Abstract
Microtubules, composed of α- and β-tubulin polymers, are essential components of the eukaryotic cytoskeleton. They maintain cellular shape and structural integrity and play critical roles in cell division and in intracellular vesicular transport. In Plasmodium, the parasite that causes malaria, nuclear replication during the liver stage is among the fastest known in eukaryotic cells and relies heavily on microtubules for DNA segregation and cytoskeletal organization. Despite their importance, the role of microtubules in liver stage development remains largely unexplored. Here, we investigated microtubule dynamics during liver stage development using a combination of cell and molecular biology techniques, expansion microscopy, and live-cell imaging. By employing antibodies specific for α-tubulin post-translational modifications (PTMs), we found that the Plasmodium sporozoites subpellicular microtubules (SSPM) persist during liver infection, giving rise to liver stage parasite microtubule bundles (LSPMB). These LSPMB form multimeric tubulin structures within hepatocytes and are redistributed to the hemi-spindle poles of parasite nuclei during schizogony. Deletion of the C-terminal region encompassing all known Plasmodium α-tubulin PTM sites prevented sporozoite migration from the mosquito midgut to the salivary glands, effectively blocking parasite transmission. Using Plasmodium microtubule-specific depolymerisation drugs, we found that while LSPMB are stable in sporozoites, they exhibit dynamic behavior during hepatocyte infection. Given the regulatory role of PTMs in microtubule dynamics, we generated parasite mutants by substituting and deleting key α-tubulin C-terminal residues involved in PTMs. Substitution of the polyglutamylation site with alanine and deletion of the C-terminal tyrosination/detyrosination motifs impaired parasite growth during liver infection. Together, our findings reveal extensive microtubule remodeling during liver stage development and establish α-tubulin C-terminal modifications as critical regulators of both intracellular development and parasite transmission of Plasmodium parasites.
Introduction
Plasmodium parasites are responsible for malaria and cause yearly half a million deaths in sub-Saharan countries. Malaria continues to be a major worldwide health concern despite recent successful attempts to lower its prevalence (1–4). The infection starts when an infected female Anopheles mosquito, during a blood meal, injects salivary gland sporozoites into the skin of the vertebrate host. Some of the motile sporozoites escape the host immune defence and invade blood vessels. They are carried by the blood flow to the liver, where they invade and infect hepatocytes (5). Approximately 50% of the parasites are often killed by host cell responses (6–8). Those that managed to establish a productive invasion form a parasitophorous vacuole (PV) in which the parasite undergoes massive growth and asexual replication known as schizogony. One single parasite gives rise to thousands of daughter parasites called merozoites in 3 to 6 days depending on the Plasmodium species (9–11). Fully developed merozoites exit the liver cells in merosome vesicles through adjacent blood vessels (11–14). The merosomes then rupture and release merozoites into the blood stream. The parasites immediately invade erythrocytes, where they undergo again asexual replication to form the blood stage merozoites. Some merozoites differentiate into sexual stages, termed gametocytes, which are ingested by female mosquitos during a blood meal. In the mosquito midgut, the gametocytes develop into microgametes and macrogametes, which fertilize and form a zygote. The later undergoes differentiation into motile ookinetes, which penetrate the gut wall, forming oocysts (15–18). The oocyst produces the midgut sporozoites from the sporoblasts. The sporozoites migrate to the salivary gland from day 11 post-infection in P. berghei (Frischknecht et al., 2004; Frischknecht & Matuschewski, 2017; jerome P. Vanderberg, 1974; J. P. Vanderberg, 1975). During the liver stage development, the parasite relies on the fast and massive asexual replication which is essential for the transmission and pathogenesis in erythrocyte stage, representing key targets for malaria control strategies. Investigating the parasite microtubule dynamics during the rapid replication could provide important insight on the parasite biology and to develop strategies for halting the massive proliferation during the clinically asymptomatic liver stage.
Microtubules are cytoskeletal filaments of about 25nm outer diameter, made up of usually 13 chains of protofilaments composed of globular α-tubulin and β-tubulin heterodimers. They are crucial components of the cytoskeleton and offer structural support to cells and also function as tracks for the movement of organelles, vesicles, and other cellular components (23,24). Microtubules are involved in important cellular activities such as cell division and the production of cilia and flagella to allow cell motility. Microtubules exhibit inherent dynamics, alternating between polymerisation (rapid growth) and spontaneous depolymerisation (shrinkage) in a process termed dynamic instability (23,25,26). The dynamics of microtubules and their interactions with other cellular structures are regulated by specific microtubule-associated proteins (MAPs), which selectively bind to either the plus or minus ends of microtubules (27–30).
Tubulin is subject to seven identified post-translational modifications (PTMs), among which are acetylation of the lysine residue at position 40 in the luminally exposed part of α-tubulin, glycylation, which entails the incorporation of polyglycine side chains of variable length, phosphorylation, palmitoylation, tyrosination/ detyrosination and polyglutamylation. These modifications are extensively preserved and enabling cells to modulate microtubule characteristics, dynamics, and interactions with microtubule-associated proteins (MAPs) and motor proteins (31,32). Tyrosinated microtubules are often associated with microtubule stability. These stable microtubules represent a specialised subset that exhibits enhanced resistance to depolymerization, even in the presence of destabilizing agents. They play a crucial role in maintaining cellular architecture, facilitating intracellular transport, and supporting specialized cellular functions. In contrast to dynamic microtubules, which continuously undergo cycles of polymerization and depolymerization due to dynamic instability, stable microtubules persist for extended periods. They are also frequently polyglutamylated, a modification that regulates their interactions with motor proteins and other cellular components. These microtubules are predominantly localized in structures requiring sustained structural integrity, such as cilia, flagella, neuronal axons, and motile cell types (33–36). Glutamylation, or polyglutamylation, is a post-translational modification characterized by the addition of multiple glutamate side chains to glutamate residues on both α- and β-tubulin. This modification occurs primarily at the C-terminal tails and is catalyzed by a family of enzymes known as polyglutamylases. Polyglutamylation plays a critical role in regulating the interactions of microtubules with MAPs and motor proteins, thereby influencing microtubule dynamics and cellular functions (37–40). The properties and functions of tubulin are largely encoded within their protein sequences, contributing to molecular diversity that influences key microtubule characteristics. This variation has been conceptualized as the “tubulin code”, a regulatory framework through which cells control microtubule diversity to achieve specific physiological functions (24). Nevertheless, the precise mechanisms by which cells establish and interpret this code remain largely unknown. Although tubulin is highly conserved across evolution, it exhibits structural and functional variations between species, cell types, and even within single cells.
In eukaryotic cells, tubulins are key components of the microtubule cytoskeleton, and their genetic complexity reflects the organism’s level of cellular specialization. In yeast, two distinct genes encode α-tubulin, while a single gene encodes β-tubulin. In contrast, the human genome contains nine genes encoding α-tubulin and an equal number encoding β-tubulin, reflecting a greater level of complexity and functional specialization in higher eukaryotes (24,41,42).
The Plasmodium genome encodes two α-tubulins (α1-tubulin and α2-tubulin) and one β-tubulin (43–47). The α1-tubulin gene encodes a protein of 450 amino acids, while α2-tubulin encodes 453 amino acids (48). The two isoforms share 94% amino acid identity. However, α2-tubulin lacks a terminal tyrosine residue, which is present in α1-tubulin. This difference suggests potential variation in post-translational modifications and functional roles (44,49). In P. berghei, the α1-tubulin gene is expressed across most stages of the Plasmodium life cycle, whereas α2-tubulin expression appears restricted to male gametocytes, gametes, and newly formed zygotes, indicating distinct functional roles for the two isoforms (32,50). Deletion of α1-tubulin blocks sporozoite formation in oocysts and lowering α1-tubulin expression leads to less infective sporozoites (50).
There is a significant degree of amino acid conservation between human and Plasmodium tubulins, with approximately 83.7% identity for α-tubulin and 88.5% for β-tubulin. However, Plasmodium tubulin shows greater similarity to plant tubulin than to mammalian tubulin. This raises the potential for developing compounds that selectively target parasite microtubules while sparing the host cell cytoskeleton, offering a promising avenue for therapeutic intervention (43,45–47,51).
Recent studies on Plasmodium falciparum blood stage schizonts have revealed that hemi-spindles lack polyglutamylation, whereas subpellicular microtubules do exhibit this post-translational modification (52–54). Similarly, SPMTs appear to be stabilized by microtubule inner proteins, while spindle microtubules do not show these luminal proteins (55). These differences may reflect functional specialisation of microtubule subpopulations in different cellular structures and developmental stages.
The liver stage of Plasmodium exhibits one of the fastest rates of replication known among eukaryotic cells, a process likely dependent on microtubule dynamics. However, to date, no studies have investigated microtubule dynamics during the liver stage. This study was therefore designed to explore the dynamics of parasite tubulin during the asexual liver stage, using recently developed expansion microscopy protocols (54,56–58).
Results
Expansion microscopy reveals the presence of the sporozoites subpellicular microtubules during the liver stage of infection
To investigate microtubule organization during the Plasmodium parasite liver stage, HeLa cells were infected with sporozoites and fixed them at different time points using cold methanol. We then performed IFA to stain host and parasite microtubules using an α-tubulin antibody (Figure 1A). The IFA revealed a marked accumulation of microtubules in proximity of the PVM (stained with anti-UIS4) from early infection (24 hours post-infection (hpi)) to the schizogony (30 to 54 hpi) (Figure 1A inlets). To examine these structures in greater detail, we applied the recently developed pre-staining expansion microscopy (PS-ExM) (56). By expanding the cells five-fold, PS-ExM enabled visualization of a large multimeric tubulin structure within the parasite, detectable from 6 hours post-infection (hpi) to 48 hpi (Figure 1B). Based on observations of sporozoites prior to liver infection, we hypothesize that this structure originates from the sporozoite subpellicular microtubules (SSPM) (Figure 1C top panel). We refer to this structure as the liver stage parasite microtubule bundles (LSPMB). Notably, the LSPMB progressively shrank over time and was no longer detectable by the end of schizogony (54 hpi) (Figure 1A, B). Using the 3D and 4D imaging software IMARIS, we quantified the LSPMB volume at various stages of liver infection. The results confirmed a consistent decrease in LSPMB size between 6 and 56 hpi (Figure 1D).

Immunofluorescence assay and expansion microscopy (PS-ExM) reveals liver stage parasite subpellicular microtubule.
(A), and (B), show maximum projection of z-stacks confocal images of infected HeLa cells and (C) sporozoites. Host and parasite microtubules were stained with anti-α-tubulin (magenta), and nuclei with DAPI (blue). The parasitophorous membrane vacuole (PVM) was stained with anti-UIS4 (green). (A) HeLa cells were infected with Plasmodium salivary glands sporozoites and methanol-fixed at 24, 30, 48, and 56 hpi. IFA was performed. (B) Fixed infected HeLa cells were expanded using the PS-ExM protocol. Inlets show LSPMB structures at 6, 30, and 48 hpi. Scale bar: 10 µm. (C) To determine whether the LSPMB originates from the sporozoite stage, PS-ExM was performed on salivary gland sporozoites, and parasite liver stage at 2 and 6 hpi. (D) LSPMB volume was quantified from PS-ExM z-stacks using 3D and 4D analysis in IMARIS software. Normalised mean volumes were: 1294.12 µm³ in sporozoites, 628.34 µm³ at 2 hpi, 414.84 µm³ at 24 hpi, 231.4 µm³ at 48 hpi, to 5.6 µm³ at 54 hpi. (E) Live cell confocal imaging of infected HeLa cells incubated with tubulin tracker SPY650. Images show microtubules at 10, 30, 36, and 48 hpi. Host and parasite microtubules are shown in magenta; the parasite expresses cytosolic GFP (green). Scale bar: 5 µm.
To verify that the LSPMB is not an artifact of the IFA and PS-ExM, we performed live cell imaging of infected HeLa cells. Cells were incubated with the tubulin tracker SPY650 for one hour prior to imaging. The LSPMB was clearly visible in sporozoites before invasion (Movie 1, ∼10 hpi) and persisted until late liver stage development, gradually shrinking during schizogony (Figure 1E; Movies 2–4, ∼36 hpi to 48 hpi). These live imaging results corroborate our findings from IFA and expansion microscopy, validating the presence and dynamic nature of the LSPMB during liver stage development.
The liver stage parasite subpellicular microtubules are dynamic
The SSPM has previously been described as highly stable structures, enabling sporozoite formation and invasion (50,55,59). These microtubules, similar to the corresponding microtubules in T. gondii, are resistant to conventional depolymerizing agents (59). To investigate whether the LSPMB retain this stability during liver stage development, we treated infected HeLa cells with the microtubule-depolymerizing agents Amiprophos-methyl (AMP) and oryzalin herbicide. These compounds are known to selectively target dynamic Plasmodium microtubules without affecting mammalian cells (43,45,47,51,60) (Figure 2 and Supplementary Figure 1A and B). We first validated drug specificity and cytotoxicity by testing various concentrations on HeLa cells over one week. Neither compound affected host cell viability (Supplementary Figure 1A and B) or disrupted host microtubules (Supplementary Figure 1D and E). However, oryzalin showed a stronger inhibitory effect on parasite growth compared to AMP (Supplementary Figure C) and was therefore selected for further experiments (Figure 2A). Sporozoites were dissected into medium containing 10 µM or 20 µM oryzalin and then used to infect HeLa cells (Figure 2A and B), which were incubated with the same drug concentrations. At 48 hpi, the cells were fixed and analyzed by IFA and PS-ExM. To test reversibility, oryzalin was washed out at 24 hpi prior to the onset of schizogony (Figure 2B). At 10 µM, oryzalin significantly impaired nuclear replication, spindle formation, and LSPMB volume when present the entire cultivation time (Figure 2C, D and E). Partial recovery of hemi-spindles and nuclear replication was observed, suggesting that the effect of 10 µM oryzalin is reversible at this stage (Figure 2C, D, and E). However, at 20 µM, neither the hemi-spindles nor the LSPMB recovered after drug removal, indicating irreversible microtubule disruption at higher concentrations (Figure 2A and B). To investigate whether the microtubule depolymerization by oryzalin affects the parasite growth and survival during the liver stage, we measured parasite size within infected HeLa cells at 6, 24 and 48 hpi. Treatment with 10 µM oryzalin led to a significant reduction in parasite size and survival (Figure 2 F, G, H, and I). We further examined merozoite formation by staining for MSP1 at 56 hpi. Parasites treated with 10 µM oryzalin failed to express MSP1, indicating a block in merozoite development and suggesting that intact microtubules are essential for DNA replication and merozoite formation (Figure 2J). Interestingly, at lower concentrations of 0.5 µM, oryzalin treatment led to an unexpected elongation of the LSPMB and hemi-spindles up to four times longer than controls suggesting that microtubule turnover is tightly regulated (Supplementary Figure 1F, G and H). In conclusion, oryzalin-mediated depolymerization confirms that both the LSPMB and the hemi-spindles are dynamic structures during the liver stage. This dynamic behaviour was further supported by live-cell imaging using the SPY650 tubulin tracker (Movie 5, 6, and 7).

Microtubules are essential for Plasmodium liver stage development.
(A and B) show the effect of oryzalin on parasite microtubules during liver stage development. Sporozoites isolated from mosquito salivary glands were incubated in medium containing either 10 or 20 µM of oryzalin. HeLa cells were infected and fixed at 48 hpi with cold methanol prior to PS-ExM. (A) confocal z-stack projections of control cells (DMSO) and cells treated with 10 and 20 µM of Oryzalin. (B) confocal z-stack projections of infected cells where oryzalin was washed out at 24 hpi. Microtubule was stained with anti-α-tubulin (magenta), the PVM with anti UIS4 (green), and nuclei with DAPI (blue). Scale bar: 10 µm. ‘’+’’ indicates cells treated with Oryzalin; ‘’ - ‘’ indicates cells from which the drug has been removed at 24 hpi. (C) Normalized total parasite DNA volume at 48 hpi. Mean DNA volume was 1952.02 µm³ in the DMSO control, 827.26 µm³ in 10 µM oryzalin-treated cells, and 1411.73 µm³ after drug removal. (D) Normalized total hemi-spindle volume. The mean volume was 539.3 µm ³ in the control, 0 µm³ with oryzalin treatment, and 89.08 µm³ after drug removal. (E) Normalized LSPMB volume at 48 hpi. Mean volume was 5.92 µm³ in the control, 2.3 µm³ with oryzalin, and 1.65 µm³ after drug removal. (F, G and H) Parasite size measurement at 6, 24, and 48 hpi, respectively. At 24 and 48 hpi, mean parasite size was 145.9 µm² in the control, 71.75 µm² with oryzalin, and 115.3 µm² after drug removal. (I) Parasite survival rate from 6 to 48 hpi. A slight increase in parasite death was observed in oryzalin-treated cells compared to the DMSO control. The red line on the graphs represents the mean and standard deviation. (C, D, E, F, and G), and (H): each dot represents an individual quantified parasite. (J) Confocal z-stack projection of IFA images at 56 hpi in cells treated with 10 µM oryzalin and DMSO control. Cells were fixed with cold methanol and stained for MSP1 (magenta), α-tubulin (grey) and nuclei (DAPI, blue). Scale bar: 5 µm. Statistical comparisons were performed using one-way ANOVA with Dunnett’s multiple comparisons test (*p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001).
Given the observed microtubule remodeling in response to oryzalin, we next sought to determine whether known microtubule-associated proteins, specifically those regulating plus-end dynamics, are expressed during the liver stage and might contribute to the observed plasticity. Only a limited number of motor proteins and microtubule associated proteins, such as centrin and the kinesin protein family, have been identified during the Plasmodium liver stage (11,61–63). The recently characterized EB1 protein, known to regulate microtubules plus-end dynamics and length, has so far been reported for being expressed during the sexual blood stage (Mauer et al., 2023; Yang et al., 2023; Zeeshan et al., 2023). However, these studies did not assess EB1 expression during the asexual mosquito and liver stages. Given our observations of dynamic microtubule behavior and excessive polymerization following oryzalin treatment, we explored whether EB1 might be expressed during the liver stage. To this end, we generated a parasite line expressing endogenous EB1 fused to eGFP via double homologous recombination (Supplementary Figure 2C and D). As expected, EB1-eGFP was detected in sexual blood stages (Supplementary Figure 2E and F) consistent with previous reports (64,65). Interestingly, we also observed EB1 expression in both midgut and salivary gland sporozoites (Supplementary Figure 2G). Importantly, EB1 was also detected during liver stage development, where it localized to the LSPMB and hemi-spindles (Supplementary Figure 2H–I).
These findings suggest that EB1 may play a broader role in Plasmodium microtubule regulation than previously recognized, potentially contributing to the dynamic remodeling of microtubules during the liver stage.
The liver stage parasite subpellicular microtubule carries tubulin post-translational modifications
Microtubule functions are often regulated by different post-translational modifications (PTMs), which influence their stability, interactions, and dynamics. To gain insight into the dynamic nature of LSPMB during the liver stage development, we examined α-tubulin PTMs using immunofluorescence (IFA) and expansion microscopy (Figure 3 and Movie 5, 6 and 7). To investigate whether the LSPMB is present in cell types other than HeLa cells, we infected primary hepatocytes with Plasmodium sporozoites and fixed the cells at different timepoints (6, 24, 36, 48, 54 and 56 hpi). We also analyzed sporozoites isolated from mosquito salivary glands. Using antibodies specific for polyglutamylated (anti-IN105) (66) (Figure 3A) and tyrosinated (anti-Ty) α-tubulin (Figure 3B), we observed that sporozoite microtubules, including the LSPMB, are both polyglutamylated and tyrosinated. Quantitative colocalization analysis confirmed a strong overlap between LSPMB and polyglutamylated tubulin in both sporozoites and liver stage parasites (Supplementary Figure 2A). Interestingly, following hepatocyte invasion, the LSPMB lost tyrosination (i.e., became detyrosinated) while retaining polyglutamylation. This detyrosination persisted throughout liver stage development, including early and late schizogony (24–56 hpi). During this period, polyglutamylation was observed on the LSPMB and at the poles of the hemi-spindles, but not along the polymerizing hemi-spindles themselves, which eventually disappeared along with the LSPMB (Figure 3A and C). Given previous findings implicating polyglutamylation in hemi-spindle pole formation in erythrocytic merozoites (31), we investigated whether there is a structural relationship between the LSPMB and the hemi-spindle poles. Using PS-ExM and the tubulin tracker SPY650, we visualized direct connections between LSPMB filaments and hemi-spindle poles. Live cell imaging confirmed this connection between LSPMB and hemi-spindle poles, mainly during the shrinkage of the LSPMB (Fig 3 F; Movie 8, 9 and 10, Supplementary Movie 1). These observations suggest that the LSPMB may be redistributed to form or support the hemi-spindle poles during schizogony. We also stained for γ-tubulin and observed its localization at the hemi-spindle poles in both the parasite and host cell (Supplementary Figure 2B), supporting the idea that Plasmodium may utilize γ-tubulin for nuclear division and microtubule assembly, as reported in other systems (67–70).

LSPMB is polyglutamylate.
(A and B) show maximum projection of confocal z-stacks form sporozoites and infected primary hepatocytes at different time points during liver stage development. Primary hepatocytes were isolated from mice and infected with sporozoites. Cells were fixed and stained with antibodies against (A) polyglutamylated tubulin (anti-IN105), (B) tyrosinated tubulin (anti-Ty), and α-tubulin (anti-α-tubulin). Nuclei were stained with DAPI (blue), and the PVM with anti-UIS4 (grey). ROIs in (A) show the LSPMB at 6 and 36 hpi and polyglutamylated hemi-spindle poles at 48 hpi. ROIs in (B) display LSPMB and α-tubulin evolution across liver stages. (C) Normalized LSPMB volume and the hemi-spindle pole counts from 6 hpi to 56 hpi, quantified using IMARIS 3D/4D analysis. LSPMB volume decreased from 1038.9 µm³ at 6 hpi to 0 µm³ at 56 hpi, while hemi-spindle poles count increased from 0 at 6 hpi to a median of 928 at 56 hpi, showing a perfect Spearman anti-correlation coefficient of -1. n = 15 per time point. (D) Single-plane PS-ExM confocal image of infected HeLa cells at 36 hpi with parasites expressing PMP1::GFP. Cells were PFA-fixed and stained with anti-GFP (magenta), anti-IN105 (green), and anti-UIS4 (PVM). The yellow arrow indicates the direction of Cell RGB Profiler analysis from outside the PVM to inside the parasite. (E) Pixel intensity profiles (arbitrary units) for PVM (grey), LSPMB (green), and parasite plasma membrane (magenta) in expanded (PS-ExM) cells (E) and non-expanded (NE) cells. Overlap was observed in NE cells, while PS-ExM resolved LSPMB localization between the PVM and the parasite plasma membrane. n=10, in triplicate. Scare bar: 10 µm. (F) Single-plane confocal image of infected primary hepatocytes fixed with cold methanol. Cells were stained with anti-IN105 (green), α-tubulin (magenta), and DAPI (blue). The ROI shows a LSPMB tubule in direct contact with a parasite hemi-spindle pole. MZP: Maximum Z projection. Scale bars: 10 µm.
To further define the spatial localization of the LSPMB, we used P. berghei parasites expressing the plasma membrane protein PMP1 fused to superfolder GFP (sfGFP) previously generated (71). ImageJ RGB profiling revealed that the LSPMB lies between the parasite plasma membrane (PM) and the parasitophorous vacuole membrane (PVM), suggesting that the sporozoite subpellicular microtubules undergo subcellular relocalization during liver stage development (Figure 3D and E). At later stages, particularly the cytomere stage, we observed reappearance of tyrosinated tubulin near the parasite nuclei (Figure 3B). During merozoite formation, this tyrosinated tubulin appeared as punctate structures within the forming merozoites (Figure 3B).
Tyrosination has previously been associated with stable microtubules involved in motility, such as those in sporozoites, flagella, and spermatozoa (33,34,50,55). The absence of tyrosination during early intracellular liver stage development when the parasite is non-motile may facilitate its transformation from a banana-shaped sporozoite to a rounded liver stage form (Figure 1 and 3). LSPMB detyrosination may contribute to microtubule dynamic instability, providing both energy and tubulin monomers for rapid growth and spindle formation during schizogony. Finally, the reappearance of tyrosinated microtubules during merozoite formation may serve to stabilize the microtubules required for merozoite motility and erythrocyte invasion.
The Plasmodium α-tubulin C-terminal modification are important for sporozoite formation
To investigate the functional role of parasite α-tubulin PTMs during the liver stage development, we generated P. berghei mutant parasite lines with targeted mutations in the α-tubulin C-terminal region, where most PTMs are predicted to occur (Figure 4A and Supplementary Figure 3A and B). Specifically, we created three mutant lines: PsΔ: a point mutation replacing the polyglutamylation site glutamic acid (E445) with alanine (A), TyΔ: deletion of the tyrosination/detyrosination motif (YEADY) from position 449, CtΔ: deletion of the entire C-terminal tail (EGEDEGYEADY) to prevent all possible C-terminal tail PTMs (Supplementary Figure 3A to C).

Plasmodium α1-tubulin C-terminal post-translational modification is important for the mosquito stage development.
(A) Schematic of the double homologous recombination (DHR) construct targeting the Plasmodium α1-tubulin gene (PBANKA_0417700) for C-terminal mutagenesis. The cassette (blue box) includes the 3’UTR P. berghei dihydrofolate reductase (PbDHFR), elongation factor-1 alpha (ef1α) promoter, human dihydrofolate reductase (hdhfr), yeast FCU (yfcu), 3’UTR PbDHFR, Hsp70 promoter, mCherry and the 3’ UTR Hsp70, with the 3’UTR region of the PBANKA_0417700 gene. This cassette was flanked by a 5′ homologous region (5′HR) containing the α1-tubulin coding region including the stop codon (with introduced mutations in the C-terminal region, cyan box) and a part of the 3′UTR of the gene (3’HR). The construct was transfected into the PbWT (P. berghei ANKA line cl15cy1). Black arrows indicate PCR primer binding sites and expected product sizes. Primer sequences are listed in Table 2 and 3. (B) Integration PCR results for the mutant parasite lines, showing expected band sizes on a 1% agarose gel: 5’HR = 4.4 kb, 3’HR = 2.7 kb, whole locus (WL) = 8.8 kb. Ladder: 1 kb DNA marker. Genotypes: WT, (wild-type); PsΔ (glutamine-to-alanine substitution); TyΔ (deletion the C-terminal tyrosination/detyrosination residues); CtΔ (complete C-terminal deletion), and NoΔ (construct control with no mutation). To ensure that all samples contained the same amount of DNA, a control gene representing a portion of tubulin (Ctrl tub about 470 bp) was used for integration PCR. (C) Representative confocal images of live oocysts at 11 days post infection (dpi). Nuclei were stained with Hoechst 33342 (hot cyan) and cytosolic mCherry (red) was expressed under the HSp70 promoter. BF, brightfield. (D) Normalized salivary sporozoite counts at 18 dpi. The control (NoΔ) averaged 14,033 sporozoites count per mosquito (C./mosq.), compared to 7,300 (PsΔ), 8,066 (TyΔ), and 200 (CtΔ). Experiment performed in triplicate; n = 20 per replicate. (E and F) Wide-field microscope motility assay images of salivary gland sporozoites showing the typical circular movement (maximum z projection in Fiji) of the control, PsΔ, and TyΔ salivary gland (SG) sporozoites (E) and the control and CtΔ hemolymph (H) sporozoites (F). For CtΔ, hemolymph sporozoites were used due to the absence of salivary gland sporozoites; these lacked typical motility. (G)) Quantification of sporozoite gliding velocity for PsΔ, and TyΔ salivary gland sporozoites. Mean velocities: NoΔ = 1.3 µm/s, PsΔ = 1.38 µm/s, TyΔ = 1.9 µm/s, CtΔ = 0.5 µm/s. Statistical analysis: one-way ANOVA with Dunnett’s multiple comparisons test (*p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001). (H) Quantification of sporozoite gliding velocity of CtΔ haemolymph sporozoites. The average gliding speed for control hemolymph sporozoites was 1.4 µm/s, whereas CtΔ sporozoites exhibited a slower speed of 0.5 µm/s. Statistical comparison tests were performed using two tailed unpaired t tests (*p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001).
Using AlphaFold predictions, we confirmed that the mutations were unlikely to affect tubulin folding (Supplementary Figure 3C). The mutants were generated via double homologous recombination and verified by integration PCR and sequencing of the mutated α-tubulin gene (Figure 4A and B; Supplementary Figure 3B). The development of mutant parasites was investigated during the mosquito, and liver stages. Significant defects emerged during mosquito stage development. The PsΔ and TyΔ mutants were able to form oocysts (Figure 4 C, Supplementary Fig 4A) and gliding sporozoites, albeit in reduced numbers (Figure 4D, E and G; Supplementary Fig 4B; and Supplementary Movie 2, 3, 4 and 5). Quantification of salivary gland sporozoites at 18 days post-blood feeding showed that PsΔ and TyΔ mutants produced approximately half as many sporozoites as the control line (NoΔ) (Figure 4D). We next examined sporozoite motility, which typically consists of continuous circular gliding (16,72–76). PsΔ and TyΔ salivary gland sporozoites were assessed and compared to controls. While PsΔ have a similar gliding velocity compared to controls, TyΔ sporozoites displayed increased gliding velocity (2.5 µm/s vs. 1.4 µm/s) (Figure 4G), suggesting a regulatory role of the tyrosination motif in fine-tuning motility. To assess whether these motility phenotypes influenced infectivity, we performed in vivo infection assays with PsΔ and TyΔ. All mutant lines sporozoites were able to infect mice similarly to control parasites (Supplementary Figure 4C). However, the CtΔ mutant failed to produce salivary gland sporozoites entirely (Figure 4D). Since CtΔ mutant parasites failed to reach the salivary glands, we analyzed hemolymph sporozoites for this line compared to the controls (Figure 4F). These showed severely impaired motility, lacking the typical gliding pattern and exhibiting significantly reduced speed (Figure 4F and H). Given that PsΔ and TyΔ mutants produced motile salivary gland sporozoites, we infected HeLa cells with PsΔ and TyΔ mutant sporozoites and CtΔ mutant hemolymph sporozoites to assess their liver stage development and identify any downstream phenotypic effects.
The Plasmodium α-tubulin C-terminal mutants undergo liver stage development
Using polyglutamylation-specific antibodies, we found that PsΔ mutant parasites still exhibited polyglutamylated liver stage parasite microtubule bundles (LSPMBs) in both sporozoites and liver stage forms, although the signal intensity was significantly reduced compared to controls (Figure 5A, B and C and G). As tyrosination was previously observed only in sporozoites and late liver stages, we performed PS-ExM on sporozoites and stained them with α-tubulin tyrosination-specific antibodies. TyΔ salivary gland sporozoites and CtΔ hemolymph sporozoites both showed detectable tyrosination signals, although the signal in CtΔ parasites was markedly weaker than in controls (Figure 5D, E and F). Interestingly, as PTM-specific antibodies continued to recognize signals in the PsΔ and TyΔ mutants, it is possible that the parasite compensates by relocating the PTM sites to adjacent residues within the C-terminal sequence. Alternatively, the antibody signal may stem from β-tubulin modifications rather than α-tubulin.

Liver stage development of Plasmodium α1-tubulin mutant parasites.
(A, B and C) Maximum projections of PS-ExM-acquired z-stack confocal images of infected HeLa cells at 6 and 36 hpi, showing the control (NoΔ), PsΔ, and CtΔ mutant parasites. (D and E) Maximum projections of PS-ExM-acquired z-stack confocal images of control and TyΔ salivary gland sporozoites, as well as CtΔ hemolymph sporozoites (E). In (A, B and C), cells were stained with antibodies against polyglutamylated tubulin (anti-IN105), while (D and E) antibodies against tyrosinated tubulin (anti-Ty) were used. In all panels, α-tubulin was labeled with anti-α-tubulin, nuclei with DAPI (blue), and the parasitophorous vacuole membrane (PVM) with anti-UIS4 (grey). (F and G) represent the fluorescence intensity (arbitrary units, a.u.) of anti-Ty in sporozoites (F) and anti-IN105 at 36 hpi in the liver stage (G). In (F), the mean fluorescence intensity of CtΔ (22 a.u.) was significantly reduced compared to TyΔ (33.4 a.u.) and NoΔ (49.6 a.u.), while in (G), PsΔ (79.9 a.u.) showed a significant reduction compared to NoΔ (121 a.u.). (H and L) showed infection rates of mutant parasites in HeLa cells at 6 hpi, determined by pre-counted sporozoites and quantitative microscopy. PsΔ (23.7%) and TyΔ (27.3%) exhibited typical infection rates compared to control (NoΔ) (16%) and WT (14%) (H), whereas CtΔ hemolymph sporozoites had a drastically reduced infection rate (2%) relative to controls hemolymph sporozoites (7.4%) (L). (K and O) Parasite survival from 6 to 48 hpi, normalized to 100% at 6 hpi. At 48 hpi, survival rates were 63% (NoΔ), 60% (WT), 50% (PsΔ), and 47% (TyΔ) (K). In contrast, CtΔ hemolymph sporozoites showed a significantly lower survival rate (2%) compared to controls (52%) (O), indicating impaired viability. (I and J, M and N) Parasite size measurements at 6 and 48 hpi for control, PsΔ, and TyΔ salivary gland sporozoites (I and J), as well as control and CtΔ hemolymph mutants (M and N). At 48 hpi, PsΔ (208.16 µm²) and TyΔ (203.7 µm²) were significantly smaller than NoΔ (237.9 µm²) and WT (235.7 µm²) (J). The CtΔ hemolymph mutants failed to establish liver stage infection, with an average size (88.1 µm²) markedly smaller than controls (187.4 µm²) (N). Each dot represents an individual parasite. PsΔ: Glutamine-to-alanine substitution at the polyglutamylation site; TyΔ: Deletion of C-terminal tyrosination/detyrosination residues; CtΔ: Complete deletion of PBANKA_0417700 α-tubulin C-terminal PTM residues and NoΔ: Control construct with no mutation. WT: wild type parasite. The experiments were conducted using, (F, H, I, J, K): One-way ANOVA with Dunnett’s multiple comparisons test (*p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001). (G, L, M, N, O): Two-tailed unpaired t-tests (*p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001). All experiments were performed in triplicate.
To assess parasite survival and growth during liver stage development, we performed quantitative microscopy (Figure 5H to O). Both PsΔ and TyΔ mutant parasites successfully infected HeLa cells and progressed through liver stage development (Figure 5B, D, H, I, J and K). This was further confirmed by staining the parasitophorous vacuole membrane (PVM) using anti-UIS4 antibodies (Figure 5A, B and C). Parasite size was measured at 6 hpi and 48 hpi to assess developmental progression. No significant differences were observed at 6 hpi (Figure 5I). However, by 48 hpi, mutant parasites were significantly smaller than controls, indicating impaired growth (Figure 5J). Survival rates of PsΔ and TyΔ mutants were comparable to controls (Figure 5K), with infection rates of 15% for PsΔ (matching the control) and 27% for TyΔ, exceeding the control. As expected, the CtΔ hemolymph sporozoites failed to infect and to develop liver stage forms (Figure 5C, L, M, N and O). The sporozoites failed to establish productive infections with a typical PVM at all examined time points (Figure 5C, M, N and O).
To confirm the phenotypes, we compared our mutants with previously generated P. berghei α1-tubulin mutants described by Spreng et al. (2019), including parasites with intron deletions (intronΔ), C-terminal tyrosination deletions 540ADY (C-termΔ), and α2-tubulin exon swaps. These mutants showed similar phenotypes, including a complete absence of salivary gland sporozoites in α2-tubulin exon swap parasites, mirroring the CtΔ mutant phenotype (Figure 4D and H; Supplementary Figure 3D and E). These results underscore the importance of α-tubulin C-terminal PTMs in sporozoite formation and migration from the midgut to the salivary glands.
The failure of CtΔ parasites to generate viable sporozoites highlights the essential role of α-tubulin PTMs in sporozoite development and migration. Co-mutation of both α- and β-tubulin PTM sites may prove lethal to parasite development in the mosquito stage. Future studies should focus on identifying and characterizing the enzymes responsible for tubulin PTMs in Plasmodium, and genetic knockout strategies should be pursued to further elucidate the function of these modifications.
Discussion
Our study provides the first in-depth characterization of microtubule architecture and dynamics during the Plasmodium liver stage. Using tubulin trackers and α-tubulin antibodies, we identified two major microtubule populations: a large, multimeric subpellicular network that we termed the liver stage parasite microtubule bundles (LSPMB), and the polymerizing hemi-spindles involved in nuclear division. The LSPMB appears to originate from the SSPM and persists until late schizogony. These observations align with earlier electron microscopy studies by Meis et al. (77) who reported the presence of subpellicular microtubules and inner membranes in liver stage parasites up to the point of mitotic spindle formation.
The behavior and function of microtubules are closely regulated by PTMs. We found that α1-tubulin expressed during the liver stage carries residues associated with polyglutamylation and tyrosination. Using PTM-specific antibodies, we confirmed that Plasmodium sporozoites display both modifications on their subpellicular microtubules. Tyrosination, a well-known marker of stable microtubules, has been described in highly motile systems such as cilia, flagella, and spermatozoa (33,34,50,55). This is consistent with the need for high motility in sporozoites, which must traverse multiple tissue barriers during transmission (16,72,73).
Polyglutamylation, in contrast, introduces negative charges on the tubulin surface that enhance interactions with MAPs, and is commonly associated with dynamic microtubule behavior (78–80). This modification has previously been linked to merozoite formation and intracellular trafficking in Plasmodium blood stages (31). The co-occurrence of tyrosination and polyglutamylation in sporozoites likely ensures a microtubule network that is both stable and functionally interactive during mosquito stage migration.
Upon hepatocyte invasion, we observed a loss of tyrosination while polyglutamylation was maintained, suggesting a functional switch from stable to dynamic microtubules (Figure 6). This transition may facilitate morphological transformation from a motile, slender sporozoite into a rounded liver stage form and could also support the parasite’s rapid replication. The liver stage is one of the fastest-replicating phases in the Plasmodium life cycle, with replication rates among the highest observed in eukaryotic cells (81,82). During this phase, the parasite synthesizes proteins de novo and hijacks host cell lipids and proteins for membrane biogenesis (83–86). We propose that the parasite brings its SSPM into hepatocytes to be repurposed as the LSPMB (Figure 6). The observed microtubule disassembly over time may support tubulin turnover and safes energy for intracellular development.

Model of the tubulin behavior during Plasmodium berghei salivary gland sporozoite and exo-erythrocytic stage.
The graphical abstract summarizes the key finding of this study. It illustrates the development of Plasmodium parasites within primary hepatocytes, from invasion to the cytomere stage. α-tubulin was stained with anti-α-tubulin (magenta), nuclei with DAPI (blue), and the parasitophorous vacuole membrane (PVM) with anti-UIS4 (grey). The model highlights the evolution of sporozoite subpellicular microtubules (SSPM) into liver stage subpellicular microtubules (LSPMB), along with their associated tubulin post-translational modifications (PTMs). We propose that polyglutamylated LSPMB originates from the SSPM and is recycled during liver stage development to support hemi-spindle pole formation, membrane biogenesis, and potentially other functions. Upon hepatocyte invasion, the SSPM loses tyrosination, a modification that initially confers stability and supports sporozoite motility. This detyrosination likely facilitates parasite shape transformation, increases microtubule dynamics, and enables interactions with ER structures and MAPs. Tyrosination reappears in late stages, possibly stabilizing merozoite microtubules for erythrocyte invasion. Microtubule-stabilizing drugs did not affect the SSPM but inhibited liver stage microtubule polymerization, confirming their dynamic nature. Inhibition of microtubule dynamics also prevented merozoite formation. Finally, C-terminal tubulin PTM residues were shown to be essential for salivary gland sporozoite development and successful liver stage infection.
In addition to their structural roles, LSPMB filaments were occasionally found in contact with hemi-spindle poles, suggesting that LSPMB contribute to spindle formation during schizogony. This connection, although infrequent in fixed cells, was frequently observed during live imaging suggesting a transient connection. We also detected expression of EB1, a microtubule plus-end binding protein previously believed to be restricted to the sexual blood stage (64,65,87), during mosquito and liver stages. Its localization to LSPMB and hemi-spindles suggests a broader role for EB1 in regulating microtubule dynamics throughout the parasite life cycle.
To directly test the function of tubulin PTMs, we generated P. berghei mutants with targeted C-terminal modifications of α1-tubulin. These included a glutamate-to-alanine substitution at a polyglutamylation site, deletion of the tyrosination motif, and complete deletion of the C-terminal tail. Despite the mutations, PTM-specific antibody staining remained detectable in the first two mutants, suggesting compensatory mechanisms such as modification of adjacent residues or of β-tubulin. The tyrosination antibody used (anti-Ty, T9028, Sigma-Aldrich) is a purified monoclonal antibody targeting C-terminal tyrosine residues on tubulins, whereas the monoclonal GT335 antibody (AG-20B-0020-C100, AdipoGen Life Sciences) recognizes the epitope formed at the branching point of polyglutamylation side chains (GEGE*EE, * marking the modification site) and detects both short and long glutamate side chains. The polyclonal antibody (AG-25B-0030-C050, AdipoGen Life Sciences) was raised against at least three glutamate residues and is thereby specific to long polyglutamylation side chains, though both antibodies may cross-react with β-tubulin or other proteins containing genetically encoded C-terminal modifications (66,88). The reduced fluorescence signal observed (Figure 5) in mutant parasite microtubules suggests either antibody cross-reactivity with β-tubulin or compensatory PTM relocation to adjacent α-tubulin residues. In the CtΔ mutant (lacking the entire C-terminal tail), PTM relocation is abolished, and residual signal likely stems from β-tubulin which was insufficient to sustain sporozoite motility and transmission. These possibilities warrant further investigation. Previous studies have also demonstrated partial redundancy between α1- and α2-tubulin modified (49,50,89). However, the complete C-terminal deletion mutant failed to produce salivary gland sporozoites and could not establish liver stage infection. These results are consistent with findings from Spreng et al. (2019), who showed that α1-tubulin mutants lacking the C-terminal region produced defective sporozoites with altered microtubule numbers and impaired motility.
We also observed that the LSPMB relocates during liver stage development. Using expansion microscopy on PMP1::GFP-expressing parasites (71), we found that the LSPMB localizes between the parasite plasma membrane and the PVM. This differs from earlier reports of SSPM positioning (77,90–92) but both notions are not mutually exclusive. It is possible that the LSPMB first localizes within the parasite but then re-localizes at later stages or even re-localizes dynamically. Its localization between the PM and the PVM positions it well for interaction with the host cell indicating an interaction with host cell structures such as ER tubules or host microtubules (71,84,93,94). However, whether this re-localization plays a role in nutrient acquisition or host-pathogen signaling remains to be explored.
Finally, during late schizogony and merozoite formation, we observed the reappearance of tyrosinated tubulin. This may reflect the need for stable microtubules in newly formed merozoites, which must be competent for host cell invasion.
In conclusion, our study reveals a previously unrecognized complexity in the microtubule biology of Plasmodium liver stages. The identification of the LSPMB as a dynamic structure regulated by tubulin PTMs and repurposed from the sporozoite cytoskeleton opens new avenues for understanding how the parasite controls its intracellular architecture. These PTMs appear critical for parasite shape, replication, and stage transition, and may represent valuable targets for therapeutic intervention. Future work should aim to identify the enzymes responsible for these modifications and explore their potential as drug targets. The behavior and function of microtubules are closely tied to their PTMs. We found that α1-tubulin expressed during the liver stage carries residues predicted to undergo polyglutamylation and tyrosination.
Using PTM-specific antibodies, we confirmed that sporozoite microtubules are both tyrosinated and polyglutamylated. Tyrosination is typically associated with stable microtubules and has been described in highly motile structures such as cilia and spermatozoa. This is consistent with the sporozoite’s need for high motility gliding at approximately 1.5 µm/s as it migrates from the mosquito midgut to the salivary glands and eventually traverses skin and vasculature to reach hepatocytes.
Methods
Ethics statement
All experimental procedures were performed in full compliance with the Swiss Animal Protection Act (TSchG) and were approved by the Canton of Bern’s Animal Experimentation Commission (Authorization, BE118/22). Personnel involved in animal studies held the Federation of European Laboratory Animal Science Associations (FELASA) accredited certifications. Female C57BL/6 and BALB/c mice of 6 to 10 weeks old and 20-30 g at infection were sourced from Janvier Labs (France) or bred in the animal facility at the University of Bern’s Institute of Cell Biology. Animals were maintained under standard housing conditions with daily health monitoring by trained staff.
P. berghei lines used in this study and the generation of transgenic parasites
The P. berghei ANKA parasite line was utilized for both murine and cellular infection models. To generate the α1-tubulin C-terminal mutant and eb1::eGFP parasite lines, the C-terminal coding and 3’ untranslated regions (UTRs) were cloned into the double homologous recombination vector pDHR, which contains the 3’UTR PbDHFR, the ef1α promoter region, the selectable markers hdhfr::yfcu, and additional 3’UTR PbDHFR sequences. The pDHR vector backbone originated from the plasmid pG362 (kindly provided by the Waters lab) (95).
First, the pG362 plasmid was digested with SacII and PstI to remove the AID, selectable marker, and GFP sequences. The amplified product (linker-eGFP-3’UTR PbDHFR) from the pOB277 vector was then cloned using T4 DNA ligase (NEB, M0202L). For subsequent cloning, restriction sites were added to the forward (KpnI and ApaI) and reverse (EcoRI and SmaI) primers. The plasmid was then digested with EcoRI and SmaI to clone the amplified product: ef1α promoter region-hdhfr::yfcu marker (a fusion gene of the positive selection marker human dihydrofolate reductase (hdhfr) and the negative selection marker yeast cytosine deaminase-uridyl phosphoribosyl transferase (yfcu) 3’UTR PbDHFR from the genomic DNA of the P. berghei GIMO parasites (RMgm-687) parental line (96).
The complete α1-tubulin gene sequence (PBANKA_0417700), including 5’ and 3’ UTRs, was retrieved from PlasmoDB (https://plasmodb.org). The linker-eGFP region of the pDHR vector was removed using ApaI and NotI restriction enzymes. The 5’UTR of the α1-tubulin gene was then subcloned into the digested pDHR using Gibson assembly. The resulting vector was digested with SmaI, and the PbHsp70 promoter and mCherry regions from RMgm-687, along with the 3’UTR of the α1-tubulin gene, were subcloned into the vector at the ApaI and SmaI restriction sites, respectively, using Gibson assembly.
Site-directed mutagenesis (QuickChange II XL Kit) was used to introduce the C-terminal modifications, with primer sequences listed in Tables 2 and 3. The final plasmid was linearized with ScaI before being transfected into the PbWT line (P. berghei ANKA line cl15cy1) (97). Similarly, the end-binding protein eB1 (PBANKA_0405600) with an eGFP tag was engineered using identical cloning strategies. The 5’ and 3’ UTRs were retrieved from PlasmoDB (https://plasmodb.org) and subcloned into the pDHR vector at the ApaI and SmaI restriction sites, respectively, using Gibson assembly. The final vector was linearized with NdeI before being transfected into the Pb1868 line (constitutively expressing Hsp70-mCherry-ef1α-NLuc) (98). Primer sequences are listed in Table 1.

Plasmodium berghei eB1egfp parasites integration PCR primer

Plasmodium berghei α1-tubulin mutant parasites integration PCR primer

Plasmodium berghei α1-tubulin mutagenesis QuickChangeII primers sequences
Vector transfections were performed in schizont cultures as previously described (16,99). Briefly, P. berghei schizonts were cultured and purified (16,99,100). Electroporation of schizonts from the respective parasite lines was performed using the Amaxa® Human T Cell Nucleofector® system with the U33 program, followed by intravenous injection into naïve mice. The construct was integrated by double-crossover homologous recombination at the locus of interest. Pyrimethamine drug selection was administered via drinking water, and parasitemia was monitored daily from day 7 post-infection until mice tested positive (99).
Parasitaemia measurement and cloning of transgenic parasite lines
To measure the parasitemia ratio of infected mice, approximately 2 μL of blood from the tail vein of each infected mouse was added to 500 μL of PBS and analyzed by flow cytometry using the NovoCyte (Agilent, 2023) as described (101). The data were analyzed using NovoExpress Flow Cytometry Analysis Software (version 1.6.2, Build 230717.50551; Agilent Technologies).
To clone the different engineered transgenic parasite lines, mice were infected with the transgenic parasites. Three days post-infection, parasitemia was assessed and when parasitemia reached at least 1%, blood was collected from the infected mouse via heart puncture following euthanasia as described (16). The collected blood was passaged into a new naïve mouse. When parasitemia in the newly infected mouse reached 1-2%, 2 μL of tail vein blood was collected and mixed with RPMI-1640. Maintaining low parasitemia was crucial to prevent both double erythrocyte infection by P. berghei and gametocyte formation. Single P. berghei-infected erythrocytes were then sorted into individual wells of 96-well plates (Cellvis) containing 200 μL of RPMI-1640 supplemented with 10% FCS using a BD FACSDiscover S8 with CellView. Supplementing the medium with 10% FCS allowed the sorted infected erythrocytes to recover from the sorting process. Five mice were then intravenously infected with a single sorted infected erythrocyte each. The mice underwent drug selection via drinking water, and parasitemia was monitored daily from day 7 post-infection until parasites were detected. Clonality of the parasites in positive mice was verified by integration PCR using primers listed in Tables 1, 2, and 3.
Cell culture
Human epithelial HeLa cells (European Cell Culture Collection) were maintained in Minimum Essential Medium (MEM; 1-31F01-I BioConcept) supplemented with Earle’s salts and 10% heat-inactivated fetal bovine serum (FBS; Sigma-Aldrich), hereafter referred to as complete MEM (cMEM). Primary hepatocytes were cultured in William’s Medium E (WME) complete medium. All cells were incubated at 37°C under 5% CO₂. For infection assays, 40,000 HeLa cells were seeded onto coverslips in 24-well plates and allowed to adhere overnight. Cells were then infected with P. berghei sporozoites and maintained in cMEM containing 2.5 µg/mL amphotericin B (PAA Laboratories, P11-001) to prevent fungal contamination. Medium was exchanged every 24 hours.
Primary hepatocyte isolation
The mice primary hepatocytes were isolated according to the method described (8). Briefly, the cells were isolated from 12 to16 week old Balb/c or C57BL/6 mice using a two-step perfusion technique (8,83,102). Mice were euthanized via CO₂ inhalation (1–2 minutes), and after securing the animal in a supine position, a midline abdominal incision was made to expose the portal vein or inferior vena cava (IVC). A peristaltic pump was pre-rinsed with 70% ethanol and HEPES buffer, then set to 50 for initial cannulation of the portal vein (or IVC) using a Teflon-coated needle. Upon successful insertion, the flow was increased to maximum (999), and the opposing vein was severed to allow perfusion with HEPES buffer for 5 minutes, during which periodic clamping (10 seconds per minute) of the cut vein was performed. The tubing was subsequently changed to a collagenase solution, and perfusion was held for 7 minutes with repeated clamping. The liver was excised, transferred to a collagenase-containing well, and mechanically dissociated using filter tips and gentle shaking with forceps. Cell suspensions were sequentially collected, pooled, and washed with William’s Medium E (WME) by centrifugation at 50g for 2 minutes. The pellet was resuspended in WME, optionally passed through a 70 µm nylon strainer, and washed twice more. The final pellet was resuspended in WME complete medium (WME supplemented with L-glutamine, penicillin-streptomycin, amphotericin B, insulin-transferrin, and aprotinin), and cells were counted and seeded at densities appropriate for T25 flasks, GB dishes, or 24-well plates. After initial adhesion (1 hour), culture medium was supplemented or exchanged after 2–4 hours.
Immunofluorescence assay (IFA) of fixed primary hepatocytes and HeLa cells
Immunofluorescence was carried out as previously described (56,86,103). Cells were seeded onto coverslips in culture 24 well-plates and allowed to adhere overnight before infection. Infection was maintained for 24 hours post-infection (hpi), after which cells were fixed using cold methanol at -20°C for 15 minutes and subsequently washed twice with Phosphate buffered saline (PBS). Blocking was performed by incubating cells in 10% FCS in PBS for 20 minutes at room temperature to reduce non-specific binding. Primary antibodies staining was carried out by incubating cells in 10% of FCS diluted in PBS for 1 hour at room temperature, followed by three washes with PBS. Secondary antibodies incubation was performed in 10% of FCS as well for 1 hour at room temperature in the dark. The antibodies used are listed in the enumerated session bellow. After three additional washes with PBS, nuclei were counterstained with DAPI. Coverslips were then mounted onto slides with DAKO mounting medium and fluorescence imaging was performed using either Nikon W1 or LIPSI and Crest V3 confocal microscope equipped with filters for DAPI (4,6-Diamidin-2-phenylindol), AF488, AF594, and Cy5. Single-stain controls were carried out for every staining to ensure that bleed-through was negligible.
Sporozoite isolation from mosquito salivary glands and haemolymph
Anopheles mosquitoes were fed on Plasmodium-infected mice exhibiting a gametocytaemia of 0.2 to 1% and the sporozoite isolations were performed from day 18 post-infection as previously described (16,61). For salivary gland sporozoites, mosquitoes were anesthetized with chloroform, sterilized in 70% ethanol, and rinsed twice in PBS. The mosquitoes were then dissected, and salivary glands were extracted under a binocular microscope in Iscove’s Modified Dulbecco’s Medium (IMDM) pooled in 50 µL of ice-cold IMDM and mechanically homogenized using a pestle (Sigma-Aldrich, Z359947) powered with cordless motor (Sigma-Aldrich, Z359971) as described (16,61). For hemolymph sporozoite extraction, glass capillaries were heat-pulled to fine tips. The abdominal terminal segment of the mosquito was excised, and hemolymph was flushed from the thorax by PBS perfusion via capillary insertion (75,104) . Hemolymphs extracted were centrifuged (11,000 × g, 1 min), and pellets were resuspended in PBS or IMDM. Sporozoite quantification was performed using a Neubauer hemacytometer. Midgut, hemolymph, or salivary gland suspensions were mixed 1:1 with RPMI-1640 supplemented 10% FCS, and sporozoites within four corner grids were counted. Total yields (sporozoites/mL and per mosquito) were calculated based on dilution factors and dissection numbers.
Sporozoites IFA and gliding motility assay
To perform the sporozoite immunofluorescence assay (IFA), 12 mm cover glasses (0111530, Paul Marienfeld, Germany) were coated with 0.1 mg/mL poly-D-lysine (A38904-01, Gibco, USA) (56,105) and incubated overnight at room temperature. The following day, sporozoites were isolated from mosquito midguts, hemolymph, or salivary glands as described above. The extracted sporozoites were suspended in RPMI-1640 medium to a final volume of 200 µL in an Eppendorf tube and centrifuged at 1000 rpm for 3 min. The pellet was fixed by resuspension in 200 µL of cold methanol and incubated for 15 min at −20°C. After fixation, sporozoites were washed twice with PBS (1000 rpm centrifugation) and transferred onto poly-D-lysine-coated cover glasses in a 24-well plate (400024, Bioswisstec, Switzerland). The plate was incubated for 30 min at room temperature to facilitate sporozoite adhesion before proceeding with either the IFA or expansion microscopy protocol.
For the gliding motility assay, 5 µL of the sporozoite suspension in RPMI-1640 (supplemented with 10% FCS) was added to 96-well glass-bottom plates (P96-1.5H-N, Cellvis, USA) (56,72,73). The plate was centrifuged (1000 rpm, 1 min) and immediately live imaged using a Leica DM5500 widefield fluorescence microscope.
Expansion microscopy
Expansion microscopy (PS-ExM) was performed as previously established (56–58). Infected cells (HeLa cells, primary hepatocytes and sporozoites) were seeded on 13 mm coverslips in 24-well plates and fixed with either 4% paraformaldehyde (PFA; F8775, Sigma) or cold methanol. The cells were anchored by incubation in PBS containing 0.7% formaldehyde and 1% acrylamide at 37°C for 2 hours. Gelation was then performed by adding 5 µL each of 10% tetramethylethylenediamine (TEMED; 17919, Thermo Fisher) and 10% ammonium persulfate (APS) to 90 µL of monomer solution containing 19% sodium acrylate (408220, Sigma), 10% acrylamide (A4058, Sigma), and 0.1% N, N’-methylenebisacrylamide (M1533, Sigma). A 35 µL of monomer solution was dispensed onto parafilm in a humidity chamber, and coverslips containing the anchored cells were inverted onto droplets. Following 5 minutes incubation on ice to improve gel penetration, gels were polymerized at 37°C for 30 minutes. Gel-embedded samples were transferred to denaturation buffer (200 mM SDS, 200 mM NaCl, 50 mM Tris-HCl, pH 9) in 6-well plates and agitated (200 rpm) at room temperature for 15 minutes, followed by incubation in fresh buffer at 95°C for 1 hour with gentle shaking. Gel expansion was performed in autoclaved MilliQ water (50 mL) for 1 hour with optional buffer exchange. For immunostaining, gels were incubated with primary antibodies in 2% BSA for 2 hours at 37°C, washed with PBS-Tween 0.1%, then labelled with secondary antibodies plus 1 µg/mL DAPI for nuclear staining in 2% BSA for 2 hours at 37°C. Final gel expansion was conducted in MilliQ water containing 0.2% propyl gallate (02370, Sigma) to prevent photobleaching. Expanded samples were mounted on poly-D-lysine-coated (A38904, Gibco USA) glass-bottom dishes with cellular surfaces facing downward and imaged with confocal microscopy.
Antibodies used for IFA and expansion microscopy
The primary antibodies used were: anti-UIS4 chicken (1:1000 IFA/1:500 expansion; Proteogenix), anti-UIS4 rabbit (1:1000 IFA/1:500 expansion; P. Sinnis, Baltimore), polyglutamylation IN105 rabbit (1:1000 IFA/1:500 expansion; AG-25B-0030-C050, AdipoGen Life Sciences), anti-GFP rabbit (1:1000 IFA/1:500 expansion; SP3005P, Origene), anti-α-tubulin guinea pig (1:500 IFA/1:125 expansion; AA345, ABCD), anti-β-tubulin guinea pig (1:500 IFA/1:125 expansion; AA344, ABCD), anti-tyrosinated tubulin mouse (1:1000 IFA/1:500 expansion; T9028, Sigma Aldrich), anti-polyglutamylation GT335 mouse (AG-20B-0020-C100, AdipoGen Life Sciences), anti-γ-tubulin mouse (1:1000 IFA/1:500 expansion; T6557, Sigma Aldrich), and anti-GFP mouse (1:1000 IFA/1:500 expansion; 814460001, Roche).
Secondary antibodies included anti-chicken IgY Alexa Fluor 594 (1:1000 IFA/1:500 expansion; A11042, Invitrogen Molecular Probes), anti-rabbit IgG Alexa Fluor 594 (1:1000 IFA/1:500 expansion; A21207, Life Technologies) and 488 (1:1000 IFA/1:500 expansion; A11034, Sigma Aldrich), anti-rabbit IgG ATTO 647N (1:1000 expansion only; 40839, Sigma Aldrich), anti-guinea pig IgG Alexa Fluor 647 (1:1000 IFA/1:500 expansion; SAB4600033, Sigma Aldrich) and 594 (1:1000 IFA/1:500 expansion; A11076, Invitrogen Molecular Probes), and anti-mouse IgG ATTO 647N (1:1000 expansion only; 50185, Sigma Aldrich) and Alexa Fluor 488 (1:1000 IFA/1:500 expansion; A11001, Invitrogen Molecular Probes).
In vivo assay
To assess the infectivity of parasite mutant sporozoites to hepatocytes, Anopheles stephensi mosquitoes infected with different parasite mutants were allowed to feed on anesthetized naïve mice as previously described (Atchou et al., 2025). Three mice per parasite line and five mosquitoes per mouse were used. The blood stage infection progression was monitored daily via flow cytometry, starting from the day of infection until mice tested positive.
Oocyst size measurement
To quantify oocyst size, Anopheles stephensi mosquitoes were allowed to feed on anesthetized mice infected with the parasite (Rogerio et al. 2006; Hegge et al., 2009). At 7- and 11-days post-feeding, ten mosquitoes per time point were dissected, and their midguts were extracted and placed in 1× PBS. The midguts were then transferred to a 21-well microscopy slide, covered with a coverslip, and imaged using a Nikon Crest V3 confocal microscope (10× objective). The oocyst area was measured using Fiji software (ImageJ). Each experiment was performed in triplicate for every parasite line.
Liver stage parasite size measurement
To quantify the parasite size during liver stage development, HeLa cells (40,000 cells/well, triplicate) were seeded in 96-well plates and infected 24 hours later with 20,000 sporozoites per well. After a 2-hour incubation at 37°C, infected cells from each well were washed and redistributed into eight wells across two 96 well plates (4 wells/plate) to optimize cell density for imaging (Pino et al., 2017; Atchou et al., 2025; Sturm et al., 2006). Parasite development was analyzed at 6, 24, and 48 hpi using an InCell Analyzer 2000 with 10x objective. The image processing parameters such as kernel size, sensitivity, and size thresholds were optimized in InCell Developer software to distinguish individual parasites from background signals. Parasite growth was assessed based on size measurements, while survival rates were calculated as percentages relative to 6 hpi baseline counts.
Live cell imaging of Plasmodium microtubule dynamism
HeLa cells were seeded in a 96-well plate and infected the following day with P. berghei salivary gland sporozoites. After a 2-hour incubation at 37°C, cells were washed with MEM medium to remove uninfected parasites and residual mosquito salivary gland materials. The infected cells were then reseeded in a dark 24-well glass-bottom plate (Cellvis) and maintained in fresh cMEM at 37°C with 5% CO₂ (16,106). Prior to live imaging, cells were incubated for 1 hour in cMEM containing the SPY650-tubulin probe (1:1000 dilution; SC503, Spirochrome, Switzerland). Images were acquired using a Nikon W1 LIPSI spinning disk microscope with a 100X objective. Marked infected HeLa cells were imaged at defined time points to generate time-lapse movies capturing tubulin dynamics and parasite development.
Image processing and data analysis
All the live cell imaging, expansion microscopy, and IFA images in this study were acquired with Nikon W1 LIPSI and Crest V3, both in spinning disk mode. The microscope images were processed and analyzed using FIJI software, Huygens deconvolution Professional version (Huygens Remote Manager v3.8, Scientific Volume Imaging, The Netherlands), or, when specified, the 3D and 4D IMARIS software (Imaris 10.2.0; June 18, 2024). The 3D/4D image analysis software was used to segment the parasite LSPMB, hemi-spindle, and poles into either isospots or isosurfaces, as previously described (16,56). The counts, volumes, and coefficients of the isospots or isosurfaces were retrieved in .csv files, which were then imported into GraphPad Prism for statistical analysis. All data were analyzed using GraphPad Prism version 10.4.1 for macOS (GraphPad Software, San Diego, California, USA). Statistical comparisons were performed using one-way ANOVA with Dunn’s multiple comparisons test (*p ≤ 0.05, p ≤ 0.01, *p ≤ 0.001). All experiments in the study were carried out in triplicate.
Supplementary figures

Effect of Plasmodium-specific microtubule inhibitors on HeLa cell.
HeLa cells were seeded in 6-well plates and treated the following day with various concentrations of microtubule-targeting drugs. Media were replaced daily with fresh drug-supplemented medium. After five days, surviving cells were detached and counted. (A) Effect of amiprophos-methyl (AMP) on HeLa cell viability. A significant reduction in cell count was observed only at 50 µM compared to the DMSO control. (B) effect of the oryzalin on HeLa cells viability. No significant differences in cell count were observed at any tested concentrations tested relative to DMSO. (C) represent the parasite size at 48 hpi in infected HeLa cells treated with oryzalin and AMP at 50 µM. oryzalin showed a significant inhibitory effect on parasite growth compared to AMP and DMSO control. Statistical comparison tests were performed using a one-way ANOVA test (*p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001). (D and E) Maximum projection confocal IFA z-stacks of HeLa cells treated with amiprophos-methyl (D) and oryzalin (E) tested at different concentrations, respectively. Cells were fixed with cold and stained with anti-α-tubulin (magenta) and DAPI (blue). (F) Confocal IFA z-projection of HeLa cells treated with low-dose oryzalin (0.5 µM), showing unusually elongated microtubule structures. (G) PS-ExM confocal z-stack of oryzalin-treated cells showing the very long LSPMB and hemi-spindles. (H) Quantification of LSPMB and hemi-spindle lengths. Oryzalin-treated microtubules were on average four times longer than those in DMSO-treated controls. Each dot represents a measurement from a single cell. LMT, Long microtubule; NMT, normal microtubule. Statistical comparison tests were performed using two tailed unpaired t tests (*p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001).

Plasmodium end-binding protein 1 (eB1) is expressed in mosquito and liver stag.
(A) Pearson correlation coefficients between polyglutamylated LSPMB and α-tubulin LSPMB at 6, 36, and 48 hpi during liver stage development. On average, 92% of polyglutamylated tubulin colocalized with the LSPMB. (B) Maximum projection of PS-ExM confocal z-stacks of infected HeLa cells fixed at 30 and 48 hpi. Cells were stained with anti-γ-tubulin (green), anti-α-tubulin (magenta), anti-UIS4 (grey, PVM), and DAPI (blue, nuclei). White arrows indicate host centrosomes; yellow arrows highlight the LSPMB. Parasite hemi-spindle poles were also detected with γ-tubulin. (C) Schematic of the double homologous recombination construct used to tag Plasmodium eB1 (PBANKA_0405600) with eGFP at the C-terminus. The expression cassette (blue box) includes 3′UTR PbDHFR, EF1α promoter, hDHFR, yeast fcu, and the 3′UTR of PBANKA_0405600, flanked by the 5′HR (eB1 coding region) and 3′UTR. The construct was transfected into the PbmCherry 1868 mother line. (D) Integration PCR confirming correct insertion of the eGFP-tagged eB1 cassette. PCR product sizes: 5′HR = 1.5 kb, 3′HR = 1.3 kb, whole locus (WL) = 6 kb. Primer sequences listed in Table 1. Lane labels: Tr (transgenic), WT (wild type), ctrl (control). Ladder: 1 kb DNA marker. (E) Flow cytometry profile of erythrocytes infected with eB1-GFP-expressing parasites. Gate P1 includes all analyzed cells; P11 identifies mCherry-positive (PE-Texas Red-H) infected erythrocytes; R10 identifies GFP+/mCherry+ gametocytes. (F) Summary table showing parasitemia percentages across gates defined in (E). (G) Representative confocal live imaging of P. berghei eB1-GFP oocysts at 7 and 12 days post-infection (dpi), showing eB1-GFP (green), parasite mCherry (red), and Hoechst-stained nuclei (blue). Scale bar: 5 µm. (H) Confocal IFA image of P. berghei eB1-GFP sporozoites stained with anti-GFP (green), anti-α-tubulin (magenta), and anti-circumsporozoite protein (CSP, grey). (I) Maximum projection of PS-ExM confocal z-stacks of HeLa cells infected with P. berghei eB1-GFP parasites at 36 and 48 hpi. Cells were stained with anti-GFP (green), anti-α-tubulin (magenta), and anti-UIS4 (grey). Scale bar: 10 µm.

Plasmodium α-tubulin proteins and mutant parasite characterization.
(A) Comparison of Plasmodium α1- and α2-tubulin protein sequences highlighting predicted post-translational modification (PTM) sites. Polyglutamylation site E445 is conserved in both isoforms (green). α1-tubulin contains a predicted tyrosination site at Y453 (magenta), while α2-tubulin has a predicted detyrosination site at E450 (magenta). Neither isoform has the conserved acetylation site K40 (yellow). C-terminal PTM regions are underlined in red. (B) Sequence alignment of PBANKA_0417700 gene showing mutations introduced in the tubulin mutant parasites. Red highlights indicate substitutions or deletions. Dashes (—) indicate deleted nucleotides. Ps″: glutamine (E) substituted with alanine (A) at the polyglutamylation site; Ty″: deletion of the C-terminal tyrosination/detyrosination motif; Ct″: deletion of the entire C-terminal PTM region; No″: unmodified construct control. (C) Structural comparison of α1-tubulin mutant proteins using AlphaFold 3 predictions, aligned with PyMOL (Version 2.6.0). Green: wild type (No″); cyan: Ps″ mutant; magenta: Ty″ mutant; and yellow: Ct″ mutant. All predicted structures retained overall folding. (D) Maximum projection of confocal z-stacks of HeLa cells infected with mutant parasites (adapted from Freedy et al.). Microtubules were stained with anti-α-tubulin (magenta) and polyglutamylated tubulin with anti-IN105 (green). Nuclei were stained with DAPI (blue); the parasitophorous vacuole membrane (PVM) was stained with anti-UIS4 (grey). IntronΔ: line with deleted introns in the Plasmodium α1-tubulin gene; C-termΔ: line with deletion of C-terminal residues 540ADY. (E) In vivo infection kinetics in mice infected with mutant parasites. Parasitemia was measured daily from day 1 to day 5 post-infection. No significant differences were observed between mutant lines (intronΔ and C-termΔ) and wild type (WT). ExonΔ: mutant line in which α1-tubulin exons were replaced with α2-tubulin introns. This line failed to produce salivary gland sporozoites. Liver stage development in this line was delayed by 4 days due to hemolymph sporozoites used. Experiments were performed in triplicate.

Plasmodium α-tubulin mutant parasite oocyst formation and sporozoite infectivity.
(A) Comparison of oocyst area at day 7 and 11 post-infection. No significant difference was observed between the WT (NoΔ) and the PsΔ or TyΔ mutants at day 7 post-infection. However, by day 11, the CtΔ mutant exhibited a significantly reduced oocyst size compared to the WT. (B) Normalized midgut sporozoites count per mosquito (C./mosq.) at day 14 post-feeding. No significant difference in sporozoite numbers was observed between NoΔ and PsΔ, whereas CtΔ showed a drastic reduction. In contrast, TyΔ displayed both a significantly larger oocyst size at day 11 and a higher sporozoite count at day 14 compared to the WT. (C) Blood parasitemia levels in mice infected with mutant parasites, measured from day 0 to day 7 post-infection. All mutants (PsΔ and TyΔ) established infections and progressed similarly to the WT (NoΔ). PsΔ: glutamine-to-alanine substitution at polyglutamylation site; TyΔ: deletion of C-terminal tyrosination/detyrosination residues; CtΔ: complete deletion of PBANKA_0417700 α-tubulin C-terminal PTM residues; NoΔ: control construct with no mutation. The experiments were conducted in triplicate. Statistical multiple comparisons were performed using one-way ANOVA with Dunnett’s multiple comparisons test (*p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001).
Acknowledgements
The authors gratefully acknowledge the Microscopy Imaging Center (MIC) at the University of Bern, Switzerland, for their technical support and access to microscopy equipment. K.A. acknowledges the Swiss Confederation for the award of a Government Excellence Scholarship. This work was funded by the Swiss National Science Foundation (SNSF) (grant number 310030_212795) and an Multidisciplinary Center for Infectious Diseases (MCID), University of Bern, Switzerland, grant to Volker Heussler (MA-09).
Additional files
References
- 1.Impact of Malaria Diagnostic Technologies on the Disease Burden in the Sub-Saharan Africahttps://onlinelibrary.wiley.com/doi/10.1155/2022/7324281
- 2.MalariaLancet 402:2328–45PubMedGoogle Scholar
- 3.Update on malariaMed Clin (Barc) 155:395–402PubMedGoogle Scholar
- 4.WHO world malaria report 2024Lancet Microbe 6:101073https://www.thelancet.com/action/showFullText?pii=S2666524725000011Google Scholar
- 5.Role of host cell traversal by the malaria sporozoite during liver infectionJournal of Experimental Medicine 210:905–15https://www.jem.org/cgi/doi/10.1084/jem.20121130Google Scholar
- 6.Plasmodium berghei sporozoites in nonreplicative vacuole are eliminated by a PI3P-mediated autophagy-independent pathwayCell Microbiol 23PubMedGoogle Scholar
- 7.Hepatocytes and the art of killing Plasmodium softlyTrends Parasitol 40:466–76https://www.cell.com/action/showFullText?pii=S1471492224000862Google Scholar
- 8.Long-term live imaging reveals cytosolic immune responses of host hepatocytes against Plasmodium infection and parasite escape mechanismsAutophagy 11:1561–79PubMedGoogle Scholar
- 9.Plasmodium vivax pre-erythrocytic stages and the latent hypnozoiteParasitol Int 85:102447Google Scholar
- 10.The silent path to thousands of merozoites: the Plasmodium liver stageNature Reviews Microbiology 4:849–56https://www.nature.com/articles/nrmicro1529Google Scholar
- 11.Same, same but different: Exploring Plasmodium cell division during liver stage developmentPLoS Pathog 19:e1011210Google Scholar
- 12.Proteomic Analysis of Plasmodium Merosomes: The Link between Liver and Blood Stages in MalariaJ Proteome Res 18:3404–18https://pubs.acs.org/doi/full/10.1021/acs.jproteome.9b00324Google Scholar
- 13.Rodent malaria models: insights into human disease and parasite biologyCurr Opin Microbiol 46:93–101PubMedGoogle Scholar
- 14.Manipulation of host hepatocytes by the malaria parasite for delivery into liver sinusoidsScience 313:1287–90https://www.science.org/doi/10.1126/science.1129720Google Scholar
- 15.PlasmodiumMolecular Parasitology: Protozoan Parasites and their Molecules :241–84https://link.springer.com/chapter/10.1007/978-3-7091-1416-2_9Google Scholar
- 16.Expanding the fluorescent toolkit: Blue fluorescent protein-expressing Plasmodium berghei for enhanced multiplex microscopyPLoS One 20:e0308055PubMedGoogle Scholar
- 17.Genome-Scale Identification of Essential Metabolic Processes for Targeting the Plasmodium Liver StageCell 179:1112–1128PubMedGoogle Scholar
- 18.Biology of Malaria TransmissionCold Spring Harb Perspect Med 7PubMedGoogle Scholar
- 19.Imaging movement of malaria parasites during transmission by Anopheles mosquitoesCell Microbiol 6:687–94https://onlinelibrary.wiley.com/doi/full/10.1111/j.1462-5822.2004.00395.xGoogle Scholar
- 20.Development of infectivity by the Plasmodium berghei sporozoiteJournal of Parasitology 61:43–50Google Scholar
- 21.Studies on the Motility of Plasmodium Sporozoites*J Protozool 21:527–37https://onlinelibrary.wiley.com/doi/full/10.1111/j.1550-7408.1974.tb03693.xGoogle Scholar
- 22.Plasmodium Sporozoite BiologyCold Spring Harb Perspect Med 7:a025478Google Scholar
- 23.Dynamic instability of microtubule growthNature 312:237–42https://www.nature.com/articles/312237a0Google Scholar
- 24.The tubulin code and its role in controlling microtubule properties and functionsNat Rev Mol Cell Biol 21:307–26Google Scholar
- 25.Structural insights into microtubule functionAnnu Rev Biophys Biomol Struct 30:397–420https://www.annualreviews.org/content/journals/10.1146/annurev.biophys.30.1.397Google Scholar
- 26.The Tubulin Code in Microtubule Dynamics and Information EncodingDev Cell 54:7–20Google Scholar
- 27.Microtubule-Associated Proteins: Structuring the CytoskeletonTrends Cell Biol 29:804–19Google Scholar
- 28.Non-motor microtubule-associated proteinsCurr Opin Cell Biol 5:88–91Google Scholar
- 29.Microtubule-associated proteins and enzymes modifying tubulinCytoskeleton 80:60–76https://onlinelibrary.wiley.com/doi/full/10.1002/cm.21748Google Scholar
- 30.Microtubule-associated proteins and enzymes modifying tubulinCytoskeleton (Hoboken) 80:60–76PubMedGoogle Scholar
- 31.Isotype expression, post-translational modification and stage-dependent production of tubulins in erythrocytic Plasmodium falciparumInt J Parasitol 38:527–39Google Scholar
- 32.α-Tubulin II is a male-specific protein in Plasmodium falciparumMol Biochem Parasitol 56:239–50Google Scholar
- 33.Tyrosinated and detyrosinated microtubules in axonal processes of cerebellar macroneurons grown in cultureJ Neurosci Res 28:171–81PubMedGoogle Scholar
- 34.Differential turnover of tyrosinated and detyrosinated microtubulesProc Natl Acad Sci U S A 84:9040–4PubMedGoogle Scholar
- 35.The detyrosination/re-tyrosination cycle of tubulin and its role and dysfunction in neurons and cardiomyocytesSemin Cell Dev Biol 137:46–62PubMedGoogle Scholar
- 36.α-tubulin detyrosination fine-tunes kinetochore-microtubule attachmentsNat Commun PubMedGoogle Scholar
- 37.The polyglutamylated lateral chain of alpha-tubulin plays a key role in flagellar motilityJ Cell Sci 109:1545–53https://doi.org/10.1242/jcs.109.6.1545Google Scholar
- 38.Polyglutamylated alpha-tubulin can enter the tyrosination/detyrosination cycleBiochemistry 31:403–10PubMedGoogle Scholar
- 39.Polyglutamylation: biology and analysisAmino Acids 54:529–42PubMedGoogle Scholar
- 40.ER proteins decipher the tubulin code to regulate organelle distributionNature 601:132–8PubMedGoogle Scholar
- 41.Isolation of the beta-tubulin gene from yeast and demonstration of its essential function in vivoCell 33:211–9PubMedGoogle Scholar
- 42.Identification of tubulin from the yeast Saccharomyces cerevisiaeProc Natl Acad Sci U S A 75:4962–6Google Scholar
- 43.Purification of functional Plasmodium falciparum tubulin allows for the identification of parasite-specific microtubule inhibitorsCurrent Biology 32:919–926Google Scholar
- 44.The tubulin genes of the human malaria parasite Plasmodium falciparum, their chromosomal location and sequence analysis of the α-tubulin II geneMol Biochem Parasitol 43:257–70Google Scholar
- 45.Cellular and molecular actions of dinitroaniline and phosphorothioamidate herbicides on Plasmodium falciparum: tubulin as a specific antimalarial targetMol Biochem Parasitol 145:226–38PubMedGoogle Scholar
- 46.Plasmodium: assessment of the antimalarial potential of trifluralin and related compounds using a rat model of malaria, Rattus norvegicusExp Parasitol 100:155–60PubMedGoogle Scholar
- 47.Antimitotic herbicides bind to an unidentified site on malarial parasite tubulin and block development of liver-stage Plasmodium parasitesMol Biochem Parasitol 188:116–27PubMedGoogle Scholar
- 48.Plasmodium berghei alpha-tubulin II: a role in both male gamete formation and asexual blood stagesMol Biochem Parasitol 144:16–26PubMedGoogle Scholar
- 49.Isolation of alpha-tubulin genes from the human malaria parasite, Plasmodium falciparum: sequence analysis of alpha-tubulinMol Microbiol 3:1501–10PubMedGoogle Scholar
- 50.Microtubule number and length determine cellular shape and function in PlasmodiumEMBO J 38https://www.embopress.org/doi/10.15252/embj.2018100984Google Scholar
- 51.Targeting plasmodium α-tubulin-1 to block malaria transmission to mosquitoesFront Cell Infect Microbiol 13:1132647Google Scholar
- 52.Graded Control of Microtubule Severing by Tubulin GlutamylationCell 164:911–21https://www.cell.com/action/showFullText?pii=S0092867416000593Google Scholar
- 53.Differential Binding Regulation of Microtubule-associated Proteins MAP1A, MAP1B, and MAP2 by Tubulin PolyglutamylationJournal of Biological Chemistry 276:12839–48https://www.jbc.org/action/showFullText?pii=S0021925819343935Google Scholar
- 54.Expansion microscopy provides new insights into the cytoskeleton of malaria parasites including the conservation of a conoidPLoS Biol 19PubMedGoogle Scholar
- 55.Microtubule inner proteins of Plasmodium are essential for transmission of malaria parasitesProceedings of the National Academy of Sciences 122:e2421737122https://www.pnas.org/doi/abs/10.1073/pnas.2421737122Google Scholar
- 56.Pre-gelation staining expansion microscopy for visualisation of the Plasmodium liver stageJ Cell Sci 136:jcs261377Google Scholar
- 57.Expansion microscopy of apicomplexan parasitesMol Microbiol 121:619–35PubMedGoogle Scholar
- 58.Atlas of Plasmodium falciparum intraerythrocytic development using expansion microscopyElife 12:RP88088PubMedGoogle Scholar
- 59.Variable microtubule architecture in the malaria parasiteNat Commun 14:1216Google Scholar
- 60.Unveiling the Possible Oryzalin-Binding Site in the α-Tubulin of Toxoplasma gondiiACS Omega 7:18434–42https://pubs.acs.org/doi/full/10.1021/acsomega.2c00729Google Scholar
- 61.Putative prefoldin complex subunit 5 of Plasmodium berghei is crucial for microtubule formation and parasite development in the mosquitoMol Microbiol 121:481–96PubMedGoogle Scholar
- 62.Plasmodium centrin PbCEN-4 localizes to the putative MTOC and is dispensable for malaria parasite proliferationBiol Open 8:bio036822Google Scholar
- 63.Plasmodium kinesin-8X associates with mitotic spindles and is essential for oocyst development during parasite proliferation and transmissionPLoS Pathog 15:e1008048Google Scholar
- 64.EB1 decoration of microtubule lattice facilitates spindle-kinetochore lateral attachment in Plasmodium male gametogenesisNature Communications 14:1–20https://www.nature.com/articles/s41467-023-38516-3Google Scholar
- 65.Plasmodium microtubule-binding protein EB1 is critical for partitioning of nuclei in male gametogenesismBio 14:e00822–23Google Scholar
- 66.Investigating Tubulin Posttranslational Modifications with Specific AntibodiesMethods Cell Biol 115:247–67Google Scholar
- 67.gamma-TubulinCurr Top Dev Biol 49:27–54PubMedGoogle Scholar
- 68.The γ-tubulin gene of the malaria parasite Plasmodium falciparumMol Biochem Parasitol 60:27–35Google Scholar
- 69.γ-tubulin complex controls the nucleation of tubulin-based structures in ApicomplexaMol Biol Cell 35:ar121Google Scholar
- 70.Host–Parasite Interactions in Toxoplasma gondii-Infected Cells: Roles of Mitochondria, Microtubules, and the Parasitophorous Vacuole. Int J Mol Sci 25:13459Google Scholar
- 71.Manipulation of the Host Cell Membrane during Plasmodium Liver Stage EgressmBio 8PubMedGoogle Scholar
- 72.Longitudinal analysis of plasmodium sporozoite motility in the dermis reveals component of blood vessel recognitioneLife 4Google Scholar
- 73.Limited Plasmodium sporozoite gliding motility in the absence of TRAP family adhesinsMalar J 20:1–12https://malariajournal.biomedcentral.com/articles/10.1186/s12936-021-03960-3Google Scholar
- 74.Motility precedes egress of malaria parasites from oocystseLife 6Google Scholar
- 75.Conformational change of Plasmodium TRAP is essential for sporozoite migration and transmissionEMBO Rep 24https://www.embopress.org/doi/10.15252/embr.202357064Google Scholar
- 76.Screening for potential prophylactics targeting sporozoite motility through the skinMalar J 17PubMedGoogle Scholar
- 77.Transformation of sporozoites of Plasmodium berghei into exoerythrocytic forms in the liver of its mammalian hostCell Tissue Res 241:353–60https://link.springer.com/article/10.1007/BF00217180Google Scholar
- 78.Polyglutamylation makes the cutJ Cell Biol 189:920Google Scholar
- 79.Tubulin polyglutamylation differentially regulates microtubule-interacting proteinsEMBO J 42:e112101Google Scholar
- 80.Polyglutamylation and the fleer geneMethods Cell Biol 94:317–32Google Scholar
- 81.Metamorphosis of the malaria parasite in the liver is associated with organelle clearanceCell Res 20:1043Google Scholar
- 82.Chronicle of a death foretold: Plasmodium liver stage parasites decide on the fate of the host cellFEMS Microbiol Rev 36:111–30https://doi.org/10.1111/j.1574-6976.2011.00297.xGoogle Scholar
- 83.Hijacking of the host cell Golgi by Plasmodium berghei liver stage parasitesJ Cell Sci 134:jcs252213Google Scholar
- 84.Lipid hijacking: A unifying theme in vector-borne diseaseseLife 9:1–31Google Scholar
- 85.Harnessing cholesterol uptake of malaria parasites for therapeutic applicationsEMBO Mol Med 16:1515–32https://www.embopress.org/doi/10.1038/s44321-024-00087-1Google Scholar
- 86.Plasmodium berghei liver stage parasites exploit host GABARAP proteins for TFEB activationCommunications Biology 7:1–13https://www.nature.com/articles/s42003-024-07242-xGoogle Scholar
- 87.Plasmodium ARK2 and EB1 drive unconventional spindle dynamics, during chromosome segregation in sexual transmission stagesNature Communications 14:1–19https://www.nature.com/articles/s41467-023-41395-3Google Scholar
- 88.Polyglutamylation Is a Post-translational Modification with a Broad Range of SubstratesJournal of Biological Chemistry 283:3915–22Google Scholar
- 89.The Tubulin Superfamily in Apicomplexan ParasitesMicroorganisms 11PubMedGoogle Scholar
- 90.The Plasmodium sporozoite journey: a rite of passageTrends Parasitol 19:135–43Google Scholar
- 91.The Dynamic Roles of the Inner Membrane Complex in the Multiple Stages of the Malaria ParasiteFront Cell Infect Microbiol 10:611801Google Scholar
- 92.The inner membrane complex through development of Toxoplasma gondii and PlasmodiumCell Microbiol 16:632Google Scholar
- 93.A Plasmodium Phospholipase Is Involved in Disruption of the Liver Stage Parasitophorous Vacuole MembranePLoS Pathog 11:e1004760Google Scholar
- 94.High resolution microscopy reveals an unusual architecture of the Plasmodium berghei endoplasmic reticulumMol Microbiol 102:775–91PubMedGoogle Scholar
- 95.Conditional degradation of plasmodium calcineurin reveals functions in parasite colonization of both host and vectorCell Host Microbe 18:122–31https://www.cell.com/action/showFullText?pii=S1931312815002504Google Scholar
- 96.A novel “Gene Insertion/Marker Out” (GIMO) method for transgene expression and gene complementation in rodent malaria parasitesPLoS One 6Google Scholar
- 97.High-efficiency transfection and drug selection of genetically transformed blood stages of the rodent malaria parasite Plasmodium bergheiNature Protocols 1:346–56https://www.nature.com/articles/nprot.2006.53Google Scholar
- 98.An ultrasensitive NanoLuc-based luminescence system for monitoring Plasmodium berghei throughout its life cycleMalaria Journal 15:1–24https://malariajournal.biomedcentral.com/articles/10.1186/s12936-016-1291-9Google Scholar
- 99.Generation of transgenic rodent malaria parasites expressing human malaria parasite proteinsMethods in Molecular Biology 1325:257–86Google Scholar
- 100.Lyse-Reseal Erythrocytes for Transfection of Plasmodium falciparumSci Rep 9Google Scholar
- 101.A multistage antimalarial targets the plasmepsins IX and X essential for invasion and egressScience (1979) 358:522–8https://www.science.org/doi/10.1126/science.aaf8675Google Scholar
- 102.Primary Hepatocyte Isolation and Cultures: Technical Aspects, Challenges and AdvancementsBioengineering 10:131Google Scholar
- 103.LC3B labeling of the parasitophorous vacuole membrane of Plasmodium berghei liver stage parasites depends on the V-ATPase and ATG16L1Mol Microbiol 121:1095–111https://onlinelibrary.wiley.com/doi/full/10.1111/mmi.15259Google Scholar
- 104.Dissection-independent production of Plasmodium sporozoites from whole mosquitoesLife Sci Alliance 4:e202101094Google Scholar
- 105.Mosquito Tissue Ultrastructure-Expansion Microscopy (MoTissU-ExM) enables ultrastructural and anatomical analysis of malaria parasites and their mosquitoBMC Methods 1:1–15https://bmcmethods.biomedcentral.com/articles/10.1186/s44330-024-00013-4Google Scholar
- 106.Imaging liver-stage malaria parasitesCell Microbiol 12:569–79PubMedGoogle Scholar
Article and author information
Author information
Version history
- Sent for peer review:
- Preprint posted:
- Reviewed Preprint version 1:
Cite all versions
You can cite all versions using the DOI https://doi.org/10.7554/eLife.108209. This DOI represents all versions, and will always resolve to the latest one.
Copyright
© 2025, Atchou et al.
This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.
Metrics
- views
- 0
- downloads
- 0
- citations
- 0
Views, downloads and citations are aggregated across all versions of this paper published by eLife.