Next Article in Journal
Innate Lymphoid Cells and Their Role in the Immune Response to Infections
Previous Article in Journal
Regulation of the Activity of the Dual Leucine Zipper Kinase by Distinct Mechanisms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

P. falciparum Invasion and Erythrocyte Aging

by
María Fernanda Alves-Rosa
,
Nicole M. Tayler
,
Doriana Dorta
,
Lorena M. Coronado
and
Carmenza Spadafora
*
Center of Cellular and Molecular Biology of Diseases, Instituto de Investigaciones Científicas y Servicio de Alta Tecnología (INDICASAT AIP), City of Knowledge, Panama City 0843-01103, Panama
*
Author to whom correspondence should be addressed.
Current address: Estée Lauder Panamá, PH Plaza del Este Piso 7, Boulevard Costa del Este, Juan Díaz, Panama City 0843-01103, Panama.
Cells 2024, 13(4), 334; https://doi.org/10.3390/cells13040334
Submission received: 16 January 2024 / Revised: 1 February 2024 / Accepted: 6 February 2024 / Published: 12 February 2024
(This article belongs to the Section Cellular Aging)

Abstract

:
Plasmodium parasites need to find red blood cells (RBCs) that, on the one hand, expose receptors for the pathogen ligands and, on the other hand, maintain the right geometry to facilitate merozoite attachment and entry into the red blood cell. Both characteristics change with the maturation of erythrocytes. Some Plasmodia prefer younger vs. older erythrocytes. How does the life evolution of the RBC affect the invasion of the parasite? What happens when the RBC ages? In this review, we present what is known up until now.

Graphical Abstract

1. Introduction: The Malaria Burden

After more than 120 years of research, malaria is still on the list of the deadliest tropical diseases. Most commonly found in tropical and subtropical countries, the African region bears the highest burden of malaria cases and fatalities, which are primarily attributable to the Plasmodium falciparum species, but every year nearly half of the world population is at risk of contracting malaria. In 2021, there were an estimated 247 million cases of malaria worldwide and the estimated number of malaria deaths stood at 619,000 [1].
As if these numbers were not daunting enough, the increase in the average temperature and precipitation patterns is leading to an increase in the vulnerability to the disease, turning vectors with historically limited range of transmission into high-reaching ones, rendering populations more vulnerable due to their reduced acquired immunity [2].
The use of drugs creates pressure on the parasites, forcing them to be selected for the presence of resistance genes, for example, the pfcrt gene, which is known to confer resistance against chloroquine [3,4,5], and the pfmdr1 gene [6,7], which enable the parasite to expel multiple drugs. A study from the last decade reported that approximately 2 million known drugs were screened for antimalarial activity, of which more than 13,000 were found to be effective against the intraerythrocytic stage of the parasite [8].

2. Malarial Erythrocyte Receptors and Plasmodium Ligands

The identification of crucial ligand–receptor interactions involved in parasite erythrocyte entry is a venue that has been explored to tackle the disease, as their blockage may lead to the development of multivalent and more effective vaccines [9,10].
In studies focused on determining the proteins involved in the invasion process of P. falciparum, it was made clear that the physical interaction of the merozoite (the invading stage) with its proper receptor in the red blood cell (RBC) is a well-orchestrated process and that the integrity of the RBC membrane with its scaffold of proteins is crucial for this interaction to be successful. Invasion mechanisms used by the merozoite involve several steps that have been well described throughout the years [11].
As far as it is known, the first merozoite–host interaction is reversible, but it becomes more stable as the merozoite starts expressing some proteins on its surface. Microneme proteins form strong linkages between the parasite and the erythrocyte, helping rhoptry proteins in the final entry of the merozoite into the host cell. These initial steps in Plasmodium invasion are followed by the deformation of the erythrocyte membrane to form a parasitophorous vacuole membrane, in which the merozoite encapsulates itself after the shedding of surface proteins into the extracellular environment [12,13].
Based on the host molecules used by the parasite and their response to enzymatic treatment, two main invasion pathways for P. falciparum have been defined. One is based on the interaction of the parasite with sialic acid (SA) residues and the other pathway works in a manner that is independent of these molecules [14].
The known receptors for the SA-dependent pathways are Glycophorin (Gly) A [15,16,17,18]. Glycophorins are heavily glycosylated transmembrane sialo glycoproteins, which partly explains why these multi-abundant proteins in the erythrocyte membrane are responsible for the SA-dependent invasion by P. falciparum. These molecules have been characterized as carrying the antigens for several human blood groups: Gly A and Gly B carry the MN and SS blood groups, and Gly C carries the Gerbich blood group system [19,20]. Given the abundance of Glys on the RBC cell surface, it is likely that they also serve as substrates for glycosylation, which provides the RBC with a negatively charged complex glycan “coat” that allows for their circulation without adherence to other cells or walls of blood vessels [21].
As expected, the SA-dependent pathway efficiency is reduced upon enzymatic treatment of the erythrocyte with neuraminidase and the subsequent removal of sialic acid residues [22]. For this pathway, there is a redundancy in the parasite, comprising mainly several ligands of P. falciparum belonging to the Erythrocyte Binding Ligand (EBL) proteins and Erythrocyte Binding Antigens (EBAs).
Four main P. falciparum ligands have been identified for the SA-independent invasion: erythrocyte-binding antigen-175 (EBA-175), erythrocyte-binding antigen-181 (EBA-181), erythrocyte-binding ligand-1 (EBL-1), and erythrocyte-binding antigen-140 (EBA-140) [23]. It is generally accepted that Gly A is the receptor for the EBA-175 ligand, but P. falciparum can also invade erythrocytes using Gly B through EBL-1 [24] and through P. falciparum EBA-140 using Gly C as a receptor [25]. Regarding these receptors, Dankwa et al. reported that GPA and GPB are the key ones involved in the P. falciparum invasion route into human erythrocytes [26], probably due in part to their abundance on the surface of the erythrocyte.
The SA-independent parasite invasion ligands are dominated by the reticulocyte binding homologs family (PfRBL or PfRh) [18,27]. The family of sialic acid-independent or neuraminidase-resistant receptors [28,29,30] includes Receptor Z [27], Complement Receptor 1 (CR1) [31,32], Basigin (BSG) [33,34,35], and CD55 [36].
Receptor Z is used by the Plasmodium falciparum W2-mef and 3D7 strains. In related studies, the P. falciparum reticulocyte binding protein homolog 2b (PfRH2b) bound to RBC via this putative receptor, which was resistant to trypsin and neuraminidase treatment but sensitive to chymotrypsin [11]. Another ligand proposed for receptor Z is the Erythrocyte binding antigen-181 (EBA-181), for which no receptor is known [37].
The Complement Receptor 1 (CR1), also known as CD35 (cluster of differentiation 35), is an important polymorphic glycoprotein on the membrane surface of erythrocytes and many other nucleated cells. It is highly sensitive to treatment with trypsin. Along with other proteins, CR1 is also a regulator of the complement system, where it helps the RBC to avoid autologous complement attack [38].
The activation of a complement on the RBC membrane may occur due to the deposition of naturally occurring IgG autoantibodies, leading to the accumulation of C3b/C4b on the cell surface. In this scenario, C3b molecules bound to CR1 are deactivated by Complement Factor I, preventing erythrocytes against complement or phagocytosis-mediated destruction [38,39].
This feature also takes place in P. falciparum-infected erythrocytes. In ring-stage infections, phagocytosis is almost entirely dependent on the intervention of CR1 complement activation. This role of CR1 is reduced, however, when more mature forms of the parasite become present.
CR1 levels have been associated with malarial susceptibility and/or severity of the disease in different population groups [40]. Spadafora et al. observed that the amount of these molecules per erythrocyte varies depending on the donor and that levels of CR1 decrease in older erythrocytes when compared with younger ones [31]. The P. falciparum protein reticulocyte homology 4 (PfRh4) was reported as the CR1 ligand on the parasite [41].
Basigin (BSG), also called extracellular matrix metalloproteinase inducer (EMMPRIN) or CD147, is a type I transmembrane protein of the immunoglobulin (Ig) superfamily. This molecule was shown to associate with monocarboxylate transporters (MCTs) or plasma membrane calcium ATPases (PMCAs) expressed on the RBC surface [42,43]. MCTs are involved in a proton-coupled exchange of lactate or pyruvate across plasma membranes. The actions of both MCTs and PMCAs have been associated with the development of severe malaria [44,45], especially the presence of polymorphisms in PMCA [46].
The PfRh5 protein was found to bind specifically to BSG [33] at two highly glycosylated extracellular Ig domains [42], both of which contact PfRh5 [47]. The specific blockade of the PfRH5 binding to Basigin, coupled with PMCA or Basigin-MCT1, inhibits the parasitic invasion process [35]. PfRh5 is the only essential invasion ligand within the PfRh family. Both laboratory-adapted and field strains of P. falciparum follow a Basigin-dependent invasion route, corroborating the critical nature of these receptor-ligand interactions in erythrocyte invasion [48].
Decay-Accelerating Factor (DAF), also known as CD55, is a GPI-linked complement-regulatory protein that protects cells from lysis by complement [49]. DAF is an essential host factor for P. falciparum invasion. CD55-null erythrocytes are refractory to invasion in vitro by isolates of P. falciparum due to the lack of parasite attachment to the erythrocyte surface [36]. In addition, CD55 is a receptor for bacterial and viral pathogens on epithelial cells [50]. Figure 1 displays the main ligand and receptor couples described here.
While it is known that there are other molecules involved in the parasite entry process into the RBC, the identity of some of their cellular receptors or ligands in the parasite is uncertain, and in some other cases, they belong to the same family of the ones depicted in Figure 1 [51,52,53,54,55].
All these data suggest that the more receptors available on the RBC surface, the more efficient the invasion process, and thus, the parasite virulence. As a result, any qualitative or quantitative change in the RBC cell surface could negatively modulate the parasite invasion efficiency.

3. Erythrocyte Age-Related Markers

The aging of RBCs is a natural process that occurs during their circulation in the vascular system. Normal human erythrocytes have an average life span of about 115 days in circulation, after which they are engulfed by macrophages [56,57].
The molecular mechanism by which macrophages discriminate between new and aged erythrocytes is a subject of study. There is, at least, a consensus that this process involves the appearance in the erythrocyte of different age-related markers, including changes in the composition of the cell membrane [58].
One of the most commonly accepted erythrocyte aging signals is the aggregation of the anion exchanger Band 3 [59]. These molecules cluster together on the membrane due to the association of hemichromes on the cytoplasmic domain of Band 3 [60]. This protein undergoes conformational dynamics localized in a region previously identified as an erythrocyte senescence epitope and may act as the “molecular clock” in erythrocyte senescence [61].
Naturally occurring antibodies that bind to clustered Band 3 were proposed as mediators of senescent RBC clearance [62,63]. The elimination of aged RBCs also involves phagocytosis by liver and spleen macrophages triggered by the binding of autologous IgGs to the aggregated Band 3 [64,65].
Band 3 plays an extremely important role as the organizational center of erythrocytes. This protein is linked to multiple complexes through adaptor proteins and constitutes a component of the cytoskeletal spectrin-based network [66]. It also serves as a binding site for several membrane-associated proteins [67], including P. falciparum glutamic acid–rich protein (PfGARP) [68] and Glycosyl-phosphatidylinositol-anchored micronemal antigen (GAMA) [69], both of which are erythrocyte-binding proteins known to be involved in malarial parasite invasion. PfGARPs are potential ligands that bind to the surface of intact RBCs with Band 3 as their receptor [68]. They are detected in the trophozoite and schizont stages of the parasite, but not in the ring stage.
Like other proteins on the surface of erythrocytes, CR1 levels also decrease as a result of senescence [70]. Ripoche et al. analyzed the loss of these molecules from erythrocytes by measuring the CR1 activity and found that older erythrocytes showed a decrease in activity relative to younger ones [71].
CR1 loss has also been tied to vesicular liberation from the membrane of erythrocytes. Observing the natural degradation of the erythrocytic plasma membrane, Pascual et al. proposed that intact CR1 molecules were released in phospholipid vesicles [72], which contributes to the loss of erythrocyte surface and volume. Nowadays, the discovery of ubiquitous extracellular vesicles and microvesicles, which are also shed by RBCs, can explain this observation. The release of MVs is considered a strategy of the erythrocyte to remodel, condense, and change the surface area to volume ratio, in the case of younger, bigger reticulocytes [73] and to dispose of some wasted proteins and contents in the case of adult erythrocytes [74]. Among the proteins liberated with MVs are membrane proteins, such as glycophorins [75]. However, when studying EVs released from P. falciparum-infected erythrocytes, Correa et al. observed that RBC CR1, in the same way as glycophorin A, is also grabbed from the RBC membrane into shed particles, confirming the report of Pascual et al. This finding suggests that this protein is regularly lost during MV biogenesis, together with glycophorins: a process that should deeply change the shape of the erythrocyte [76].
Trautsch C et al. reported lower levels of glycoproteins and acetylcholine in older RBC populations in vitro and in vivo [77]. Miyahara K et al. demonstrated an in vivo loss of glycoconjugate molecules, i.e., sialoglycoproteins, macroglycolipids, low-molecular-weight glycolipids, and Band 3 glycolipids on the surface of normal aging or diabetic human erythrocytes [78].
The presence of cell adhesion molecules (CAMs), such as CD44, CD47, BSG, Gly-A, and phosphatidylserine (PS), decreases upon erythrocyte aging [79,80].
Regarding CD44, this molecule exists in several isoforms, but RBCs only express the shortest one. Its role is not fully understood, aside from that of encoding the Indian blood group antigens [81]. When erythroid cell CD44 expression was abrogated by CRISPR/Cas9 genome editing, erythropoiesis and enucleation of orthochromatic erythroblasts were not affected [82]. However, CD44 was shown to play a role in Plasmodium falciparum invasion. The parasite requires CD44 as a co-receptor for both Gly-A ↔ EBA-175 and Gly-C ↔ EBA-140 interactions. Moreover, EBA-175 triggers the phosphorylation of cytoskeletal proteins through a CD44-dependent pathway [83].
The highly glycosylated RBC surface protein CD47 signals the regulatory protein alpha (SIRPα) on the macrophage. SIRPα signals for inhibition of both phagocytosis and inflammatory response. This interaction leads to erythrocytes phagocytosis suppression [84]. Physiologically, CD47 functions to prevent RBCs from undergoing phagocytosis [85].
During the aging of red blood cells, surface components, such as sialoglycoproteins, specifically sialic acids (SAs), undergo detachment [86,87]. Human erythrocyte SAs were found to contain 95% N-acetylneuraminic acid (NANA) [88]. The electrical charge that arises from NANA carboxyls and other ionizable chemicals is one of the major physicochemical factors that govern cell interactions. Negatively charged RBC membrane promotes intercellular electrostatic repulsion avoiding erythrocyte aggregation [89].
An enhancement of the phagocytosis of P. falciparum-infected red blood cells was observed when CD47-SIRPα engagement was disrupted using either anti-SIRPα antibodies or SIRPα-Fc fusion protein. These findings highlight the significance of CD47-SIRPα interactions in the innate regulation of malaria [90].
Basigin, which is expressed on erythrocytes, plays a critical role in recirculating mature erythrocytes from the spleen into the general circulation. Coste et al. showed that erythrocytes accumulate in the red pulp of spleens of anti-CD147-treated mice [91,92].
Another feature of RBC aging is changes in the intracellular Ca2+ concentration and translocation of PS from the inner to the outer membrane, leading to PS exposure. This is attributable to a decreased flippase activity in senescent erythrocytes [93] and the activation of scramblase [94].
As mentioned elsewhere, senescent erythrocytes show a shrinkage effect caused by the loss of cell membrane, which is manifested in the form of microvesicles shed into the surrounding media [95,96]. As observed both in vivo and in vitro, under various conditions, vesicles usually contain hemoglobin, lipids, band 3, glycophorins, actin, lipid raft proteins, caspases, Fas, and extracellular PS [97,98,99]. The proteins contained in microvesicles can be derived from plasma or small G proteins and they are rapidly removed from circulation, as signaled by the presence of PS and immunoglobulin G (IgG) on their surface [100,101]. Cell contents and membrane parts, carried outside by microparticles, result in a diminished ability of the erythrocyte to deform.
A reduction in deformability is a feature of an RBC’s physiological aging and it is associated with mostly uncharacterized metabolic and structural alterations [102]. To assess deformability, the first method used was simple observation under the microscope, but over time, different methods have been developed, which have increased in complexity and accuracy: micropipette aspiration [103,104], simple and microfluidic filtration [105,106], atomic force microscopy [107], erythrocyte shape recovery [108,109], optical tweezers [110,111], and laser diffractometry [112].
Using a range of the above techniques, a strong and meaningful intercorrelation was established between the deformability of RBCs and the presence of certain proteins [113].
Senescent cells, which are unable to squeeze through narrow splenic slits, are retained and cleared from the blood circulation, preventing microvessels from being clogged by aged, rigid RBCs. The biconcave shape that is natural to erythrocytes is an important contributor to maintaining adequate blood flow [114] because it contributes to this flexibility. Certainly, the retention of less deformable RBCs is a crucial factor that influences the pathogenesis of malaria [115]. Studies using optical tweezers revealed mechanical changes in P. falciparum-infected RBCs. In fact, an increase of up to 10-fold in the continuous force–displacement curves was detected for different intracellular developmental stages of the parasite [116].

4. Effect of Plasmodium spp. Invasion on Mechanical and Molecular Erythrocyte Properties

When erythrocytes are infected with P. falciparum, their natural aging process is accelerated and many of the RBC-aging features appear in infected RBCs, even though they are young [117]. The intra-erythrocytic phase of Plasmodium infection is initiated by erythrocyte invasion by merozoites, followed by the asexual replication cycle, which progresses through the ring, trophozoite, and schizont stages, until the new release of merozoites, a step which is associated with the clinical symptoms of malaria [118].
It has been well documented that erythrocytes that have been infected with the Plasmodium parasite undergo changes in their membrane composition, particularly in components such as phospholipids and cholesterol, and how these are organized [119,120]. In addition, P. falciparum also elicits the formation of hemichromes and the aggregation of Band 3 molecules for further opsonization and phagocytosis of the infected RBCs [121,122,123].
There is also an effect seen on the osmotic, antigenic, transportation, and deformation properties of P. falciparum-infected erythrocytes [114,124]. This is also true for uninfected red blood cells when malaria infection takes place. In fact, infected red blood cells (iRBCs) cause a bystander effect, wherein RBCs hosting the parasite provoke changes in the physical properties of the surrounding non-hosting RBCs [125,126]. These rigidified, uninfected red blood cells are mainly removed by splenic macrophages [127].
Changes in the membrane of the erythrocyte that are induced by malaria infection also affect their deformability. As previously explained, this property is crucial for their intrinsic ability to pass through capillaries and other narrow passages of the vascular system.
As the parasite grows inside the RBC, the latter becomes rounder and wrinkled. During asexual stages, the membrane of iRBCs presents knobs, or protrusions, elicited by parasitic proteins known as Knob Associated Histidine–Rich proteins [128,129,130]. Knobs act as a scaffold for the presentation of PfEMP1, which is a protein known as the main actor for the adhesion of the infected RBCs to the endothelium [131].
Red blood cells containing mature parasites exhibit unusual rigidity, and the primary factor responsible for their absence in the bloodstream is not their splenic uptake but an increase in their adherence to endothelial cells. The increased cytoadherence of the iRBCs to the endothelial lining of capillaries and deep tissues through Intercellular Adhesion Molecule-1 (ICAM-1), vascular cellular adhesion molecule (VCAM), Thrombospondin (TSP), P-selectin (CD36), E-selectin, and other molecules [132] is facilitated by specific parasite proteins exposed on the surface of the iRBCs (e.g., PfEMP1) [133]. Their increased adherence is responsible for the clinical occlusion of deep blood vessels in the brain when infected erythrocyte rosettes are formed, leading to the dreaded condition of cerebral malaria, which is a major complication in the malarial pathology [134,135] (Figure 2).
Most of the RBC deformability studies were performed on erythrocytes infected with the asexual forms of the parasites [136,137,138]. However, other lines of investigation on deformability were based on the effects that the sexual forms of the parasite exert on the erythrocyte [129,130]. It is worth noting that throughout their development, P. falciparum gametocytes alter the structural and mechanical characteristics of their host erythrocyte membrane.
The gametocyte has at least five (I–V) morphologically distinct stages in which it transforms; each one of them has different characteristics and effects on the host cell [139]. Through mathematical modeling, 3D imaging, and the use of transgenic parasites, Aingaran et al. demonstrated that early gametocytes increase the rigidity of the erythrocyte stages I to IV [129] and that immature sexual stages are enriched in proximity to erythroblastic islands [140]. Thus, the increased stiffness of immature gametocyte-infected erythrocytes could play a role in their entrapment within the bone marrow due to mechanical retention, favoring their maturation in the hematopoietic system. Mature gametocytes (stage V) exit this microenvironment possibly due to the restoration of their deformability [130], regaining their capacity to pass through narrow openings and be released into the bloodstream. This strategy can enable them to be available for ingestion by mosquitoes only once they have matured [131,141,142].
Many studies provided evidence about the role of several proteins expressed during the sexual stages of the parasite, such as Knob-associated histidine-rich protein, PfEMP3 [143], and P. falciparum Ring infected Erythrocyte Surface Antigen (RESA) [144] in the reduction of the erythrocyte deformability [131] (see a detailed list in Neveu et al., 2019). Furthermore, the expression of another family of proteins called STEVOR (Subtelomeric Variable Open Reading Frame) was studied in both the sexual and asexual stages of the parasite regarding this issue [145,146,147,148].
Varied functions in different parasite life cycle stages have been reported for the STEVORs and the Repetitive Interspersed Family (RIFIN) of proteins, such as rosetting, alteration of iRBC rigidity, and immune evasion [149,150].
Experiments performed on trophozoite-infected erythrocytes showed that endogenous expression of the STEVOR genes provokes important deformability effects on the erythrocytes, with the likely participation of RESA proteins [144,150]. This family of proteins is also expressed during the early stages of gametocytogenesis. STEVOR proteins are exported to the erythrocyte membrane during immature gametocyte stages [148,151,152,153].
The significance of these proteins lies in their potential involvement in, or induction of, the sequestration of gametocytes as a result of stiffness. This hypothesis is supported by data that reflect a high expression of these proteins in field samples, where parasite transmission is highly necessary for survival. Oddly, the expression of STEVOR proteins seems to be undetectable in in vitro clones, possibly because the mosquito stage of gametocytes is not necessary [147,148,154].

5. Changes in Plasmodium Invasion Strategies during Erythrocyte Senescence

As stated before, as the erythrocyte ages, changes in its membrane become evident, making this cell susceptible to opsonization and eventual phagocytosis. At first sight, erythrocyte senescence could impair the ability of parasites to survive due to decreased receptor availability and loss of deformability.
Indeed, the loss of SA receptors would pose another obstacle for the malaria parasite to overcome since most strains of P. falciparum primarily invade through the SA-dependent pathway, using alternate ways of invasion only when these glycoproteins are not available due to mutations, blockage with antibodies, enzymatic cleavage, or natural loss due to aging. However, Huang et al. showed that the RBC charge density is affected by the loss of NANA (SA) during erythrocyte aging [155]. The loss of this negative charge favors a stronger adhesion of RBCs to other cells and tissues.
The parasite, thus, maximizes the above scenario, adding the concomitant help of a reduced negative charge in the RBC to the endothelium-adhering action of its exported proteins, which provides an opportunity to better stick to capillaries and other vessels and avoid the spleen clearance.
To add to the remarkable resilience of this parasite, P. falciparum can infect red blood cells at different stages of the erythrocyte, including earlier maturation stages [156,157]. This observation could be added to the many resources it has, which might explain its high virulence and set it apart from other Plasmodium spp. that invade red blood cells at specific differentiation stages, such as P. knowlesi, P. vivax, and P. ovale. The entry of the latter into the cell is almost restricted to the very youngest circulating class of RBCs [158,159]. P. malariae, in contrast, prefers to invade older erythrocytes [160]. Receptors expressed in younger or older erythrocytes, for which each species probably expresses more ligands, might help to determine their invasion preference.
Just as P. falciparum relies on the presence of erythrocyte receptors, Plasmodium vivax [161,162] and Plasmodium knowlesi [22] also depend on the presence of the Duffy antigen (Fy) in its different polymorphic expressions. Fy is an almost obligatory receptor for the invasion of these parasites into host reticulocytes, wherein it is expressed at higher levels than in older RBCs [163,164]. This could be one of the reasons why P. vivax and P. knowlesi show a marked preference for the invasion of reticulocytes rather than the invasion of the more mature erythrocytes. This is not the case for P. falciparum, which is sufficiently resilient to survive a myriad of challenges, including that of erythrocyte aging.
Regarding the molecular changes experienced by senescent erythrocytes, which involve the loss of parasite receptors, either through aggregation and subsequent loss of function (e.g., Band 3), vesicle release, or downregulation, the parasite exhibits enough flexibility to compensate for their disappearance by adopting alternative entry pathways. While the diversity of receptors is reduced due to the aging process, the molecular availability of ligands remains high enough to promote infection, thereby conferring the characteristic virulence of P. falciparum. The extensive range of molecular options that enable the parasite to invade red blood cells complicates the development of effective blocking vaccines.
In terms of mechanical changes, the loss of elasticity cannot, by itself, be considered a marker of erythrocyte maturity, for this loss is also observed in young red blood cells infected with Plasmodium or even uninfected red blood cells subjected to bystander effects during infection [128]. Under normal circumstances, erythrocytes that have lost their deformability are captured by splenic macrophages, leading to their rapid removal. However, during infection, the parasite evades this process by expressing surface proteins on erythrocytes, allowing them to bind to the molecules present on endothelial cells. This enables the parasite to remain in vascular niches of organs, such as the heart, bone marrow, gastric mucosa, and even the brain, until reaching maturity [165], reducing its circulation in the bloodstream.
Under these circumstances, senescent red blood cells, or young red blood cells subjected to bystander effects when there is infection with Plasmodium, or even infected red blood cells that have not reached enough stiffness and remain in circulation, may act as decoys to partially saturate the phagocytic capacity of splenic macrophages, favoring the progression of the infectious process.
Some questions linger regarding the strategies of P. falciparum and other Plasmodia to select their receptors: Do they depend solely on their availability? How much of this preference could be attributed to changes in the geometry of the RBC due to the aging process? Do their preferred invasion receptors change over the lifespan of the RBC? Could other factors influence their invasion receptor selection?
Butcher et al., using P. knowlesi, reported that the invasion of RBCs might be affected by structural changes on the erythrocyte surface, but at the same time, they also showed that these changes are intrinsic to age progression [166]. Studies performed in P. gallinaceum-infected White Leghorn chickens demonstrated that this parasite prefers to invade young erythrocytes [167] and that this may be because young erythrocytes have larger amounts of phospholipids, which are required by the parasite invasion [168].
It is also known that choline, which is a precursor of the phospholipid composition of cells, is avidly taken in by P. knowlesi-infected simian red blood cells. The permeability of simian erythrocytes to choline was found to be considerably increased after infection by the malaria parasite [169,170]. In addition, it was reported that young malaria-infected erythrocytes showed an increase in choline permeability [171].
The increased expression of the key P. falciparum receptor, namely, Basigin, on early reticulocytes [172,173,174] could augment P. falciparum merozoite binding and invasion into reticulocytes, although this effect has not been directly demonstrated [156].
McQueen and McKenzie hypothesized that the different age-related preferences of RBCs according to species could be a strategy of Plasmodium spp. in areas where co-infection with different species takes place to keep a low-density parasitemia in balance with the ongoing availability of young, adult, and senescent RBCs [159].

6. Conclusions

From the concerted interactions between the merozoite and the RBC surface, it is evident that changes in an erythrocyte’s physical and molecular composition are crucial for the success or failure of the parasite entry and the development of its infective cycle.
Given that the aging of the erythrocyte may influence the number and availability of the receptors and that the deformability of the erythrocyte is critical for the invasion and development of Plasmodium parasites, an analysis of the relationship between RBC aging and malaria is very relevant to knowledge regarding the malaria pathogen. Understanding these dynamic changes holds substantial importance in the development of therapies to control malaria and vaccines to prevent it, and provides further insight into the comprehension of the pathogenesis of this disease.
For Plasmodium, the aging of red blood cells is not a determinant issue for its survival; on the contrary, it executes its plan for the survival of the species in a way that humans have not been able to overcome yet.

Author Contributions

Writing—review and editing: M.F.A.-R. and C.S.; writing—original draft preparation, N.M.T.; writing: L.M.C. and D.D. All authors read and agreed to the published version of the manuscript.

Funding

C.S. and L.M.C. were partially funded by the Sistema Nacional de Investigación de Panamá (SNI).

Acknowledgments

The authors would like to acknowledge INDICASAT AIP support.

Conflicts of Interest

Nicole M. Tayler is employed by the Estée Lauder Panamá. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The Estée Lauder Panamá, nor SNI, had any role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. World Health Organization. World malaria report 2022; World Health Organization: Geneva, Switzerland, 2022; Licence: CC BY-NC-SA 3.0 IGO. [Google Scholar]
  2. Leal Filho, W.; May, J.; May, M.; Nagy, G.J. Climate change and malaria: Some recent trends of malaria incidence rates and average annual temperature in selected sub-Saharan African countries from 2000 to 2018. Malar. J. 2023, 22, 248. [Google Scholar] [CrossRef] [PubMed]
  3. Wellems, T.E.; Plowe, C.V. Chloroquine-Resistant Malaria. J. Infect. Dis. 2001, 184, 770–776. [Google Scholar] [CrossRef] [PubMed]
  4. Gomez, G.M.; D’Arrigo, G.; Sanchez, C.P.; Berger, F.; Wade, R.C.; Lanzer, M. PfCRT mutations conferring piperaquine resistance in falciparum malaria shape the kinetics of quinoline drug binding and transport. PLoS Pathog. 2023, 19, e1011436. [Google Scholar] [CrossRef] [PubMed]
  5. Ross, L.S.; Dhingra, S.K.; Mok, S.; Yeo, T.; Wicht, K.J.; Kümpornsin, K.; Takala-Harrison, S.; Witkowski, B.; Fairhurst, R.M.; Ariey, F.; et al. Emerging Southeast Asian PfCRT mutations confer Plasmodium falciparum resistance to the first-line antimalarial piperaquine. Nat. Commun. 2018, 9, 3314. [Google Scholar] [CrossRef] [PubMed]
  6. Veiga, M.I.; Dhingra, S.K.; Henrich, P.P.; Straimer, J.; Gnädig, N.; Uhlemann, A.C.; Martin, R.E.; Lehane, A.M.; Fidock, D.A. Globally prevalent PfMDR1 mutations modulate Plasmodium falciparum susceptibility to artemisinin-based combination therapies. Nat. Commun. 2016, 7, 11553. [Google Scholar] [CrossRef] [PubMed]
  7. Wurtz, N.; Fall, B.; Pascual, A.; Fall, M.; Baret, E.; Camara, C.; Nakoulima, A.; Diatta, B.; Fall, K.B.; Mbaye, P.S.; et al. Role of Pfmdr1 in in vitro Plasmodium falciparum susceptibility to chloroquine, quinine, monodesethylamodiaquine, mefloquine, lumefantrine, and dihydroartemisinin. Antimicrob. Agents Chemother. 2014, 58, 7032–7040. [Google Scholar] [CrossRef]
  8. Alvarez, E.; Lavandera, J.L.; Vanderwall, D.E.; Green, D.V.; Kumar, V.; Hasan, S.; Brown, J.R.; Peishoff, C.E.; Cardon, L.R.; Garcia-Bustos, J.F.; et al. Thousands of chemical starting points for antimalarial lead identification. Nature 2010, 465, 305–310. [Google Scholar] [CrossRef]
  9. Spiegel, H.; Boes, A.; Kastilan, R.; Kapelski, S.; Edgue, G.; Beiss, V.; Chubodova, I.; Scheuermayer, M.; Pradel, G.; Schillberg, S.; et al. The stage-specific in vitro efficacy of a malaria antigen cocktail provides valuable insights into the development of effective multi-stage vaccines. Biotechnol. J. 2015, 10, 1651–1659. [Google Scholar] [CrossRef]
  10. Vijayan, A.; Chitnis, C.E. Development of Blood Stage Malaria Vaccines. Methods Mol. Biol. 2019, 2013, 199–218. [Google Scholar] [CrossRef]
  11. Molina-Franky, J.; Patarroyo, M.E.; Kalkum, M.; Patarroyo, M.A. The Cellular and Molecular Interaction Between Erythrocytes and Plasmodium falciparum Merozoites. Front. Cell. Infect. Microbiol. 2022, 12, 816574. [Google Scholar] [CrossRef]
  12. Dluzewski, A.R.; Mitchell, G.H.; Fryer, P.R.; Griffiths, S.; Wilson, R.J.; Gratzer, W.B. Origins of the parasitophorous vacuole membrane of the malaria parasite, Plasmodium falciparum, in human red blood cells. J. Cell Sci. 1992, 102(Pt3), 527–532. [Google Scholar] [CrossRef] [PubMed]
  13. Cowman, A.F.; Tonkin, C.J.; Tham, W.H.; Duraisingh, M.T. The Molecular Basis of Erythrocyte Invasion by Malaria Parasites. Cell Host Microbe 2017, 22, 232–245. [Google Scholar] [CrossRef] [PubMed]
  14. Mitchell, G.H.; Hadley, T.J.; McGinniss, M.H.; Klotz, F.W.; Miller, L.H. Invasion of erythrocytes by Plasmodium falciparum malaria parasites: Evidence for receptor heterogeneity and two receptors. Blood 1986, 67, 1519–1521. [Google Scholar] [CrossRef] [PubMed]
  15. Sim, B.K.; Chitnis, C.E.; Wasniowska, K.; Hadley, T.J.; Miller, L.H. Receptor and ligand domains for invasion of erythrocytes by Plasmodium falciparum. Science 1994, 264, 1941–1944. [Google Scholar] [CrossRef] [PubMed]
  16. Li, X.; Marinkovic, M.; Russo, C.; McKnight, C.J.; Coetzer, T.L.; Chishti, A.H. Identification of a specific region of Plasmodium falciparum EBL-1 that binds to host receptor glycophorin B and inhibits merozoite invasion in human red blood cells. Mol. Biochem. Parasitol. 2012, 183, 23–31. [Google Scholar] [CrossRef] [PubMed]
  17. Lin, D.H.; Malpede, B.M.; Batchelor, J.D.; Tolia, N.H. Crystal and Solution Structures of Plasmodium falciparum Erythrocyte-binding Antigen 140 Reveal Determinants of Receptor Specificity during Erythrocyte Invasion. J. Biol. Chem. 2012, 287, P36830–P36836. [Google Scholar] [CrossRef]
  18. Lobo, C.A.; Rodriguez, M.; Reid, M.; Lustigman, S. Glycophorin C is the receptor for the Plasmodium falciparum erythrocyte binding ligand PfEBP-2 (baebl). Blood 2003, 101, 4628–4631. [Google Scholar] [CrossRef]
  19. Daniels, G. Functional aspects of red cell antigens. Blood Rev. 1999, 13, 14–35. [Google Scholar] [CrossRef]
  20. Lopez, G.H.; Wei, L.; Ji, Y.; Condon, J.A.; Luo, G.; Hyland, C.A.; Flower, R.L. GYP*Kip, a novel GYP(B-A-B) hybrid allele, encoding the MNS48 (KIPP) antigen. Transfusion 2016, 56, 539–541. [Google Scholar] [CrossRef]
  21. Hollox, E.J.; Louzada, S. Genetic variation of glycophorins and infectious disease. Immunogenetics 2023, 75, 201–206. [Google Scholar] [CrossRef]
  22. Miller, L.H.; Haynes, J.D.; McAuliffe, F.M.; Shiroishi, T.; Durocher, J.R.; McGinniss, M.H. Evidence for differences in erythrocyte surface receptors for the malarial parasites, Plasmodium falciparum and Plasmodium knowlesi. J. Exp. Med. 1977, 146, 277–281. [Google Scholar] [CrossRef]
  23. Ochola-Oyier, L.I.; Wamae, K.; Omedo, I.; Ogola, C.; Matharu, A.; Musabyimana, J.P.; Njogu, F.K.; Marsh, K. Few Plasmodium falciparum merozoite ligand and erythrocyte receptor pairs show evidence of balancing selection. Infect. Genet. Evol. 2019, 69, 235–245. [Google Scholar] [CrossRef]
  24. Mayer, D.C.; Cofie, J.; Jiang, L.; Hartl, D.L.; Tracy, E.; Kabat, J.; Mendoza, L.H.; Miller, L.H. Glycophorin B is the erythrocyte receptor of Plasmodium falciparum erythrocyte-binding ligand, EBL-1. Proc. Natl. Acad. Sci. USA 2009, 106, 5348–5352. [Google Scholar] [CrossRef]
  25. Jaskiewicz, E.; Jodłowska, M.; Kaczmarek, R.; Zerka, A. Erythrocyte glycophorins as receptors for Plasmodium merozoites. Parasit. Vectors 2019, 12, 317. [Google Scholar] [CrossRef] [PubMed]
  26. Dankwa, S.; Chaand, M.; Kanjee, U.; Jiang, R.H.Y.; Nobre, L.V.; Goldberg, J.M.; Bei, A.K.; Moechtar, M.A.; Grüring, C.; Ahouidi, A.D.; et al. Genetic Evidence for Erythrocyte Receptor Glycophorin B Expression Levels Defining a Dominant Plasmodium falciparum Invasion Pathway into Human Erythrocytes. Infect. Immun. 2017, 85, e00074-17. [Google Scholar] [CrossRef] [PubMed]
  27. Duraisingh, M.T.; Maier, A.G.; Triglia, T.; Cowman, A.F. Erythrocyte-binding antigen 175 mediates invasion in Plasmodium falciparum utilizing sialic acid-dependent and -independent pathways. Proc. Natl. Acad. Sci. USA 2003, 100, 4796–4801. [Google Scholar] [CrossRef] [PubMed]
  28. Tham, W.H.; Wilson, D.W.; Reiling, L.; Chen, L.; Beeson, J.G.; Cowman, A.F. Antibodies to reticulocyte binding protein-like homologue 4 inhibit invasion of Plasmodium falciparum into human erythrocytes. Infect. Immun. 2009, 77, 2427–2435. [Google Scholar] [CrossRef] [PubMed]
  29. Gaur, D.; Singh, S.; Jiang, L.; Diouf, A.; Miller, L.H. Recombinant Plasmodium falciparum reticulocyte homology protein 4 binds to erythrocytes and blocks invasion. Proc. Natl. Acad. Sci. USA 2007, 104, 17789–17794. [Google Scholar] [CrossRef] [PubMed]
  30. Stubbs, J.; Simpson, K.M.; Triglia, T.; Plouffe, D.; Tonkin, C.J.; Duraisingh, M.T.; Maier, A.G.; Winzeler, E.A.; Cowman, A.F. Molecular mechanism for switching of P. falciparum invasion pathways into human erythrocytes. Science 2005, 309, 1384–1387. [Google Scholar] [CrossRef] [PubMed]
  31. Spadafora, C.; Awandare, G.A.; Kopydlowski, K.M.; Czege, J.; Moch, J.K.; Finberg, R.W.; Tsokos, G.C.; Stoute, J.A. Complement receptor 1 is a sialic acid-independent erythrocyte receptor of Plasmodium falciparum. PLoS Pathog. 2010, 6, e1000968. [Google Scholar] [CrossRef]
  32. Awandare, G.A.; Spadafora, C.; Moch, J.K.; Dutta, S.; Haynes, J.D.; Stoute, J.A. Plasmodium falciparum field isolates use complement receptor 1 (CR1) as a receptor for invasion of erythrocytes. Mol. Biochem. Parasitol. 2011, 177, 57–60. [Google Scholar] [CrossRef] [PubMed]
  33. Rodriguez, M.; Lustigman, S.; Montero, E.; Oksov, Y.; Lobo, C.A. PfRH5: A novel reticulocyte-binding family homolog of Plasmodium falciparum that binds to the erythrocyte, and an investigation of its receptor. PLoS ONE 2008, 3, e3300. [Google Scholar] [CrossRef]
  34. Wanaguru, M.; Liu, W.; Hahn, B.H.; Rayner, J.C.; Wright, G.J. RH5-Basigin interaction plays a major role in the host tropism of Plasmodium falciparum. Proc. Natl. Acad. Sci. USA 2013, 110, 20735–20740. [Google Scholar] [CrossRef] [PubMed]
  35. Jamwal, A.; Constantin, C.F.; Hirschi, S.; Henrich, S.; Bildl, W.; Fakler, B.; Draper, S.J.; Schulte, U.; Higgins, M.K. Erythrocyte invasion-neutralising antibodies prevent Plasmodium falciparum RH5 from binding to basigin-containing membrane protein complexes. Elife 2023, 5, e83681. [Google Scholar] [CrossRef] [PubMed]
  36. Egan, E.S.; Jiang, R.H.; Moechtar, M.A.; Barteneva, N.S.; Weekes, M.P.; Nobre, L.V.; Gygi, S.P.; Paulo, J.A.; Frantzreb, C.; Tani, Y.; et al. A forward genetic screen identifies erythrocyte CD55 as essential for Plasmodium falciparum invasion. Science 2015, 348, 711–714. [Google Scholar] [CrossRef] [PubMed]
  37. Diédhiou, C.K.; Moussa, R.A.; Bei, A.K.; Daniels, R.; Papa Mze, N.; Ndiaye, D.; Faye, N.; Wirth, D.; Amambua-Ngwa, A.; Mboup, S.; et al. Temporal changes in Plasmodium falciparum reticulocyte binding protein homolog 2b (PfRh2b) in Senegal and The Gambia. Malar. J. 2019, 18, 239. [Google Scholar] [CrossRef] [PubMed]
  38. Stoute, J.A. Complement-regulatory proteins in severe malaria: Too little or too much of a good thing? Trends Parasitol. 2005, 21, 218–223, Erratum in: Trends Parasitol. 2005, 21, 358. [Google Scholar] [CrossRef] [PubMed]
  39. Eggleton, P.; Tenner, A.J.; Reid, K.B. C1q receptors. Clin. Exp. Immunol. 2012, 120, 406–412. [Google Scholar] [CrossRef]
  40. Kosoy, R.; Ransom, M.; Chen, H.; Marconi, M.; Macciardi, F.; Glorioso, N.; Gregersen, P.K.; Cusi, D.; Seldin, M.F. Evidence for malaria selection of a CR1 haplotype in Sardinia. Genes Immun. 2011, 12, 582–588. [Google Scholar] [CrossRef]
  41. Tham, W.H.; Wilson, D.W.; Lopaticki, S.; Schmidt, C.Q.; Tetteh-Quarcoo, P.B.; Barlow, P.N.; Richard, D.; Corbin, J.E.; Beeson, J.G.; Cowman, A.F. Complement receptor 1 is the host erythrocyte receptor for Plasmodium falciparum PfRh4 invasion ligand. Proc. Natl. Acad. Sci. USA 2010, 107, 17327–17332. [Google Scholar] [CrossRef]
  42. Muramatsu, T. Basigin (CD147), a multifunctional transmembrane glycoprotein with various binding partners. J. Biochem. 2016, 159, 481–490. [Google Scholar] [CrossRef] [PubMed]
  43. Supper, V.; Schiller, H.B.; Paster, W.; Forster, F.; Boulègue, C.; Mitulovic, G.; Leksa, V.; Ohradanova-Repic, A.; Machacek, C.; Schatzlmaier, P.; et al. Association of CD147 and Calcium Exporter PMCA4 Uncouples IL-2 Expression from Early TCR Signaling. J. Immunol. 2016, 196, 1387–1399. [Google Scholar] [CrossRef] [PubMed]
  44. Bedu-Addo, G.; Meese, S.; Mockenhaupt, F.P. An ATP2B4 polymorphism protects against malaria in pregnancy. J. Infect. Dis. 2013, 207, 1600–1603. [Google Scholar] [CrossRef] [PubMed]
  45. Mariga, S.T.; Kolko, M.; Gjedde, A.; Bergersen, L.H. Lactate transport and receptor actions in cerebral malaria. Front. Neurosci. 2014, 8, 125. [Google Scholar] [CrossRef] [PubMed]
  46. Ndila, C.M.; Uyoga, S.; Macharia, A.W.; Nyutu, G.; Peshu, N.; Ojal, J.; Shebe, M.; Awuondo, K.O.; Mturi, N.; Tsofa, B.; et al. MalariaGEN Consortium. Human candidate gene polymorphisms and risk of severe malaria in children in Kilifi, Kenya: A case-control association study. Lancet Haematol. 2018, 5, e333–e345. [Google Scholar] [CrossRef]
  47. Wright, K.E.; Hjerrild, K.A.; Bartlett, J.; Douglas, A.D.; Jin, J.; Brown, R.E.; Illingworth, J.J.; Ashfield, R.; Clemmensen, S.B.; de Jongh, W.A.; et al. Structure of malaria invasion protein RH5 with erythrocyte basigin and blocking antibodies. Nature 2014, 515, 427–430. [Google Scholar] [CrossRef]
  48. Crosnier, C.; Bustamante, L.Y.; Bartholdson, S.J.; Bei, A.K.; Theron, M.; Uchikawa, M.; Mboup, S.; Ndir, O.; Kwiatkowski, D.P.; Duraisingh, M.T.; et al. Basigin is a receptor essential for erythrocyte invasion by Plasmodium falciparum. Nature 2011, 480, 534–537. [Google Scholar] [CrossRef]
  49. Storry, J.R.; Reid, M.E.; Yazer, M.H. The Cromer blood group system: A review. Immunohematology 2010, 26, 109–118. [Google Scholar] [CrossRef]
  50. Coyne, C.B.; Bergelson, J.M. Virus-induced Abl and Fyn kinase signals permit coxsackievirus entry through epithelial tight junctions. Cell 2006, 124, 119–131. [Google Scholar] [CrossRef] [PubMed]
  51. Sahar, T.; Reddy, K.S.; Bharadwaj, M.; Pandey, A.K.; Singh, S.; Chitnis, C.E.; Gaur, D. Plasmodium falciparum reticulocyte binding-like homologue protein 2 (PfRH2) is a key adhesive molecule involved in erythrocyte invasion. PLoS ONE 2011, 6, e17102. [Google Scholar] [CrossRef] [PubMed]
  52. Lin, C.S.; Uboldi, A.D.; Marapana, D.; Czabotar, P.E.; Epp, C.; Bujard, H.; Taylor, N.L.; Perugini, M.A.; Hodder, A.N.; Cowman, A.F. The merozoite surface protein 1 complex is a platform for binding to human erythrocytes by Plasmodium falciparum. J. Biol. Chem. 2014, 289, 25655–25669. [Google Scholar] [CrossRef] [PubMed]
  53. Siddiqui, F.A.; Dhawan, S.; Singh, S.; Singh, B.; Gupta, P.; Pandey, A.; Mohmmed, A.; Gaur, D.; Chitnis, C.E. A thrombospondin structural repeat containing rhoptry protein from Plasmodium falciparum mediates erythrocyte invasion. Cell. Microbiol. 2013, 15, 1341–1356. [Google Scholar] [CrossRef] [PubMed]
  54. Hayton, K.; Dumoulin, P.; Henschen, B.; Liu, A.; Papakrivos, J.; Wellems, T.E. Various PfRH5 polymorphisms can support Plasmodium falciparum invasion into the erythrocytes of owl monkeys and rats. Mol. Biochem. Parasitol. 2013, 187, 103–110. [Google Scholar] [CrossRef] [PubMed]
  55. Salinas, N.D.; Tolia, N.H. Red cell receptors as access points for malaria infection. Curr. Opin. Hematol. 2016, 23, 215–223. [Google Scholar] [CrossRef] [PubMed]
  56. Mock, D.M.; Matthews, N.I.; Zhu, S.; Strauss, R.G.; Schmidt, R.L.; Nalbant, D.; Cress, G.A.; Widness, J.A. Red blood cell (RBC) survival determined in humans using RBCs labeled at multiple biotin densities. Transfusion 2011, 51, 1047–1057. [Google Scholar] [CrossRef]
  57. Thiagarajan, P.; Parker, C.J.; Prchal, J.T. How Do Red Blood Cells Die? Front. Physiol. 2021, 12, 655393. [Google Scholar] [CrossRef]
  58. Antonelou, M.H.; Kriebardis, A.G.; Papassideri, I.S. Aging and death signalling in mature red cells: From basic science to transfusion practice. Blood Transfus. 2010, 8 (Suppl. S3), s39–s47. [Google Scholar] [CrossRef]
  59. Lutz, H.U.; Bogdanova, A. Mechanisms tagging senescent red blood cells for clearance in healthy humans. Front. Physiol. 2013, 4, 387. [Google Scholar] [CrossRef]
  60. Waugh, S.M.; Walder, J.A.; Low, P.S. Partial characterization of the copolymerization reaction of erythrocyte membrane band 3 with hemichromes. Biochemistry 1987, 26, 1777–1783. [Google Scholar] [CrossRef] [PubMed]
  61. Badior, K.; Casey, J.R. Large conformational dynamics in Band 3 protein: Significance for erythrocyte senescence signalling, Biochim. Biophys. Acta BBA Biomembr. 2021, 1863, 183678. [Google Scholar] [CrossRef] [PubMed]
  62. Kay, M.M. Aging of cell membrane molecules leads to appearance of an aging antigen and removal of senescent cells. Gerontology 1985, 31, 215–235. [Google Scholar] [CrossRef] [PubMed]
  63. Lutz, H.U. Naturally occurring autoantibodies in mediating clearance of senescent red blood cells. Adv. Exp. Med. Biol. 2012, 750, 76–90. [Google Scholar] [CrossRef] [PubMed]
  64. Kay, M.M. Mechanism of removal of senescent cells by human macrophages in situ. Proc. Natl. Acad. Sci. USA 1975, 72, 3521–3525. [Google Scholar] [CrossRef] [PubMed]
  65. Safeukui, I.; Buffet, P.A.; Deplaine, G.; Perrot, S.; Brousse, V.; Ndour, A.; Nguyen, M.; Mercereau-Puijalon, O.; David, P.H.; Milon, G.; et al. Quantitative assessment of sensing and sequestration of spherocytic erythrocytes by the human spleen. Blood 2012, 120, 424–430. [Google Scholar] [CrossRef] [PubMed]
  66. Satchwell, T.J.; Toye, A.M. Band 3, an essential red blood cell hub of activity. Haematologica 2021, 106, 2792–2793. [Google Scholar] [CrossRef] [PubMed]
  67. Lutz, H.U.; Bussolino, F.; Flepp, R.; Fasler, S.; Stammler, P.; Kazatchkine, M.D.; Arese, P. Naturally occurring anti-band-3 antibodies and complement together mediate phagocytosis of oxidatively stressed human erythrocytes. Proc. Natl. Acad. Sci. USA 1987, 84, 7368–7372. [Google Scholar] [CrossRef] [PubMed]
  68. Almukadi, H.; Schwake, C.; Kaiser, M.M.; Mayer, D.C.G.; Schiemer, J.; Baldwin, M.R.; Hegde, S.; Lu, Y.; Hanada, T.; Chishti, A.H. Human erythrocyte band 3 is a host receptor for Plasmodium falciparum glutamic acid-rich protein. Blood 2019, 133, 470–480. [Google Scholar] [CrossRef]
  69. Lu, J.; Chu, R.; Yin, Y.; Yu, H.; Xu, Q.; Yang, B.; Sun, Y.; Song, J.; Wang, Q.; Xu, J.; et al. Glycosylphosphatidylinositol-anchored micronemal antigen (GAMA) interacts with the band 3 receptor to promote erythrocyte invasion by malaria parasites. J. Biol. Chem. 2022, 298, 101765. [Google Scholar] [CrossRef]
  70. Fishelson, Z.; Marikovsky, Y. Reduced CR1 expression on aged human erythrocytes: Immuno-electron microscopic and functional analysis. Mech. Ageing Dev. 1993, 72, 25–35. [Google Scholar] [CrossRef]
  71. Ripoche, J.; Sim, R.B. Loss of complement receptor type 1 (CR1) on ageing of erythrocytes. Studies of proteolytic release of the receptor. Biochem. J. 1986, 235, 815–821. [Google Scholar] [CrossRef]
  72. Pascual, M.; Lutz, H.U.; Steiger, G.; Stammler, P.; Schifferli, J.A. Release of vesicles enriched in complement receptor 1 from human erythrocytes. J. Immunol. 1993, 151, 397–404. [Google Scholar] [CrossRef] [PubMed]
  73. Blanc, L.; De Gassart, A.; Géminard, C.; Bette-Bobillo, P.; Vidal, M. Exosome release by reticulocytes—An integral part of the red blood cell differentiation system. Blood Cells Mol. Dis. 2005, 35, 21–26. [Google Scholar] [CrossRef] [PubMed]
  74. Willekens, F.L.A.; Werre, J.M.; Groenen-Döpp, Y.A.M.; Roerdinkholder-Stoelwinder, B.; de Pauw, B.; Bosman, G.J.C.G.M. Erythrocyte vesiculation: A self-protective mechanism? Br. J. Haematol. 2008, 141, 549–556. [Google Scholar] [CrossRef] [PubMed]
  75. Thangaraju, K.; Neerukonda, S.N.; Katneni, U.; Buehler, P.W. Extracellular Vesicles from Red Blood Cells and Their Evolving Roles in Health, Coagulopathy and Therapy. Int. J. Mol. Sci. 2020, 22, 153. [Google Scholar] [CrossRef] [PubMed]
  76. Correa, R.; Coronado, L.M.; Garrido, A.C.; Durant-Archibald, A.A.; Spadafora, C. Volatile organic compounds associated with Plasmodium falciparum infection in vitro. Parasit. Vectors 2017, 10, 215. [Google Scholar] [CrossRef]
  77. Trautsch, C.; Tannert, C.; Maretzki, D. Disproportional loss of membrane constituents in the course of erythrocyte aging. Acta Biol. Medica Ger. 1981, 40, 743–746. [Google Scholar]
  78. Miyahara, K.; Spiro, M.J. Nonuniform loss of membrane glycoconjugates during in vivo aging of human erythrocytes: Studies of normal and diabetic red cell saccharides. Arch. Biochem. Biophys. 1984, 232, 310–322. [Google Scholar] [CrossRef] [PubMed]
  79. Sparrow, R.L.; Healey, G.; Patton, K.A.; Veale, M.F. Red blood cell age determines the impact of storage and leukocyte burden on cell adhesion molecules, glycophorin A and the release of annexin V. Transfus. Apher. Sci. 2006, 34, 15–23. [Google Scholar] [CrossRef]
  80. Biasini, G.M.; Botrè, F.; de la Torre, X.; Donati, F. Age-Markers on the Red Blood Cell Surface and Erythrocyte Microparticles may Constitute a Multi-parametric Strategy for Detection of Autologous Blood Transfusion. Sports Med. Open 2023, 9, 113. [Google Scholar] [CrossRef]
  81. Lux, S.E. 4th. Anatomy of the red cell membrane skeleton: Unanswered questions. Blood 2016, 127, 187–199. [Google Scholar] [CrossRef]
  82. Mohandas, N.; Blanc, L. Parasite hijacks red cell membrane proteins. Blood 2023, 142, 1942–1944. [Google Scholar] [CrossRef]
  83. Baro, B.; Kim, C.Y.; Lin, C.; Kongsomboonvech, A.K.; Tetard, M.; Peterson, N.A.; Salinas, N.D.; Tolia, N.H.; Egan, E.S. Plasmodium falciparum exploits CD44 as a coreceptor for erythrocyte invasion. Blood 2023, 142, 2016–2028. [Google Scholar] [CrossRef]
  84. Morrissey, M.A.; Kern, N.; Vale, R.D. CD47 Ligation Repositions the Inhibitory Receptor SIRPA to Suppress Integrin Activation and Phagocytosis. Immunity 2020, 53, 290–302.e6, Erratum in: Immunity 2023, 56, 2172. [Google Scholar] [CrossRef] [PubMed]
  85. Jaiswal, S.; Jamieson, C.H.; Pang, W.W.; Park, C.Y.; Chao, M.P.; Majeti, R.; Traver, D.; van Rooijen, N.; Weissman, I.L. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell 2009, 138, 271–285. [Google Scholar] [CrossRef] [PubMed]
  86. Balduini, C.; Balduini, C.L.; Ascari, E. Membrane glycopeptides from old and young human erythrocytes. Biochem. J. 1974, 140, 557–560. [Google Scholar] [CrossRef] [PubMed]
  87. Gattegno, L.; Bladier, D.; Garnier, M.; Cornillot, P. Changes in carbohydrate content of surface membranes of human erythrocytes during aging. Carbohydr. Res. 1976, 52, 197–208. [Google Scholar] [CrossRef] [PubMed]
  88. Huang, Y.X.; Tuo, W.W.; Wang, D.; Kang, L.L.; Chen, X.Y.; Luo, M. Restoring the youth of aged red blood cells and extending their lifespan in circulation by remodeling membrane sialic acid. J. Cell. Mol. Med. 2016, 20, 294–301. [Google Scholar] [CrossRef] [PubMed]
  89. Fernandes, H.P.; Cesar, C.L.; Barjas-Castro, M. de L. Electrical properties of the red blood cell membrane and immunohematological investigation. Rev. Bras. Hematol. Hemoter. 2011, 33, 297–301. [Google Scholar] [CrossRef]
  90. Ayi, K.; Lu, Z.; Serghides, L.; Ho, J.M.; Finney, C.; Wang, J.C.Y.; Liles, W.C.; Kain, K.C. CD47-SIRPα Interactions Regulate Macrophage Uptake of Plasmodium falciparum-Infected Erythrocytes and Clearance of Malaria in vivo. Infect. Immun. 2016, 84, 2002–2011. [Google Scholar] [CrossRef]
  91. Coste, I.; Gauchat, J.F.; Wilson, A.; Izui, S.; Jeannin, P.; Delneste, Y.; MacDonald, H.R.; Bonnefoy, J.Y.; Renno, T. Unavailability of CD147 leads to selective erythrocyte trapping in the spleen. Blood 2001, 97, 3984–3988. [Google Scholar] [CrossRef]
  92. Bhardwaj, N.; Singh, A. Splenectomy Modulates the Erythrocyte Turnover and Basigin (CD147) Expression in Mice. Indian J. Hematol. Blood Transfus. 2020, 36, 711–718. [Google Scholar] [CrossRef]
  93. Seki, M.; Arashiki, N.; Takakuwa, Y.; Nitta, K.; Nakamura, F. Reduction in flippase activity contributes to surface presentation of phosphatidylserine in human senescent erythrocytes. J. Cell. Mol. Med. 2020, 24, 13991–14000. [Google Scholar] [CrossRef]
  94. Bernhardt, I.; Nguyen, D.B.; Wesseling, M.C.; Kaestner, L. Intracellular Ca2+ Concentration and Phosphatidylserine Exposure in Healthy Human Erythrocytes in Dependence on in vivo Cell Age. Front. Physiol. 2020, 10, 1629. [Google Scholar] [CrossRef]
  95. Arese, P.; Turrini, F.; Schwarzer, E. Band 3/complement-mediated recognition and removal of normally senescent and pathological human erythrocytes. Cell. Physiol. Biochem. 2005, 16, 133–146. [Google Scholar] [CrossRef]
  96. Gao, Y.; Lv, L.; Liu, S.; Ma, G.; Su, Y. Elevated levels of thrombin-generating microparticles in stored red blood cells. Vox Sang. 2013, 105, 11–17. [Google Scholar] [CrossRef]
  97. Greenwalt, T.J. Autologous and aged blood donors. JAMA 1987, 257, 1220–1221. [Google Scholar] [CrossRef] [PubMed]
  98. Kriebardis, A.G.; Antonelou, M.H.; Stamoulis, K.E.; Economou-Petersen, E.; Margaritis, L.H.; Papassideri, I.S. RBC-derived vesicles during storage: Ultrastructure, protein composition, oxidation, and signaling components. Transfusion 2008, 48, 1943–1953. [Google Scholar] [CrossRef] [PubMed]
  99. Koshiar, R.L.; Somajo, S.; Norström, E.; Dahlbäck, B. Erythrocyte-derived microparticles supporting activated protein C-mediated regulation of blood coagulation. PLoS ONE 2014, 9, e104200. [Google Scholar]
  100. Bosman, G.J.; Cluitmans, J.C.; Groenen, Y.A.; Werre, J.M.; Willekens, F.L.; Novotný, V.M. Susceptibility to hyperosmotic stress-induced phosphatidylserine exposure increases during red blood cell storage. Transfusion 2011, 51, 1072–1078. [Google Scholar] [CrossRef] [PubMed]
  101. Koshkaryev, A.; Livshits, L.; Pajic-Lijakovic, I.; Gural, A.; Barshtein, G.; Yedgar, S. Non-oxidative band-3 clustering agents cause the externalization of phosphatidylserine on erythrocyte surfaces by a calcium-independent mechanism. Biochim. Biophys. Acta Biomembr. 2020, 1862, 183231. [Google Scholar] [CrossRef] [PubMed]
  102. Livshits, L.; Barshtein, G.; Arbell, D.; Gural, A.; Levin, C.; Guizouarn, H. Do We Store Packed Red Blood Cells under “Quasi-Diabetic” Conditions? Biomolecules 2021, 11, 992. [Google Scholar] [CrossRef]
  103. Rand, R.P. Mechanical properties of the red cell membrane: II. Viscoelastic breakdown of the membrane. Biophys. J. 1964, 4, 303–316. [Google Scholar] [CrossRef]
  104. Kozlov, M.M.; Lerche, D.; Meier, W. RBC membrane instability for large pipette deformation. A theoretical approach. Biorheology 1988, 25, 843–856. [Google Scholar] [CrossRef]
  105. Gueguen, M.; Bidet, J.M.; Durand, F.; Driss, F.; Joffre, A.; Genetet, B. Filtration pressure and red blood cell deformability: Evaluation of a new device: Erythrometre. Biorheol. Suppl. 1984, 1, 261–265. [Google Scholar] [CrossRef] [PubMed]
  106. Islamzada, E.; Matthews, K.; Guo, Q.; Santoso, A.T.; Duffy, S.P.; Scott, M.D.; Ma, H. Deformability based sorting of stored red blood cells reveals donor-dependent aging curves. Lab. Chip 2020, 20, 226–235. [Google Scholar] [CrossRef] [PubMed]
  107. Dulinska, I.; Targosz, M.; Strojny, W.; Lekka, M.; Czuba, P.; Balwierz, W.; Szymonski, M. Stiffness of normal and pathological erythrocytes studied by means of atomic force microscopy. J. Biochem. Biophys. Methods 2006, 66, 1–11. [Google Scholar] [CrossRef] [PubMed]
  108. Baskurt, O.K.; Meiselman, H.J. Determination of red blood cell shape recovery time constant in a Couette system by the analysis of light reflectance and ektacytometry. Biorheology 1996, 33, 489–503. [Google Scholar] [CrossRef] [PubMed]
  109. Sonmez, M.; Ince, H.Y.; Yalcin, O.; Ajdžanović, V.; Spasojević, I.; Meiselman, H.J.; Baskurt, O.K. The effect of alcohols on red blood cell mechanical properties and membrane fluidity depends on their molecular size. PLoS ONE 2013, 8, e76579. [Google Scholar] [CrossRef] [PubMed]
  110. Brandão, M.M.; Fontes, A.; Barjas-Castro, M.L.; Barbosa, L.C.; Costa, F.F.; Cesar, C.L.; Saad, S.T. Optical tweezers for measuring red blood cell elasticity: Application to the study of drug response in sickle cell disease. Eur. J. Haematol. 2003, 70, 207–211. [Google Scholar] [CrossRef] [PubMed]
  111. Agrawal, R.; Smart, T.; Nobre-Cardoso, J.; Richards, C.; Bhatnagar, R.; Tufail, A.; Shima, D.H.; Jones, P.; Pavesio, C. Assessment of red blood cell deformability in type 2 diabetes mellitus and diabetic retinopathy by dual optical tweezers stretching technique. Sci. Rep. 2016, 6, 15873. [Google Scholar] [CrossRef]
  112. Rabai, M.; Meiselman, H.J.; Wenby, R.B.; Detterich, J.A.; Feinberg, J. Analysis of light scattering by red blood cells in ektacytometry using global pattern fitting. Biorheology 2012, 49, 317–328. [Google Scholar] [CrossRef]
  113. Barshtein, G.; Gural, A.; Arbell, D.; Barkan, R.; Livshits, L.; Pajic-Lijakovic, I.; Yedgar, S. Red Blood Cell Deformability Is Expressed by a Set of Interrelated Membrane Proteins. Int. J. Mol. Sci. 2023, 24, 12755. [Google Scholar] [CrossRef]
  114. Sherman, I.W.; Eda, S.; Winograd, E. Erythrocyte aging and malaria. Cell. Mol. Biol. 2004, 50, 159–169. [Google Scholar] [PubMed]
  115. Duez, J.; Holleran, J.P.; Ndour, P.A.; Pionneau, C.; Diakité, S.; Roussel, C.; Dussiot, M.; Amireault, P.; Avery, V.M.; Buffet, P.A. Mechanical clearance of red blood cells by the human spleen: Potential therapeutic applications of a biomimetic RBC filtration method. Transfus. Clin. Biol. 2015, 22, 151–157. [Google Scholar] [CrossRef] [PubMed]
  116. Suresh, S.; Spatz, J.; Mills, J.P.; Micoulet, A.; Dao, M.; Lim, C.T.; Beil, M.; Seufferlein, T. Reprint of: Connections between single-cell biomechanics and human disease states: Gastrointestinal cancer and malaria. Acta Biomater. 2015, 1, 15–30. [Google Scholar] [CrossRef] [PubMed]
  117. Omodeo-Salè, F.; Motti, A.; Basilico, N.; Parapini, S.; Olliaro, P.; Taramelli, D. Accelerated senescence of human erythrocytes cultured with Plasmodium falciparum. Blood 2003, 102, 705–711. [Google Scholar] [CrossRef] [PubMed]
  118. Salinas, N.D.; Paing, M.M.; Adhikari, J.; Gross, M.L.; Tolia, N. Moderately Neutralizing Epitopes in Nonfunctional Regions Dominate the Antibody Response to Plasmodium falciparum EBA-140. Infect. Immun. 2019, 87, e00716-18. [Google Scholar] [CrossRef] [PubMed]
  119. Fraser, M.; Matuschewski, K.; Maier, A.G. Of membranes and malaria: Phospholipid asymmetry in Plasmodium falciparum-infected red blood cells. Cell. Mol. Life Sci. 2021, 78, 4545–4561. [Google Scholar] [CrossRef] [PubMed]
  120. Ahiya, A.I.; Bhatnagar, S.; Morrisey, J.M.; Beck, J.R.; Vaidya, A.B. Dramatic Consequences of Reducing Erythrocyte Membrane Cholesterol on Plasmodium falciparum. Microbiol. Spectr. 2022, 10, e0015822. [Google Scholar] [CrossRef] [PubMed]
  121. Turrini, F.; Arese, P.; Yuan, J.; Low, P.S. Clustering of integral membrane proteins of the human erythrocyte membrane stimulates autologous IgG binding, complement deposition, and phagocytosis. J. Biol. Chem. 1991, 266, 23611–23617. [Google Scholar] [CrossRef]
  122. Turrini, F.; Mannu, F.; Arese, P.; Yuan, J.; Low, P.S. Characterization of the autologous antibodies that opsonize erythrocytes with clustered integral membrane proteins. Blood 1993, 81, 3146–3152. [Google Scholar] [CrossRef]
  123. Turrini, F.; Mannu, F.; Cappadoro, M.; Ulliers, D.; Giribaldi, G.; Arese, P. Binding of naturally occurring antibodies to oxidatively and nonoxidatively modified erythrocyte band 3. Biochim. Biophys. Acta 1994, 1190, 297–303. [Google Scholar] [CrossRef]
  124. Dondorp, A.M.; Kager, P.A.; Vreeken, J.; White, N.J. Abnormal blood flow and red blood cell deformability in severe malaria. Parasitol. Today 2000, 16, 228–232. [Google Scholar] [CrossRef]
  125. Paul, A.; Pallavi, R.; Tatu, U.S.; Natarajan, V. The bystander effect in optically trapped red blood cells due to Plasmodium falciparum infection. Trans. R. Soc. Trop. Med. Hyg. 2013, 107, 220–223. [Google Scholar] [CrossRef]
  126. Barber, B.E.; Russell, B.; Grigg, M.J.; Zhang, R.; William, T.; Amir, A.; Lau, Y.L.; Chatfield, M.D.; Dondorp, A.M.; Anstey, N.M.; et al. Reduced red blood cell deformability in Plasmodium knowlesi malaria. Blood Adv. 2018, 2, 433–443. [Google Scholar] [CrossRef]
  127. Drenckhahn, D. Removal of Old and Abnormal Red Blood Cells from Circulation: Mechanical and Immunologic Mechanisms. In Blood Cells, Rheology, and Aging; Platt, D., Ed.; Springer: Berlin/Heidelberg, Germany, 1988. [Google Scholar] [CrossRef]
  128. Hosseini, S.M.; and Feng, J.J. How malaria parasites reduce the deformability of infected red blood cells. Biophys. J. 2012, 103, 1–10. [Google Scholar] [CrossRef] [PubMed]
  129. Aingaran, M.; Zhang, R.; Law, S.K.; Peng, Z.; Undisz, A.; Meyer, E.; Diez-Silva, M.; Burke, T.A.; Spielmann, T.; Lim, C.T.; et al. Host cell deformability is linked to transmission in the human malaria parasite Plasmodium falciparum. Cell. Microbiol. 2012, 14, 983–993. [Google Scholar] [CrossRef] [PubMed]
  130. Tiburcio, M.; Niang, M.; Deplaine, G.; Perrot, S.; Bischoff, E.; Ndour, P.A.; Silvestrini, F.; Khattab, A.; Milon, G.; David, P.H.; et al. A switch in infected erythrocyte deformability at the maturation and blood circulation of Plasmodium falciparum transmission stages. Blood 2012, 119, e172–e180. [Google Scholar] [CrossRef] [PubMed]
  131. Neveu, G.; Lavazec, C. Erythrocyte Membrane Makeover by Plasmodium falciparum Gametocytes. Front. Microbiol. 2019, 10, 2652. [Google Scholar] [CrossRef] [PubMed]
  132. Metwally, N.G.; Tilly, A.K.; Lubiana, P.; Roth, L.K.; Dörpinghaus, M.; Lorenzen, S.; Schuldt, K.; Witt, S.; Bachmann, A.; Tidow, H.; et al. Characterisation of Plasmodium falciparum populations selected on the human endothelial receptors P-selectin, E-selectin, CD9 and CD151. Sci. Rep. 2017, 7, 4069. [Google Scholar] [CrossRef] [PubMed]
  133. Depond, M.; Henry, B.; Buffet, P.; Ndour, P.A. Methods to Investigate the Deformability of RBC During Malaria. Front. Physiol. 2020, 10, 1613. [Google Scholar] [CrossRef] [PubMed]
  134. Hawkes, M.; Elphinstone, R.E.; Conroy, A.L.; Kain, K.C. Contrasting pediatric and adult cerebral malaria: The role of the endothelial barrier. Virulence 2013, 4, 543–555. [Google Scholar] [CrossRef] [PubMed]
  135. Stoute, J.A. Complement receptor 1 and malaria. Cell. Microbiol. 2011, 13, 1441–1450. [Google Scholar] [CrossRef] [PubMed]
  136. Miller, L.H.; Usami, S.; Chien, S. Alteration in the rheologic properties of Plasmodium knowlesi-infected red cells. A possible mechanism for capillary obstruction. J. Clin. Investig. 1971, 50, 1451–1455. [Google Scholar] [CrossRef] [PubMed]
  137. Metcalf, C.J.; Graham, A.L.; Huijben, S.; Barclay, V.C.; Long, G.H.; Grenfell, B.T.; Read, A.F.; Bjørnstad, O.N. Partitioning regulatory mechanisms of within-host malaria dynamics using the effective propagation number. Science 2011, 333, 984–988. [Google Scholar] [CrossRef] [PubMed]
  138. Eraky Mohamed, T.; Abd El-Rahman Ahmed, I.; Shazly Mostafa, H.; Abdelrahman Mohamed, M. Mechanics of deformation of malaria-infected red blood cells. Mech. Res. Commun. 2021, 113, 103666. [Google Scholar] [CrossRef]
  139. Hawking, F.; Wilson, M.E.; Gammage, K. Evidence for cyclic development and short-lived maturity in the gametocytes of Plasmodium falciparum. Trans. R. Soc. Trop. Med. Hyg. 1971, 65, 549–559. [Google Scholar] [CrossRef] [PubMed]
  140. Joice, R.; Nilsson, S.K.; Montgomery, J.; Dankwa, S.; Egan, E.; Morahan, B.; Seydel, K.B.; Bertuccini, L.; Alano, P.; Williamson, K.C.; et al. Plasmodium falciparum transmission stages accumulate in the human bone marrow. Sci. Transl. Med. 2014, 6, 244re5. [Google Scholar] [CrossRef]
  141. Bachmann, A.; Esser, C.; Petter, M.; Predehl, S.; von Kalckreuth, V.; Schmiedel, S.; Bruchhaus, I.; Tannich, E. Absence of erythrocyte sequestration and lack of multicopy gene family expression in Plasmodium falciparum from a splenectomized malaria patient. PLoS ONE 2009, 4, e7459. [Google Scholar] [CrossRef]
  142. Meibalan, E.; Marti, M. Biology of Malaria Transmission. Cold Spring Harb. Perspect. Med. 2017, 7, a025452. [Google Scholar] [CrossRef]
  143. Glenister, F.K.; Coppel, R.L.; Cowman, A.F.; Mohandas, N.; Cooke, B.M. Contribution of parasite proteins to altered mechanical properties of malaria-infected red blood cells. Blood 2002, 99, 1060–1063. [Google Scholar] [CrossRef] [PubMed]
  144. Mills, J.P.; Diez-Silva, M.; Quinn, D.J.; Dao, M.; Lang, M.J.; Tan, K.S.; Lim, C.T.; Milon, G.; David, P.H.; Mercereau-Puijalon, O.; et al. Effect of plasmodial RESA protein on deformability of human red blood cells harboring Plasmodium falciparum. Proc. Natl. Acad. Sci. USA 2007, 104, 9213–9217. [Google Scholar] [CrossRef] [PubMed]
  145. Kaviratne, M.; Khan, S.M.; Jarra, W.; Preiser, P.R. Small variant STEVOR antigen is uniquely located within Maurer’s clefts in Plasmodium falciparum-infected red blood cells. Eukaryot. Cell 2002, 1, 926–935. [Google Scholar] [CrossRef] [PubMed]
  146. Sam-Yellowe, T.Y.; Florens, L.; Johnson, J.R.; Wang, T.; Drazba, J.A.; Le Roch, K.G.; Zhou, Y.; Batalov, S.; Carucci, D.J.; Winzeler, E.A.; et al. A Plasmodium gene family encoding Maurer’s cleft membrane proteins: Structural properties and expression profiling. Genome Res. 2004, 14, 1052–1059. [Google Scholar] [CrossRef]
  147. Lavazec, C.; Sanyal, S.; Templeton, T.J. Hypervariability within the Rifin, Stevor and Pfmc-2TM superfamilies in Plasmodium falciparum. Nucleic Acids Res. 2006, 34, 6696–6707. [Google Scholar] [CrossRef]
  148. Blythe, J.E.; Yam, X.Y.; Kuss, C.; Bozdech, Z.; Holder, A.A.; Marsh, K.; Langhorne, J.; Preiser, P.R. Plasmodium falciparum STEVOR proteins are highly expressed in patient isolates and located in the surface membranes of infected red blood cells and the apical tips of merozoites. Infect. Immun. 2008, 76, 3329–3336. [Google Scholar] [CrossRef]
  149. Kaur, J.; Hora, R. ‘2TM proteins’: An antigenically diverse superfamily with variable functions and export pathways. PeerJ 2018, 6, e4757. [Google Scholar] [CrossRef]
  150. Sanyal, S.; Egee, S.; Bouyer, G.; Perrot, S.; Safeukui, I.; Bischoff, E.; Buffet, P.; Deitsch, K.W.; Mercereau-Puijalon, O.; David, P.H.; et al. Plasmodium falciparum STEVOR proteins impact erythrocyte mechanical properties. Blood 2012, 119, e1–e8. [Google Scholar] [CrossRef]
  151. McRobert, L.; Preiser, P.; Sharp, S.; Jarra, W.; Kaviratne, M.; Taylor, M.C.; Renia, L.; Sutherland, C.J. Distinct trafficking and localization of STEVOR proteins in three stages of the Plasmodium falciparum life cycle. Infect. Immun. 2004, 72, 6597–6602. [Google Scholar] [CrossRef] [PubMed]
  152. Sharp, S.; Lavstsen, T.; Fivelman, Q.L.; Saeed, M.; McRobert, L.; Templeton, T.J.; Jensen, A.T.; Baker, D.A.; Theander, T.G.; Sutherland, C.J. Programmed transcription of the var gene family, but not of stevor, in Plasmodium falciparum gametocytes. Eukaryot. Cell 2006, 5, 1206–1214. [Google Scholar] [CrossRef] [PubMed]
  153. Petter, M.; Haeggström, M.; Khattab, A.; Fernandez, V.; Klinkert, M.Q.; Wahlgren, M. Variant proteins of the Plasmodium falciparum RIFIN family show distinct subcellular localization and developmental expression patterns. Mol. Biochem. Parasitol. 2007, 156, 51–61. [Google Scholar] [CrossRef] [PubMed]
  154. Daily, J.P.; Le Roch, K.G.; Sarr, O.; Ndiaye, D.; Lukens, A.; Zhou, Y.; Ndir, O.; Mboup, S.; Sultan, A.; Winzeler, E.A.; et al. . In vivo transcriptome of Plasmodium falciparum reveals overexpression of transcripts that encode surface proteins. J. Infect. Dis. 2005, 191, 1196–1203. [Google Scholar] [CrossRef]
  155. Huang, Y.X.; Wu, Z.J.; Mehrishi, J.; Huang, B.T.; Chen, X.Y.; Zheng, X.J.; Liu, W.J.; Luo, M. Human red blood cell aging: Correlative changes in surface charge and cell properties. J. Cell. Mol. Med. 2011, 15, 2634–2642. [Google Scholar] [CrossRef]
  156. Leong, Y.W.; Russell, B.; Malleret, B.; Rénia, L. Erythrocyte tropism of malarial parasites: The reticulocyte appeal. Front. Microbiol. 2022, 13, 1022828. [Google Scholar] [CrossRef] [PubMed]
  157. Kerlin, D.H.; Gatton, M.L. Preferential invasion by Plasmodium merozoites and the self-regulation of parasite burden. PLoS ONE 2013, 8, e57434. [Google Scholar] [CrossRef]
  158. Amir, A.; Russell, B.; Liew, J.; Moon, R.W.; Fong, M.Y.; Vythilingam, I.; Subramaniam, V.; Snounou, G.; Lau, Y.L. Invasion characteristics of a Plasmodium knowlesi line newly isolated from a human. Sci. Rep. 2016, 6, 24623. [Google Scholar] [CrossRef]
  159. McQueen, P.G.; McKenzie, F.E. Age-structured red blood cell susceptibility and the dynamics of malaria infections. Proc. Natl. Acad. Sci. USA 2004, 101, 9161–9166. [Google Scholar] [CrossRef] [PubMed]
  160. McKenzie, F.E.; Jeffery, G.M.; Collins, W.E. Plasmodium malariae blood-stage dynamics. J. Parasitol. 2001, 87, 626–637. [Google Scholar] [CrossRef]
  161. Abate, A.; Bouyssou, I.; Mabilotte, S.; Doderer-Lang, C.; Dembele, L.; Menard, D.; Golassa, L. Vivax malaria in Duffy-negative patients shows invariably low asexual parasitaemia: Implication towards malaria control in Ethiopia. Malar. J. 2022, 21, 230. [Google Scholar] [CrossRef]
  162. Ferreira, N.S.; Mathias, J.L.S.; Albuquerque, S.R.L.; Almeida, A.C.G.; Dantas, A.C.; Anselmo, F.C.; Lima, E.S.; Lacerda, M.V.G.; Nogueira, P.A.; Ramasawmy, R.; et al. Duffy blood system and G6PD genetic variants in vivax malaria patients from Manaus, Amazonas, Brazil. Malar. J. 2022, 21, 144. [Google Scholar] [CrossRef]
  163. Woolley, I.J.; Wood, E.M.; Sramkoski, R.M.; Zimmerman, P.A.; Miller, J.P.; Kazura, J.W. Expression of Duffy antigen receptor for chemokines during reticulocyte maturation: Using a CD71 flow cytometric technique to identify reticulocytes. Immunohematology 2005, 21, 15–20. [Google Scholar] [CrossRef] [PubMed]
  164. Moras, M.; Lefevre, S.D.; Ostuni, M.A. From Erythroblasts to Mature Red Blood Cells: Organelle Clearance in Mammals. Front. Physiol. 2017, 8, 1076. [Google Scholar] [CrossRef] [PubMed]
  165. Coban, C.; Lee, M.S.J.; Ishii, K.J. Tissue-specific immunopathology during malaria infection. Nat. Rev. Immunol. 2018, 18, 266–278. [Google Scholar] [CrossRef] [PubMed]
  166. Butcher, G.A.; Mitchell, G.H.; Cohen, S. Mechanism of host specificity in malarial infection. Nature 1973, 244, 40–42. [Google Scholar] [CrossRef]
  167. Swann, A.I. The relationship of erythrocyte age and parasitization with Plasmodium gallinaceum in chickens. Can. J. Comp. Med. 1974, 38, 391–397. [Google Scholar]
  168. Ben Mamoun, C.; Prigge, S.T.; Vial, H. Targeting the Lipid Metabolic Pathways for the Treatment of Malaria. Drug Dev. Res. 2010, 71, 44–55. [Google Scholar] [CrossRef]
  169. Ancelin, M.L.; Parant, M.; Thuet, M.J.; Philippot, J.R.; Vial, H.J. Increased permeability to choline in simian erythrocytes after Plasmodium knowlesi infection. Biochem. J. 1991, 273 Pt 3, 701–709. [Google Scholar] [CrossRef] [PubMed]
  170. Ramaprasad, A.; Burda, P.C.; Calvani, E.; Sait, A.J.; Palma-Duran, S.A.; Withers-Martinez, C.; Hackett, F.; Macrae, J.; Collinson, L.; Gilberger, T.W.; et al. A choline-releasing glycerophosphodiesterase essential for phosphatidylcholine biosynthesis and blood stage development in the malaria parasite. eLife 2022, 11, e82207. [Google Scholar] [CrossRef]
  171. Kirk, K.; Poli de Figueiredo, C.E.; Elford, B.C.; Ellory, J.C. Effect of cell age on erythrocyte choline transport: Implications for the increased choline permeability of malaria-infected erythrocytes. Biochem. J. 1992, 283, 617–619. [Google Scholar] [CrossRef]
  172. Griffiths, R.E.; Kupzig, S.; Cogan, N.; Mankelow, T.J.; Betin, V.M.S.; Trakarnsanga, K.; Massey, E.J.; Lane, J.D.; Parsons, S.F.; Anstee, D.J. Maturing reticulocytes internalize plasma membrane in glycophorin A–containing vesicles that fuse with autophagosomes before exocytosis. Blood 2012, 119, 6296–6306. [Google Scholar] [CrossRef]
  173. Malleret, B.; Xu, F.; Mohandas, N.; Suwanarusk, R.; Chu, C.; Leite, J.A.; Low, K.; Turner, C.; Sriprawat, K.; Zhang, R.; et al. Significant biochemical, biophysical and metabolic diversity in circulating human cord blood reticulocytes. PLoS ONE 2013, 8, e76062. [Google Scholar] [CrossRef] [PubMed]
  174. Aniweh, Y.; Gao, X.; Hao, P.; Meng, W.; Lai, S.K.; Gunalan, K.; Chu, T.T.; Sinha, A.; Lescar, J.; Chandramohanadas, R.; et al. P. falciparum RH5-Basigin interaction induces changes in the cytoskeleton of the host RBC. Cell. Microbiol. 2017, 19, e12747. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Main receptors and ligands involved in P. falciparum invasion of human erythrocytes. Gly A: glycophorin A. Gly B: glycophorin B. Gly C: Glycophorin C. CR1: Complement Receptor 1. PMCA: plasma membrane calcium ATPase. MCT: monocarboxylate transporter. CD55: Decay-Accelerating Factor (DAF). EBA-175: Erythrocyte Binding Antigen-175. EBL-1: Erythrocyte Binding Ligand-1. EBA-140: Erythrocyte Binding Antigen-140. EBA-181: Erythrocyte Binding Antigen-181. PfRh2b: P. falciparum Rhoptry homolog 2b. PfRh4: P. falciparum Rhoptry homolog 4. PfRh5: P. falciparum Rhoptry homolog 5. SA: Sialic Acid. The illustration is not at scale. This figure is created with BioRender.com.
Figure 1. Main receptors and ligands involved in P. falciparum invasion of human erythrocytes. Gly A: glycophorin A. Gly B: glycophorin B. Gly C: Glycophorin C. CR1: Complement Receptor 1. PMCA: plasma membrane calcium ATPase. MCT: monocarboxylate transporter. CD55: Decay-Accelerating Factor (DAF). EBA-175: Erythrocyte Binding Antigen-175. EBL-1: Erythrocyte Binding Ligand-1. EBA-140: Erythrocyte Binding Antigen-140. EBA-181: Erythrocyte Binding Antigen-181. PfRh2b: P. falciparum Rhoptry homolog 2b. PfRh4: P. falciparum Rhoptry homolog 4. PfRh5: P. falciparum Rhoptry homolog 5. SA: Sialic Acid. The illustration is not at scale. This figure is created with BioRender.com.
Cells 13 00334 g001
Figure 2. Cytoadherence events and main actors in the vascular system that takes place during P. falciparum infection and development in humans. ICAM-1: Adhesion Molecule-1, VCAM: Vascular cellular adhesion molecule, TSP: Thrombospondin, CD62P: P-selectin, CD62E: E-selectin, PfEMP1: P. falciparum Erythrocyte Membrane Protein 1. The illustrations are not at scale. The inset at the top of the figure represents a protein expressed in the membrane of each stage of P. falciparum: Gametocyte stages I–IV, trophozoite stage, and schizont stages. This figure is created with BioRender.com.
Figure 2. Cytoadherence events and main actors in the vascular system that takes place during P. falciparum infection and development in humans. ICAM-1: Adhesion Molecule-1, VCAM: Vascular cellular adhesion molecule, TSP: Thrombospondin, CD62P: P-selectin, CD62E: E-selectin, PfEMP1: P. falciparum Erythrocyte Membrane Protein 1. The illustrations are not at scale. The inset at the top of the figure represents a protein expressed in the membrane of each stage of P. falciparum: Gametocyte stages I–IV, trophozoite stage, and schizont stages. This figure is created with BioRender.com.
Cells 13 00334 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alves-Rosa, M.F.; Tayler, N.M.; Dorta, D.; Coronado, L.M.; Spadafora, C. P. falciparum Invasion and Erythrocyte Aging. Cells 2024, 13, 334. https://doi.org/10.3390/cells13040334

AMA Style

Alves-Rosa MF, Tayler NM, Dorta D, Coronado LM, Spadafora C. P. falciparum Invasion and Erythrocyte Aging. Cells. 2024; 13(4):334. https://doi.org/10.3390/cells13040334

Chicago/Turabian Style

Alves-Rosa, María Fernanda, Nicole M. Tayler, Doriana Dorta, Lorena M. Coronado, and Carmenza Spadafora. 2024. "P. falciparum Invasion and Erythrocyte Aging" Cells 13, no. 4: 334. https://doi.org/10.3390/cells13040334

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop