, U, Moll, K, Albrecht, L, Ahmed Ismail, H, Normark, J, Flaberg, E, Szekely, L, Hultenby, K, Persson, KE, Egwang, TG and Wahlgren, M (2013) Improved in vitro culture of Plasmodium falciparum permits establishment of clinical isolates with preserved multiplication, invasion and rosetting phenotypes. PLoS ONE 8, e69781.CrossRefGoogle ScholarPubMed
Ringwald, P, Peyron, F, Lepers, JP, Rabarison, P, Rakotomalala, C, Razanamparany, M, Rabodonirina, M, Roux, J and Le Bras, J (1993) Parasite virulence factors during falciparum malaria: rosetting, cytoadherence, and modulation of cytoadherence by cytokines. Infection and Immunity 61, 51985204.CrossRefGoogle ScholarPubMed
Robinson, BA, Welch, TL and Smith, JD (2003) Widespread functional specialization of Plasmodium falciparum erythrocyte membrane protein 1 family members to bind CD36 analysed across a parasite genome. Molecular Microbiology 47, 12651278.CrossRefGoogle ScholarPubMed
Rogerson, SJ, Reeder, JC, al-Yaman, F and Brown, GV (1994) Sulfated glycoconjugates as disrupters of Plasmodium falciparum erythrocyte rosettes. American Journal of Tropical Medicine and Hygiene 51, 198203.CrossRefGoogle ScholarPubMed
Rout, R, Dhangadamajhi, G, Mohapatra, BN, Kar, SK and Ranjit, M (2011) High CR1 level and related polymorphic variants are associated with cerebral malaria in eastern-India. Infection Genetics and Evolution 11, 139144.CrossRefGoogle ScholarPubMed
Rout, R, Dhangadamajhi, G, Ghadei, M, Mohapatra, BN, Kar, SK and Ranjit, M (2012) Blood group phenotypes A and B are risk factors for cerebral malaria in Odisha, India. Transactions of the Royal Society of Tropical Medicine and Hygiene 106, 538543.CrossRefGoogle Scholar
Rowe, A, Berendt, AR, Marsh, K and Newbold, CI (1994) Plasmodium falciparum: a family of sulphated glycoconjugates disrupts erythrocyte rosettes. Experimental Parasitology 79, 506516.CrossRefGoogle ScholarPubMed
Rowe, A, Obeiro, J, Newbold, CI and Marsh, K (1995) Plasmodium falciparum rosetting is associated with malaria severity in Kenya. Infection and Immunity 63, 23232326.CrossRefGoogle ScholarPubMed
Rowe, JA, Moulds, JM, Newbold, CI and Miller, LH (1997) P. falciparum rosetting mediated by a parasite-variant erythrocyte membrane protein and complement-receptor 1. Nature 388, 292295.CrossRefGoogle ScholarPubMed
Rowe, JA, Rogerson, SJ, Raza, A, Moulds, JM, Kazatchkine, MD, Marsh, K, Newbold, CI, Atkinson, JP and Miller, LH (2000) Mapping of the region of complement receptor (CR) 1 required for Plasmodium falciparum rosetting and demonstration of the importance of CR1 in rosetting in field isolates. Journal of Immunology 165, 63416346.CrossRefGoogle ScholarPubMed
Rowe, JA, Obiero, J, Marsh, K and Raza, A (2002) Short report: positive correlation between rosetting and parasitemia in Plasmodium falciparum clinical isolates. American Journal of Tropical Medicine and Hygiene 66, 458460.CrossRefGoogle ScholarPubMed
Rowe, JA, Handel, IG, Thera, MA, Deans, AM, Lyke, KE, Koné, A, Diallo, DA, Raza, A, Kai, O, Marsh, K, Plowe, CV, Doumbo, OK and Moulds, JM (2007) Blood group O protects against severe Plasmodium falciparum malaria through the mechanism of reduced rosetting. Proceedings of the National Academy of Sciences USA 104, 1747117476.CrossRefGoogle ScholarPubMed
Rowe, JA, Claessens, A, Corrigan, RA and Arman, M (2009 a) Adhesion of Plasmodium falciparum-infected erythrocytes to human cells: molecular mechanisms and therapeutic implications. Expert Reviews in Molecular Medicine 11, e16.CrossRefGoogle ScholarPubMed
Rowe, JA, Opi, DH and Williams, TN (2009 b) Blood groups and malaria: fresh insights into pathogenesis and identification of targets for intervention. Current Opinion in Hematology 16, 480487.CrossRefGoogle ScholarPubMed
Saiwaew, S, Sritabal, J, Piaraksa, N, Keayarsa, S, Ruengweerayut, R, Utaisin, C, Sila, P, Niramis, R, Udomsangpetch, R, Charunwatthana, P, Pongponratn, E, Pukrittayakamee, S, Leitgeb, AM, Wahlgren, M, Lee, SJ, Day, NP, White, NJ, Dondorp, AM and Chotivanich, K (2017) Effects of sevuparin on rosette formation and cytoadherence of Plasmodium falciparum infected erythrocytes. PLoS ONE 12, e0172718.CrossRefGoogle ScholarPubMed
Schmidt, CQ, Kennedy, AT and Tham, WH (2015) More than just immune evasion: Hijacking complement by Plasmodium falciparum. Molecular Immunology 67, 7184.CrossRefGoogle ScholarPubMed
Scholander, C, Treutiger, CJ, Hultenby, K and Wahlgren, M (1996) Novel fibrillar structure confers adhesive property to malaria-infected erythrocytes. Nature Medicine 2, 204208.CrossRefGoogle ScholarPubMed
Scully, EJ, Shabani, E, Rangel, GW, Gruring, C, Kanjee, U, Clark, MA, Chaand, M, Kurita, R, Nakamura, Y, Ferreira, MU and Duraisingh, MT (2019) Generation of an immortalized erythroid progenitor cell line from peripheral blood: a model system for the functional analysis of Plasmodium spp. invasion. American Journal of Hematology 94, 963974.CrossRefGoogle Scholar
Semblat, JP, Ghumra, A, Czajkowsky, DM, Wallis, R, Mitchell, DA, Raza, A and Rowe, JA (2015) Identification of the minimal binding region of a Plasmodium falciparum IgM binding PfEMP1 domain. Molecular and Biochemical Parasitology 201, 7682.CrossRefGoogle ScholarPubMed
Silverstein, RL and Febbraio, M (2009) CD36, a scavenger receptor involved in immunity, metabolism, angiogenesis, and behavior. Science Signaling 2, re3.CrossRefGoogle Scholar
Sim, BK, Chitnis, CE, Wasniowska, K, Hadley, TJ and Miller, LH (1994) Receptor and ligand domains for invasion of erythrocytes by Plasmodium falciparum. Science 264, 19411944.CrossRefGoogle ScholarPubMed
Sinha, S, Jha, GN, Anand, P, Qidwai, T, Pati, SS, Mohanty, S, Mishra, SK, Tyagi, PK, Sharma, SK, Venkatesh, V and Habib, S (2009) CR1 levels and gene polymorphisms exhibit differential association with falciparum malaria in regions of varying disease endemicity. Human Immunology 70, 244250.CrossRefGoogle ScholarPubMed
Song, B, Fan, Y, He, W, Zhu, D, Niu, X, Wang, D, Ou, Z, Luo, M and Sun, X (2015) Improved hematopoietic differentiation efficiency of gene-corrected beta-thalassemia induced pluripotent stem cells by CRISPR/Cas9 system. Stem Cells and Development 24, 10531065.CrossRefGoogle ScholarPubMed
Stevenson, L, Huda, P, Jeppesen, A, Laursen, E, Rowe, JA, Craig, A, Streicher, W, Barfod, L and Hviid, L (2015 a) Investigating the function of Fc-specific binding of IgM to Plasmodium falciparum erythrocyte membrane protein 1 mediating erythrocyte rosetting. Cellular Microbiology 17, 819831.CrossRefGoogle ScholarPubMed
Stevenson, L, Laursen, E, Cowan, GJ, Bandoh, B, Barfod, L, Cavanagh, DR, Andersen, GR and Hviid, L (2015 b) α2-Macroglobulin can crosslink multiple Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) molecules and may facilitate adhesion of parasitized erythrocytes. PLoS Pathogens 11, e1005022.CrossRefGoogle ScholarPubMed
Stoler-Barak, L, Moussion, C, Shezen, E, Hatzav, M, Sixt, M and Alon, R (2014) Blood vessels pattern heparan sulfate gradients between their apical and basolateral aspects. PLoS ONE 9, e85699.CrossRefGoogle ScholarPubMed
Teeranaipong, P, Ohashi, J, Patarapotikul, J, Kimura, R, Nuchnoi, P, Hananantachai, H, Naka, I, Putaporntip, C, Jongwutiwes, S and Tokunaga, K (2008) A functional single-nucleotide polymorphism in the CR1 promoter region contributes to protection against cerebral malaria. Journal of Infectious Diseases 198, 18801891.CrossRefGoogle ScholarPubMed
Tekeste, Z and Petros, B (2010) The ABO blood group and Plasmodium falciparum malaria in Awash, Metehara and Ziway areas, Ethiopia. Malaria Journal 9, 280.CrossRefGoogle ScholarPubMed
Tetteh-Quarcoo, PB, Schmidt, CQ, Tham, WH, Hauhart, R, Mertens, HD, Rowe, A, Atkinson, JP, Cowman, AF, Rowe, JA and Barlow, PN (2012) Lack of evidence from studies of soluble protein fragments that Knops blood group polymorphisms in complement receptor-type 1 are driven by malaria. PLoS ONE 7, e34820.CrossRefGoogle ScholarPubMed
Thathy, V, Moulds, JM, Guyah, B, Otieno, W and Stoute, JA (2005) Complement receptor 1 polymorphisms associated with resistance to severe malaria in Kenya. Malaria Journal 4, 54.CrossRefGoogle ScholarPubMed
Theron, M, Cross, N, Cawkill, P, Bustamante, LY and Rayner, JC (2018) An in vitro erythrocyte preference assay reveals that Plasmodium falciparum parasites prefer Type O over Type A erythrocytes. Scientific Reports 8, 8133.CrossRefGoogle Scholar
Thielen, AJF, Zeerleder, S and Wouters, D (2018) Consequences of dysregulated complement regulators on red blood cells. Blood Reviews 32, 280288.CrossRefGoogle ScholarPubMed
Trakarnsanga, K, Griffiths, RE, Wilson, MC, Blair, A, Satchwell, TJ, Meinders, M, Cogan, N, Kupzig, S, Kurita, R, Nakamura, Y, Toye, AM, Anstee, DJ and Frayne, J (2017) An immortalized adult human erythroid line facilitates sustainable and scalable generation of functional red cells. Nature Communications 8, 14750.CrossRefGoogle ScholarPubMed
Treutiger, CJ, Hedlund, I, Helmby, H, Carlson, J, Jepson, A, Twumasi, P, Kwiatkowski, D, Greenwood, BM and Wahlgren, M (1992) Rosette formation in Plasmodium falciparum isolates and anti-rosette activity of sera from Gambians with cerebral or uncomplicated malaria. American Journal of Tropical Medicine and Hygiene 46, 503510.CrossRefGoogle ScholarPubMed
Treutiger, CJ, Scholander, C, Carlson, J, McAdam, KP, Raynes, JG, Falksveden, L and Wahlgren, M (1999) Rouleaux-forming serum proteins are involved in the rosetting of Plasmodium falciparum-infected erythrocytes. Experimental Parasitology 93, 215224.CrossRefGoogle ScholarPubMed
Udomsangpetch, R, Wåhlin, B, Carlson, J, Berzins, K, Torii, M, Aikawa, M, Perlmann, P and Wahlgren, M (1989) Plasmodium falciparum-infected erythrocytes form spontaneous erythrocyte rosettes. Journal of Experimental Medicine 169, 18351840.CrossRefGoogle ScholarPubMed
Udomsangpetch, R, Webster, HK, Pattanapanyasat, K, Pitchayangkul, S and Thaithong, S (1992) Cytoadherence characteristics of rosette-forming Plasmodium falciparum. Infection and Immunity 60, 44834490.CrossRefGoogle ScholarPubMed
Udomsangpetch, R, Todd, J, Carlson, J and Greenwood, BM (1993) The effects of hemoglobin genotype and ABO blood group on the formation of rosettes by Plasmodium falciparum-infected red blood cells. American Journal of Tropical Medicine and Hygiene 48, 149153.CrossRefGoogle ScholarPubMed
Udomsanpetch, R, Thanikkul, K, Pukrittayakamee, S and White, NJ (1995) Rosette formation by Plasmodium vivax. Transactions of the Royal Society of Tropical Medicine and Hygiene 89, 635637.CrossRefGoogle ScholarPubMed
Uyoga, S, Skorokhod, OA, Opiyo, M, Orori, EN, Williams, TN, Arese, P and Schwarzer, E (2012) Transfer of 4-hydroxynonenal from parasitized to non-parasitized erythrocytes in rosettes. Proposed role in severe malaria anemia. British Journal of Haematology 157, 116124.CrossRefGoogle ScholarPubMed
van Schravendijk, M, Handunnetti, S, Barnwell, J and Howard, R (1992) Normal human erythrocytes express CD36, an adhesion molecule of monocytes, platelets and endothelial cells. Blood 80, 21052114.CrossRefGoogle ScholarPubMed
Vigan-Womas, I, Guillotte, M, Le Scanf, C, Igonet, S, Petres, S, Juillerat, A, Badaut, C, Nato, F, Schneider, A, Lavergne, A, Contamin, H, Tall, A, Baril, L, Bentley, GA and Mercereau-Puijalon, O (2008) An in vivo and in vitro model of Plasmodium falciparum rosetting and autoagglutination mediated by varO, a group A var gene encoding a frequent serotype. Infection and Immunity 76, 55655580.CrossRefGoogle Scholar
Vigan-Womas, I, Guillotte, M, Juillerat, A, Vallieres, C, Lewit-Bentley, A, Tall, A, Baril, L, Bentley, GA and Mercereau-Puijalon, O (2011) Allelic diversity of the Plasmodium falciparum erythrocyte membrane protein 1 entails variant-specific red cell surface epitopes. PLoS ONE 6, e16544.CrossRefGoogle ScholarPubMed
Vigan-Womas, I, Guillotte, M, Juillerat, A, Hessel, A, Raynal, B, England, P, Cohen, JH, Bertrand, O, Peyrard, T, Bentley, GA, Lewit-Bentley, A and Mercereau-Puijalon, O (2012) Structural basis for the ABO blood-group dependence of Plasmodium falciparum rosetting. PLoS Pathogens 8, e1002781.CrossRefGoogle ScholarPubMed
Vogt, AM, Barragan, A, Chen, Q, Kironde, F, Spillmann, D and Wahlgren, M (2003) Heparan sulfate on endothelial cells mediates the binding of Plasmodium falciparum-infected erythrocytes via the DBL1alpha domain of PfEMP1. Blood 101, 24052411.CrossRefGoogle ScholarPubMed
Vogt, AM, Winter, G, Wahlgren, M and Spillmann, D (2004) Heparan sulphate identified on human erythrocytes: a Plasmodium falciparum receptor. Biochemical Journal 381, 593597.CrossRefGoogle ScholarPubMed
Wahlgren, M, Carlson, J, Udomsangpetch, R and Perlmann, P (1989) Why do Plasmodium falciparum-infected erythrocytes form spontaneous erythrocyte rosettes? Parasitology Today 5, 183185.CrossRefGoogle Scholar
Wang, CW and Hviid, L (2015) Rifins, rosetting, and red blood cells. Trends in Parasitology 31, 285286.CrossRefGoogle ScholarPubMed
Warimwe, GM, Fegan, G, Musyoki, JN, Newton, CR, Opiyo, M, Githinji, G, Andisi, C, Menza, F, Kitsao, B, Marsh, K and Bull, PC (2012) Prognostic indicators of life-threatening malaria are associated with distinct parasite variant antigen profiles. Science Translational Medicine 4, 129ra145.CrossRefGoogle ScholarPubMed
White, NJ, Turner, GD, Day, NP and Dondorp, AM (2013) Lethal malaria: Marchiafava and Bignami were right. Journal of Infectious Diseases 208, 192198.CrossRefGoogle ScholarPubMed
Wilson, JG, Murphy, EE, Wong, WW, Klickstein, LB, Weis, JH and Fearon, DT (1986) Identification of a restriction fragment length polymorphism by a CR1 cDNA that correlates with the number of CR1 on erythrocytes. Journal of Experimental Medicine 164, 5059.CrossRefGoogle ScholarPubMed
Wilson, MC, Trakarnsanga, K, Heesom, KJ, Cogan, N, Green, C, Toye, AM, Parsons, SF, Anstee, DJ and Frayne, J (2016) Comparison of the proteome of adult and cord erythroid cells, and changes in the proteome following reticulocyte maturation. Molecular and Cellular Proteomics 15, 19381946.CrossRefGoogle ScholarPubMed
Wolofsky, KT, Ayi, K, Branch, DR, Hult, AK, Olsson, ML, Liles, WC, Cserti-Gazdewich, CM and Kain, KC (2012) ABO blood groups influence macrophage-mediated phagocytosis of Plasmodium falciparum-infected erythrocytes. PLoS Pathogens 8, e1002942.CrossRefGoogle ScholarPubMed
World Health Organisation (1986) Severe and complicated malaria. World Health Organization Malaria Action Programme. Transactions of the Royal Society of Tropical Medicine and Hygiene 80(suppl.), 3–50.Google Scholar
Yam, XY, Niang, M, Madnani, KG and Preiser, PR (2017) Three is a crowd – new insights into rosetting in Plasmodium falciparum. Trends in Parasitology 33, 309320.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Plasmodium falciparum rosetting in an in vitro culture. Rosettes consisting of clusters of infected and uninfected erythrocytes are shown. Inset image shows a single infected erythrocyte (centre) and three adherent uninfected erythrocytes. Images were taken using a Yenway microscope camera on a Leica DM LB2 fluorescent microscope using the ×40 and ×100 (inset) objectives.

Figure 1

Fig. 2. Parasite-derived adhesion ligands and host receptors that interact to form rosettes. UE, uninfected erythrocyte; IE, infected erythrocyte; GAGs, glycosaminoglycans; HS, heparan sulphate; CS, chondroitin sulphate; CR1, complement receptor 1; GYPA, glycophorin A; GYPC, glycophorin C. Dotted lines represent proposed host receptors for each parasite ligand.

Figure 2

Table 1. Summary of host erythrocyte receptors for Plasmodium falciparum rosetting

Figure 3

Fig. 3. Diagram of the ABO blood group sugars. Schematic representation of the terminal structure of the A (blue square), B (purple) H (green; H is the antigen carried on blood group O erythrocytes) and Bombay (orange) antigens. Yellow circle: D-Galactose (Gal), yellow square: N-acetyl-D-galactosamine (GalNac), red triangle: L-Fucose (Fuc). The symbols α and β indicate the position of the hydroxyl group and the numbers indicate the specific carbon atoms that are linked between the sugars. The H, A and B antigens are synthesized by a series of glycosyltransferase enzymes that add monosaccharides to create oligosaccharide chains attached to lipids and proteins in the erythrocyte membrane.

Figure 4

Table 2. Key areas for future research on rosetting receptors