Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

The transcriptional regulator HDP1 controls expansion of the inner membrane complex during early sexual differentiation of malaria parasites

A Publisher Correction to this article was published on 08 February 2022

This article has been updated

Abstract

Transmission of Plasmodium falciparum and other malaria parasites requires their differentiation from asexual blood stages into gametocytes, the non-replicative sexual stage necessary to infect the mosquito vector. This transition involves changes in gene expression and chromatin reorganization that result in the activation and silencing of stage-specific genes. However, the genomes of malaria parasites have been noted for their limited number of transcriptional and chromatin regulators, and the molecular mediators of these changes remain largely unknown. We recently identified homeodomain protein 1 (HDP1) as a DNA-binding protein, first expressed in gametocytes, that enhances the expression of key genes critical for early sexual differentiation. The discovery of HDP1 marks a new class of transcriptional regulator in malaria parasites outside of the better-characterized ApiAP2 family. Here, using molecular biology, biochemistry and microscopy techniques, we show that HDP1 is essential for gametocyte maturation, facilitating the necessary upregulation of inner membrane complex components during early gametocytogenesis that gives P. falciparum gametocytes their characteristic shape.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Loss of HDP1 function disrupts gametocyte maturation.
Fig. 2: HDP1 is a chromatin-associated protein expressed in gametocytes.
Fig. 3: Disruption of HDP1 results in leaky expression of heterochromatin-associated genes and reduced expression of IMC genes in early gametocytes.
Fig. 4: HDP1 binds near the TSS of genes expressed in early gametocytes.
Fig. 5: HDP1 is essential for expansion of the IMC in early gametocytes.

Similar content being viewed by others

Data availability

Processed and raw high-throughput sequencing data have been deposited in the NCBI Gene Expression Omnibus under accession nos. GSE189197 (RNA-seq) and GSE189151 (ChIP–seq). The pipelines for RNA-seq and ChIP–seq data analysis are available at https://github.com/KafsackLab/HDP1. All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Information. Additional data related to this paper may be requested from the authors. P. falciparum lines generated in this study are available at cost on request. Source data are provided with this paper.

Change history

References

  1. Park, B. O., Ahrends, R. & Teruel, M. N. Consecutive positive feedback loops create a bistable switch that controls preadipocyte-to-adipocyte conversion. Cell Rep. 2, 976–990 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Bhattacharya, S. et al. A bistable switch underlying B-cell differentiation and its disruption by the environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol. Sci. 115, 51–65 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Norman, T. M., Lord, N. D., Paulsson, J. & Losick, R. Stochastic switching of cell fate in microbes. Annu. Rev. Microbiol. 69, 381–403 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Satory, D., Gordon, A. J., Halliday, J. A. & Herman, C. Epigenetic switches: can infidelity govern fate in microbes? Curr. Opin. Microbiol. 14, 212–217 (2011).

    Article  CAS  PubMed  Google Scholar 

  5. Kafsack, B. F. C. et al. A transcriptional switch underlies commitment to sexual development in malaria parasites. Nature 507, 248–252 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Sinha, A. et al. A cascade of DNA-binding proteins for sexual commitment and development in Plasmodium. Nature 507, 253–257 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lopez-Rubio, J. J., Mancio-Silva, L. & Scherf, A. Genome-wide analysis of heterochromatin associates clonally variant gene regulation with perinuclear repressive centers in malaria parasites. Cell Host Microbe 5, 179–190 (2009).

    Article  CAS  PubMed  Google Scholar 

  8. Brancucci, N. M. B. et al. Heterochromatin protein 1 secures survival and transmission of malaria parasites. Cell Host Microbe 16, 165–176 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. Poran, A. et al. Single-cell RNA sequencing reveals a signature of sexual commitment in malaria parasites. Nature 551, 95–99 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Josling, G. A. et al. Dissecting the role of PfAP2-G in malaria gametocytogenesis. Nat. Commun. 11, 1503 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Coleman, B. I. et al. A Plasmodium falciparum histone deacetylase regulates antigenic variation and gametocyte conversion. Cell Host Microbe 16, 177–186 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Filarsky, M. et al. GDV1 induces sexual commitment of malaria parasites by antagonizing HP1-dependent gene silencing. Science 359, 1259–1263 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Bunnik, E. M. et al. Changes in genome organization of parasite-specific gene families during the Plasmodium transmission stages. Nat. Commun. 9, 1910 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Fraschka, S. A. et al. Comparative heterochromatin profiling reveals conserved and unique epigenome signatures linked to adaptation and development of malaria parasites. Cell Host Microbe 23, 407–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Bancells, C. et al. Revisiting the initial steps of sexual development in the malaria parasite Plasmodium falciparum. Nat. Microbiol. 4, 144–154 (2019).

    Article  CAS  PubMed  Google Scholar 

  16. Young, J. A. et al. The Plasmodium falciparum sexual development transcriptome: a microarray analysis using ontology-based pattern identification. Mol. Biochem Parasitol. 143, 67–79 (2005).

    Article  CAS  PubMed  Google Scholar 

  17. Painter, H. J., Carrasquilla, M. & Llinás, M. Capturing in vivo RNA transcriptional dynamics from the malaria parasite Plasmodium falciparum. Genome Res. 27, 1074–1086 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. van Biljon, R. et al. Hierarchical transcriptional control regulates Plasmodium falciparum sexual differentiation. BMC Genomics 20, 920 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Kent, R. S. et al. Inducible developmental reprogramming redefines commitment to sexual development in the malaria parasite Plasmodium berghei. Nat. Microbiol. 3, 1206–1213 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Balaji, S., Babu, M. M., Iyer, L. M. & Aravind, L. Discovery of the principal specific transcription factors of Apicomplexa and their implication for the evolution of the AP2-integrase DNA binding domains. Nucleic Acids Res. 33, 3994–4006 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Modrzynska, K. et al. A knockout screen of ApiAP2 genes reveals networks of interacting transcriptional regulators controlling the Plasmodium life cycle. Cell Host Microbe 21, 11–22 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yuda, M., Iwanaga, S., Kaneko, I. & Kato, T. Global transcriptional repression: an initial and essential step for Plasmodium sexual development. Proc. Natl Acad. Sci. USA 112, 12824–12829 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Singh, S. et al. The PfAP2-G2 transcription factor is a critical regulator of gametocyte maturation. Mol. Microbiol. 115, 1005–1024 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Zhang, C. et al. Systematic CRISPR-Cas9-mediated modifications of Plasmodium yoelii ApiAP2 genes reveal functional insights into parasite development. mBio 8, e01986-17 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Yuda, M., Kaneko, I., Iwanaga, S., Mura, Y. & Kato, T. Female‐specific gene regulation in malaria parasites by an AP2‐family transcription factor. Mol. Microbiol. 113, 40–51 (2019).

    Article  PubMed  Google Scholar 

  26. Bürglin, T. R. & Affolter, M. Homeodomain proteins: an update. Chromosoma 125, 497–521 (2016).

    Article  PubMed  Google Scholar 

  27. Llorà-Batlle, O. et al. Conditional expression of PfAP2-G for controlled massive sexual conversion in Plasmodium falciparum. Sci. Adv. 6, eaaz5057 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Prommana, P. et al. Inducible knockdown of Plasmodium gene expression using the glmS ribozyme. PLoS ONE 8, e73783 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Nixon, C. P. et al. Antibodies to PfsEGXP, an early gametocyte-enriched phosphoprotein, predict decreased Plasmodium falciparum gametocyte density in humans. J. Infect. Dis. 218, 1792–1801 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Pradhan, L. et al. Crystal structure of the human NKX2.5 homeodomain in complex with DNA target. Biochemistry 51, 6312–6319 (2012).

    Article  CAS  PubMed  Google Scholar 

  31. Parkyn Schneider, M. et al. Disrupting assembly of the inner membrane complex blocks Plasmodium falciparum sexual stage development. PLoS Pathog. 13, e1006659 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Flueck, C. et al. A major role for the Plasmodium falciparum ApiAP2 protein PfSIP2 in chromosome end biology. PLoS Pathog. 6, e1000784 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Campbell, T. L., De Silva, E. K., Olszewski, K. L., Elemento, O. & Llinás, M. Identification and genome-wide prediction of DNA binding specificities for the ApiAP2 family of regulators from the malaria parasite. PLoS Pathog. 6, e1001165 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Santos, J. M. et al. Red blood cell invasion by the malaria parasite is coordinated by the PfAP2-I transcription factor. Cell Host Microbe 21, 731–741 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Brodsky, S. et al. Intrinsically disordered regions direct transcription factor in vivo binding specificity. Mol. Cell 79, 459–471 (2020).

    Article  CAS  PubMed  Google Scholar 

  36. Furuya, T. et al. Disruption of a Plasmodium falciparum gene linked to male sexual development causes early arrest in gametocytogenesis. Proc. Natl Acad. Sci. USA 102, 16813–16818 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hedgethorne, K. et al. Homeodomain-like DNA binding proteins control the haploid-to-diploid transition in Dictyostelium. Sci. Adv. 3, e1602937 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  38. López-Barragán, M. J. et al. Directional gene expression and antisense transcripts in sexual and asexual stages of Plasmodium falciparum. BMC Genomics 12, 587 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Bushell, E. et al. Functional profiling of a Plasmodium genome reveals an abundance of essential genes. Cell 170, 260–272 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Moll, K., Ljungström, I., Perlmann, H. & Scherf, A. Methods in Malaria Research (MR4/ATCC, 2008).

  41. Morlon-Guyot, J. et al. A proteomic analysis unravels novel CORVET and HOPS proteins involved in Toxoplasma gondii secretory organelles biogenesis. Cell Microbiol. 20, e12870 (2018).

    Article  PubMed  Google Scholar 

  42. Rug, M. & Maier, A. G. in Malaria (ed. Menard R.) 75–98 (Humana Press, 2012).

  43. Ghorbal, M. et al. Genome editing in the human malaria parasite Plasmodium falciparum using the CRISPR-Cas9 system. Nat. Biotechnol. 32, 819–821 (2014).

    Article  CAS  PubMed  Google Scholar 

  44. Tanaka, T. Q. & Williamson, K. C. A malaria gametocytocidal assay using oxidoreduction indicator, alamarBlue. Mol. Biochem. Parasitol. 177, 160–163 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Flueck, C. et al. Plasmodium falciparum heterochromatin protein 1 marks genomic loci linked to phenotypic variation of exported virulence factors. PLoS Pathog. 5, e1000569 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Saini, E. et al. Photosensitized INA-labelled protein 1 (PhIL1) is novel component of the inner membrane complex and is required for Plasmodium parasite development. Sci. Rep. 7, 15577 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Mehnert, A.-K., Simon, C. S. & Guizetti, J. Immunofluorescence staining protocol for STED nanoscopy of Plasmodium-infected red blood cells. Mol. Biochem. Parasitol. 229, 47–52 (2019).

    Article  CAS  PubMed  Google Scholar 

  48. RR, B. Use of polyethyleneimine in purification of DNA-binding proteins. Meth. Enzymol. 208, 3–10 (1991).

    Article  Google Scholar 

  49. Berger, M. F. et al. Compact, universal DNA microarrays to comprehensively determine transcription-factor binding site specificities. Nat. Biotechnol. 24, 1429–1435 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Berger, M. F. & Bulyk, M. L. Universal protein-binding microarrays for the comprehensive characterization of the DNA-binding specificities of transcription factors. Nat. Protoc. 4, 393–411 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 17, 10–12 (2011).

    Article  Google Scholar 

  52. Warrenfeltz, S. et al. EuPathDB: the Eukaryotic Pathogen Genomics Database Resource. Methods Mol. Biol. 1757, 69–113 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Trapnell, C. et al. Differential analysis of gene regulation at transcript resolution with RNA-seq. Nat. Biotechnol. 31, 46–53 (2013).

    Article  CAS  PubMed  Google Scholar 

  55. Korotkevich, G., Sukhov, V. & Sergushichev, A. Fast gene set enrichment analysis. Preprint at bioRxiv https://www.biorxiv.org/content/10.1101/060012v3 (2021).

  56. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Li, H. et al. The sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Lawrence, M. et al. Software for computing and annotating genomic ranges. PLoS Comput. Biol. 9, e1003118 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Hahne, F. & Ivanek, R. Visualizing genomic data using Gviz and Bioconductor. Methods Mol. Biol. 1418, 335–351 (2016).

    Article  PubMed  Google Scholar 

  61. Zhu, L. J. et al. ChIPpeakAnno: a Bioconductor package to annotate ChIP-seq and ChIP-chip data. BMC Bioinformatics 11, 237 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Huber, W. et al. Orchestrating high-throughput genomic analysis with Bioconductor. Nat. Methods 12, 115–121 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank P. Malhotra for the generous gift of anti-PhIL1 antibodies, V. Carruthers for the generous gift of anti-Ty1 antibodies and the Weill Cornell Medicine genomics core for technical support; and K. Deitsch and J. King for valuable feedback on the manuscript. This work was supported by startup funds from Weill Cornell Medicine: to B.F.C.K., nos. 1R01 AI141965 and 1R01 AI138499; to M.L., no. 1R01 AI125565; and to K.G.L.R., nos. 1R01 AI136511 and R21 AI142506-01; by the University of California, Riverside (no. NIFA-Hatch-225935, to K.G.L.R.); and by support from the Mathers Foundation (to D.J.P.).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization was the responsibility of B.F.C.K. Methodology was performed by B.F.C.K., R.A.C.M., W.X. and K.G.L.R. R.A.C.M., X.T., W.X., L.M.O. and W.D. carried out investigations. Software, formal analysis and data curation were provided by B.F.C.K. R.A.C.M. wrote the original draft. B.F.C.K. and R.A.C.M. wrote, reviewed and edited the article. Visualization was conducted by R.A.C.M. and B.F.C.K. B.F.C.K., K.G.L.R., M.L. and D.J.P. supervised the project. Project administration and funding acquisition were carried out by B.F.C.K.

Corresponding author

Correspondence to Björn F. C. Kafsack.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review information

Nature Microbiology thanks Leann Tilley and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 The predicted DNA-binding protein HDP1 is expressed in gametocytes.

(a) The single exon locus Pf3D7_1466200 encodes a large 3078aa protein with a predicted C-terminal Helix-Turn-Helix DNA-binding domain (DBD) and two nuclear localization signals (NLS). Multiple AP2-G binding sites are found in its 2 kb promoter region. Black bracket indicates the antigen used for generating antisera used in29. (b) Alignment of the helix-turn-helix domain for homologs from other apicomplexan parasites. (c) Quantitative RT-PCR of hdp1 transcripts found minimal expression in asexual blood stages with upregulation during gametocyte development (data are presented as mean values of n = 2 biologically independent samples). Transcript levels were normalized to Seryl-tRNA synthetase.

Source data

Extended Data Fig. 2 Validation of engineered parasite lines.

(a) Generation of halo-hdp1 parasites by CAS9 genome editing. Insertion of the N-terminal HALO tag at the 5’ end of hdp1 coding sequence was confirmed by PCR and checked for mutations by Sanger sequencing of the 3.8 kb PCR product (not shown). (b) Generation of hdp1-gfp parasites by CAS9 genome editing. Insertion of the C-terminal GFP tag at the 3’ end the hdp1 coding sequence was confirmed by PCR and checked for mutations by Sanger sequencing of the 1.9 kb PCR product (not shown). (c) Generation of hdp1-Ty1 parasites by CAS9 genome editing. Insertion of the C-terminal triple Ty1 epitope tag at the 3’ end the hdp1 coding sequence was confirmed by PCR and checked for mutations by Sanger sequencing of the 1.1 kb PCR product (not shown). (d) Generation of ∆hdp1 parasites by CAS9 genome editing. Replacement of 1.4 kb flanking the hdp1 start codon by a hDHFR selectable marker cassette was confirmed by PCR. (e) Generation of hdp1-glmS parasites by CAS9 genome editing. Insertion of the C-terminal triple Ty1 epitope tag and the glmS ribozyme at the 3’ end the hdp1 coding sequence was confirmed by PCR and checked for mutations by Sanger sequencing of the 1.4 kb PCR product (not shown).

Source data

Extended Data Fig. 3 Loss of HDP1 does not alter the sexual commitment frequency or Stage I gametocyte viability.

(a) The sexual commitment frequency (day 5 gametocytes per day 1 ring stages) is not significantly affected in halo-hdp1 and ∆hdp1 parasites. Data are presented as mean values ± s.e.m. of n = 3 biologically independent samples. (b) Mitochondrial membrane potential of gametocytes (GCs), as measured by DilC(1)5 staining, indicates similar viability on day 2, but not days 5 or 10, for NF54 (orange) and ∆hdp1 (green) gametocytes. Flow cytometry plots are representative of n = 2 biologically independet samples.

Source data

Extended Data Fig. 4 Inducible knockdown of HDP1 reduces gametocyte maturation in early but not late gametocytes.

(a) Percentage of falciform gametocytes on Day 10 in response to 5 mM glucosamine on days 1-5, 3-8, 5-10, or in the absence of glucosamine for hdp1-Ty1 or hdp1-glmS parasites. Data are presented as mean values ± s.e.m of n = 2 biologically independent samples. (b) Representative morphology of hdp1-glmS gametocytes in response to 5 mM glucosamine on days 1-5, 3-8, 5-10, or in the absence of glucosamine, among two independent experiments. Scale bar, 3 μm.

Source data

Extended Data Fig. 5 Immunofluorescence microscopy localizes the HA-tagged ortholog TGME49_233160 to the nucleus of Toxoplasma gondii tachyzoites.

Staining of TgME49_233160-HA intracellular tachyzoites with anti-HA (TgME49_23316, green), anti-GAP45 (inner membrane complex, red), and DAPI (DNA, blue). Scale bar, 5 μm. Image is representative of n = 2 independent experiments.

Extended Data Fig. 6 HDP1-GFP localized to the nucleus of hdp1-gfp gametocytes.

No signal was observed in hdp1-gfp asexual blood stages or gametocytes of the untagged NF54 parent line. Exposure and brightness/contrast settings are uniform across the images shown. Images are representative of n = 2 biologically independent experiments. Scale bar, 3 µm.

Extended Data Fig. 7 The HDP1 DNA-binding domain tandem GC-rich motif.

(a) Coomassie stain of the recombinant GST-HDP1-DBD used for PBM analysis. (b) The three most highly enriched DNA motifs for the GST-HDP1 DBD domain on the protein-binding microarray. (c) Top three secondary motif hits after removal of primary hits.

Source data

Extended Data Fig. 8 Genes encoding inner membrane complex genes with significantly reduced expression in HDP1 knockout parasites have upstream HDP1 binding sites.

Remaining HDP1 binding sites upstream of genes encoding inner membrane complex proteins. Histogram track shows the significance of enrichment by position. Regions of significant enrichment are shown as boxes with black vertical lines indicating peak summits within each peak. Instances of Motif A, Motif B, or overlapping motifs within peaks are shown in red, blue and purple, respectively. Genes encoded in forward or reverse orientation are shown in blue or red, respectively. Combined estimate of two biologically independent samples.

Extended Data Fig. 9 Gating schema for viable gametocytes.

Populations were gated for single cells based on forward (FSC) and side scatter (SSC). Viable gametocytes were identified based on DNA content and mitochondrial membrane potential based on Hoechst33342 and DilC(1)5 staining.

Supplementary information

Reporting Summary

Supplementary Data

Processed RNA-seq and ChIP–seq data.

Supplementary Table 1

List of primers used.

Source data

Source Data Fig. 1

Numerical source data.

Source Data Fig. 1

Unprocessed immunoblots.

Source Data Fig. 2

Unprocessed immunoblots.

Source Data Fig. 4

Unprocessed EMSA.

Source Data Fig. 5

Numerical source data.

Source Data Fig. 5

Unprocessed immunoblots.

Source Data Extended Data Fig. 1

Numerical source data.

Source Data Extended Data Fig. 2

Unprocessed agarose gels.

Source Data Extended Data Fig. 3

Numerical source data.

Source Data Extended Data Fig. 4

Numerical source data.

Source Data Extended Data Fig. 7

Unprocessed acrylamide gel stained with Coomassie blue.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Campelo Morillo, R.A., Tong, X., Xie, W. et al. The transcriptional regulator HDP1 controls expansion of the inner membrane complex during early sexual differentiation of malaria parasites. Nat Microbiol 7, 289–299 (2022). https://doi.org/10.1038/s41564-021-01045-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-021-01045-0

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing