Skip to main content
PLOS Pathogens logoLink to PLOS Pathogens
. 2023 Feb 13;19(2):e1010890. doi: 10.1371/journal.ppat.1010890

PbAP2-FG2 and PbAP2R-2 function together as a transcriptional repressor complex essential for Plasmodium female development

Tsubasa Nishi 1, Izumi Kaneko 1, Shiroh Iwanaga 2, Masao Yuda 1,*
Editor: Ron Dzikowski3
PMCID: PMC9956629  PMID: 36780562

Abstract

Gametocyte development is a critical step in the life cycle of Plasmodium. Despite the number of studies on gametocyte development that have been conducted, the molecular mechanisms regulating this process remain to be fully understood. This study investigates the functional roles of two female-specific transcriptional regulators, PbAP2-FG2 and PbAP2R-2, in P. berghei. Knockout of pbap2-fg2 or pbap2r-2 impairs female gametocyte development, resulting in developmental arrest during ookinete development. ChIP-seq analyses of these two factors indicated their colocalization on the genome, suggesting that they function as a complex. These analyses also revealed that their target genes contained a variety of genes, including both male and female-enriched genes. Moreover, differential expression analyses showed that these target genes were upregulated through the disruption of pbap2-fg2 or pbap2r-2, indicating that these two factors function as a transcriptional repressor complex in female gametocytes. Formation of a complex between PbAP2-FG2 and PbAP2R-2 was confirmed by RIME, a method that combines ChIP and MS analysis. In addition, the analysis identified a chromatin regulator PbMORC as an interaction partner of PbAP2-FG2. Comparative target analysis between PbAP2-FG2 and PbAP2-G demonstrated a significant overlap between their target genes, suggesting that repression of early gametocyte genes activated by PbAP2-G is one of the key roles for this female transcriptional repressor complex. Our results indicate that the PbAP2-FG2-PbAP2R-2 complex-mediated repression of the target genes supports the female differentiation from early gametocytes.

Author summary

Gametocyte development in Plasmodium parasites, a causative agent of malaria, is an essential step for their transmission from vertebrate hosts to mosquitoes. Gametocytes are sexual precursor cells produced from a subpopulation of asexual blood-stage parasites. Upon uptake by mosquitoes through blood feeding, the male and female gametocytes become microgametes and macrogametes, respectively, and then they fertilize and develop into the mosquito midgut invasive stage, called ookinete. Therefore, it is crucial to understand the underlying mechanisms regulating this developmental process. This study revealed that the two female transcriptional regulators, PbAP2-FG2 and PbAP2R-2, function together as an essential transcriptional repressor complex in P. berghei, the target genes of which include male, female, and early gametocyte genes activated by PbAP2-G. Our findings suggest that PbAP2-FG2 and PbAP2R-2 play multiple roles in supporting the development of female gametocytes from early gametocytes.

Introduction

Plasmodium parasites are the causative agent of malaria, one of the most severe infectious diseases worldwide. The spread of the parasites among individuals occurs through mosquito bites, resulting in more than 200 million cases and 500 thousand deaths yearly [1]. Parasite transmission from vertebrate hosts to mosquitoes is involved in the sexual development of the parasite [2,3]. During asexual reproduction in the host blood, subpopulations of parasites differentiate into gametocytes to prepare for transmission to mosquitoes [4,5]. When the gametocytes are taken up by mosquitoes through blood feeding, they egress from red blood cells, form gametes, and fertilize. The fertilized cells then develop into ookinetes and invade the midgut of mosquitoes, completing the transmission [6]. Parasite transmission is a critical event in the propagation of malaria, and thus understanding the molecular mechanisms regulating these developmental steps is crucial for malaria epidemiology.

Along with several transcriptomic [710] and proteomic studies [11,12], studies of gametocyte-specific transcriptional regulators have been conducted to elucidate the mechanisms of Plasmodium gametocyte development. In Plasmodium spp., sexual development is triggered by AP2-G [1315]. It is an AP2-family transcription factor that is expressed in a subpopulation of blood-stage parasites, and disruption of the gene results in complete loss of the parasite’s ability to produce gametocytes [16,17]. Furthermore, it has been reported that intentional conversion of parasites into the sexual stage could be achieved by conditional induction of AP2-G in both P. falciparum and P. berghei [18,19]. We previously conducted chromatin immunoprecipitation (ChIP) followed by high-throughput sequencing (ChIP-seq) analysis of PbAP2-G and identified its target genes [20]. Among these targets of PbAP2-G, we found several important transcription factors for sexual development. These targets include pbap2-g2, a transcription factor that is expressed in both male and female gametocytes. In both P. berghei and P. falciparum, AP2-G2 functions as a transcriptional repressor to ensure the alteration of cell fate from the asexual blood stage to the sexual stage [21,22]. The target genes of PbAP2-G also include a female-specific transcription factor gene, pbap2-fg [23,24]. PbAP2-FG activates most female-enriched genes, and disruption of this gene results in the formation of abnormal female gametocytes [23]. In addition to these gametocyte-specific transcription factors, the zygote transcription factor gene pbap2-z was also identified as a target of PbAP2-G [25]. Moreover, a transcriptional regulator gene, pfhdp1, that is essential for early gametocyte development was included in the target genes of PfAP2-G, and its ortholog was also a target of PbAP2-G [26,27]. Therefore, it was considered that PbAP2-G comprehensively activates transcriptional regulator genes essential for Plasmodium sexual development.

pbap2-o3 and pbap2r-2 are also a target gene of PbAP2-G. It has been reported that in both P. berghei and P. yoelii, disruption of ap2-o3 results in arrest of parasite development during ookinete development, thereby helping derive the name ap2-o3 [28,29]. PbAP2R-2, on the other hand, is expressed in female gametocytes, and disruption of this gene impairs ookinete development [20]. Here, we report that PbAP2-O3 and PbAP2R-2 are expressed in female gametocytes and function together as a transcriptional repressor complex in P. berghei. Accordingly, we renamed PbAP2-O3 PbAP2-FG2. Moreover, differential expression analyses and ChIP-seq analyses revealed that PbAP2-FG2 and PbAP2R-2 repress various genes, including female, male, and early gametocyte genes, to support female development. Recently, Li et al. reported that the ortholog of PbAP2-FG2 in another rodent malaria parasite, P. yoelii, (PyAP2-O3) also functions as a transcriptional repressor in female gametocytes [30]. Although some of their conclusions were inconsistent with those in this study, reassessment of their data led to similar conclusions in both P. berghei and P. yoelii.

Results

PbAP2-FG2 is expressed in female gametocytes and is essential for their development

PbAP2-FG2 (encoded by PBANKA_1015500), previously named PbAP2-O3, is an AP2-family transcription factor, conserved across the Plasmodium species. It possesses a single AP2 domain near its N-terminus and an AP2-coincident domain mostly at the C-terminus (ACDC) domain (Fig 1A). Modrzynska et al. demonstrated that disruption of this gene results in the failure of the majority of zygotes to form an apical protrusion in P. berghei [28]. This result indicated that PbAP2-FG2 has a role in zygote/ookinete development or regulating female transcriptome because female gametocytes store large amount of mRNAs that are essential for development after fertilization. Our previous ChIP-seq data demonstrated that the upstream region of pbap2-fg2 harbored binding sites of PbAP2-G and PbAP2-FG (encoded by PBANKA_1437500 and PBANKA_1415700, respectively), which are transcriptional activators expressed in early gametocytes and female gametocytes, respectively (S1 Fig) [20,23]. This indicates that PbAP2-FG2 is possibly expressed in female gametocytes. To identify the stage at which PbAP2-FG2 functions, we first generated a parasite line expressing GFP-fused PbAP2-FG2 (PbAP2-FG2::GFP, S2A Fig) and assessed its expression pattern. In the blood stage, PbAP2-FG2 expression was observed in the nucleus of female gametocytes but not in the other stages including male gametocytes (Fig 1B). When cultured in an ookinete culture medium, PbAP2-FG2::GFP parasites produced banana-shaped ookinetes, confirming that the GFP fusion did not impair PbAP2-FG2 function. During ookinete development, no fluorescence was detected at any stage of the parasites (Fig 1B). These results collectively indicated that like PyAP2-O3 (encoded by PY17X_1017000), PbAP2-FG2 is only expressed in female gametocytes during sexual development and presumably functions in females instead of zygotes.

Fig 1. Expression pattern and knockout phenotype of pbap2-fg2.

Fig 1

(A) Schematic diagram of protein features in PbAP2-FG2. (B) Fluorescence analysis of PbAP2-FG2::GFP during blood-stage and sexual development. Nuclei were stained with Hoechst 33342. Scale bar = 5 μm. (C) Top: representative Giemsa-stained images showing gametocytes of WT and pbap2-fg2(-) parasites. Scale bar = 5 μm. Bottom: ratio of normal/abnormal female and male gametocytes in WT and pbap2-fg2(-). Error bars indicate the standard error of the mean (n = 3). (D) Number of exflagellation centers per 1 μL of peripheral blood from mice infected with WT and pbap2-fg2(-). The infected mice were treated with sulfadiazine in their drinking water to enrich gametocyte population in their blood stream. Error bars indicate the standard error of the mean (n = 3). (E) Left: representative Giemsa-stained images showing ookinetes of WT and pbap2-fg2(-) at 20 h after starting ookinete cultures. Scale bar = 5 μm. Right: Rate of conversion to zygote, retort-form ookinete, and mature ookinete against all female-derived cells in WT and pbap2-fg2(-) at 20 hoc. Zygote and retort-form ookinete include morphologically abnormal cells without and with an apical protrusion, respectively. Error bars indicate the standard error of the mean (n = 3). (F) Number of midgut oocysts at 14 days post infection for WT and pbap2-fg2(-). Lines indicate the mean values and the standard error (n = 20). (G) Cross-fertilization assay among pbap2-fg2(-), p48/45(-) and p47(-). The number of normal and abnormal ookinetes are indicated as white and grey bars, respectively. Error bars indicate the standard error of the mean (n = 3).

To investigate the function of PbAP2-FG2, we developed pbap2-fg2 knockout parasites [pbap2-fg2(-), S2B Fig] and evaluated the phenotype in detail. pbap2-fg2 was disrupted by double cross-over homologous recombination of an hdhfr expression cassette, with the transfection conducted twice to obtain two independent clonal lines. For both of the clones, the resultant parasites formed morphologically normal female and male gametocytes (Fig 1C), and the male gametocytes showed normal exflagellation (Fig 1D). In the ookinete culture medium, pbap2-fg2(-) parasites failed to produce banana-shaped ookinetes; more than half of the fertilized population stopped developing at round zygotes, and the others, at retort-form ookinetes (Fig 1E). Furthermore, both of the clones failed to infect mosquitoes through blood feeding (Fig 1F), confirming that pbap2-fg2(-) parasites completely lost the ability to produce normal ookinetes. These results corroborated the findings of Modrzynska et al. [28]. Next, we performed a cross-fertilization assay using the clone 1 to evaluate whether female or male gametocytes of pbap2-fg2(-) were capable of forming banana-shaped ookinetes upon fertilization with normal gametocytes. We observed that crossing of pbap2-fg2(-) with a line that produced infertile females [p47(-)] [31] led to no female gametocytes being converted to banana-shaped ookinetes (Fig 1G). Development of their fertilized females was arrested at the round zygote or retort-form ookinete, recapitulating the scenario of pbap2-fg2(-) parasites cultured alone. In contrast, when pbap2-fg2(-) was crossed with a line that produces infertile males [p48/45(-)] [32], approximately 30% of female gametocytes were converted to banana-shaped ookinetes, which was approximately as much as when p48/45(-) and p47(-) were crossed (Fig 1G), demonstrating the ability of pbap2-fg2(-) to produce normal male gametocytes. Collectively, these results revealed that only female gametocytes were abnormal in pbap2-fg2(-), which, in turn, affected their ookinete development. Together with the fluorescence analysis, these results strongly suggested that PbAP2-FG2 is essential for the development of normal female gametocytes, which is consistent with the study of the PyAP2-O3 reported by Li et al. [30].

Disruption of pbap2-fg2 affected the female transcriptome

To further investigate the effect of disrupting pbap2-fg2 on female development, we performed RNA-seq analysis on gametocyte-enriched populations of wild-type ANKA strain (WT) and pbap2-fg2(-), and compared their transcriptomes. The total RNA was harvested from parasites enriched with gametocytes, which were prepared by killing asexual parasites with sulfadiazine treatment, and then sequenced using next-generation sequencing. Differentially expressed genes (DEGs) between WT and pbap2-fg2(-) were identified by analyzing the sequence data using DESeq2 after excluding genes with reads per kilobase of transcript per million mapped reads (RPKM) < 10 (S1A Table). In pbap2-fg2(-) parasites, 180 genes were significantly downregulated [log2(fold change) < -1, p-value adjusted for multiple testing with the Benjamini-Hochberg procedure (p-value adj) < 0.05], and 96 genes were significantly upregulated [log2(fold change) > 1, p-value adj < 0.05] compared to the WT (Fig 2A, S1B and S1C Table). To evaluate how disruption of pbap2-fg2 affects gametocyte transcriptome, we assessed the expression of these DEGs in previously reported sex-specific RNA-seq data [10]. We identified genes more than fourfold enriched with p-value adj < 0.001 in each sexual stage compared to the other and asexual blood stages as sex-enriched genes and obtained 504 female-enriched genes and 438 male-enriched genes (all the other genes were assigned as “other” in this context) (S2A and S2B Table). The genes downregulated in pbap2-fg2(-) contained 54 female-enriched genes and nine male-enriched genes, highlighting the dominance of female-enriched genes with p-value = 7.0 × 10−14 by Fisher’s exact test (Fig 2A and 2B). Furthermore, in the total transcriptome, log2(fold change) of female-enriched genes tended to be lower than the other genes (p-value = 1.4 × 10−18 by two-tailed Student’s t-test). In contrast, the upregulated genes showed no specific enrichment in the female- or male-enriched genes (Fig 2A and 2C). These results indicated that disruption of pbap2-fg2 impaired the female transcriptome, causing downregulation of female-enriched genes.

Fig 2. Differential expression analysis between WT and pbap2-fg2(-).

Fig 2

(A) A volcano plot showing differential expression of genes in pbap2-fg2(-) compared to WT. Purple and orange dots represent female and male-enriched genes, respectively. A red dot indicates pbap2-fg2. A horizontal line indicates p-value of 0.05, and two vertical lines indicate log2(Fold Change) of -1 and 1. (B) Classification of significantly downregulated genes into sexual stage-enriched gene sets. All genes not enriched in either “female” or “male” are classified as “other”. (C) Classification of significantly upregulated genes into sexual stage-enriched gene sets.

PbAP2-FG2 targets a wide variety of genes, binding to specific sequences

Differential expression analysis between WT and pbap2-fg2(-) suggested that PbAP2-FG2 was involved in transcriptional regulation in female gametocytes. Therefore, we employed ChIP-seq analysis to identify the binding motif of PbAP2-FG2 and its target genes. We performed ChIP experiments with PbAP2-FG2::GFP using an anti-GFP antibody, followed by the sequencing of the DNA fragments purified from the immunoprecipitated chromatin and the input cell lysate. From the sequence data, peaks were called with fold enrichment > 3.0 and q-value < 0.01 using the macs2 program, setting the sequence data of input DNA as a control. Two biologically independent experiments were performed, the results of which were comparable based on the genome-wide peak pattern of the data (Fig 3A). We identified 1321 and 1648 peaks in Experiments 1 and 2, respectively, and the locations of 1231 peaks (93.2% of the peaks from Experiment 1) overlapped between the two experiments, suggesting that the data had high reproducibility. To further evaluate the reproducibility of each peak, IDR1D analysis was performed on the two data [33,34]. In this analysis, peaks were ranked according to their p-values within each replicate, and the ranks were compared between the two experiments. According to the consistency of ranks across the two experiments, the irreproducible discovery rate (IDR) score, which defines the reproducibility of each peak, was calculated for each one. As the ranks of each peak lose consistency between the replicates, the peaks have higher IDR scores; hence, peaks with small IDR scores are considered reliable. The results depicted that 638 peaks had an IDR < 0.01 (Fig 3B, S3A and S3B Table), and we decided to utilize these peaks for further analysis as they are highly reproducible.

Fig 3. ChIP-seq analysis using PbAP2-FG2::GFP.

Fig 3

(A) IGV images showing peaks identified in the ChIP-seq experiment 1 and 2 of PbAP2-FG2 on the chromosome 1. Read coverage for the ChIP data is shown. (B) IDR1D analysis between the ChIP-seq experiment 1 and 2. The rank of peaks according to their p-value for each experiment is plotted against each other. Peaks with IDR < 0.01 are indicated as blue dots. (C) Motifs enriched within 50 bp from peak summits identified in the ChIP-seq of PbAP2-FG2. The logos were depicted using WebLogo 3 (http://weblogo.threeplusone.com/). Percentage of the peaks that had at least one of these motifs within 300 bp from the summit is indicated as a pie graph in the top left corner. (D) Distance between peak summits and the nearest major or variant motifs. (E) Classification of target genes into 17 groups according to their functional annotation. (F) Gene ontology analysis for target genes of PbAP2-FG2. Terms with p-value < 0.01 are shown. (G) Classification of target genes of PbAP2-FG2 into sexual stage-enriched gene sets.

We first attempted to identify the binding motif of PbAP2-FG2 by searching for statistically enriched sequences around the highly reproducible peaks. We searched for 6-bp motifs on sequences within 100 bp from each summit and found enrichment of several motifs using Fisher’s exact test. These motifs were unified, and RGAGAR (R = A or G) was identified as the most significantly enriched motif in the peak regions with a p-value of 5.2 × 10−89 (Fig 3C). In addition to the RGAGAR motifs, we found GAGARA and ARGAGA as enriched motifs with a p-value of 2.6 × 10−68 and 3.5 × 10−53, respectively (Fig 3C). These motifs appeared to be variants of the most enriched motif, sharing GAGA within their sequences. Accordingly, we hereafter refer to the RGAGAR motif as the major motif and the GAGARA and ARGAGA motifs as the variant motifs 1 and 2, respectively. Searching for these three motifs around peak summits revealed that 85% of the peaks had at least one of these enriched motifs within 300 bp of the summit (Fig 3C). Moreover, for more than half of these peaks, the distance between the peak summit and the nearest motif was within 50 bp (Fig 3D). These results indicated that PbAP2-FG2 binds to the major motif RGAGAR and its variant motifs.

Next, we analyzed the genomic location of the peaks identified by ChIP-seq analysis to determine the potential targets of PbAP2-FG2. The analysis revealed that of the 638 highly reproducible peaks, 558 peaks located in intergenic regions. These intergenic peaks located in the upstream regions (within 1200 bp from ATG) of 517 genes, suggesting that these genes are a target gene of PbAP2-FG2 (S3C Table). Of the 517 target genes, 350 have been functionally annotated on PlasmoDB (https://plasmodb.org). We classified these 350 genes into functional groups to evaluate functional characteristics of the target genes (Fig 3E and S3C Table). The target genes contained some groups that included many genes highly transcribed in female gametocytes, such as “cytoskeleton” and “secretory protein”. The group “cytoskeleton” had some genes encoding inner membrane complex and myosin proteins [35,36], and the group “secretory protein” contained warp and some secreted ookinete protein genes [37,38]. Meanwhile, some other functional groups, such as “DNA replication and repair” and “cell division”, did not seem to be related to female development. To further investigate whether genes of any specific function were enriched in the targets, we performed a gene ontology (GO) analysis. The GO analysis revealed that the target genes were most enriched in the term “DNA replication,” which included putative DNA replication licensing factor genes, DNA polymerase subunit genes and so on, with p-value of 8.0 × 10−4 (Fig 3F). In addition, genes that belong to the GO terms “entry into host cell,” “transmembrane transport,” “DNA metabolic process,” and “microtubule-based process” were also found to be enriched (p-value < 0.01) (Fig 3F).

Next, we evaluated the composition of sex-enriched genes among the targets of PbAP2-FG2. Of the 517 target genes, 90 were female-enriched (Fig 3G and S3C Table). However, in these female-enriched target genes, genes of any specific function did not appear enriched. The targets also contained 101 male-enriched genes (Fig 3G and S3C Table), including most of the genes that were classified into the functional groups “DNA replication and repair” and “cell division”. Because PbAP2-FG2 is a female-specific transcription factor, the considerable number of male-enriched genes in the targets may imply the possible role of PbAP2-FG2 as a transcriptional repressor. Concordantly, when the association between the target genes of PbAP2-FG2 and genes downregulated in pbap2-fg2(-) was assessed, only four target genes were significantly downregulated. Therefore, we considered that the downregulation of genes in pbap2-fg2(-) was not a direct effect of disrupting pbap2-fg2.

Target genes of PbAP2-FG2 were upregulated in pbap2-fg2(-)

To evaluate how the disruption of PbAP2-FG2 affected the transcription of its target genes, we compared the target genes and DEGs identified in the RNA-seq analysis. Intriguingly, the target genes were enriched in the genes significantly upregulated in pbap2-fg2(-) (40 of the 96 upregulated genes, p-value = 6.9 × 10−15 by Fisher’s exact test), while only four targets were included in the significantly downregulated genes, as mentioned above (Fig 4A and S3C Table). In addition, although not more than 2-fold, the other 47 target genes were upregulated, with a p-value adj < 0.05. Furthermore, comparing the log2(fold change) distribution of the target genes with that of the other genes revealed that the target genes tended to be upregulated in pbap2-fg2(-) with a p-value of 8.2 × 10−58 by two-tailed Student’s t-test (Fig 4A). Therefore, we considered that PbAP2-FG2 repressed the transcription of its target genes in female gametocytes. An upregulation pattern of the target genes was also observed in female and male-enriched genes with a p-value of 3.9 × 10−13 and 1.2 × 10−4 by two-tailed Student’s t-test, respectively (Fig 4B and 4C), indicating a role of PbAP2-FG2 in repressing its target genes regardless of their sex-enriched expression property.

Fig 4. Relationship between target genes of PbAP2-FG2 and DEGs in pbap2-fg2(-).

Fig 4

(A) A volcano plot showing DEGs in pbap2-fg2(-) compared to WT. Blue dots represent the target genes of PbAP2-FG2, and a red dot indicates pbap2-fg2. A horizontal line indicates p-value of 0.05, and two vertical lines indicate log2(Fold Change) of -1 and 1. All genes with RPKM ≥ 10 are depicted in the left panel. For the right panel, region from log2(Fold Change) of -2 to 2 and from -log10(p-value) of 0 to 7.5 is magnified. (B) Volcano plots showing DEGs in pbap2-fg2(-) for female and male-enriched genes (the left and right panel, respectively). Blue dots represent the target genes of PbAP2-FG2. A horizontal line indicates a p-value of 0.05, and a vertical line indicates log2(Fold Change) of 0. (C) Distribution of log2(Fold Change) values for female and male-enriched target genes of PbAP2-FG2 (the left and right graph, respectively). A green line indicates log2(Fold Change) of 0. (D) Six-bp DNA motifs enriched within the upstream region (300 to 1200 bp from ATG) of genes upregulated in pbap2-fg2(-). The major and variant motifs are each indicated in different color boxes. The ranks were assigned to all enriched motifs according to their p-values.

Considering the above results, we examined whether the binding motifs of PbAP2-FG2 were enriched in the upstream region (300–1200 bp from ATG) of the upregulated genes compared to that of the other genes. Through this analysis, we found that one of the motifs that belong to the major motifs, GGAGAG, was found to be the most enriched by Fisher’s exact test (p-value = 4.4 × 10−6, Fig 4D). Additionally, two other major motifs (CTCTCT and CTCTCC) and one of the variant motifs 1 and 2 (TTTCTC and AGGAGA, respectively) were also found to be enriched, with a p-value < 0.005 (Fig 4D). These results strongly suggested that the upregulation of genes in pbap2-fg2(-) was primarily a direct effect of its disruption. In addition, this comparison of the differential expression analysis and target analysis indicated the importance of target analysis by ChIP-seq for identifying direct targets of a transcriptional regulator because it is difficult to distinguish direct and indirect effects resulted from a gene knockout by RNA-seq analysis alone.

The binding motifs of PbAP2-FG2 functioned as a cis-acting repressive element

To confirm that PbAP2-FG2 functions as a transcriptional repressor upon binding to the major and variant motifs, we evaluated the effects of disrupting motifs in the upstream region of target genes. First, we generated parasites expressing GFP-fused PbAP2-FG2 by the CRISPR/Cas9 system (PbAP2-FG2::GFPC, S2C Fig) using Cas9-expressing parasites called Pbcas9 [39]. After developing PbAP2-FG2::GFPC, we introduced point mutations in the motif upstream of psh3 (PBANKA_1223500). Parasite-specific helicase 3 (PSH3) is a helicase conserved in apicomplexan parasites, which in P. falciparum is reported to be expressed in trophozoites and schizonts and is essential for asexual stage development [40]. In addition, psh3 is also reported to be essential for asexual development of P. berghei in the PlasmoGEM study [41]. The female and male gametocyte RNA-seq data indicated that psh3 is transcribed in male but not female gametocytes. In the differential expression analysis between WT and pbap2-fg2(-), psh3 was significantly upregulated in pbap2-fg2(-) with log2(fold change) of 2.0, assuring that it is a true target gene of PbAP2-FG2. Using the CRISPR/Cas9 system, we changed the motif in the peak region upstream of psh3 from GGAGAA to atAtAt (Fig 5A).

Fig 5. Disruption of the binding motif of PbAP2-FG2 in the upstream region of psh3.

Fig 5

(A) Genomic sequence around the binding motif of PbAP2-FG2 located upstream of psh3, and Sanger sequence result of the region in motif_mutated parasites. (B) ChIP-qPCR analysis of PbAP2-FG2 at the mutated site upstream of psh3. Grey and white bars indicate %input for motif_WT and motif_mutated, respectively. Error bars indicate the standard error of the mean %input values from three independent experiments. [PSH3: Parasite-Specific Helicase 3, MyoE: Myosin E, DNA Primase: DNA Primase large subunit, TRAP: Thrombospondin-Related Anonymous Protein] (C) RT-qPCR analysis of psh3 in motif_WT and motif_mutated parasites. The relative transcript level of psh3 against p28 and dynein is presented at the left and right, respectively. Error bars indicate the standard error of the mean from three independent experiments.

To evaluate the binding of PbAP2-FG2 at the mutated site, we first performed ChIP coupled with quantitative PCR (ChIP-qPCR) analysis using PbAP2-FG2::GFPC with the wild-type (motif_WT) or mutated (motif_mutated) motif upstream of psh3. In the motif_mutated parasites, the amount of immunoprecipitated DNA fragments relative to input DNA (%input value) was significantly decreased at the mutated site compared to the motif_WT (Fig 5B). In contrast, at the other sites, the upstream region of myoE and the DNA primase large subunit gene as positive controls and trap as a negative control, the %input values were comparable between the motif_WT and motif_mutated parasites (Fig 5B). Together with the ChIP-seq results, these results strongly indicated that the ChIP-seq-identified motifs are the binding motifs of PbAP2-FG2.

Subsequently, using these mutants, we assessed how the binding of PbAP2-FG2 affects downstream transcription by reverse transcription quantitative PCR (RT-qPCR) analysis. Total RNA was harvested from motif_WT and motif_mutated parasites treated with sulfadiazine, and the relative amount of psh3 transcripts to p28 transcripts was analyzed. The results demonstrated that the relative transcript level of psh3 in motif_mutated parasites was more than 2.5-fold higher than in motif_WT (Fig 5C, left graph). Since psh3 is usually expressed in male gametocytes but not in females, we supposed that it is essential to exclude the possibility that the male-to-female ratio affected the result. Accordingly, we evaluated the amount of psh3 transcripts relative to that of a male-specific gene, the dynein heavy chain gene (PBANKA_0416100). The result was comparable to when p28 was used as a control; the relative transcript level of psh3 was more than 3-fold higher in motif_mutated than in motif_WT (Fig 5C, right graph). Collectively, these results indicated that the major motif functions as a cis-regulatory element for repressing downstream genes.

PbAP2-FG2 requires a co-repressor PbAP2R-2 to repress its target genes

We previously identified two putative transcriptional regulator genes, pbap2r-1 (PBANKA_0612400) and pbap2r-2 (PBANKA_1418100), as a target gene of PbAP2-G and PbAP2-FG [20]. Of these, pbap2r-1 functions as a transcriptional activator in zygotes and is renamed pbap2-z [25], but the functional role of pbap2r-2 remains unknown. PbAP2R-2 has an ACDC domain at its C-terminus. A previous study indicated that PfAP2R-2 has two putative AP2 domain [42], one of which is not highly conserved in the Plasmodium species (S3 Fig). The other one had AP2-like structure in both P. berghei and P. falciparum, but only the third beta-sheet and the alpha-helix were highly conserved between the two species. Because three-stranded antiparallel beta-sheets in AP2 are the structure that interacts with DNA [43,44], it was considered that this AP2-like domain is probably not involved in binding to DNA of a specific sequence. PbAP2R-2 is expressed in females, and pbap2r-2 knockout parasites [pbap2r-2(-)] are not able to form banana-shaped ookinetes [20]. Furthermore, in the recent study by Russell et al., pbap2r-2 was identified as one of the genes critical for female differentiation (referred as fd3) [45]. Therefore, we hypothesized that PbAP2R-2 might play an essential role in female development and assessed its function. First, we performed a cross-fertilization assay to confirm the role of PbAP2R-2 in female gametocyte development. When crossed with p47(-), pbap2r-2(-) could not produce normal ookinetes, confirming that development of female gametocytes in pbap2r-2(-) was impaired (Fig 6A). On the other hand, when crossed with p48/45(-), pbap2r-2(-) produced mature ookinetes nearly as many as when p47(-) and p48/45(-) were crossed (Fig 6A). These results revealed that as suggested by the previous studies, PbAP2R-2 is essential for female development. Next, we developed a parasite line expressing GFP-fused PbAP2R-2 using CRISPR/Cas9 (PbAP2R-2::GFPC, S2D Fig). The female gametocytes of PbAP2R-2::GFPC showed nuclear-localized fluorescence, consistent with our previous study; therefore, we performed ChIP-seq analysis at the gametocyte stage. Through this analysis, we obtained 944 highly reproducible peaks (IDR < 0.01) from 1597 and 1459 peaks identified in Experiments 1 and 2, respectively (S4A and S4B Table). Intriguingly, the genome-wide peak patterns for ChIP-seq of PbAP2-FG2 and PbAP2R-2 seemed highly similar (Fig 6B), and more than 70% of the PbAP2-FG2 peaks were overlapped with the PbAP2R-2 peaks (Fig 6C). To evaluate the consistency of the ChIP peak pattern over the whole genome, we assessed the read coverage in the ChIP-seq data of PbAP2-FG2 at the peak summits identified in the ChIP-seq of PbAP2R-2 and vice versa. The results depicted that read coverage for PbAP2-FG2 was enriched at the PbAP2R-2 peaks, and higher fold enrichment of PbAP2R-2 peaks correlated with higher read count detected in the PbAP2-FG2 ChIP-seq (Figs 6D S4). The same was true for the read coverage of the PbAP2R-2 ChIP-seq at the PbAP2-FG2 peaks, indicating that the peak patterns in the ChIP-seq of PbAP2-FG2 and PbAP2R-2 were genome-widely consistent. Consistently, motif enrichment analysis by Fisher’s exact test revealed that the major and two variant motifs were all enriched within 100 bp from the summits of peaks identified in the ChIP-seq of PbAP2R-2 (p-value = 7.3 × 10−151, 2.1 × 10−116 and 2.7 × 10−76 for the major motif, variant motifs 1 and 2, respectively, Fig 6E). Collectively, these results suggested that PbAP2R-2 colocalizes with PbAP2-FG2 on the genome and may cooperatively work with PbAP2-FG2 during female development.

Fig 6. ChIP-seq of PbAP2R-2 and differential expression analysis between WT and pbap2r-2(-).

Fig 6

(A) Cross-fertilization assay among pbap2r-2(-), p48/45(-) and p47(-). The number of normal and abnormal ookinetes are indicated as white and grey bars, respectively. Error bars indicate the standard error of the mean (n = 3). (B) IGV images showing peaks identified in the ChIP-seq analysis of PbAP2-FG2 and PbAP2R-2. Read coverage for the ChIP data is shown. (C) A Venn diagram showing the overlap between peaks detected in the ChIP-seq of PbAP2-FG2 and PbAP2R-2. (D) Heat maps showing coverage in ChIP-seq of PbAP2-FG2 at PbAP2R-2 peaks (left) and coverage in ChIP-seq of PbAP2R-2 at PbAP2-FG2 peaks (right). Peak regions are aligned in ascending order of their fold enrichment value. Graphs on top of the heat maps show the mean coverage of all peak regions. (E) Motifs enriched within 50 bp from peak summits identified in the ChIP-seq of PbAP2R-2. The logos were depicted using WebLogo 3. (F) A volcano plot showing DEGs in pbap2r-2(-) compared to WT. Blue dots represent the target genes of PbAP2-FG2. A horizontal line indicates a p-value of 0.05, and two vertical lines indicate log2(Fold Change) of -1 and 1. (G) Six-bp DNA motifs enriched within upstream region of genes upregulated in pbap2r-2(-). (H) A scatter plot showing relationship between log10(RPKM) in pbap2-fg2(-) and pbap2r-2(-) for each gene. A red line indicates a linear approximation of the plots.

To evaluate the functions of PbAP2R-2 in the expression of its targets, we performed differential expression analysis by comparing WT and pbap2r-2(-). We identified 95 significantly upregulated and 222 significantly downregulated genes (Fig 6F, S5A, S5B, and S5C Table). Among the upregulated genes, 36 targets of PbAP2-FG2 were detected, and overall, the target genes tended to be upregulated in pbap2r-2(-) with a p-value of 1.6 × 10−53 by two-tailed Student’s t-test. This upregulation tendency was also detected within female- and male-enriched genes with a p-value of 5.6 × 10−5 and 4.2 × 10−8, respectively. Moreover, in the upstream region of the genes upregulated in pbap2r-2(-), one of the major motifs, GGAGAG, was most enriched with a p-value of 1.6 × 10−5 by Fisher’s exact test, and some other binding motifs of PbAP2-FG2/PbAP2R-2 were also significantly enriched (Fig 6G). Therefore, we considered that PbAP2R-2 functions as a transcriptional repressive factor in female gametocytes. The upregulated and downregulated genes identified in pbap2-fg2(-) and pbap2r-2(-) presented a significant overlap with a p-value of 3.4 × 10−37 and 4.1 × 10−62 by Fisher’s exact test, respectively. To further evaluate the relationship between pbap2-fg2(-) and pbap2r-2(-) in detail, we plotted RPKM value in pbap2-fg2(-) against that in pbap2r-2(-) for each gene. We then constructed a linear approximation of the plots, which depicted a line with a slope of 1.07 and a mean square correlation coefficient of 0.92 (Fig 6H). This result indicated that the change in the expression level for each gene was mostly comparable between pbap2-fg2(-) and pbap2r-2(-); hence, disruption of pbap2-fg2 and pbap2r-2 had highly similar effects on the female transcriptome. Therefore, we hypothesized that PbAP2R-2 may function as an essential co-repressor of PbAP2-FG2 in female gametocytes.

PbAP2-FG2 and PbAP2R-2 form a transcriptional repressor complex

To investigate whether PbAP2-FG2 and PbAP2R-2 function as a transcriptional repressor complex on the genome, we performed rapid immunoprecipitation mass spectrometry of endogenous proteins (RIME) [46,47]. RIME is a method combining ChIP and MS to identify proteins that form a complex with a target protein of ChIP. We performed ChIP experiments with PbAP2-FG2::GFP and WT, as a negative control, by the same method as ChIP-seq analysis and then on-beads digested the immunoprecipitated proteins with trypsin. The released peptides were then analyzed by liquid chromatography-tandem MS (LC-MS/MS) analysis. Four biologically independent experiments were performed for each parasite line, and immunoprecipitated proteins were identified. The analysis detected 482 and 463 proteins for PbAP2-FG2::GFP and WT, respectively (Fig 7A and S6 Table). As a possible interaction partner of PbAP2-FG2, proteins that were unique or more than fivefold enriched in PbAP2-FG2 compared to WT were searched according to the criteria used in the previous RIME study [47], and 40 unique and 12 fivefold enriched proteins were identified (Fig 7A). Of the proteins that were unique for PbAP2-FG2::GFP, PbAP2-FG2 was detected with the highest quantitative value as expected (Fig 7B). The unique protein with the second highest value was PbAP2R-2. This result revealed that PbAP2-FG2 and PbAP2R-2 actually form a complex on the genome. The third unique protein was PbAP2-O2 (encoded by PBANKA_1231600), an AP2 transcription factor essential for ookinete development [28]. A study in P. yoelii reported that PyAP2-O2 was expressed at several stages including gametocyte [29]. Therefore, PbAP2-O2 might function as a transcriptional repressor in gametocyte together with PbAP2-FG2.

Fig 7. RIME using PbAP2-FG2::GFP.

Fig 7

(A) A Venn diagram showing the overlap between proteins detected in the RIME using PbA2-FG2 and WT. Number of proteins that were unique and more than fivefold enriched with p-value < 0.05 in PbAP2-FG2::GFP compared to WT was indicated in a yellow and green circle, respectively. (B) A list of possible interaction partners of PbAP2-FG2 identified in the RIME. Those with average quantitative value > 5 for PbAP2-FG2::GFP (average QV) are shown.

In the proteins identified as an interaction partner of PbAP2-FG2, we also found PbMORC (encoded by PBANKA_1331400) with average quantitative value of 37.8, which was the highest in the fivefold enriched proteins, and log2(fold enrichment) of 3.64 (Fig 7B). Microrchidia (MORC) is a nuclear protein that contains a GHKL (gyrase, Hsp90, histidine kinase, and MutL)-ATPase domain [48]. The MORC family proteins in animals and plants are involved in chromatin condensation and remodeling [49,50]. In an apicomplexan parasite Toxoplasma gondii, MORC interacts with AP2 transcription factors and plays a role in transcriptional repression of genes downstream of its binding sites [51]. Therefore, PbMORC might be recruited by PbAP2-FG2 and play a role in the transcriptional repression through remodeling of chromatins.

PbAP2-FG2 and PbAP2R-2 repress the target genes of AP2-G

In previous studies, we observed the expression patterns of PbAP2-G and PbAP2-FG and found that the expression of PbAP2-FG begins during the period when the expression of PbAP2-G decreases [20,23]. This result indicates that a major transcriptional activator switches from PbAP2-G to PbAP2-FG as early gametocytes develop into females. Considering this scenario, we hypothesized that the PbAP2-FG2-PbAP2R-2 complex might support stage conversion from early gametocytes to female gametocytes by repressing early gametocyte genes activated by PbAP2-G. To address this possibility, we assessed whether the target genes of PbAP2-FG2 include those of PbAP2-G. Consistent with our hypothesis, we found a significant overlap (105 genes) between the target genes of PbAP2-G and PbAP2-FG2 with a p-value of 4.5 × 10−7 by Fisher’s exact test (Fig 8A). In addition, the distribution of log2(fold change) values for these common target genes in the differential expression analysis between WT and pbap2-fg2(-) was higher than that for the non-target genes with p-value = 1.4 × 10−13 by two-tailed Student’s t-test, suggesting that these genes are actually repressed in female gametocytes.

Fig 8. Relationship between target genes of PbAP2-G and PbAP2-FG2.

Fig 8

(A) A Venn diagram showing the overlap between target genes of PbAP2-G and PbAP2-FG2. (B) IGV images showing representative peaks of ChIP-seq for PbAP2-G, PbAP2-FG2, and PbAP2R-2 on the upstream of target genes common for PbAP2-G and PbAP2-FG2. Read coverage for the ChIP data is shown. Positions of the PbAP2-G and PbAP2-FG2 binding motifs are indicated in green and blue, respectively.

Most of these common target genes between PbAP2-FG2 and PbAP2-G have not been assessed for their function during Plasmodium gametocyte development. In the common target genes, we found T-complex protein 1 subunit (TCP-1) genes (Fig 8B, S3C Table). Plasmodium parasites possess eight TCP-1 genes, all of which are a target of PbAP2-G. Although the target genes of PbAP2-FG2 contained only four of these genes, peaks with fold enrichment > 2.5 were found upstream of three other TCP-1 genes in the ChIP-seq of PbAP2-FG2. TCP-1s comprise a type 2 chaperonin, tailless complex polypeptide 1 ring complex (TRiC), which has been indicated to play essential roles in folding diverse polypeptides, including actin and tubulin [5255]. In P. falciparum, it has been reported that TRiC is essential for the asexual blood-stage development [56,57]. Thus, this complex is presumed to be widely required in the Plasmodium life cycle, which, for P. berghei, includes early gametocyte development, but not during female development. Notably, the common target genes of PbAP2-FG2 and PbAP2-G also included the actin I gene, alpha-tubulin 2 gene, and a putative tubulin beta chain gene (S3C Table). These findings suggested the possibility that for female gametocytes of P. berghei, cytoskeletal development is mostly completed during early gametocyte development.

The function of PyAP2-O3 is highly similar to that of PbAP2-FG2

The present study revealed that transcriptional repression by the PbAP2-FG2-PbAP2R-2 complex is essential for regulating the female transcriptome. Recently, Li et al. reported that PyAP2-O3 is also a transcriptional repressor in female gametocytes, but their other conclusions differed from what we have revealed here [30]. The first is regarding the role of PyAP2-O3, which was primarily based on their RNA-seq data. The authors performed RNA-seq analyses with female gametocytes collected by cell sorting using a female-specific fluorescent marker and compared the female transcriptome of P. yoelii 17XNL (PyWT) and pyap2-o3-null parasites. The analysis detected significant upregulation of 1141 genes in pyap2-o3-null parasites, more than half of which were specifically or preferentially expressed in males. Accordingly, the authors concluded that PyAP2-O3 globally represses male genes to safeguard the female transcriptome. This statement differed from our conclusion that PbAP2-FG2 and PbAP2R-2 repress various genes, which include some early gametocyte and female genes. The second is the conclusions derived from their ChIP-seq analysis of PyAP2-O3. For example, the binding motif of PyAP2-O3 predicted in their study was considerably different from that of PbAP2-FG2 identified in this study; the binding motif of PyAP2-O3 was predicted to be TRTRTGCA. As P. berghei and P. yoelii are phylogenetically very close, such discrepancies in the roles of orthologous genes seems unlikely. Therefore, we reassessed the ChIP-seq and RNA-seq data deposited from the PyAP2-O3 study to clarify the inconsistency between the two studies.

First, we reanalyzed the ChIP-seq data for PyAP2-O3. We mapped their sequence data onto the P. yoelii reference genome (downloaded from PlasmoDB) using bowtie2 and removed reads aligned onto more than two sites of the genome, as in our ChIP-seq analysis. Then, we called peaks using macs2 with the criteria used by Li et al. (fold enrichment > 2.0 and p-value < 1.0 × 10−5) and obtained 1309 peaks that were common in the duplicates (S7A and S7B Table). Within 100 bp from these peak summits, we found enrichment of TRTRTGCA with a p-value of 7.0 × 10−52 by Fisher’s exact test (Fig 9A). On the other hand, the major motif identified in our study, RGAGAR, was also enriched with a p-value of 2.2 × 10−52, comparable to that of the TRTRTGCA motif (Fig 9A). In addition, the variant motifs 1 and 2 were also enriched with p-values of 4.0 × 10−48 and 9.4 × 10−26, respectively. We further performed IDR1D analysis and found that only 122 peaks had an IDR < 0.01, which implies the low reproducibility of the ChIP-seq data (Fig 9B and 9C, S7A and S7B Table). Within these highly reliable peaks (IDR < 0.01), the RGAGAR motif was much more enriched (p-value = 2.1 × 10−12) than the TRTRTGCA motif (p-value = 1.1 × 10−4, Fig 9A). Therefore, in contrast to the major motif, the TRTRTGCA motif appeared to have been mainly derived from unreliable peaks (IDR ≥ 0.01).

Fig 9. Reassessment of the ChIP-seq and RNA-seq data for the study of PyAP2-O3.

Fig 9

(A) Enrichment of RGAGAR and TRTRTGCA motif in the peak region identified by ChIP-seq analysis of PyAP2-O3. The logos were depicted by WebLogo 3. (B) IDR1D analysis between the ChIP-seq experiment 1 and 2 of PyAP2-O3. Peaks with IDR < 0.01 are indicated as blue dots. (C) Ratio of peaks with IDR < 0.01 in all peaks identified in ChIP-seq of PbAP2-FG2 and PyAP2-O3. (D) A Venn diagram showing number of genes common in the targets of PbAP2-FG2 and PyAP2-O3. (E) Classification of the target genes of PyAP2-O3 into sexual stage-enriched gene sets. The number of target genes common for PyAP2-O3 and PbAP2-FG2 are indicated with a blue star for each set. (F) A volcano plot showing DEGs in pyap2-o3-null parasite compared to PyWT. Blue dots represent orthologs of the target genes of PbAP2-FG2, and a red dot indicates pyap2-o3. A horizontal line indicates a p-value of 0.05, and two vertical lines indicate log2(Fold Change) of -1 and 1. (G) Six-bp DNA motifs enriched within the upstream region (300 to 1200 bp from ATG) of genes upregulated in pyap2-o3-null parasite. (H) Classification of significantly upregulated and downregulated genes into sexual stage-enriched gene sets (top and bottom, respectively). (I) Histograms showing distribution of log2(Fold Change) values in pyap2-o3-null parasite for female and male-enriched genes (top and bottom, respectively). Green lines indicate log2(Fold Change) of -1 and 1.

We further predicted the target genes of PyAP2-O3 from the 1309 peaks common in duplicates. The analysis identified 781 target genes, 271 of which were included in the target genes of PbAP2-FG2 as an orthologous gene (Fig 9D and S7C Table). Although there was a significant overlap between the two target sets (p-value = 2.4 × 10−85 by Fisher’s exact test), approximately 70% of the PyAP2-O3 targets were not included in the PbAP2-FG2 targets. Given the low reproducibility of the ChIP-seq data, we supposed that some targets might have been derived from unreliable peaks and hence not a true target. We further assessed the sex-enriched expression of the target genes of PyAP2-O3 considering orthologous genes of the female- and male-enriched genes defined above as sex-enriched genes for P. yoelii. We found that the target genes contained only 94 male-enriched genes, which contradicted the conclusion that PyAP2-O3 globally represses male genes (Fig 9E). Moreover, the targets also contained 128 female-enriched genes, 58 orthologous genes of which were included in the targets of PbAP2-FG2, implying that, similar to PbAP2-FG2, PyAP2-O3 also plays a role in repressing a substantial number of female genes.

Next, we reassessed the RNA-seq data of the PyWT and pyap2-o3-null parasites. In their RNA-seq analysis, no threshold of fragments per kilobase of transcript per million mapped reads (FPKM) was set to exclude genes with low expression levels. Such an analytical process may detect DEGs derived from artificial variances and yield large DEG lists with large variance. In the analysis performed by Li et al., more than one-third of the upregulated genes (445/1,141 genes) had FPKM < 10 for pyap2-o3-null parasites. Although these genes with low scores could demonstrate high-fold enrichment, the actual upregulation was low and could be false positives; thus, it is not appropriate to conclude the function of PyAP2-O3 from such analyses. To obtain more robust results, we analyzed the RNA-seq data according to the analytical process performed in this study, setting a minimum FPKM threshold of ≥ 10 (S8A Table). This analysis identified 418 significantly upregulated and 230 significantly downregulated genes (Fig 9F, S8B and S8C Table). In the upregulated genes, the target genes of PyAP2-O3, which we obtained above, were enriched with a p-value of 7.7 × 10−9 by Fisher’s exact test. On the other hand, the orthologous genes of PbAP2-FG2 targets were more enriched (p-value = 3.4 × 10−17) (Fig 9F), again suggesting that the target list of PyAP2-O3 may contain several pseudo-targets. We next investigated whether the binding motifs of AP2-FG2 identified in this study and that by Li et al. were enriched in the upstream of these upregulated genes. Consistent with our results, the major motif and two variant motifs were highly enriched in the upstream region compared to that of the other genes (Fig 9G). In fact, all 6-bp motifs that belonged to the major motif, variant motif 1 or 2 were detected in the 20 most enriched motifs, suggesting that these motifs functioned as a cis-acting repressive element in P. yoelii as well. However, when enrichment of any 8-bp motif was searched in the same region, the TRTRTGCA motif was not found to be significantly enriched; that is, the most enriched motif that corresponds to TRTRTGCA was detected as the 776th enriched motif with a p-value of 0.019 (Fig 9G), indicating that this motif is not related to the upregulation of genes detected in pyap2-o3-null parasites.

According to our analysis, the majority of male-enriched genes were not significantly upregulated in pyap2-o3-null parasites; the upregulated genes only contained 103 male-enriched genes (Fig 9H and 9I). Moreover, nearly half of the upregulated male-enriched genes were not a target gene of PbAP2-FG2 or PyAP2-O3. Therefore, based on our analysis of their RNA-seq data, it seemed not appropriate to conclude that PyAP2-O3 globally represses male genes. For female-enriched genes, only nine genes were significantly upregulated, and most genes tended to be downregulated, as discussed by Li et al. (Fig 9H and 9I). The downregulation of female genes was considered to be caused by impairment of the female transcriptome upon disruption of pyap2-o3. Therefore, we hypothesized that such an effect caused by the disruption of pyap2-o3 might have masked the upregulation of female-enriched target genes in the RNA-seq analysis. In fact, despite the overall downregulation of female-enriched genes, most of the female-enriched target genes were not downregulated; there was a significant difference in the distribution of log2(fold change) between the female-enriched target genes and the other female-enriched genes (p-value = 1.7 × 10−4 by two-tailed Student’s t-test) (Fig 9I). These results suggested that consistent with the results for PbAP2-FG2, PyAP2-O3 also targets female genes and represses their expression. Collectively, our analysis revealed that PbAP2-FG2 and PyAP2-O3 both repress not only male genes but also a wide-variety of genes to support female differentiation.

Discussion

This study highlights how PbAP2-FG2 and PbAP2R-2 function cooperatively as a transcriptional repressor complex during female development. Their target genes contained variable genes regarding functional annotation and expression patterns, which indicated that this repressor complex might play distinct roles for each group of the target genes during female development. As suggested by the comparison between the target genes of PbAP2-FG2 and PbAP2-G, one of the roles of the repressor complex could be repression of the early gametocyte genes to promote female differentiation. During early gametocyte development, the female transcriptional activator PbAP2-FG begins to be expressed as PbAP2-G expression decreases [20,23]. Thus, we considered that the repression of PbAP2-G targets was vital for completing this switch of major transcriptional activators. On the other hand, the target genes of PbAP2-FG2 also included a significant number of female-enriched genes. This result appeared unreasonable because it would mean that such genes are activated and repressed during the same period. Nevertheless, the differential expression analysis suggested that these female-enriched target genes were indeed repressed by AP2-FG2/AP2-O3; for both P. berghei and P. yoelii, expression of the female-enriched target genes was not downregulated in ap2-fg2/ap2-o3-knockout parasites despite that the other female-enriched genes were predominantly downregulated. This observation implied that transcriptional activators alone could not precisely control gene expression for female gametocyte development, thereby requiring repressors for its modulation. Another possible role of the PbAP2-FG2-PbAP2R-2 repressor complex was the repression of male genes, as suggested in the study of PyAP2-O3 [30]. However, the results in this study did not corroborate their conclusion that the global repression of male genes by PyAP2-O3 was required for balancing the female-specific transcriptome. The ChIP-seq analyses of AP2-FG2/AP2-O3 in P. berghei and P. yoelii both showed that the target genes of AP2-FG2/AP2-O3 contained only a subset of male-enriched genes. These included only a limited number of genes related to the major characteristic features of male gametocytes/microgametes, such as flagella formation and DNA replication. Moreover, although a significant number of male-enriched genes were upregulated in pyap2-o3-null parasites, nearly half of them were not a target gene of PyAP2-O3. From this result, we speculated that the target genes of AP2-FG2/AP2-O3 could contain male-specific transcriptional regulator genes, especially activators; the non-target genes were indirectly upregulated in pyap2-o3-null parasites, as the expression of male transcriptional activators was released from the repression by PyAP2-O3. Therefore, we proposed that the repressor complex repressed a subset of male genes that include those essential for regulating male differentiation. Very recently, Russel et al. identified five genes that are essential for male differentiation [45], but these genes were not included in the target gene list of PbAP2-FG2 and PbAP2R-2. Collectively, we concluded that the PbAP2-FG2-PbAP2R-2 complex could play three roles: promoting female differentiation by repressing early gametocyte genes, modulating the expression level of female genes, and suppressing male differentiation.

In this study, we performed RIME on the PbAP2-FG2::GFP parasite and identified PbMORC as one of the proteins that form a complex with PbAP2-FG2. Previous studies in apicomplexan parasites reported that MORC interacts with diverse AP2 transcription factors [22,51,58]. In P. falciparum, PfMORC was found to interact with an AP2 transcriptional repressor, PfAP2-G2 [22]. In addition, a study in Toxoplasma gondii reported that MORC forms a complex with several AP2 transcription factors and plays a role in transcriptional repression [51]. These studies were consistent with our result that PbMORC is an interaction partner of a transcriptional repressor PbAP2-FG2 and hence indicate possibility that in Apicomplexa, transcriptional repression occurs in a similar mechanism involving MORC and AP2 transcription factors. The study in Toxoplasma reported that MORC is also involved in recruitment of histone deacetylase 3, which plays a role in gene repression through reversing histone acetylation, a mark for transcriptional activation [59,60]. However, any histone deacetylase was not found as an interaction partner of PbAP2-FG2 in our RIME or that of PfAP2-G2 by IP-MS [22]. Given that MORC has a conserved ATPase domain and is known to play a role in chromatin regulation in animals and plants, a major function of MORC in Plasmodium might be related to ATPase activity and chromatin regulation but not recruitment of other factors, such as histone deacetylase.

Our result demonstrated that the RIME method is a powerful tool for identifying proteins in a transcriptional complex in Plasmodium. By cross-linking complexes in their native state using formaldehyde, transient protein-protein interactions can be preserved in the RIME [46]. Furthermore, the formaldehyde fixation allows stringent washing regimes, which reduce background nonspecific noise in the MS analysis [46]. Although recent studies in AP2 and other sequence specific transcription factors have advanced our knowledge on stage specific transcriptional regulation of Plasmodium, the mechanisms of transcriptional activation/repression by the transcription factors and their cofactors have been poorly investigated. We believe that further use of the RIME method on Plasmodium transcription factors would identify components of transcriptional complexes and considerably help us understand detailed mechanisms of transcriptional regulation in the parasites.

In P. berghei, another AP2 transcriptional repressor gene, pbap2-g2, is activated by PbAP2-G [20], a transcriptional activator that triggers sexual differentiation, similar to pbap2-fg2, which is activated by the female transcriptional activator PbAP2-FG. PbAP2-G2 induces global gene repression, and disruption of this gene results in the aberrant development of gametocytes. The target genes of PbAP2-G2 include various genes, like those of PbAP2-FG2 do, suggesting that PbAP2-G2 also plays multiple roles, possibly repressing trophozoite genes, modulating the expression of early gametocyte genes, and suppressing asexual fate. Studies of Plasmodium AP2-family proteins have shown that transcriptional regulation by AP2 transcription factors is remarkably simple; one transcription factor comprehensively activates certain stage-specific genes [20,23,25,61,62]. On the other hand, recent studies of repressors have indicated that to regulate stage conversion, such as sexual differentiation and sex determination, transcriptional repressors also play an essential role [21,22,30,63]. In addition, studies in another apicomplexan parasite, Toxoplasma, have reported an essential role for transcriptional repressors in conversion from tachyzoite to bradyzoite [64,65]. Therefore, it was suggested that optimal transcriptional regulation could not be controlled by transcriptional activators alone during stage conversion in Apicomplexa. This observation suggested the possibility of finding transcriptional repressors during the other stage conversion processes as well, and investigating these factors would help us understand the mechanisms promoting the life cycle of Plasmodium.

Materials and methods

Ethics statement

All experiments in this study were performed following the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health to minimize animal suffering and were approved by the Animal Research Ethics Committee of Mie University, Mie, Japan (permit number 23–29).

Parasite preparation

pbap2-fg2(-), PbAP2-FG2::GFP, and pbap2r-2(-) were derived from the P. berghei ANKA strain. The other transgenic parasites were derived from Pbcas9 [39]. Parasites were inoculated in Balb/c or ddY mice. Ookinete cultures were performed at 20°C using RPMI1640 medium, pH 8.0, supplemented with fetal calf serum and penicillin/streptomycin at final concentrations of 20% and 100 U/mL, respectively. Cross-fertilization assays were conducted as previously described [66].

Generation of mutant parasites

The DNA constructs for tagging PbAP2-FG2 with GFP and knocking out pbap2-fg2 were prepared as previously reported [66,67]. Briefly, for gfp-tagging, two homologous regions were cloned into the gfp-fusion vector to fuse pbap2-fg2 in-frame with gfp. The gfp-fusion vector was previously generated on the backbone of pBluescript SK (+) (Stratagene, La Jolla, CA, USA). In this vector, 3’ untranslated region (UTR) of hsp70 and a hdhfr expression cassette (under the control of pbef-1α promoter and 3’ UTR of pbdhfr) were located 3’-side of the gfp gene in this order. Of the two homologous regions, one was cloned to 5’-side of the gfp using XhoI/NheI site, and the other was cloned to 3’-side of the hdhfr cassette using BamHI/NotI site (S2A Fig). The plasmid was linearized by XhoI and NotI digestion before use in transfection experiments. To knock out pbap2-fg2, the targeting construct was prepared using overlap PCR. The construct had two homologous regions around the pbap2-fg2 locus flanking a hdhfr expression cassette, which was derived from the gfp-fusion vector described above. pbap2r-2(-) was generated in a previous study [20].

The other transgenic parasites were generated by the CRISPR/Cas9 system using the parasites expressing Cas9 [39]. The Cas9-expressing parasite Pbcas9 has a cas9 cassette at the p230p locus. The hsp70 promoter controls the expression of Cas9, and Pbcas9 constitutively expresses Cas9 throughout the asexual blood cycle. Donor DNA for transfection was constructed by overlap PCR, cloned into pBluescript KS (+) using the XhoI and BamHI sites by In-Fusion cloning, and then amplified by PCR from the constructed plasmid. sgRNA vectors were constructed as previously described [39]. Target sites of sgRNA were designed using the online tool CHOPCHOP (https://chopchop.cbu.uib.no/).

Transfection was performed using the Amaxa Basic Parasite Nucleofector Kit 2 (LONZA). All transfectants were selected by treating mice with 70 μg/mL pyrimethamine in their drinking water. Recombination was confirmed by PCR and, for the motif_mutated parasite, Sanger sequencing. The PCRs were performed using PrimeSTAR GXL DNA Polymerase (Takara). PCR products were amplified for 30 cycles, and annealing temperature was set at 55°C. Clonal parasites were obtained by limiting dilution. All primers used in this study are listed in S9 Table (No. 1–48).

Fluorescence analysis

Expression of GFP-fused PbAP2-FG2 was analyzed by live fluorescence microscopy. Cells were stained with 1 ng/mL Hoechst 33342 for 10 min. Images were taken by Olympus BX51 microscope with Olympus DP74 camera.

RNA-seq and sequence data analysis

The total RNA was extracted from parasites enriched with gametocytes by treating infected mice with 10 mg/L sulfadiazine in their drinking water for two days, using the Isogen II reagent (Nippon gene). Briefly, whole blood was withdrawn from infected mice and passed through the Plasmodipur filter to remove white blood cells, and the red blood cells (RBCs) were lysed in an ice-cold 1.5 M NH4Cl solution. After the lysis, the cells were subjected to Isogen II (NIPPON GENE), and the total RNA was extracted according to the manufacturer’s instructions. RNA-seq libraries were prepared from the total RNA using the KAPA mRNA HyperPrep Kit (Kapa Biosystems) and sequenced using Illumina NextSeq. Three biologically independent experiments were conducted for each parasite line. The obtained sequence data were mapped onto the reference genome sequence version 3 of P. berghei, downloaded from PlasmoDB 46, using HISAT2, setting the parameter for maximum intron length to 1000. The mapping data for each sample were analyzed using featureCounts and compared using DESeq2. Genes in the subtelomeric regions were removed from the differential expression analysis. The parameters for all programs were set as the default unless otherwise specified. For comparison between results for P. berghei in this study and P. yoelii in the previous study, 4426 orthologues were considered according to the orthology and synteny data in PlasmoDB.

ChIP-seq and sequencing data analysis

Whole blood was withdrawn from the infected mice treated with sulfadiazine and passed through the Plasmodipur filter to remove white blood cells. The blood was diluted in a complete medium (RPMI1640 supplemented with 20% fetal calf serum) and immediately fixed with 1% formalin at 30°C. After fixing, RBCs were lysed in ice-cold 1.5 M NH4Cl solution. This step was performed several times until the supernatant became clear, and the cells were lysed in SDS lysis buffer (50 mM Tris-HCl, 1% SDS, 10 mM EDTA). The samples were sonicated at 4°C using Bioruptor (Cosmo Bio) for 20 cycles of 30 sec on/30 sec off to shear the chromatin. Input samples were collected at this point from the sonicated cell lysate. For IP samples, chromatins were immunoprecipitated with anti-GFP polyclonal antibodies (5 mg/mL; Abcam, ab290), which were bound to Protein A Magnetic Beads (Invitrogen) before the ChIP step (2 μL of antibody was mixed with 20 μL of beads and incubated for > 1 h). After 12 h of incubation at 4°C, the beads were washed with low-salt buffer (20mM Tris-HCl, 0.1% SDS, 1% Triton X-100, 2 mM EDTA, 150 mM NaCl) for five times and high-salt buffer (20mM Tris-HCl, 0.1% SDS, 1% Triton X-100, 2 mM EDTA, 500 mM NaCl) for three times. Immunoprecipitated chromatin was then eluted in elution buffer (10mM Tris-HCl, 1% SDS, 5 mM EDTA, 300 mM NaCl), heated at 65°C for 8 h, and processed with RNase H for 1 h and proteinase K for 2 h. DNA fragments were purified from the immunoprecipitated chromatin by phenol/chloroform extraction and ethanol precipitation and used for library construction. Libraries for NGS were prepared using the KAPA HyperPrep Kit (Kapa Biosystems) according to the manufacturer’s instructions and sequenced using Illumina NextSeq. Two biologically independent experiments were performed for each sample and used for the following analysis.

The obtained sequence data were mapped onto the reference genome sequence version 3 of P. berghei, downloaded from PlasmoDB 46, using Bowtie 2. Reads aligned onto more than two sites were removed from the mapping data. Using the trimmed mapping data, peaks were called with macs2 callpeak with fold enrichment > 3.0 and q-value < 0.01. To identify reliable peaks, the data obtained from two biologically independent experiments were compared using IDR1D analysis (https://idr2d.mit.edu/) setting max gap to 100. Briefly, the peaks were ranked for each replicate according to their p-value. The peaks from each replicate were then compared and scored based on their respective ranks. Highly reproducible peaks were defined as those with an IDR score < 0.01. Binding motifs were predicted by analyzing the enrichment of motifs within 50 bp of peak summits using Fisher’s exact test (the method was previously described in detail [61]). Genes with peaks within upstream of 1200 bp from ATG were identified as target genes. The parameters for all programs were set as the default unless otherwise specified.

ChIP-qPCR and RT-qPCR for reporter experiments

ChIP experiments were performed as described for ChIP-seq analysis. IP and input samples were each prepared from 200 μL and 10 μL of sonicated cell lysate, respectively. Quantification of DNA fragments of interest was performed by real-time qPCR using TB Green Fast qPCR Mix (Takara) and Thermal Cycler Dice Real Time System II (Takara). Amplification cycles were performed for 40 cycles, and cycle threshold (Ct) was detected between 20 to 35 cycles. From the Ct values, %input values were calculated as 2^(CtIP-Ctinput) × (1/20) × 100. Three biologically independent experiments were performed and used for the analysis.

For RT-qPCR analyses, cDNA was synthesized from 1 μg of total RNA, extracted as described for RNA-seq analysis, using the PrimeScript RT reagent Kit with gDNA Eraser (Takara), and 1/200 of the synthesized cDNA was used for qPCR. Real-time qPCR experiments were performed as described above. All primers used are listed in S9 Table (No. 49–62).

RIME

RIME was performed according to the previous studies [46,47] with some modifications. ChIP was performed as described for ChIP-seq analysis until washing beads with high salt buffer. After the wash, beads were washed twice with 100 mM ammonium hydrogen carbonate (AMBIC) solution. The bead-bound proteins were digested with 10 μl of trypsin (Promega) in 100 mM AMBIC at an enzyme-to-protein ratio of 1:100 (wt/wt) for overnight at 37°C. After the overnight digest, additional 10 μl of trypsin was added, and the beads were further incubated for 4 h at 37°C. The supernatant, which contains the digested peptides, was added to 100% formic acid, resulting in the final concentration of formic acid 5% (vol/vol). The digested peptides were purified with C18 tip (GL-Science, Tokyo, Japan) and then subjected to nanocapillary reversed-phase LC-MS/MS analysis using a C18 column (12 cm × 75 μm, 1.9 μm, Nikkyo technos, Tokyo, Japan) on a nanoLC system (Bruker Daltoniks, Bremen, Germany) connected to a timsTOF Pro mass spectrometer (Bruker Daltoniks) and a modified nano-electrospray ion source (CaptiveSpray; Bruker Daltoniks). The mobile phase consisted of water containing 0.1% formic acid (solvent A) and acetonitrile containing 0.1% formic acid (solvent B). Linear gradient elution was carried out from 2% to 35% solvent B for 20 min at a flow rate of 250 nL/min. The ion spray voltage was set at 1.6 kV in the positive ion mode. Ions were collected in the trapped ion mobility spectrometry (TIMS) device over 100 ms and MS and MS/MS data were acquired over an m/z range of 100–2,000. During the collection of MS/MS data, the TIMS cycle was adjusted to 0.53 s and included 1 MS plus 4 parallel accumulation serial fragmentation (PASEF)-MS/MS scans, each containing on average 12 MS/MS spectra (>100 Hz) [68,69], and nitrogen gas was used as collision gas.

Data analysis for RIME

The MS/MS data obtained by RIME was processed using DataAnalysis version 5.2 (Bruker Daltoniks), and proteins were identified using MASCOT version 2.7.0 (Matrix Science, London, UK) against the Uniprot_Plasmodium_berghei_ANKA_strain database (4,948 sequences; 3,412,795 residues). Protease specificity was set for trypsin (C-term, KR; Restrict, P; Independent, no; Semispecific, no; two missed and/or nonspecific cleavages permitted). Variable modifications considered were N-terminal Gln to pyro-Glu, and oxidation of methionine. The mass tolerance for precursor ions was ±15 ppm. The mass tolerance for fragment ions was ±0.05 Da. The threshold score/expectation value for accepting individual spectra was p < 0.05. Quantitative value and fold enrichment were calculated by Scaffold5 version5.1.2 (Proteome Software, Portland, OR, USA) [70] and Microsoft Excel, respectively, for MS/MS-based proteomic studies. Proteins that were unique or more than fivefold enriched with p-value < 0.05 by two-tailed Student’s t-test in PbAP2-FG2 compared to WT were identified as a possible interaction partner of PbAP2-FG2.

Supporting information

S1 Fig. An Integrative Genomics Viewer (IGV) image showing peaks identified in ChIP-seq analysis of PbAP2-G and PbAP2-FG in the upstream region of pbap2-fg2.

The grey bar indicates the gene body of pbap2-fg2.

(TIF)

S2 Fig. Genotyping of transgenic parasites developed in this study.

(A) PbAP2-FG2::GFP. (B) pbap2-fg2(-). (C) PbAP2-FG2::GFPC. (D) PbAP2R-2::GFPC.

(TIF)

S3 Fig. Alignment of amino acid sequences for previously reported putative AP2 domains for PbAP2R-2 and PfAP2R-2.

The sequences were aligned using the ClustalW program in Mega X. Asterisks indicate amino acids conserved between the two species.

(TIF)

S4 Fig. Heat maps showing coverage in both IP and input samples for ChIP-seq of PbAP2-FG2 at PbAP2R-2 peaks (left) and for ChIP-seq of PbAP2R-2 at PbAP2-FG2 peaks (right).

Peak regions are aligned in ascending order of their fold enrichment value.

(TIF)

S1 Table. List of differentially expressed genes in pbap2-fg2(-).

(A) RPKM values in each data. (B) Significantly downregulated genes. (C) Significantly upregulated genes.

(XLSX)

S2 Table. List of sex-enriched genes.

(A) Female-enriched genes. (B) Male-enriched genes.

(XLSX)

S3 Table. List of peaks and target genes identified in the ChIP-seq experiments of PbAP2-FG2.

(A) Peaks in Experiment 1. (B) Peaks in Experiment 2. (C) Target genes.

(XLSX)

S4 Table. List of peaks identified in the ChIP-seq experiments of PbAP2R-2.

(A) Peaks in Experiment 1. (B) Peaks in Experiment 2.

(XLSX)

S5 Table. List of differentially expressed genes in pbap2r-2(-).

(A) RPKM values in each data. (B) Significantly downregulated genes. (C) Significantly upregulated genes.

(XLSX)

S6 Table. List of proteins identified in the RIME using PbAP2-FG2::GFP and WT.

(XLSX)

S7 Table. List of peaks and target genes identified in the ChIP-seq experiments of PyAP2-O3.

(A) Peaks in Experiment 1. (B) Peaks in Experiment 2. (C) Target genes.

(XLSX)

S8 Table. List of differentially expressed genes in pyap2-o3-null parasite.

(A) FPKM values in each data. (B) Significantly downregulated genes. (C) Significantly upregulated genes.

(XLSX)

S9 Table. List of primers used in this study.

(XLSX)

Data Availability

The authors confirm that all data underlying the findings are fully available without restriction. All fastq files for ChIP-seq and RNA-seq experiments are available from the GEO database (accession numbers GSE198588, GSE213776).

Funding Statement

This work was supported by the Japan Society for the Promotion of Science (17H01542 to YM; 20K07462 to IK; 21K06986 to TN). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.World Health Organization. World Malaria Report. World Health. 2021. ISBN 978 92 4 1564403
  • 2.Bousema T, Drakeley C. Epidemiology and infectivity of Plasmodium falciparum and Plasmodium vivax gametocytes in relation to malaria control and elimination. Clinical Microbiology Reviews. American Society for Microbiology 1752 N St., N.W., Washington, DC; 2011. pp. 377–410. doi: 10.1128/CMR.00051-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Guttery DS, Holder AA, Tewari R. Sexual development in plasmodium: Lessons from functional analyses. PLoS Pathog. 2012;8: e1002404. doi: 10.1371/journal.ppat.1002404 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Baker DA. Malaria gametocytogenesis. Molecular and Biochemical Parasitology. Elsevier; 2010. pp. 57–65. doi: 10.1016/j.molbiopara.2010.03.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Josling GA, Llinás M. Sexual development in Plasmodium parasites: Knowing when it’s time to commit. Nature Reviews Microbiology. Nature Publishing Group; 2015. pp. 573–587. doi: 10.1038/nrmicro3519 [DOI] [PubMed] [Google Scholar]
  • 6.Shahabuddin M. Plasmodium ookinete development in the mosquito midgut: A case of reciprocal manipulation. Parasitology. 1998;116. doi: 10.1017/s0031182000084973 [DOI] [PubMed] [Google Scholar]
  • 7.Yeoh LM, Goodman CD, Mollard V, McFadden GI, Ralph SA. Comparative transcriptomics of female and male gametocytes in Plasmodium berghei and the evolution of sex in alveolates. BMC Genomics. 2017;18: 1–16. doi: 10.1186/S12864-017-4100-0/FIGURES/4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Poran A, Nötzel C, Aly O, Mencia-Trinchant N, Harris CT, Guzman ML, et al. Single-cell RNA sequencing reveals a signature of sexual commitment in malaria parasites. Nature. 2017;551: 95–99. doi: 10.1038/nature24280 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Reid AJ, Talman AM, Bennett HM, Gomes AR, Sanders MJ, Illingworth CJR, et al. Single-cell RNA-seq reveals hidden transcriptional variation in malaria parasites. Elife. 2018;7. doi: 10.7554/eLife.33105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Witmer K, Fraschka SA, Vlachou D, Bártfai R, Christophides GK. An epigenetic map of malaria parasite development from host to vector. Sci Rep. 2020;10: 1–19. doi: 10.1038/s41598-020-63121-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Khan SM, Franke-Fayard B, Mair GR, Lasonder E, Janse CJ, Mann M, et al. Proteome Analysis of Separated Male and Female Gametocytes Reveals Novel Sex-Specific Plasmodium Biology. Cell. 2005;121: 675–687. doi: 10.1016/j.cell.2005.03.027 [DOI] [PubMed] [Google Scholar]
  • 12.Tao D, Ubaida-Mohien C, Mathias DK, King JG, Pastrana-Mena R, Tripathi A, et al. Sex-partitioning of the Plasmodium falciparum stage V gametocyte proteome provides insight into falciparum-specific cell biology. Mol Cell Proteomics. 2014;13: 2705–2724. doi: 10.1074/mcp.M114.040956 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Guttery DS, Roques M, Holder AA, Tewari R. Commit and Transmit: Molecular Players in Plasmodium Sexual Development and Zygote Differentiation. Trends Parasitol. 2015;31: 676–685. doi: 10.1016/j.pt.2015.08.002 [DOI] [PubMed] [Google Scholar]
  • 14.Waters AP. Epigenetic Roulette in Blood Stream Plasmodium: Gambling on Sex. PLOS Pathog. 2016;12: e1005353. doi: 10.1371/journal.ppat.1005353 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Prajapati SK, Ayanful-Torgby R, Pava Z, Barbeau MC, Acquah FK, Cudjoe E, et al. The transcriptome of circulating sexually committed Plasmodium falciparum ring stage parasites forecasts malaria transmission potential. Nat Commun 2020 111. 2020;11: 1–11. doi: 10.1038/s41467-020-19988-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Sinha A, Hughes KR, Modrzynska KK, Otto TD, Pfander C, Dickens NJ, et al. A cascade of DNA-binding proteins for sexual commitment and development in Plasmodium. Nature. 2014;507: 253–257. doi: 10.1038/nature12970 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kafsack BFC, Rovira-Graells N, Clark TG, Bancells C, Crowley VM, Campino SG, et al. A transcriptional switch underlies commitment to sexual development in malaria parasites. Nature. 2014;507: 248–252. doi: 10.1038/nature12920 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kent RS, Modrzynska KK, Cameron R, Philip N, Billker O, Waters AP. Inducible developmental reprogramming redefines commitment to sexual development in the malaria parasite Plasmodium berghei. Nat Microbiol. 2018;3: 1206–1213. doi: 10.1038/s41564-018-0223-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Llorà-Batlle O, Michel-Todó L, Witmer K, Toda H, Fernández-Becerra C, Baum J, et al. Conditional expression of PfAP2-G for controlled massive sexual conversion in Plasmodium falciparum. Sci Adv. 2020;6: eaaz5057. doi: 10.1126/sciadv.aaz5057 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Yuda M, Kaneko I, Murata Y, Iwanaga S, Nishi T. Mechanisms of triggering malaria gametocytogenesis by AP2-G. Parasitol Int. 2021;84: 102403. doi: 10.1016/j.parint.2021.102403 [DOI] [PubMed] [Google Scholar]
  • 21.Yuda M, Iwanaga S, Kaneko I, Kato T. Global transcriptional repression: An initial and essential step for Plasmodium sexual development. Proc Natl Acad Sci U S A. 2015;112: 12824–12829. doi: 10.1073/pnas.1504389112 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Singh S, Santos JM, Orchard LM, Yamada N, van Biljon R, Painter HJ, et al. The PfAP2-G2 transcription factor is a critical regulator of gametocyte maturation. Mol Microbiol. 2021;115: 1005–1024. doi: 10.1111/mmi.14676 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Yuda M, Kaneko I, Iwanaga S, Murata Y, Kato T. Female-specific gene regulation in malaria parasites by an AP2-family transcription factor. Mol Microbiol. 2020;113: 40–51. doi: 10.1111/mmi.14334 [DOI] [PubMed] [Google Scholar]
  • 24.Ralph SA, Cortés A. Plasmodium sexual differentiation: how to make a female. Mol Microbiol. 2019;112: 1627–1631. doi: 10.1111/mmi.14340 [DOI] [PubMed] [Google Scholar]
  • 25.Nishi T, Kaneko I, Yuda M. Identification of a novel AP2 transcription factor in zygotes with an essential role in Plasmodium ookinete development. Blackman MJ, editor. PLOS Pathog. 2022;18: e1010510. doi: 10.1371/journal.ppat.1010510 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Campelo Morillo RA, Tong X, Xie W, Abel S, Orchard LM, Daher W, et al. The transcriptional regulator HDP1 controls expansion of the inner membrane complex during early sexual differentiation of malaria parasites. Nat Microbiol 2022 72. 2022;7: 289–299. doi: 10.1038/s41564-021-01045-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.McLean KJ, Niles JC. A transcription factor helps Plasmodium falciparum gametocytogenesis take shape. Trends Parasitol. 2022;38: 722–723. doi: 10.1016/j.pt.2022.07.004 [DOI] [PubMed] [Google Scholar]
  • 28.Modrzynska K, Pfander C, Chappell L, Yu L, Suarez C, Dundas K, et al. A Knockout Screen of ApiAP2 Genes Reveals Networks of Interacting Transcriptional Regulators Controlling the Plasmodium Life Cycle. Cell Host Microbe. 2017;21: 11–22. doi: 10.1016/j.chom.2016.12.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Zhang C, Li Z, Cui H, Jiang Y, Yang Z, Wang X, et al. Systematic CRISPR-Cas9-mediated modifications of plasmodium yoelii ApiAP2 genes reveal functional insights into parasite development. MBio. 2017;8: 1–17. doi: 10.1128/mBio.01986-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Li Z, Cui H, Guan J, Liu C, Yang Z, Yuan J. Plasmodium transcription repressor AP2-O3 regulates sex-specific identity of gene expression in female gametocytes. EMBO Rep. 2021;22: 1–18. doi: 10.15252/embr.202051660 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.van Dijk MR, van Schaijk BCL, Khan SM, van Dooren MW, Ramesar J, Kaczanowski S, et al. Three members of the 6-cys protein family of plasmodium play a role in gamete fertility. PLoS Pathog. 2010;6: 1–13. doi: 10.1371/journal.ppat.1000853 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Van Dijk MR, Janse CJ, Thompson J, Waters AP, Braks JAM, Dodemont HJ, et al. A Central Role for P48/45 in Malaria Parasite Male Gamete Fertility. Cell. 2001;104: 153–164. doi: 10.1016/s0092-8674(01)00199-4 [DOI] [PubMed] [Google Scholar]
  • 33.Li Q, Brown JB, Huang H, Bickel PJ. Measuring reproducibility of high-throughput experiments. Ann Appl Stat. 2011;5: 1752–1779. doi: 10.1214/11-AOAS466 [DOI] [Google Scholar]
  • 34.Krismer K, Guo Y, Gifford DK. IDR2D identifies reproducible genomic interactions. Nucleic Acids Res. 2020;48: e31–e31. doi: 10.1093/nar/gkaa030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Green JL, Martin SR, Fielden J, Ksagoni A, Grainger M, Yim Lim BYS, et al. The MTIP–Myosin A Complex in Blood Stage Malaria Parasites. J Mol Biol. 2006;355: 933–941. doi: 10.1016/j.jmb.2005.11.027 [DOI] [PubMed] [Google Scholar]
  • 36.Ferreira JL, Heincke D, Wichers JS, Liffner B, Wilson DW, Gilberger TW. The Dynamic Roles of the Inner Membrane Complex in the Multiple Stages of the Malaria Parasite. Frontiers in Cellular and Infection Microbiology. Frontiers Media S.A.; 2021. p. 841. doi: 10.3389/fcimb.2020.611801 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Yuda M, Yano K, Tsuboi T, Torii M, Chinzei Y. von Willebrand factor A domain-related protein, a novel microneme protein of the malaria ookinete highly conserved throughout Plasmodium parasites. Mol Biochem Parasitol. 2001;116: 65–72. doi: 10.1016/s0166-6851(01)00304-8 [DOI] [PubMed] [Google Scholar]
  • 38.Ecker A, Bushell ESC, Tewari R, Sinden RE. Reverse genetics screen identifies six proteins important for malaria development in the mosquito. Mol Microbiol. 2008;70: 209–220. doi: 10.1111/j.1365-2958.2008.06407.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Shinzawa N, Nishi T, Hiyoshi F, Motooka D, Yuda M, Iwanaga S. Improvement of CRISPR/Cas9 system by transfecting Cas9-expressing Plasmodium berghei with linear donor template. Commun Biol 2020 31. 2020;3: 1–13. doi: 10.1038/s42003-020-01138-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Chauhan M, Tarique M, Tuteja R. Plasmodium falciparum specific helicase 3 is nucleocytoplasmic protein and unwinds DNA duplex in 3′ to 5′ direction. Sci Rep. 2017;7: 1–14. doi: 10.1038/s41598-017-12927-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Bushell E, Gomes AR, Sanderson T, Anar B, Girling G, Herd C, et al. Functional Profiling of a Plasmodium Genome Reveals an Abundance of Essential Genes. Cell. 2017;170: 260–272.e8. doi: 10.1016/j.cell.2017.06.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Oehring SC, Woodcroft BJ, Moes S, Wetzel J, Dietz O, Pulfer A, et al. Organellar proteomics reveals hundreds of novel nuclear proteins in the malaria parasite Plasmodium falciparum. Genome Biol. 2012;13: 1–21. doi: 10.1186/gb-2012-13-11-r108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Allen MD, Yamasaki K, Ohme-Takagi M, Tateno M, Suzuki M. A novel mode of DNA recognition by a β-sheet revealed by the solution structure of the GCC-box binding domain in complex with DNA. EMBO J. 1998;17: 5484–5496. doi: 10.1093/EMBOJ/17.18.5484 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Painter HJ, Campbell TL, Llinás M. The Apicomplexan AP2 family: Integral factors regulating Plasmodium development. Molecular and Biochemical Parasitology. NIH Public Access; 2011. pp. 1–7. doi: 10.1016/j.molbiopara.2010.11.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Russell AJC, Sanderson T, Bushell E, Talman AM, Anar B, Girling G, et al. Regulators of male and female sexual development are critical for the transmission of a malaria parasite. Cell Host Microbe. 2023. [cited 26 Jan 2023]. doi: 10.1016/j.chom.2022.12.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Mohammed H, Taylor C, Brown GD, Papachristou EK, Carroll JS, D’Santos CS. Rapid immunoprecipitation mass spectrometry of endogenous proteins (RIME) for analysis of chromatin complexes. Nat Protoc 2015 112. 2016;11: 316–326. doi: 10.1038/nprot.2016.020 [DOI] [PubMed] [Google Scholar]
  • 47.Scholtes C, Dufour CR, Giguère V. Rapid immunoprecipitation mass spectrometry of endogenous protein (RIME) to identify chromatin-interactome in prostate cancer cells. STAR Protoc. 2022;3: 101434. doi: 10.1016/j.xpro.2022.101434 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Inoue N, Hess KD, Moreadith RW, Richardson LL, Handel MA, Watson ML, et al. New Gene Family Defined by MORC, a Nuclear Protein Required for Mouse Spermatogenesis. Hum Mol Genet. 1999;8: 1201–1207. doi: 10.1093/hmg/8.7.1201 [DOI] [PubMed] [Google Scholar]
  • 49.Koch A, Kang HG, Steinbrenner J, Dempsey DA, Klessig DF, Kogel KH. MORC proteins: Novel players in plant and animal health. Front Plant Sci. 2017;8: 1720. doi: 10.3389/fpls.2017.01720 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wang H, Zhang L, Luo Q, Liu J, Wang G. MORC protein family-related signature within human disease and cancer. Cell Death Dis 2021 1212. 2021;12: 1–11. doi: 10.1038/s41419-021-04393-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Farhat DC, Swale C, Dard C, Cannella D, Ortet P, Barakat M, et al. A MORC-driven transcriptional switch controls Toxoplasma developmental trajectories and sexual commitment. Nat Microbiol. 2020;5: 570. doi: 10.1038/s41564-020-0674-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Vinh DBN, Drubin DG. A yeast TCP-1-like protein is required for actin function in vivo. Proc Natl Acad Sci U S A. 1994;91: 9116. doi: 10.1073/pnas.91.19.9116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Chen X, Sullivan DS, Huffaker TC. Two yeast genes with similarity to TCP-1 are required for microtubule and actin function in vivo. Proc Natl Acad Sci U S A. 1994;91: 9111. doi: 10.1073/pnas.91.19.9111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Yam AY, Xia Y, Lin HTJ, Burlingame A, Gerstein M, Frydman J. Defining the TRiC/CCT interactome links chaperonin function to stabilization of newly made proteins with complex topologies. Nat Struct Mol Biol 2008 1512. 2008;15: 1255–1262. doi: 10.1038/nsmb.1515 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kelly JJ, Tranter D, Pardon E, Chi G, Kramer H, Happonen L, et al. Snapshots of actin and tubulin folding inside the TRiC chaperonin. Nat Struct Mol Biol 2022 295. 2022;29: 420–429. doi: 10.1038/s41594-022-00755-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Mbengue A, Vialla E, Berry L, Fall G, Audiger N, Demettre-Verceil E, et al. New Export Pathway in Plasmodium falciparum-Infected Erythrocytes: Role of the Parasite Group II Chaperonin, PfTRiC. Traffic. 2015;16: 461–475. doi: 10.1111/tra.12266 [DOI] [PubMed] [Google Scholar]
  • 57.Spillman NJ, Beck JR, Ganesan SM, Niles JC, Goldberg DE. The chaperonin TRiC forms an oligomeric complex in the malaria parasite cytosol. Cell Microbiol. 2017;19: e12719. doi: 10.1111/cmi.12719 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Hillier C, Pardo M, Yu L, Bushell E, Sanderson T, Metcalf T, et al. Landscape of the Plasmodium Interactome Reveals Both Conserved and Species-Specific Functionality. Cell Rep. 2019;28: 1635–1647.e5. doi: 10.1016/j.celrep.2019.07.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Saksouk N, Bhatti MM, Kieffer S, Smith AT, Musset K, Garin J, et al. Histone-Modifying Complexes Regulate Gene Expression Pertinent to the Differentiation of the Protozoan Parasite Toxoplasma gondii. Mol Cell Biol. 2005;25: 10301–10314. doi: 10.1128/MCB.25.23.10301-10314.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Bougdour A, Maubon D, Baldacci P, Ortet P, Bastien O, Bouillon A, et al. Drug inhibition of HDAC3 and epigenetic control of differentiation in Apicomplexa parasites. J Exp Med. 2009;206: 953–966. doi: 10.1084/jem.20082826 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kaneko I, Iwanaga S, Kato T, Kobayashi I, Yuda M. Genome-Wide Identification of the Target Genes of AP2-O, a Plasmodium AP2-Family Transcription Factor. PLoS Pathog. 2015;11: 1–27. doi: 10.1371/journal.ppat.1004905 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Santos JM, Josling G, Ross P, Joshi P, Orchard L, Campbell T, et al. Red Blood Cell Invasion by the Malaria Parasite Is Coordinated by the PfAP2-I Transcription Factor. Cell Host Microbe. 2017;21: 731–741.e10. doi: 10.1016/j.chom.2017.05.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Shang X, Shen S, Tang J, He X, Zhao Y, Wang C, et al. A cascade of transcriptional repression determines sexual commitment and development in Plasmodium falciparum. Nucleic Acids Res. 2021;49: 9264–9279. doi: 10.1093/nar/gkab683 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Radke JB, Lucas O, De Silva EK, Ma Y, Sullivan WJ, Weiss LM, et al. ApiAP2 transcription factor restricts development of the Toxoplasma tissue cyst. Proc Natl Acad Sci U S A. 2013;110: 6871–6876. doi: 10.1073/pnas.1300059110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Radke JB, Worth D, Hong D, Huang S, Sullivan WJ, Wilson EH, et al. Transcriptional repression by ApiAP2 factors is central to chronic toxoplasmosis. PLOS Pathog. 2018;14: e1007035. doi: 10.1371/journal.ppat.1007035 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Yuda M, Iwanaga S, Shigenobu S, Mair GR, Janse CJ, Waters AP, et al. Identification of a transcription factor in the mosquito-invasive stage of malaria parasites. Mol Microbiol. 2009;71: 1402–1414. doi: 10.1111/j.1365-2958.2009.06609.x [DOI] [PubMed] [Google Scholar]
  • 67.Yuda M, Sakaida H, Chinzei Y. Targeted disruption of the Plasmodium berghei CTRP gene reveals its essential role in malaria infection of the vector mosquito. J Exp Med. 1999;190: 1711–1715. doi: 10.1084/jem.190.11.1711 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Meier F, Beck S, Grassl N, Lubeck M, Park MA, Raether O, et al. Parallel accumulation-serial fragmentation (PASEF): Multiplying sequencing speed and sensitivity by synchronized scans in a trapped ion mobility device. J Proteome Res. 2015;14: 5378–5387. doi: 10.1021/acs.jproteome.5b00932 [DOI] [PubMed] [Google Scholar]
  • 69.Meier F, Brunner AD, Koch S, Koch H, Lubeck M, Krause M, et al. Online parallel accumulation–serial fragmentation (PASEF) with a novel trapped ion mobility mass spectrometer. Mol Cell Proteomics. 2018;17: 2534–2545. doi: 10.1074/mcp.TIR118.000900 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Searle BC. Scaffold: A bioinformatic tool for validating MS/MS-based proteomic studies. Proteomics. 2010;10: 1265–1269. doi: 10.1002/pmic.200900437 [DOI] [PubMed] [Google Scholar]

Decision Letter 0

Kami Kim, Ron Dzikowski

29 Oct 2022

Dear Dr. Yuda,

Thank you very much for submitting your manuscript "PbAP2-FG2 and AP2R-2 function together as a transcriptional repressor complex essential for Plasmodium female development" for consideration at PLOS Pathogens. As with all papers reviewed by the journal, your manuscript was reviewed by members of the editorial board and by several independent reviewers. In light of the reviews (below this email), we would like to invite the resubmission of a significantly-revised version that takes into account the reviewers' comments.

All reviewers the appreciated the potential advance of the data presented to the field. However they raised major concerns that will required major revisions to answer each of their comments. Primarily, it is necessary to provide experimental data to demonstrate that there is a functional repressor complex which is formed between PbAP2-FG2 and PbAP2R-2.

We cannot make any decision about publication until we have seen the revised manuscript and your response to the reviewers' comments. Your revised manuscript is also likely to be sent to reviewers for further evaluation.

When you are ready to resubmit, please upload the following:

[1] A letter containing a detailed list of your responses to the review comments and a description of the changes you have made in the manuscript. Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out.

[2] Two versions of the revised manuscript: one with either highlights or tracked changes denoting where the text has been changed; the other a clean version (uploaded as the manuscript file).

Important additional instructions are given below your reviewer comments.

Please prepare and submit your revised manuscript within 60 days. If you anticipate any delay, please let us know the expected resubmission date by replying to this email. Please note that revised manuscripts received after the 60-day due date may require evaluation and peer review similar to newly submitted manuscripts.

Thank you again for your submission. We hope that our editorial process has been constructive so far, and we welcome your feedback at any time. Please don't hesitate to contact us if you have any questions or comments.

Sincerely,

Ron Dzikowski

Associate Editor

PLOS Pathogens

Kami Kim

Section Editor

PLOS Pathogens

Kasturi Haldar

Editor-in-Chief

PLOS Pathogens

orcid.org/0000-0001-5065-158X

Michael Malim

Editor-in-Chief

PLOS Pathogens

orcid.org/0000-0002-7699-2064

***********************

All reviewers the appreciated the potential advance of the data presented to the field. However they raised major concerns that will required major revisions to answer each of their comments. Primarily, it is necessary to provide experimental data to demonstrate that there is a functional repressor complex which is formed between PbAP2-FG2 and PbAP2R-2.

Reviewer's Responses to Questions

Part I - Summary

Please use this section to discuss strengths/weaknesses of study, novelty/significance, general execution and scholarship.

Reviewer #1: Male and female gametocytes are sexual progenitor cells essential for the transmission of the malaria parasite by the mosquito. Differentiation of gametocytes into fertile gametes depends on a sex-specific transcriptional program. How parasites establish different transcriptional repertoires in male and female gametocytes is still largely unknown. In this study, they discovered that the two female transcriptional regulators PbAP2-FG2 and AP2R-2 cooperate as a transcriptional repressor complex in P. berghei, with target genes including male, female, and early gametocyte genes activated by AP2-G. Based on their results, PbAP2-FG2 and AP2R-2 appear to play multiple roles in the development of female gametocytes from early gametocytes. This study was performed in the rodent model of P. berghei malaria and challenges a previous study of PyAP2-O3 (renamed PyAP2-FG2) in the P. yoelii model (reference 18, by Li et al.).

Reviewer #2: In their manuscript: “PbAP2-FG2 and AP2-R2 function together as a transcriptional repressor complex essential for Plasmodium female development”, Nishi et al. characterize the potential transcriptional regulatory role of two ApiAP2, DNA-binidngproteins, AP2R-2 and AP2-FG. Through phenotypic analysis it would seem the AP2-FG2 protein peaks in expression in female gametocytes and the genetic perturbation of this protein results in a decrease in female gametocyte-related genes. The protein seems to exert its function through binding upstream regions of genes, repressing male and female-related gametocyte genes alike. A similar profile was observed for AP2R-2. After thorough reexamination of previously published data for the orthologue of AP2-FG2 in Plasmodium yoelli, the authors demonstrate significant overlap between the function of the two orthologous proteins. Overall, this study confirms that the role of the newly coined “AP2-FG2” in Plasmodium berghei is consistent to what has previously been published for Plasmodium yoelii. I thank the authors for a well organized, and clearly communicated study. However, there are both major and minor concerns with this manuscript and it is requested that the authors respond to each and make edits where appropriate to strengthen the manuscript.

Reviewer #3: The gametocyte stage is a critical step to continuing the Plasmodium lifecycle from the vertebrate host to mosquito vector. However, the fine-tuned regulation of this life stage transition is not completely understood. This manuscript focuses on PbAP2-FG2 and PbAP2R-2, which are two putative female gametocyte regulators. PbAP2-FG2 (for “female gametocyte 2”) was previously named PbAP2-O3 (PBANKA_1015500; “ookinete 3”) in Modryznska et al. (2017) Cell Host & Microbe due to the ap2-o3_KO causing a growth arrest prior to the ookinete stage of mosquito development. In this submission, the authors have renamed the factor to PbAP2-FG2, due to expression in female gametocytes and dysregulation of female gametocyte maturation genes after PbAP2-FG2 disruption. The second factor, PbAP2R-2 (stands for “AP2-related factor 2”) was named in previous work by this group in Yuda et al. (2021) Parasitology International because it contained an ACDC domain and was a target of PbAP2-G and PbAP2-FG via ChIP-seq. In this work, Nishi et al. employed a variety of approaches to identify the role of both PbAP2-FG2 and PbAP2R-2, including (1) GFP-tagging PbAP2-FG2 that identified peak protein expression during female gametocyte development, (2) gene KO of PbAP2-FG2 coupled with RNA-seq that identified an enrichment of female-specific dysregulated transcripts over male-specific transcripts, (3) anti-GFP chromatin immunoprecipitation followed by sequencing (ChIP-seq) of PbAP2-FG2 that identified its genome-wide binding sites and enriched RGAGAR DNA motif, (4) integration of ChIP-seq gene targets and RNA-seq DEGs that identified an enrichment of ChIP-seq targets with the female-specific genes upregulated, (5) CRISPR-Cas9 modifications to remove the RGAGAR motif from a promoter region that identified a depletion of PbAP2-FG2 binding by ChIP-qPCR, (6) CRISPR-Cas9 to integrate a GFP tag onto the 3`-end of pbap2r-2 and an anti-GFP ChIP-seq that identified the binding sites of PbAP2R-2, which had a high overlap with PbAP2-FG2 ChIP-seq binding sites, (7) integration of PbAP2-G ChIP-seq targets that identified many gene targets of PbAP2-G as targets of PbAP2-FG2 and PbAP2R-2, and finally (8) a re-analysis of ChIP-seq data from Li et al. (2021) EMBO Reports on the PbAP2-FG2 P. yoelii ortholog, PyAP2-O3, where they identified their enriched RGAGAR motif in the highly reproducible ChIP-seq binding sites.

Overall, the results presented in this manuscript regarding PbAP2-FG2 are exciting and the data analysis is thorough. The results provide a new characterization of an ApiAP2 protein PbAP2-FG2 involved in the regulation of gametocyte development, particularly in female cell fate determination. However, despite a number of preliminary results, the suggestion that the data presented demonstrate a transcriptional regulatory complex between PbAP2-FG2 and PbAP2R-2 that work together to repress gene targets is unsubstantiated. The data clearly show that PbAP2-FG2 and PbAP2R-2 target highly similar genomic regions, but proof that those components are physically interacting in the same cell at the same time are lacking.

**********

Part II – Major Issues: Key Experiments Required for Acceptance

Please use this section to detail the key new experiments or modifications of existing experiments that should be absolutely required to validate study conclusions.

Generally, there should be no more than 3 such required experiments or major modifications for a "Major Revision" recommendation. If more than 3 experiments are necessary to validate the study conclusions, then you are encouraged to recommend "Reject".

Reviewer #1: 1- I found quite puzzling that there few if no correlation between the direct targets of PbAP2-FG2 (assessed by ChIP-seq) and the dysregulation of the transcriptome in its absence. The authors have attempted to explain these discrepancies, but their hypothesis is elusive and does not allow for the possibility that this factor could be replaced, for example, by another ApiAP2. Is it possible that each AP2 they are describing homodimerizes individually and compensates for the absence of the other.

2- With their elegant experiments on cis-mutagenesis of the repressive element, they provide only a partial answer to the lack of correlation at the genomic level between ChIPseq and RNAseq by proposing that PbAP2-FG2 and PbAP2-R2 cooperatively repress genes, probably as heterodimers. However, the possibility that both homodimerize and colocalize at the same genomic sites cannot be ruled out until it is shown that they interact biochemically or that PbAP2-R2 is released from chromatin in the absence of PbAP2-FG2 and vice versa.

Reviewer #2: 1. Secondarily, the authors abandon any notion that this protein has a role in any other stage of development quite quickly, despite it’s obvious expression in both male gametocytes and ookinetes. Could the authors clarify why neither of these were pursued further? Is it clear to them that there is no possibility of a role for this AP2 in other stages of development, why or why not? In addition, please quantify all of the phenotypes that are suggested by both the GFP-tagged and genetic disruptants of AP2-FG2

2. The authors have completed a tremendous amount of work to characterize these proteins and it will add to the already large number of ApiAP2s this lab has revealed the role of. They are to be commended for their continued efforts. However, to support the assertion that AP2-FG2 and AP2R-2 act together to co-repress genes, the authors must provide definitive data that demonstrate an interaction between the two lines. The authors should consider Co-IP or tagging AP2-FG2 with 3xHA in a parasite that has AP2R-2 tagged with GFP. Flow-cytometry can be used to select for parasites expressing AP2R-2 and ChiP-seq for either or both AP2 can be performed.

Reviewer #3: There is insufficient data presented to demonstrate that there is a complex formed between PbAP2-FG2 and PbAP2R-2 implying colocalization. Therefore, the title and many statements in the Results and the beginning and end of the Discussion need to be modified to reflect this. Although the authors do demonstrate that the PbAP2-FG2 and PbAP2R-2 appear to occupy similar genomewide binding sites, they do not test whether this is due to complex formation. At least two possibilities come to mind. It is entirely feasible that in 2 different cell populations PbAP2-FG2 is binding these loci throughout the genome, while in a different population PbAP2R-2 is binding the same loci. This can only be resolved by single cell ChIP-seq which has not been successful to date in Plasmodium parasites (to my knowledge). Although this is unlikely given the roughly 1:1 correlation of the ChIP-seq peak intensities reported, it is possible. To directly test whether there is a complex, immunoprecipitation (IP) experiments will need to be performed. Both PbAP2-FG2 and PbAP2R-2 were tagged in this manuscript and used for localization studies and ChIP-seq. Therefore, IP-MS proteomics should be feasible in principle. To claim that PbAP2-FG2 and PbAP2R-2 form a complex, the authors should produce IP/MS data using all available tagged reagents already at their disposal.

Throughout the manuscript (e.g. Line 73, Line 533), reference to P. berghei targets of AP2-G are mentioned solely in the context of previous work by the same authors, However, other studies have determined targets of PfAP2-G by inducible activation, ChIP-seq, or scRNA-seq including Llora-Battle et al. 2019, Josling et al. 2020, and Poran 2017. Two points are worth addressing. First, what is the overlap in predicted targets between these studies? Can the authors speculate why they are different between PbAP2-G (many targets) and PfAP2-G (limited target set)? For example, the authors previously reported pbap2-g2, pbap2-fg, and pbab2-z as targets of PbAP2-G. However, according to the literature this is not the case for PfAP2-G, where these other ApiAP2 genes are not direct targets.

Next, overall the manuscript is highly under-cited. Please be more inclusive in recognizing the contributions of others. One major omission in the Introduction is reference to the recent work of Russell et al. who have clearly demonstrated roles for a helix-turn-helix protein and other zinc-finger proteins in the regulation of male and female sexual development lineages of Pb parasites: BioRxiv: 2021.08.04.455056v1 This study is highly relevant to the current work and an understanding of sexual maturation and sex lineage differentiation.

Given these potential species-specific differences, it would be worthwhile to identify each ApiAP2 discussed with the relevant species designation (Pb, Py, Pf) based on where the evidence presented originates. There are clear reasons to believe that target genes in one organism may or may not be the same across species. Similarly, it has already been demonstrated that certain homologous ApiAP2 proteins are essential in some species, but not all. By definition, their functional roles must therefore be different or there is functional redundancy in one species. Therefore, please use full IDs throughout such as PbAP2-G2 or PbAP2R-2 or PfAP2-G2, etc…

Another clear issue is comparing the results in lines 389-294 between Pf and Pb and the role of the cytoskeleton. Clearly the timing and duration of these complexes must be vastly different given the 24-hour maturation of gametocytes in Pb versus 10 days in Pf.

In generating tagged parasite lines for this study (Figure 1C) it is unclear if the endogenous 3`UTR is maintained in these parasites. This same group reported that altering the endogenous 3’ UTR was found to be an issue in previous work on AP2-Z (Yuda et al. (2021) and Nishi et al. (2022) BioRxiv). This was reportedly because the replacement of the endogenous 3`UTR disrupted the normal function of DOZI and the translational repression of AP2-Z. Please label the plasmid designs in Fig S1 more clearly and please note any changes to the 3`UTR in the main text.

The authors use a nice cross-fertilization assay to demonstrate the role of pbap2-fg2. The same should be done for pbap2r-2, and the tools to do so are available.

To determine differentially expressed genes (DEGs), the authors reference “previously reported sex-specific RNA-seq data” from reference 21 (lines 168, 169). This choice appears arbitrary. There are numerous datasets available (transcriptomic, proteomic, genomic) that have catalogued putative male and female gametocyte markers. A much better approach would be to use a comprehensive table with multiple lines of evidence to categorize individual genes as “male” or “female”, especially since most of them have not been independently characterized. Further, these genes should be referred to as “male-enriched” and “female-enriched” as opposed to “male-specific” or “female-specific”, because few genes are only truly present in one lineage.

The ChIP-seq results are quite surprising with over 1000 peaks in every replicate experiment. Compared to other ChIP-seq results in the Plasmodium literature, this number is very high for an individual DNA binding protein. Looking back, virtual all papers from this group report high numbers of binding events. What number of genes does this represent? Presumably not 1/5 of the genome? If not, does the appearance of multiple binding events/motifs upstream of genes suggest something associated with regulation?

Due to the high overlap in binding sites between PbAP2-FG2 and PbAP2R-2, the authors should check whether those sites are also pulled down when performing an anti-GFP ChIP-seq in a WT parasite background (i.e., are those sites hyper ChIP-able)?

Also, how many of the more than one thousand binding sites have the motif reported in Figure 3C. It is not clear. Please prepare a figure showing the peak summits for all binding events colored by the underlying nucleotide sequence.

The statement in line 320-321 that AP2-R2 has no AP2 domain is unsubstantiated. What is this evidence based on? The few descriptions in the literature of the ACDC domain suggest that it must always co-occur with an AP2 domain. In fact, in the Pf orthologue (PF3D7_1319600), Oehring et al. 2012 (Figure 8) identified 2 AP2 domains in this “ACDC domain-containing protein”. Please provide an alignment in the Supplementary materials between the Pb and Pf orthologues for AP2R-2. Also, using AlphaFold2 to examine the predicted structure of AP2R-2 clearly demonstrates at least one well-folded AP2 domain and perhaps a cryptic second AP2 domain. The authors should provide this information. In addition to demonstrating that this protein may have 1-2 AP2 domains, it may also provide an alternative interpretation of why the binding of AP2R-2 and AP2-FG2 are so similar? Perhaps their AP2 domains are actually relatively similar and therefore bind the same motif and genome-wide sites? Otherwise, it is also unclear why the authors pursue ChIP-seq of AP2R-2 if they do not suspect that it has any DNA binding domains.

In the re-analysis of the PyAP2-FG2 ChIP-seq data from Li et al. please describe clearly how your methods of analyzing the ChIP-seq data differ. You describe how you have re-analyzed their data, but have you re-analyzed it using their approach? And which approach is better and why beside the inclusion/exclusion of low read counts?

Renaming genes is a tricky business. Is there sufficient data here to fully support this? I’m not sure. I certainly do not think it is a good idea to continue renaming ApiAP2 transcription factors? This will lead to serious confusion in the field. One simple reason is that it is possible that they may serve various functions throughout the entire life cycle, so will each gene product get a different name every time a new phenotype is published? Also, species-specific differences should be anticipated given differences in the lifecycles. Within each publication it is easy to follow which gene is being discussed, but when comparing across publications it will become increasingly difficult to make comparisons. For example in lines 94, 98, and 100: you call the P. yoelii ortholog “PyAP2-FG2” but Li et al. technically call it “PyAP2-O3”. It should be PyAP2-O3 to be in-line with Li et al. and then “PbAP2-FG2” for your publication.

**********

Part III – Minor Issues: Editorial and Data Presentation Modifications

Please use this section for editorial suggestions as well as relatively minor modifications of existing data that would enhance clarity.

Reviewer #1: 1- The authors are advised to follow the nomenclature that precedes their work and not to change the genes already named by their colleagues to suit their ideas. Li et al (reference 18) used PyAP2-O3 in their article, which has been changed to PyAP2-FG2 (lines 93-95) in this manuscript, with the authors stating that PyAP2-O3 is the bona fide ortholog of PbAP2-FG2. The reviewer found this very confusing.

2- The authors compared their data with those of Li et al (reference 18). They reanalyzed all omic data, starting with the raw data deposited by Li et al, and reinterpreted their data in a very parsimonious way, concluding that they were wrong, and finally proposing their model as the true model. In my opinion, it would be beneficial if the authors were less direct in their criticisms (perhaps they should curb their enthusiasm in "killing" a competing story) and leave room for alternative hypotheses. I suggest rewriting this part with less "enthusiasm," I would say.

3- Missing scales in Figures :

• y-axis scale in figures 1B, 7B

• bar scale in figuress 1C-E

4- Figure 1D : The parental strain must be shown in parallel to demonstrate that there is no difference in the morphology of gametocytes produced by a mutant pbap2-fg2(-) background.

5- ChIPseq figures 3A and 6A : Rather than plotting a genomic area, the authors should compare enrichment levels across the genome by plotting pairwise correlations between replicates (i.e., plotting the average tag number of rep_1 versus rep_2 enrichment).

Reviewer #2: 1. The authors pursue the characterization of PBANKA_1015500, which contains both an ApiAP2 and an ACDC domain. This protein has previously been characterized by Li et al. 2021 (PMID: 33665945) as AP2-O3. Here the authors chose to re-name this protein to AP2-FG2. Little has been done to prove this protein has no role in male gametocytes or other stages of development. Since there is no official naming convention, anyone can name them based on the role of these proteins in various stages and species. Have the authors considered not renaming the protein for consistency in the field?

2. In Figure 1B, the IFAs used to substantiate the lifecycle stage-specific expression of AP2-FG2 are very difficult to discern. However, the quality of the image makes it very hard to properly assess this claim. The authors should support this assertion via an orthogonal method, such as Western Blot analysis at every stage of development. In addition, the authors should quantify their claim of stage-specific expression. Please quantify the percentage of males and females that have GFP expression.

3. The authors pursue the characterization of PBANKA_1015500, which contains both an ApiAP2 and an ACDC domain. This protein has previously been characterized by Li et al. 2021 (PMID: 33665945) as AP2-O3. Here the authors chose to re-name this protein to AP2-FG2. Little has been done to prove this protein has no role in male gametocytes or other stages of development. Since there is no official naming convention, anyone can name them based on the role of these proteins in various stages and species. Have the authors considered not renaming the protein for consistency in the field?

4. In line 128-129 the authors state two clonal lines were produced but never clarify which of the clones were used in each experiment, with the exception of their mosquito feeding experiments in line 135-136.

5. Are the images in Figure 1D and E representative images? Did the authors quantify morphologically normal/abnormal gametocytes and ookinetes compared to the WT parent? This developmental phenotype should be quantified for each knockout clone. Additionally, the WT representative images should be shown for comparison (Figure 1D).

6. In line 131-132 the authors assert that the ap2-fg2 knock out line produces normal amounts of exflagellated parasites but do not provide any supporting data. Please quantify the number of exflagellation events for both clones.

7. Line 135-136 the authors also do not show data to support the assertion that there is a loss of ookinete production in the ap2-fg2 knock out line (either clone).

8. Figure 1F is missing from the manuscript file, the authors should also state how many biological and technical replicates were performed for the experiments whose data is presented in these panels.

9. The authors should clarify the remark, “overall, log2(fold change) of female-enriched genes tended to be lower than the other genes” (line 174-176). Is this in context of the significantly decreased transcripts only or the total transcriptome? Comparing the expression of these classes of genes with a two-tailed t-test is also not strictly appropriate, as there would be thousands of genes falling into the other category that are not significantly different between WT and KO. Perhaps the more appropriate test would be to perform a Fisher’s exact test to check for relative overrepresentation of female-enriched genes in the significantly decreased transcripts identified in the study.

10. The authors determined the genomic location of their called peaks from the ChIP-seq. Out of the almost 3,000 peaks from both experiments, what was the distribution of the peaks in intergenic vs intragenic regions? For those that did not have a recognized motif, was the distribution different?

11. The authors postulate the role of AP2-FG2 as a transcriptional repressor and the authors note in Table S3 whether or not each potential target gene is significantly up or down regulated in the AP2-FG2(-) transcriptomics. To assess the significance, the authors should present the distribution (not necessarily significantly up or down) of all the identified ChIP target genes in the transcriptomics experiments.

12. Line 308-311: To demonstrate sex-specificity using the RT-qPCR, the authors failed to use a published Why did the authors use a male and female marker to compare the RT-qPCR results to instead of a published reference gene?

13. The authors also assert in their heading line 315-316 that AP2-FG2 requires a corepressor to repress its target genes, but the authors do not show that the binding of AP2-FG2 is lost in the absence of AP2R-2.

14. The authors claim that the genome-wide distribution of AP2-FG2 and AP2R-2 are highly similar based on data shown in Figure 6A and B (Lines 329-330). However, Figure 6A shows only a small portion of the genome and Figure 6B shows read coverage data instead of log2 fold enrichment. It would be more convincing if the authors instead showed the overlapping peaks the way they show for the overlap with AP2-G (figure 7A) or alternatively use the IDR1D analysis.

15. The way the authors showed the overlap between the increased and decreased transcripts between AP2-FG2 and AP2R-2 in Figure 6E with a correlation plot is not appropriate for the distribution of the data. For a linear correlation in the data to be drawn, the data needs to follow a linear distribution, which is not the case for their increased and decreased transcripts. The authors can simply show what proportion of transcripts are differentially abundant in each of the conditions.

16. Please scale ChIP-seq tracks between groups and include y- and x-axes on ChIP-seq tracks for figures 1,3,6 and 7. When showing ChIP-seq tracks also clarify whether what is being displayed is truly “coverage” or actually log2(ChIP/input).

17. In Supplementary Figure 1: the authors should state which primer sets illustrated in the pictograms produced the PCR products shown in the figure and the conditions of the PCR should be stated: nr of cycles, annealing temperature of primers should be included in supplementary table S8.

18. Line 455-457: The authors state they set their FPKM threshold at <10, but it seems they meant to include transcripts with FPKM >10.

19. Please revise or complete the following requests for the Materials and Methods section of the paper:

a. Please include a detailed methodfor the “fluorescence analysis” of the parasites- including what microscope was used, if this was an immunofluorescence assay or live fluorescence microscopy etc.

b. For the generation of plasmid for transgenic alteration of the parasite, include more details on the plasmid background, with appropriate references, should be stated clearly, with a higher level of detail than simply naming them: ie. “gfp-fusion vector” and “targeting construct”

c. The authors stated the parasite lines were verified by PCR and/or Sanger sequencing. Please specifically state which methods were used in which cases, if Sanger sequencing was used to verify these lines please provide supplementary data to this effect.

d. The authors should consider performing next-generation whole genome sequencing on the knockout parasite lines to verify that there are no other mutations that could contribute to the phenotype.

e. For the ChIP-seq experiments- there seems to be substantial information about the experimental protocol missing without any supplementary method being cited. Please provide a more detailed protocol and ensure to include the following detail OR cite the source of the protocol that includes these details: Was a nuclear extraction performed prior to sonication? What is the composition of the SDS lysis buffer? At which temperature were the samples sonicated? What was the concentration of anti-GFP antibodies used in the ChIP-seq experiment and how long was the antibody incubation performed? Were the lysates precleared before the immunoprecipitation step? When were the input control samples taken? The authors also don’t mention a proteinase digestion step, was this omitted? How was the immunoprecipitated DNA purified?

f. There is also information missing regarding the data analysis of the ChIP-seq experiments: What version of the Plasmodium berghei genome was used to map the sequencing reads? How/why did the authors decide on a fold change enrichment threshold of >3?

g. For the ChIP-qPCR and RT-qPCR experiments, what were the starting concentrations of DNA or RNA included in the experiments respectively and how many cycles were these experiments conducted for? Did the authors include reference genes for either study for normalization? No account is given of how these data were analyzed, was the 2-delta Ct method used for quantification in the qPCR?

20. Why did the authors use the Fisher’s exact test to analyze enriched motifs in 6 bp bins instead of using available softwares (ie STREME) that scan over a wider range? Could you please justify your use of the Fisher’s exact test in this context?

21. How did the authors associate genes with Chip-seq peaks if the binding site overlaps within 1200 bp of the start codon? This method does not take into account which genes are closest to the binding site or whether these bound regions are intra- or intergenic.

Reviewer #3: In general, please check the entire document for English grammar. There are examples beginning with the second sentence of the Abstract that require further attention.

The PlasmoDB IDs for all genes mentioned in the manuscript should be provided, especially since the authors are choosing to rename pbap2-o3 as pbap2-fg2. (Such as line 317 and others throughout.)

The word “identical” is used throughout the manuscript. All instances should be replaced with “highly similar” or “analogous”, etc… In biology virtually nothing is identical.

Line 68 – Probably want to use “ability” instead of “capability”.

Please be consistent in the use of AP2R-2 instead of ap2-R2 (see lines 84 and 86). Please correct throughout the manuscript.

The last section of the Introduction (lines 96-100) should be rewritten to simply state that a re-analysis of a previous PyAP2-FG2 dataset leads to similar conclusions despite being at odds with what was previously reported in the Py study.

Lone 110-111, it is unnecessary to state what a previous study failed to do. Alternatively, they may suggest that a gene may have an earlier role leading up to the point of the observed phenotype used to name ap2 genes earlier (by Modrzynska and others). This is especially true when in lines 135-137 the authors compare their results in a confirmatory manner with this previous study.

On line 145, please clarify why 30% conversion (and not 50%) to banana-shaped ookinetes is expected?

Please clarify the sentence on lines 224-226: “The target genes contained some groups of genes seemingly expressed in female gametocytes…” What does it mean to be “seemingly expressed”?

Again, on lines 244-234, the authors write: “…the number of male-enriched genes in the targets seemed too large, again implying the possible role of PbAP2-FG2 as a transcriptional repressor.” You cannot write “seemed too large” unless you have a comparative analysis? On what basis do the authors make this claim?

In lines 248-249, the authors should note that it is very difficult to distinguish the direct vs. indirect targets by RNA-seq analysis alone, therefore target genes need to be identified within the DEGs.

The candidate gene psh3 used the section beginning on lines 277-278 was selected due to its expression and essentiality in P. falciparum parasites. Is this essentiality also true in P. berghei (PlasmoGEM or other datasets?). Also, doesn’t this imply that any regulation via PbAP2-FG2 must be exclusive to the sexual stage and that the expression in trophozoites and schizonts must be via a different factor in the asexual blood stages? I suppose that I’m not convinced as to why psh3 was used for this validation? Why not use a gene that has been well-characterized in Pb?

Line 317, please spell “Previously” correctly.

Line 348, delete “were”.

Line 362, “approximately 1” should be 0.97.

Line 365-366 should read: “Therefore we hypothesized that AP2R-2 may function as an essential co-repressor of PbAP2-FG2 in female gametocytes.”

Lines 371-372 are unclear. What “changes” is not well defined.

In lines 376-378 it is reported that the targets of AP2-G (Pb I presume?) and PbAP2-FG2 overlap, but what about the mRNA abundance in the knockouts. Does the abundance go up (activated, or loss of repression) after PbAP2-FG2 knockout? Please clarify.

Line 414, do not use “implausible”. Please replace with “unlikely”.

In line 435, how is it possible that only 271 of 781 targets genes have orthologues between Py and Pb? These organisms are remarkably similar overall. Please clarify.

On line 440, you state that “…some targets might have been falsely detected.” It is not clear how a target can be falsely detected. Surely based on the analysis used by Li et al. they did measure a target peak.

In lines 440-441, how did you “…assess the sex-specific expression of the target genes of PyAP2-FG2…”?

You should not write on lines 450-451 “… yield unreliable DEG lists.” This is demeaning to the previous study. Perhaps you mean they “… yield much larger DEG lists with larger variance.” Or something along those lines?

Line 455 change to “we re-analyzed”.

How many orthologues were considered in line 461?

Line 45574 change to “the majority”.

Figure comments:

I believe that the Figure 1B ChIP-seq data is published data and should not be in a main figure. Perhaps a supplementary figure?

Figure 1: Is the IGV image group auto scaled? Meaning is the y-axis consistent between both tracks? The y-axis ranges should be indicated on the figure or in the caption.

Figure 1F: This panel regarding the crossing experiment where you have crossed a female and a male gametocyte infertile lineage with ap2-fg2 (-) is missing. Please add this.

In Figure 2 B and C, are the other genes not assigned to male and female still gametocyte genes? Or genes expressed during other (multiple perhaps?) stages? Please clarify this in the manuscript and figure legend. The same question applies to Figure 3G.

In Figure 4, the majority of the differences are really not significant. With a reduced number of candidates, it is not surprising that motif enrichment is stronger. Also, can you explain the data that emanate from the bottom of your plots in the bottom right quadrants in Figure 4A that form a line not congruent with the rest of the volcano plot?

Figure 6B: As you know, it is more widely accepted to show ChIP-seq data as a Log2 fold enrichment of the IP over the input. Why are the data shown as read coverage? From the methods, you demonstrate that you took an input sample, so it would be a nice control to show the heatmaps with the input data as the Fold Enrichment (Log2[IP/Input]), instead of just read coverage.

In Figure 8A, I would consider switching the 2nd and 3rd columns to make your point more strongly to the reader since the p-value is higher for the GAGA motif.

In Supplemental Figure S1, please label the genotyping PCR gels more clearly so that it is clear which primers are being used to generate each product. Also it would be helpful to have the parasite line name (i.e., PbAP2-FG2::GFP) above each schematic in this figure. Is the 3`UTR modified in each of these lines?

**********

PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

Reviewer #3: No

Figure Files:

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email us at figures@plos.org.

Data Requirements:

Please note that, as a condition of publication, PLOS' data policy requires that you make available all data used to draw the conclusions outlined in your manuscript. Data must be deposited in an appropriate repository, included within the body of the manuscript, or uploaded as supporting information. This includes all numerical values that were used to generate graphs, histograms etc.. For an example see here on PLOS Biology: http://www.plosbiology.org/article/info%3Adoi%2F10.1371%2Fjournal.pbio.1001908#s5.

Reproducibility:

To enhance the reproducibility of your results, we recommend that you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option to publish peer-reviewed clinical study protocols. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols

Decision Letter 1

Kami Kim, Ron Dzikowski

26 Jan 2023

Dear Dr. Yuda,

Thank you very much for submitting your manuscript "PbAP2-FG2 and PbAP2R-2 function together as a transcriptional repressor complex essential for Plasmodium female development" for consideration at PLOS Pathogens. As with all papers reviewed by the journal, your manuscript was reviewed by members of the editorial board and by several independent reviewers. The reviewers appreciated the attention to an important topic. Based on the reviews, we are likely to accept this manuscript for publication, providing that you modify the manuscript according to the review recommendations.

Two of the reviewers pointed out that a GFP tagged unrelated line should be more appropriate negative control for unspecific interactions between the two AP2 proteins. Please address this concern in you revised version in addition to their additional comments.

Please prepare and submit your revised manuscript within 30 days. If you anticipate any delay, please let us know the expected resubmission date by replying to this email.

When you are ready to resubmit, please upload the following:

[1] A letter containing a detailed list of your responses to all review comments, and a description of the changes you have made in the manuscript.

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out

[2] Two versions of the revised manuscript: one with either highlights or tracked changes denoting where the text has been changed; the other a clean version (uploaded as the manuscript file).

Important additional instructions are given below your reviewer comments.

Thank you again for your submission to our journal. We hope that our editorial process has been constructive so far, and we welcome your feedback at any time. Please don't hesitate to contact us if you have any questions or comments.

Sincerely,

Ron Dzikowski

Academic Editor

PLOS Pathogens

Kami Kim

Section Editor

PLOS Pathogens

Kasturi Haldar

Editor-in-Chief

PLOS Pathogens

orcid.org/0000-0001-5065-158X

Michael Malim

Editor-in-Chief

PLOS Pathogens

orcid.org/0000-0002-7699-2064

***********************

Two of the reviewers pointed out that a GFP tagged unrelated line should be more appropriate negative control for unspecific interactions between the two AP2 proteins. Please address this concern in you revised version in addition to their additional comments.

Reviewer Comments (if any, and for reference):

Reviewer's Responses to Questions

Part I - Summary

Please use this section to discuss strengths/weaknesses of study, novelty/significance, general execution and scholarship.

Reviewer #1: This is a timely and well-done study. The authors responded to all the points this reviewer raised. The paper was significantly improved with the addition of new controls and complementary experiences.

Reviewer #2: In their manuscript: “PbAP2-FG2 and AP2-R2 function together as a transcriptional repressor complex essential for Plasmodium female development”, Nishi et al. characterize the potential transcriptional regulatory role of two ApiAP2,

DNA-binidngproteins, AP2R-2 and AP2-FG. Through phenotypic analysis it would seem the AP2-FG2 protein peaks in expression in female gametocytes and the genetic perturbation of this protein results in a decrease in female gametocyte-related genes. The protein seems to exert its function through binding upstream regions of genes,

repressing male and female-related gametocyte genes alike.

The authors were given review comments and I appreciate the authors efforts to address the concerns I had with their study following the initial submission. Most comments/concerns were adequately addressed and the addition of RIME to determine the interacting partners of AP2-FG2 strengthens this study. I am, however, still curious about the underlying AP2-FG2 GFP-tagged line. I have commented on the authors responses in line in the attached document and summarized below in the "Minor Issues" section

Reviewer #3: Overall the authors have addressed all of my major concerns. I have some additional suggestions and minor points raised below.

**********

Part II – Major Issues: Key Experiments Required for Acceptance

Please use this section to detail the key new experiments or modifications of existing experiments that should be absolutely required to validate study conclusions.

Generally, there should be no more than 3 such required experiments or major modifications for a "Major Revision" recommendation. If more than 3 experiments are necessary to validate the study conclusions, then you are encouraged to recommend "Reject".

Reviewer #1: (No Response)

Reviewer #2: (No Response)

Reviewer #3: (No Response)

**********

Part III – Minor Issues: Editorial and Data Presentation Modifications

Please use this section for editorial suggestions as well as relatively minor modifications of existing data that would enhance clarity.

Reviewer #1: (No Response)

Reviewer #2: In response to Reviewer #2 Comment 2, the authors suggest that the GFP-signal does not need to be quantified or more clearly demonstrated to be exclusive to female gametocytes. I think it is good scientific practice to confirm any imaging with Western Blot analysis. Part of establishing confidence in the genetic line that has been created and reagents used in the ChIP-seq is first proving that the full-length GFP-tagged PbAP2-FG2 protein is exclusively expressed in the female gametocyte and that it is cleanly identified by the antibody used for the ChIP-seq by showing a Western Blot.

In response to Comment 13, I thank the authors for performing the additional RIME experiment that empirically establishes the interaction of PbAP2-FG2 and PbAP2R-2. Could the authors speculate as to why AP2-I, AP2-G2 and AP2-O2 are also found to complex with PbAP2-FG2? This should be added to the discussion of the results of RIME. Additionally, please discuss your results with respect to the recently published P. berghei male/female gametoycte differentiation and maturation ssRNA-seq study https://doi.org/10.1016/j.chom.2022.12.011.

In response to Comment 14, I thank you for the adding the Venn diagram which shows an overlap of peaks between the two ChIP-seq experiments. While we understand your justification for presentation of data, if your data analysis strictly follows ENCODE guidelines (for coverage, quality, and reproducibility), visual analysis of the quality of IP experiments should not be necessary. If you prefer to show read coverage of your IP samples, please include the read coverage from the corresponding input in each panel so that the quality of the IP experiments can be directly evaluated as you suggest.

In response to Comment 15, thank you for attempting to address our initial comment regarding the correlation plot displayed as Figure 6H. WI apologize for not being clearer in the original request. There are a couple of reasons that this analysis is statistically inappropriate. 1) The data points on the graph are not calculated entirely independent of each other. For the FC calculation, you do not have an independent WT measurement for each knockout sample. Each WT timepoint is used in the calculation of one of each knockout and, therefore, the Log2FC calculations are not completely independent of each other, and linear regression/approximation is not appropriate. (For example: What you are currently doing is A/B to C/B, where A and C are your knockout strains and B is the WT). The correct comparison would be A/B to C/D OR directly A to C. 2) The data are already filtered for statistical significance resulting in two “clusters” of data points and skewing the data toward a linear relationship based on the extreme fold changes.

There are a few things that can be done to make your analysis appropriate:

1) To draw a statistical conclusion from a correlation plot all of the data should be independent of each other. For instance, since your denominator for the Log2FC calculation per gene is the same across the knockout genotypes, you might consider just plotting the average expression value per gene with each knockout represented on a different axis, followed by a linear regression/approximation analysis. One can use a density correlation plot for large data sets.

2) Alternatively, the Log2 expression of significant genes in either knockout line can be represented on separate graphs as a box plot where the WT expression of select significant genes is separate from your knockout expression and the upward or downward trend will support/refute the hypothesis.

3) More simplistically, you could represent the significantly up or downward FC of sets of genes in both knockout strains as a Venn Diagram to demonstrate the overlap of regulated genes.

Reviewer #3: In the revised manuscript the authors now provide evidence for a direct interaction between PbAP2-FG2 and PbAP2-R2 using Rapid Immunoprecipitation Mass Spectrometry of Endogenous Proteins (RIME), which combined ChIP and MS to identify associated proteins. Although using an unrelated GFP-tagged line to monitor unspecific interactions would have been a more appropriate negative control for this experiment, the pull-down of PbAP2-FG2 detected PbAP2-R2 demonstrating that PbAP2-FG2 and PbAP2-R2 likely interact. Other proteins shown in Figure 7 appear to be more hand selected from their IP data (based on examination of Table S6), and I am less convinced by the results based solely on enrichment (over the WT control), such as PbMORC and other chromatin remodeling complexes, although these are interesting observations.

A few additional minor points:

Line 21 of the abstract should read: “Despite the number of studies on gametocyte development that have been conducted, the molecular mechanisms regulating this process remain to be fully understood.”

If PbAP2-FG2 and PbAP2R-2 function together, why does the abstract only mention “a significant overlap between the target genes of PbAP2-FG2 and AP2-G.” Why not also add PbAP2R-2 since these two proteins overlap in target gene specificity?

Should PbAP2R-2 be renamed PbAP2-FG3? Why or why not?

Line 111, please change “AP2-family transcription factor” to “ApiAP2-family transcription factor”

The authors should modify their citation of Russell et al. since it has now been published in Cell Host & Microbe and is no longer solely a bioRxiv preprint.

The authors have added additional support for the presence of an AP2 domain (at least one) in PbAP2-R2 (lines 340-346). Please include alignments of the regions that they find to not be well-conserved between Pb and Pf as supplemental data. Also, I would again encourage the authors to use AlphaFold2, which clearly demonstrates at least one well-folded AP2 domain. Given that this protein is likely to contain at least one AP2 domain, it would be good to speculate what the role of this AP2 domain might be given that the binding matches that of PbAP2-FG2?

Finally, I have an alternative title suggestion: “PbAP2-FG2 and AP2R-2 function together as an essential transcriptional repressor complex during female gametocyte development in Plasmodium”

**********

PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: Yes: HAKIMI Mohamed-Ali

Reviewer #2: No

Reviewer #3: No

Figure Files:

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email us at figures@plos.org.

Data Requirements:

Please note that, as a condition of publication, PLOS' data policy requires that you make available all data used to draw the conclusions outlined in your manuscript. Data must be deposited in an appropriate repository, included within the body of the manuscript, or uploaded as supporting information. This includes all numerical values that were used to generate graphs, histograms etc.. For an example see here: http://www.plosbiology.org/article/info%3Adoi%2F10.1371%2Fjournal.pbio.1001908#s5.

Reproducibility:

To enhance the reproducibility of your results, we recommend that you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option to publish peer-reviewed clinical study protocols. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols

References:

Please review your reference list to ensure that it is complete and correct. If you have cited papers that have been retracted, please include the rationale for doing so in the manuscript text, or remove these references and replace them with relevant current references. Any changes to the reference list should be mentioned in the rebuttal letter that accompanies your revised manuscript. If you need to cite a retracted article, indicate the article’s retracted status in the References list and also include a citation and full reference for the retraction notice.

Attachment

Submitted filename: PPATHOGENS-D-22-01649_R1_responses.docx

Decision Letter 2

Kami Kim, Ron Dzikowski

2 Feb 2023

Dear Dr. Yuda,

We are pleased to inform you that your manuscript 'PbAP2-FG2 and PbAP2R-2 function together as a transcriptional repressor complex essential for Plasmodium female development' has been provisionally accepted for publication in PLOS Pathogens.

Before your manuscript can be formally accepted you will need to complete some formatting changes, which you will receive in a follow up email. A member of our team will be in touch with a set of requests.

Please note that your manuscript will not be scheduled for publication until you have made the required changes, so a swift response is appreciated.

IMPORTANT: The editorial review process is now complete. PLOS will only permit corrections to spelling, formatting or significant scientific errors from this point onwards. Requests for major changes, or any which affect the scientific understanding of your work, will cause delays to the publication date of your manuscript.

Should you, your institution's press office or the journal office choose to press release your paper, you will automatically be opted out of early publication. We ask that you notify us now if you or your institution is planning to press release the article. All press must be co-ordinated with PLOS.

Thank you again for supporting Open Access publishing; we are looking forward to publishing your work in PLOS Pathogens.

Best regards,

Ron Dzikowski

Academic Editor

PLOS Pathogens

Kami Kim

Section Editor

PLOS Pathogens

Kasturi Haldar

Editor-in-Chief

PLOS Pathogens

orcid.org/0000-0001-5065-158X

Michael Malim

Editor-in-Chief

PLOS Pathogens

orcid.org/0000-0002-7699-2064

***********************************************************

Reviewer Comments (if any, and for reference):

Acceptance letter

Kami Kim, Ron Dzikowski

7 Feb 2023

Dear Dr. Yuda,

We are delighted to inform you that your manuscript, "PbAP2-FG2 and PbAP2R-2 function together as a transcriptional repressor complex essential for Plasmodium female development," has been formally accepted for publication in PLOS Pathogens.

We have now passed your article onto the PLOS Production Department who will complete the rest of the pre-publication process. All authors will receive a confirmation email upon publication.

The corresponding author will soon be receiving a typeset proof for review, to ensure errors have not been introduced during production. Please review the PDF proof of your manuscript carefully, as this is the last chance to correct any scientific or type-setting errors. Please note that major changes, or those which affect the scientific understanding of the work, will likely cause delays to the publication date of your manuscript. Note: Proofs for Front Matter articles (Pearls, Reviews, Opinions, etc...) are generated on a different schedule and may not be made available as quickly.

Soon after your final files are uploaded, the early version of your manuscript, if you opted to have an early version of your article, will be published online. The date of the early version will be your article's publication date. The final article will be published to the same URL, and all versions of the paper will be accessible to readers.

Thank you again for supporting open-access publishing; we are looking forward to publishing your work in PLOS Pathogens.

Best regards,

Kasturi Haldar

Editor-in-Chief

PLOS Pathogens

orcid.org/0000-0001-5065-158X

Michael Malim

Editor-in-Chief

PLOS Pathogens

orcid.org/0000-0002-7699-2064

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Fig. An Integrative Genomics Viewer (IGV) image showing peaks identified in ChIP-seq analysis of PbAP2-G and PbAP2-FG in the upstream region of pbap2-fg2.

    The grey bar indicates the gene body of pbap2-fg2.

    (TIF)

    S2 Fig. Genotyping of transgenic parasites developed in this study.

    (A) PbAP2-FG2::GFP. (B) pbap2-fg2(-). (C) PbAP2-FG2::GFPC. (D) PbAP2R-2::GFPC.

    (TIF)

    S3 Fig. Alignment of amino acid sequences for previously reported putative AP2 domains for PbAP2R-2 and PfAP2R-2.

    The sequences were aligned using the ClustalW program in Mega X. Asterisks indicate amino acids conserved between the two species.

    (TIF)

    S4 Fig. Heat maps showing coverage in both IP and input samples for ChIP-seq of PbAP2-FG2 at PbAP2R-2 peaks (left) and for ChIP-seq of PbAP2R-2 at PbAP2-FG2 peaks (right).

    Peak regions are aligned in ascending order of their fold enrichment value.

    (TIF)

    S1 Table. List of differentially expressed genes in pbap2-fg2(-).

    (A) RPKM values in each data. (B) Significantly downregulated genes. (C) Significantly upregulated genes.

    (XLSX)

    S2 Table. List of sex-enriched genes.

    (A) Female-enriched genes. (B) Male-enriched genes.

    (XLSX)

    S3 Table. List of peaks and target genes identified in the ChIP-seq experiments of PbAP2-FG2.

    (A) Peaks in Experiment 1. (B) Peaks in Experiment 2. (C) Target genes.

    (XLSX)

    S4 Table. List of peaks identified in the ChIP-seq experiments of PbAP2R-2.

    (A) Peaks in Experiment 1. (B) Peaks in Experiment 2.

    (XLSX)

    S5 Table. List of differentially expressed genes in pbap2r-2(-).

    (A) RPKM values in each data. (B) Significantly downregulated genes. (C) Significantly upregulated genes.

    (XLSX)

    S6 Table. List of proteins identified in the RIME using PbAP2-FG2::GFP and WT.

    (XLSX)

    S7 Table. List of peaks and target genes identified in the ChIP-seq experiments of PyAP2-O3.

    (A) Peaks in Experiment 1. (B) Peaks in Experiment 2. (C) Target genes.

    (XLSX)

    S8 Table. List of differentially expressed genes in pyap2-o3-null parasite.

    (A) FPKM values in each data. (B) Significantly downregulated genes. (C) Significantly upregulated genes.

    (XLSX)

    S9 Table. List of primers used in this study.

    (XLSX)

    Attachment

    Submitted filename: Responce_Letter.pdf

    Attachment

    Submitted filename: PPATHOGENS-D-22-01649_R1_responses.docx

    Attachment

    Submitted filename: Responce_Letter2.docx

    Data Availability Statement

    The authors confirm that all data underlying the findings are fully available without restriction. All fastq files for ChIP-seq and RNA-seq experiments are available from the GEO database (accession numbers GSE198588, GSE213776).


    Articles from PLOS Pathogens are provided here courtesy of PLOS

    RESOURCES