1
|
Musabyimana JP, Musa S, Manti J, Distler U, Tenzer S, Ngwa CJ, Pradel G. The Plasmodium falciparum histone methyltransferase SET10 participates in a chromatin modulation network crucial for intraerythrocytic development. mSphere 2024:e0049524. [PMID: 39445823 DOI: 10.1128/msphere.00495-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/17/2024] [Indexed: 10/25/2024] Open
Abstract
The lifecycle progression of the malaria parasite Plasmodium falciparum requires precise tuning of gene expression including histone methylation. The histone methyltransferase PfSET10 was previously described as an H3K4 methyltransferase involved in var gene regulation, making it a prominent antimalarial target. In this study, we investigated the role of PfSET10 in the blood stages of P. falciparum in more detail, using tagged PfSET10-knockout (KO) and -knockdown (KD) lines. We demonstrate a nuclear localization of PfSET10 with peak protein levels in schizonts. PfSET10 deficiency reduces intraerythrocytic growth but has no effect on gametocyte commitment and maturation. Screening of the PfSET10-KO line for histone methylation variations reveals that lack of PfSET10 renders the parasites unable to mark H3K18me1, while no reduction in the H3K4 methylation status could be observed. Comparative transcriptomic profiling of PfSET10-KO schizonts shows an upregulation of transcripts particularly encoding proteins linked to red blood cell remodeling and antigenic variation, suggesting a repressive function of the histone methylation mark. TurboID coupled with mass spectrometry further highlights an extensive nuclear PfSET10 interaction network with roles in transcriptional regulation and mRNA processing, DNA replication and repair, and chromatin remodeling. The main interactors of PfSET10 include ApiAP2 transcription factors, epigenetic regulators like PfHDAC1, chromatin modulators like PfMORC and PfISWI, mediators of RNA polymerase II, and DNA replication licensing factors. The combined data pinpoint PfSET10 as a histone methyltransferase essential for H3K18 methylation that regulates nucleic acid metabolic processes in the P. falciparum blood stages as part of a comprehensive chromatin modulation network.IMPORTANCEThe fine-tuned regulation of DNA replication and transcription is particularly crucial for the rapidly multiplying blood stages of malaria parasites and proteins involved in these processes represent important drug targets. This study demonstrates that contrary to previous reports the histone methyltransferase PfSET10 of the malaria parasite Plasmodium falciparum promotes the methylation of histone 3 at lysine K18, a histone mark to date not well understood. Deficiency of PfSET10 due to genetic knockout affects genes involved in intraerythrocytic development. Furthermore, in the nuclei of blood-stage parasites, PfSET10 interacts with various protein complexes crucial for DNA replication, remodeling, and repair, as well as for transcriptional regulation and mRNA processing. In summary, this study highlights PfSET10 as a methyltransferase affecting H3K18 methylation with critical functions in chromatin maintenance during the development of P. falciparum in red blood cells.
Collapse
Affiliation(s)
| | - Sherihan Musa
- Division of Cellular and Applied Infection Biology, RWTH Aachen University, Aachen, Germany
| | - Janice Manti
- Division of Cellular and Applied Infection Biology, RWTH Aachen University, Aachen, Germany
| | - Ute Distler
- Institute of Immunology, University Medical Centre of the Johannes-Gutenberg University, Mainz, Germany
| | - Stefan Tenzer
- Institute of Immunology, University Medical Centre of the Johannes-Gutenberg University, Mainz, Germany
| | - Che Julius Ngwa
- Division of Cellular and Applied Infection Biology, RWTH Aachen University, Aachen, Germany
| | - Gabriele Pradel
- Division of Cellular and Applied Infection Biology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
2
|
Bansal A, Sharma M, Choudhury H. Generation of a new DiCre expressing parasite strain for functional characterization of Plasmodium falciparum genes in blood stages. Sci Rep 2024; 14:24076. [PMID: 39402380 PMCID: PMC11473785 DOI: 10.1038/s41598-024-75657-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Conditional regulation is a highly beneficial system for studying the function of essential genes in Plasmodium falciparum and dimerizable Cre recombinase (DiCre) is a recently adapted conditional regulation system suitable for this purpose. In the DiCre system, two inactive fragments of Cre are reconstituted to form a functionally active enzyme in the presence of rapamycin. Different loci have been targeted to generate parasite lines that express the DiCre enzyme. Here, we have used marker-free CRISPR-Cas9 gene editing to integrate the DiCre cassette in a redundant cg6 locus. We have shown the utility of the newly generated ∆cg6DC4 parasites in mediating robust, rapid, and highly specific excision of exogenously encoded gfp sequence. The ∆cg6DC4 parasites are also capable of conditional excision of an endogenous parasite gene, PF3D7_1246000. Conditional deletion of PF3D7_1246000 did not cause any inhibition in the asexual proliferation of the parasites. Furthermore, the health and morphology of the mutant parasites were comparable to that of the control parasites in Giemsa smears. The availability of another stable DiCre parasite strain competent for conditional excision of target genes will expedite functional characterization and validation of novel drug and vaccine targets against malaria.
Collapse
Affiliation(s)
- Abhisheka Bansal
- Molecular Parasitology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| | - Manish Sharma
- Molecular Parasitology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Himashree Choudhury
- Molecular Parasitology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
3
|
Blackwell AM, Jami-Alahmadi Y, Nasamu AS, Kudo S, Senoo A, Slam C, Tsumoto K, Wohlschlegel JA, Caaveiro JMM, Goldberg DE, Sigala PA. Malaria parasites require a divergent heme oxygenase for apicoplast gene expression and biogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596652. [PMID: 38853871 PMCID: PMC11160694 DOI: 10.1101/2024.05.30.596652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Malaria parasites have evolved unusual metabolic adaptations that specialize them for growth within heme-rich human erythrocytes. During blood-stage infection, Plasmodium falciparum parasites internalize and digest abundant host hemoglobin within the digestive vacuole. This massive catabolic process generates copious free heme, most of which is biomineralized into inert hemozoin. Parasites also express a divergent heme oxygenase (HO)-like protein (PfHO) that lacks key active-site residues and has lost canonical HO activity. The cellular role of this unusual protein that underpins its retention by parasites has been unknown. To unravel PfHO function, we first determined a 2.8 Å-resolution X-ray structure that revealed a highly α-helical fold indicative of distant HO homology. Localization studies unveiled PfHO targeting to the apicoplast organelle, where it is imported and undergoes N-terminal processing but retains most of the electropositive transit peptide. We observed that conditional knockdown of PfHO was lethal to parasites, which died from defective apicoplast biogenesis and impaired isoprenoid-precursor synthesis. Complementation and molecular-interaction studies revealed an essential role for the electropositive N-terminus of PfHO, which selectively associates with the apicoplast genome and enzymes involved in nucleic acid metabolism and gene expression. PfHO knockdown resulted in a specific deficiency in levels of apicoplast-encoded RNA but not DNA. These studies reveal an essential function for PfHO in apicoplast maintenance and suggest that Plasmodium repurposed the conserved HO scaffold from its canonical heme-degrading function in the ancestral chloroplast to fulfill a critical adaptive role in organelle gene expression.
Collapse
Affiliation(s)
| | | | - Armiyaw S. Nasamu
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
| | - Shota Kudo
- Department of Chemistry & Biotechnology, The University of Tokyo, Tokyo, Japan
| | - Akinobu Senoo
- Department of Protein Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Celine Slam
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT
| | - Kouhei Tsumoto
- Department of Chemistry & Biotechnology, The University of Tokyo, Tokyo, Japan
- Department of Bioengineering, University of Tokyo, Tokyo, Japan
| | | | - Jose M. M. Caaveiro
- Department of Chemistry & Biotechnology, The University of Tokyo, Tokyo, Japan
| | - Daniel E. Goldberg
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
| | - Paul A. Sigala
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
4
|
Suwanakitti N, Talawanich Y, Vanichtanankul J, Taweechai S, Yuthavong Y, Kamchonwongpaisan S, Kongkasuriyachai D. folA thyA knockout E. coli as a suitable surrogate model for evaluation of antifolate sensitivity against PfDHFR-TS. Acta Trop 2024; 258:107360. [PMID: 39142549 DOI: 10.1016/j.actatropica.2024.107360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/06/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
A new superior bacteria complementation model was achieved for testing antifolate compounds and investigating antifolate resistance in the dihydrofolate reductase (DHFR) enzyme of the malaria parasite. Earlier models depended on the addition of trimethoprim (TMP) to chemically suppress the host Escherichia coli (Ec) DHFR function. However, incomplete suppression of EcDHFR and potential interference of antibiotics needed to maintain plasmids for complementary gene expression can complicate the interpretations. To overcome such limitations, the folA (F) and thyA (T) genes were genetically knocked out (Δ) in E. coli BL21(DE3). The resulting EcΔFΔT cells were thymidine auxotroph where thymidine supplementation or functional complementation with heterologous DHFR-thymidylate synthase (TS) is needed to restore the loss of gene functions. When tested against pyrimethamine (PYR) and its analogs designed to target Plasmodium falciparum (Pf) DHFR-TS, the 50 % inhibitory concentration values obtained from EcΔFΔT surrogates expressing wildtype (PfTM4) or double mutant (PfK1) DHFR-TS showed strong correlations to the results obtained from the standard in vitro P. falciparum growth inhibition assay. Interestingly, while TMP had little effect on the susceptibility to PYR and analogs in EcΔFΔT expressing PfDHFR-TS, it hypersensitized the chemically knockdown E. coli BL21(DE3) expressing PfTM4 DHFR-TS but desensitized the one carrying PfK1 DHFR-TS. The low intrinsic expression level of PfTM4 in E. coli BL21(DE3) by western blot analysis may explain the hypersensitive to antifolates of chemical knockdown bacteria surrogate. These results demonstrated the usefulness of EcΔFΔT surrogate as a new tool for antifolate antimalarial screening with potential application for investigation of antifolate resistance mechanism.
Collapse
Affiliation(s)
- Nattida Suwanakitti
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Thailand Science Park, Pathumthani, Thailand
| | - Yuwadee Talawanich
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Thailand Science Park, Pathumthani, Thailand
| | - Jarunee Vanichtanankul
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Thailand Science Park, Pathumthani, Thailand
| | - Supannee Taweechai
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Thailand Science Park, Pathumthani, Thailand
| | - Yongyuth Yuthavong
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Thailand Science Park, Pathumthani, Thailand
| | - Sumalee Kamchonwongpaisan
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Thailand Science Park, Pathumthani, Thailand
| | - Darin Kongkasuriyachai
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Thailand Science Park, Pathumthani, Thailand.
| |
Collapse
|
5
|
Wyss M, Thommen BT, Kofler J, Carrington E, Brancucci NMB, Voss TS. The three Plasmodium falciparum Aurora-related kinases display distinct temporal and spatial associations with mitotic structures in asexual blood stage parasites and gametocytes. mSphere 2024; 9:e0046524. [PMID: 39235260 PMCID: PMC11423587 DOI: 10.1128/msphere.00465-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024] Open
Abstract
Aurora kinases are crucial regulators of mitotic cell cycle progression in eukaryotes. The protozoan malaria parasite Plasmodium falciparum replicates via schizogony, a specialized mode of cell division characterized by consecutive asynchronous rounds of nuclear division by closed mitosis followed by a single cytokinesis event producing dozens of daughter cells. P. falciparum encodes three Aurora-related kinases (PfARKs) that have been reported essential for parasite proliferation, but their roles in regulating schizogony have not yet been explored in great detail. Here, we engineered transgenic parasite lines expressing GFP-tagged PfARK1-3 to provide a systematic analysis of their expression timing and subcellular localization throughout schizogony as well as in the non-dividing gametocyte stages, which are essential for malaria transmission. We demonstrate that all three PfARKs display distinct and highly specific and exclusive spatiotemporal associations with the mitotic machinery. In gametocytes, PfARK3 is undetectable, and PfARK1 and PfARK2 show male-specific expression in late-stage gametocytes, consistent with their requirement for endomitosis during male gametogenesis in the mosquito vector. Our combined data suggest that PfARK1 and PfARK2 have non-overlapping roles in centriolar plaque maturation, assembly of the mitotic spindle, kinetochore-spindle attachment and chromosome segregation, while PfARK3 seems to be exquisitely involved in daughter cell cytoskeleton assembly and cytokinesis. These important new insights provide a reliable foundation for future research aiming at the functional investigation of these divergent and possibly drug-targetable Aurora-related kinases in mitotic cell division of P. falciparum and related apicomplexan parasites.IMPORTANCEMalaria parasites replicate via non-conventional modes of mitotic cell division, such as schizogony, employed by the disease-causing stages in the human blood or endomitosis during male gametogenesis in the mosquito vector. Understanding the molecular mechanisms regulating cell division in these divergent unicellular eukaryotes is not only of scientific interest but also relevant to identify potential new antimalarial drug targets. Here, we carefully examined the subcellular localization of all three Plasmodium falciparum Aurora-related kinases (ARKs), distantly related homologs of Aurora kinases that coordinate mitosis in model eukaryotes. Detailed fluorescence microscopy-based analyses revealed distinct, specific, and exclusive spatial associations for each parasite ARK with different components of the mitotic machinery and at different phases of the cell cycle during schizogony and gametocytogenesis. This comprehensive set of results closes important gaps in our fragmentary knowledge on this important group of kinases and offers a valuable source of information for future functional studies.
Collapse
Affiliation(s)
- Matthias Wyss
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- />University of Basel, Basel, Switzerland
| | - Basil T. Thommen
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- />University of Basel, Basel, Switzerland
| | - Jacob Kofler
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- />University of Basel, Basel, Switzerland
| | - Eilidh Carrington
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- />University of Basel, Basel, Switzerland
| | - Nicolas M. B. Brancucci
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- />University of Basel, Basel, Switzerland
| | - Till S. Voss
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- />University of Basel, Basel, Switzerland
| |
Collapse
|
6
|
Jain S, Narwal M, Omair Anwar M, Prakash N, Mohmmed A. Unravelling the anti-apoptotic role of Plasmodium falciparum Prohibitin-2 (PfPhb2) in maintaining mitochondrial homeostasis. Mitochondrion 2024:101956. [PMID: 39245193 DOI: 10.1016/j.mito.2024.101956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/16/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
The functional mitochondrion is vital for the propagation of the malaria parasite in the human host. Members of the SPFH protein family, Prohibitins (PHBs), are known to play crucial roles in maintaining mitochondrial homeostasis and cellular functions. Here, we have functionally characterized the homologue of the Plasmodium falciparumProhibitin-2 (PfPhb2) protein. A transgenic parasite line, generated using the selection-linked integration (SLI) strategy for C-terminal tagging, was utilized for cellular localization as well as for inducible knock-down of PfPhb2. We show that PfPhb2 localizes in the parasite mitochondrion during the asexual life cycle. Inducible knock-down of PfPhb2 by GlmS ribozyme caused no significant effect on the growth and multiplication of parasites. However, depletion of PfPhb2 under mitochondrial-specific stress conditions, induced by inhibiting the essential mitochondrial AAA-protease, ClpQ protease, results in enhanced inhibition of parasite growth, mitochondrial ROS production, mitochondrial membrane potential loss and led to mitochondrial fission/fragmentation, ultimately culminating in apoptosis-like cell-death. Further, PfPhb2 depletion renders the parasites more susceptible to mitochondrial targeting drug proguanil. These data suggest the functional involvement of PfPhb2 along with ClpQ protease in stabilization of various mitochondrial proteins to maintain mitochondrial homeostasis and functioning. Overall, we show that PfPhb2 has an anti-apoptotic role in maintaining mitochondrial homeostasis in the parasite.
Collapse
Affiliation(s)
- Shilpi Jain
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India
| | - Monika Narwal
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India
| | - Md Omair Anwar
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India
| | - Neha Prakash
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India
| | - Asif Mohmmed
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India.
| |
Collapse
|
7
|
Gangwar U, Choudhury H, Shameem R, Singh Y, Bansal A. Recent development in CRISPR-Cas systems for human protozoan diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 208:109-160. [PMID: 39266180 DOI: 10.1016/bs.pmbts.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/14/2024]
Abstract
Protozoan parasitic diseases pose a substantial global health burden. Understanding the pathogenesis of these diseases is crucial for developing intervention strategies in the form of vaccine and drugs. Manipulating the parasite's genome is essential for gaining insights into its fundamental biology. Traditional genomic manipulation methods rely on stochastic homologous recombination events, which necessitates months of maintaining the cultured parasites under drug pressure to generate desired transgenics. The introduction of mega-nucleases (MNs), zinc-finger nucleases (ZFNs), and transcription activator-like effector nucleases (TALENs) greatly reduced the time required for obtaining a desired modification. However, there is a complexity associated with the design of these nucleases. CRISPR (Clustered regularly interspaced short palindromic repeats)/Cas (CRISPR associated proteins) is the latest gene editing tool that provides an efficient and convenient method for precise genomic manipulations in protozoan parasites. In this chapter, we have elaborated various strategies that have been adopted for the use of CRISPR-Cas9 system in Plasmodium, Leishmania and Trypanosoma. We have also discussed various applications of CRISPR-Cas9 pertaining to understanding of the parasite biology, development of drug resistance mechanism, gene drive and diagnosis of the infection.
Collapse
Affiliation(s)
- Utkarsh Gangwar
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | | | - Risha Shameem
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Yashi Singh
- Department of Biosciences & Biomedical Engineering, Indian Institute of Technology, Indore, India
| | - Abhisheka Bansal
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
8
|
Hemasa A, Spry C, Mack M, Saliba KJ. Mutation of the Plasmodium falciparum Flavokinase Confers Resistance to Roseoflavin and 8-Aminoriboflavin. ACS Infect Dis 2024; 10:2939-2949. [PMID: 38920250 DOI: 10.1021/acsinfecdis.4c00289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
The riboflavin analogues, roseoflavin and 8-aminoriboflavin, inhibit malaria parasite proliferation by targeting riboflavin utilization. To determine their mechanism of action, we generated roseoflavin-resistant parasites by in vitro evolution. Relative to wild-type, these parasites were 4-fold resistant to roseoflavin and cross-resistant to 8-aminoriboflavin. Whole genome sequencing of the resistant parasites revealed a missense mutation leading to an amino acid change (L672H) in the gene coding for a putative flavokinase (PfFK), the enzyme responsible for converting riboflavin into the cofactor flavin mononucleotide (FMN). To confirm that the L672H mutation is responsible for the phenotype, we generated parasites with the missense mutation incorporated into the PfFK gene. The IC50 values for roseoflavin and 8-aminoriboflavin against the roseoflavin-resistant parasites created through in vitro evolution were indistinguishable from those against parasites in which the missense mutation was introduced into the native PfFK. We also generated two parasite lines episomally expressing GFP-tagged versions of either the wild-type or mutant forms of PfFK. We found that PfFK-GFP localizes to the parasite cytosol and that immunopurified PfFK-GFP phosphorylated riboflavin, roseoflavin, and 8-aminoriboflavin. The L672H mutation increased the KM for roseoflavin, explaining the resistance phenotype. Mutant PfFK is no longer capable of phosphorylating 8-aminoriboflavin, but its antiplasmodial activity against resistant parasites can still be antagonized by increasing the extracellular concentration of riboflavin, consistent with it also inhibiting parasite growth through competitive inhibition of PfFK. Our findings, therefore, are consistent with roseoflavin and 8-aminoriboflavin inhibiting parasite proliferation by inhibiting riboflavin phosphorylation and via the generation of toxic flavin cofactor analogues.
Collapse
Affiliation(s)
- Ayman Hemasa
- Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia
| | - Christina Spry
- Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia
| | - Matthias Mack
- Institute for Technical Microbiology, Department of Biotechnology, Mannheim University of Applied Sciences, Mannheim 68163, Germany
| | - Kevin J Saliba
- Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia
| |
Collapse
|
9
|
Li J, Shami GJ, Liffner B, Cho E, Braet F, Duraisingh MT, Absalon S, Dixon MWA, Tilley L. Disruption of Plasmodium falciparum kinetochore proteins destabilises the nexus between the centrosome equivalent and the mitotic apparatus. Nat Commun 2024; 15:5794. [PMID: 38987258 PMCID: PMC11237077 DOI: 10.1038/s41467-024-50167-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 06/24/2024] [Indexed: 07/12/2024] Open
Abstract
Plasmodium falciparum is the causative agent of malaria and remains a pathogen of global importance. Asexual blood stage replication, via a process called schizogony, is an important target for the development of new antimalarials. Here we use ultrastructure-expansion microscopy to probe the organisation of the chromosome-capturing kinetochores in relation to the mitotic spindle, the centriolar plaque, the centromeres and the apical organelles during schizont development. Conditional disruption of the kinetochore components, PfNDC80 and PfNuf2, is associated with aberrant mitotic spindle organisation, disruption of the centromere marker, CENH3 and impaired karyokinesis. Surprisingly, kinetochore disruption also leads to disengagement of the centrosome equivalent from the nuclear envelope. Severing the connection between the nucleus and the apical complex leads to the formation of merozoites lacking nuclei. Here, we show that correct assembly of the kinetochore/spindle complex plays a previously unrecognised role in positioning the nascent apical complex in developing P. falciparum merozoites.
Collapse
Affiliation(s)
- Jiahong Li
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Gerald J Shami
- School of Medical Sciences (Molecular and Cellular Biomedicine) & Australian Centre for Microscopy and Microanalysis, The University of Sydney, Sydney, NSW, Australia
| | - Benjamin Liffner
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ellie Cho
- Biological Optical Microscopy Platform, The University of Melbourne, Parkville, VIC, Australia
| | - Filip Braet
- School of Medical Sciences (Molecular and Cellular Biomedicine) & Australian Centre for Microscopy and Microanalysis, The University of Sydney, Sydney, NSW, Australia
| | - Manoj T Duraisingh
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Sabrina Absalon
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Matthew W A Dixon
- Department of Infectious Diseases, The Peter Doherty Institute, The University of Melbourne, Parkville, VIC, Australia.
- Walter and Eliza Hall Institute, Parkville, VIC, Australia.
| | - Leann Tilley
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
10
|
Marothia M, Behl A, Maurya P, Saini M, Shoaib R, Garg S, Kumari G, Biswas S, Munjal A, Anand S, Kahlon AK, Gupta P, Biswas S, Goswami B, Abdulhameed Almuqdadi HT, Bhowmick IP, Shevtsov M, Ramalingam S, Ranganathan A, Singh S. Targeting PfProhibitin 2-Hu-Hsp70A1A complex as a unique approach towards malaria vaccine development. iScience 2024; 27:109918. [PMID: 38812541 PMCID: PMC11134565 DOI: 10.1016/j.isci.2024.109918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/13/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024] Open
Abstract
Malaria parasite invasion to host erythrocytes is mediated by multiple interactions between merozoite ligands and erythrocyte receptors that contribute toward the development of disease pathology. Here, we report a novel antigen Plasmodium prohibitin "PfPHB2" and identify its cognate partner "Hsp70A1A" in host erythrocyte that plays a crucial role in mediating host-parasite interaction during merozoite invasion. Using small interfering RNA (siRNA)- and glucosamine-6-phosphate riboswitch (glmS) ribozyme-mediated approach, we show that loss of Hsp70A1A in red blood cells (RBCs) or PfPHB2 in infected red blood cells (iRBCs), respectively, inhibit PfPHB2-Hsp70A1A interaction leading to invasion inhibition. Antibodies targeting PfPHB2 and monoclonal antibody therapeutics against Hsp70A1A efficiently block parasite invasion. Recombinant PfPHB2 binds to RBCs which is inhibited by anti-PfPHB2 antibody and monoclonal antibody against Hsp70A1A. The validation of PfPHB2 to serve as antigen is further supported by detection of anti-PfPHB2 antibody in patient sera. Overall, this study proposes PfPHB2 as vaccine candidate and highlights the use of monoclonal antibody therapeutics for future malaria treatment.
Collapse
Affiliation(s)
- Manisha Marothia
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Ankita Behl
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Preeti Maurya
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Monika Saini
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Rumaisha Shoaib
- Department of Bioscience, Jamia Millia Islamia, New Delhi, India
| | - Swati Garg
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Geeta Kumari
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Shreeja Biswas
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Akshay Munjal
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Sakshi Anand
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Amandeep Kaur Kahlon
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Pragya Gupta
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, Sukhdev Vihar, New Delhi 110025, India
| | - Saurav Biswas
- Regional Medical Research Center-Northeast Region (RMRC-NE)-ICMR, Dibrugarh 786001, India
| | - Bidhan Goswami
- Multidisciplinary Research Unit, Agartala Government Medical College, Agartala, Tripura (West), India
| | - Haider Thaer Abdulhameed Almuqdadi
- Department of Bioscience, Jamia Millia Islamia, New Delhi, India
- Department of Chemistry, College of Science, Al-Nahrain University, Baghdad, Iraq
| | - Ipsita Pal Bhowmick
- Regional Medical Research Center-Northeast Region (RMRC-NE)-ICMR, Dibrugarh 786001, India
| | - Maxim Shevtsov
- Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 St. Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str. 2, 197341 St. Petersburg, Russia
| | - Sivaprakash Ramalingam
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, Sukhdev Vihar, New Delhi 110025, India
| | - Anand Ranganathan
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
11
|
Alberione MP, González-Ruiz V, von Rohr O, Rudaz S, Soldati-Favre D, Izquierdo L, Kloehn J. N-acetylglucosamine supplementation fails to bypass the critical acetylation of glucosamine-6-phosphate required for Toxoplasma gondii replication and invasion. PLoS Pathog 2024; 20:e1011979. [PMID: 38900808 PMCID: PMC11218972 DOI: 10.1371/journal.ppat.1011979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 07/02/2024] [Accepted: 06/03/2024] [Indexed: 06/22/2024] Open
Abstract
The cell surface of Toxoplasma gondii is rich in glycoconjugates which hold diverse and vital functions in the lytic cycle of this obligate intracellular parasite. Additionally, the cyst wall of bradyzoites, that shields the persistent form responsible for chronic infection from the immune system, is heavily glycosylated. Formation of glycoconjugates relies on activated sugar nucleotides, such as uridine diphosphate N-acetylglucosamine (UDP-GlcNAc). The glucosamine-phosphate-N-acetyltransferase (GNA1) generates N-acetylglucosamine-6-phosphate critical to produce UDP-GlcNAc. Here, we demonstrate that downregulation of T. gondii GNA1 results in a severe reduction of UDP-GlcNAc and a concomitant drop in glycosylphosphatidylinositols (GPIs), leading to impairment of the parasite's ability to invade and replicate in the host cell. Surprisingly, attempts to rescue this defect through exogenous GlcNAc supplementation fail to completely restore these vital functions. In depth metabolomic analyses elucidate diverse causes underlying the failed rescue: utilization of GlcNAc is inefficient under glucose-replete conditions and fails to restore UDP-GlcNAc levels in GNA1-depleted parasites. In contrast, GlcNAc-supplementation under glucose-deplete conditions fully restores UDP-GlcNAc levels but fails to rescue the defects associated with GNA1 depletion. Our results underscore the importance of glucosamine-6-phosphate acetylation in governing T. gondii replication and invasion and highlight the potential of the evolutionary divergent GNA1 in Apicomplexa as a target for the development of much-needed new therapeutic strategies.
Collapse
Affiliation(s)
- María Pía Alberione
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-University of Barcelona, Barcelona, Spain
| | | | - Olivier von Rohr
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Serge Rudaz
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Luis Izquierdo
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Joachim Kloehn
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
12
|
Das S, Manna A, Majumdar O, Dhara L. M- O-M mediated denaturation resistant P2 tetramer on the infected erythrocyte surface of malaria parasite imports serum fatty acids. iScience 2024; 27:109760. [PMID: 38726364 PMCID: PMC11079477 DOI: 10.1016/j.isci.2024.109760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 04/01/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
In Plasmodium falciparum, DNA replication, and asynchronous nuclear divisions precede cytokinesis during intraerythrocytic schizogony. Regulation of nuclear division through the import of serum components was largely unknown. At the trophozoite stage, P. falciparum ribosomal protein P2 (PfP2) is exported to the infected erythrocyte (IE) cytosol and the surface as a denaturation-resistant tetramer. The inaccessibility of the IE surface exposed PfP2 to its bona fide ligand led to the arrest of nuclear division. Here, we show that at the onset of schizogony, denaturation-resistant PfP2 tetramer on the IE surface imports fatty acids (FAs). Blockage of import reversibly arrested parasite schizogony. In 11Met-O-Met11 mediated denaturation resistant PfP2 tetramer, the 12/53Cys-Cys12/53 redox switch regulates the binding and release of FAs based on oxidized/reduced state of disulfide linkages. This mechanistic insight of FAs import through PfP2 tetramer reveals a unique regulation of nuclear division at the onset of schizogony.
Collapse
Affiliation(s)
- Sudipta Das
- Division of infectious Disease and Immunology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C Mullick Road, Jadavpur, Kolkata 700032, India
| | - Anwesa Manna
- Division of infectious Disease and Immunology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C Mullick Road, Jadavpur, Kolkata 700032, India
| | - Oindrila Majumdar
- Division of infectious Disease and Immunology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C Mullick Road, Jadavpur, Kolkata 700032, India
| | - Lena Dhara
- Division of infectious Disease and Immunology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C Mullick Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
13
|
Wirjanata G, Lin J, Dziekan JM, El Sahili A, Chung Z, Tjia S, Binte Zulkifli NE, Boentoro J, Tham R, Jia LS, Go KD, Yu H, Partridge A, Olsen D, Prabhu N, Sobota RM, Nordlund P, Lescar J, Bozdech Z. Identification of an inhibitory pocket in falcilysin provides a new avenue for malaria drug development. Cell Chem Biol 2024; 31:743-759.e8. [PMID: 38593807 DOI: 10.1016/j.chembiol.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 09/02/2023] [Accepted: 03/12/2024] [Indexed: 04/11/2024]
Abstract
Identification of new druggable protein targets remains the key challenge in the current antimalarial development efforts. Here we used mass-spectrometry-based cellular thermal shift assay (MS-CETSA) to identify potential targets of several antimalarials and drug candidates. We found that falcilysin (FLN) is a common binding partner for several drug candidates such as MK-4815, MMV000848, and MMV665806 but also interacts with quinoline drugs such as chloroquine and mefloquine. Enzymatic assays showed that these compounds can inhibit FLN proteolytic activity. Their interaction with FLN was explored systematically by isothermal titration calorimetry and X-ray crystallography, revealing a shared hydrophobic pocket in the catalytic chamber of the enzyme. Characterization of transgenic cell lines with lowered FLN expression demonstrated statistically significant increases in susceptibility toward MK-4815, MMV000848, and several quinolines. Importantly, the hydrophobic pocket of FLN appears amenable to inhibition and the structures reported here can guide the development of novel drugs against malaria.
Collapse
Affiliation(s)
- Grennady Wirjanata
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | - Jianqing Lin
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore; NTU Institute of Structural Biology, Nanyang Technology University, Singapore 637551, Singapore; Infectious Diseases Labs & Singapore Immunology Network, Agency for Science, Technology and Research, 138648 Singapore, Singapore
| | - Jerzy Michal Dziekan
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | - Abbas El Sahili
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore; NTU Institute of Structural Biology, Nanyang Technology University, Singapore 637551, Singapore
| | - Zara Chung
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | - Seth Tjia
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | | | - Josephine Boentoro
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | - Roy Tham
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | - Lai Si Jia
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | - Ka Diam Go
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | - Han Yu
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | | | - David Olsen
- Merck & Co., Inc., West Point, PA 19486, USA
| | - Nayana Prabhu
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | - Radoslaw M Sobota
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A∗STAR), Singapore 138673, Singapore; Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Pär Nordlund
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A∗STAR), Singapore 138673, Singapore; Department of Oncology and Pathology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Julien Lescar
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore; NTU Institute of Structural Biology, Nanyang Technology University, Singapore 637551, Singapore; Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore 637551, Singapore.
| | - Zbynek Bozdech
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore.
| |
Collapse
|
14
|
Florini F, Visone JE, Hadjimichael E, Malpotra S, Nötzel C, Kafsack BF, Deitsch KW. Transcriptional plasticity of virulence genes provides malaria parasites with greater adaptive capacity for avoiding host immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.584127. [PMID: 38496509 PMCID: PMC10942408 DOI: 10.1101/2024.03.08.584127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Chronic, asymptomatic malaria infections contribute substantially to disease transmission and likely represent the most significant impediment preventing malaria elimination and eradication. Plasmodium falciparum parasites evade antibody recognition through transcriptional switching between members of the var gene family, which encodes the major virulence factor and surface antigen on infected red blood cells. This process can extend infections for up to a year; however, infections have been documented to last for over a decade, constituting an unseen reservoir of parasites that undermine eradication and control efforts. How parasites remain immunologically "invisible" for such lengthy periods is entirely unknown. Here we show that in addition to the accepted paradigm of mono-allelic var gene expression, individual parasites can simultaneously express multiple var genes or enter a state in which little or no var gene expression is detectable. This unappreciated flexibility provides parasites with greater adaptive capacity than previously understood and challenges the dogma of mutually exclusive var gene expression. It also provides an explanation for the antigenically "invisible" parasites observed in chronic asymptomatic infections.
Collapse
Affiliation(s)
| | | | - Evi Hadjimichael
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, USA
| | - Shivali Malpotra
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, USA
| | | | - Björn F.C. Kafsack
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, USA
| | - Kirk W. Deitsch
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
15
|
Farrukh A, Musabyimana JP, Distler U, Mahlich VJ, Mueller J, Bick F, Tenzer S, Pradel G, Ngwa CJ. The Plasmodium falciparum CCCH zinc finger protein MD3 regulates male gametocytogenesis through its interaction with RNA-binding proteins. Mol Microbiol 2024; 121:543-564. [PMID: 38148574 DOI: 10.1111/mmi.15215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/28/2023]
Abstract
The transmission of malaria parasites to mosquitoes is dependent on the formation of gametocytes. Once fully matured, gametocytes are able to transform into gametes in the mosquito's midgut, a process accompanied with their egress from the enveloping erythrocyte. Gametocyte maturation and gametogenesis require a well-coordinated gene expression program that involves a wide spectrum of regulatory proteins, ranging from histone modifiers to transcription factors to RNA-binding proteins. Here, we investigated the role of the CCCH zinc finger protein MD3 in Plasmodium falciparum gametocytogenesis. MD3 was originally identified as an epigenetically regulated protein of immature gametocytes and recently shown to be involved in male development in a barcode-based screen in P. berghei. We report that MD3 is mainly present in the cytoplasm of immature male P. falciparum gametocytes. Parasites deficient of MD3 are impaired in gametocyte maturation and male gametocytogenesis. BioID analysis in combination with co-immunoprecipitation assays unveiled an interaction network of MD3 with RNA-binding proteins like PABP1 and ALBA3, with translational initiators, regulators and repressors like elF4G, PUF1, NOT1 and CITH, and with further regulators of gametocytogenesis, including ZNF4, MD1 and GD1. We conclude that MD3 is part of a regulator complex crucial for post-transcriptional fine-tuning of male gametocytogenesis.
Collapse
Affiliation(s)
- Afia Farrukh
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Jean Pierre Musabyimana
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Ute Distler
- Core Facility for Mass Spectrometry, Institute of Immunology, University Medical Centre of the Johannes-Gutenberg University, Mainz, Germany
| | - Vanessa Jil Mahlich
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Julius Mueller
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Fabian Bick
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Stefan Tenzer
- Core Facility for Mass Spectrometry, Institute of Immunology, University Medical Centre of the Johannes-Gutenberg University, Mainz, Germany
| | - Gabriele Pradel
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Che Julius Ngwa
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
16
|
Rahman A, Ahmad MA, Mehmood S, Rauf A, Iqbal A, Ali B, Ullah M, Ali M, Mohamed HI, Uddin I. Isolation and Screening of Zn (Zn) Solubilizing Rhizosphere Bacteria from Different Vegetations for Their Ability to Improve Growth, Zn Uptake, and Expression of Zn Transporter Genes in Tomato. Curr Microbiol 2024; 81:83. [PMID: 38294556 DOI: 10.1007/s00284-023-03610-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 12/30/2023] [Indexed: 02/01/2024]
Abstract
Zinc-solubilizing bacteria (ZSB) can convert insoluble zinc to an accessible form and increase Zn bioavailability in soil, which helps mitigate Zn deficiency in crops. In this study, different bacterial strains were screened for different Zn solubilization and plant growth promotion traits. Two bacterial strains, Acinetobacter pittii DJ55 and Stenotrophomonas maltophilia DJ24, were tested for their Zn-solubilizing potential on plate media, and both showed variable levels of Zn solubilization. The results showed that the bacterial strains applied to the plants in the pot experiment caused improvements in growth parameters compared to control conditions. DJ55, when applied with an insoluble source, enhanced plant height, leaf number, and leaf area compared to DJ24 and control conditions, while the maximum fruit weight was noticed in plants treated with ZnSO4. An increase in chlorophyll contents was noted in plants treated with ZnSO4, while maximum carotenoid contents were observed in plants treated with DJ55 + ZnO when compared with their controls. Plants supplemented with ZnO and DJ55 showed higher zinc content and iron content as compared to their respective controls. The expression patterns of the SLZIP5 and SLZIP4 genes were changed in the root and shoot. Application of ZnO stimulates both gene expression and protein synthesis in tomato roots and shoots. Inoculation of tomato plants with ZSB and insoluble ZnO reduced the expression of the SLZIP5 and SLZIP4 genes in the root and shoot. In conclusion, both strains can be considered as potential zinc-solubilizing bioinoculants to promote the growth and production yield of tomato.
Collapse
Affiliation(s)
- Attequr Rahman
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture Peshawar, Peshawar, KP, Pakistan
| | - Mian Afaq Ahmad
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture Peshawar, Peshawar, KP, Pakistan.
| | - Shiraz Mehmood
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture Peshawar, Peshawar, KP, Pakistan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Abdur Rauf
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture Peshawar, Peshawar, KP, Pakistan
| | - Aqib Iqbal
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture Peshawar, Peshawar, KP, Pakistan
| | - Bakhtiar Ali
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture Peshawar, Peshawar, KP, Pakistan
| | - Mohib Ullah
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture Peshawar, Peshawar, KP, Pakistan
| | - Murad Ali
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture Peshawar, Peshawar, KP, Pakistan
| | - Heba I Mohamed
- Biological and Geological Sciences Department, Faculty of Education, Ain Shams University, Cairo, 11341, Egypt.
| | - Israr Uddin
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture Peshawar, Peshawar, KP, Pakistan
| |
Collapse
|
17
|
Leela N, Prommana P, Kamchonwongpaisan S, Taechalertpaisarn T, Shaw PJ. Antimalarial target vulnerability of the putative Plasmodium falciparum methionine synthase. PeerJ 2024; 12:e16595. [PMID: 38239295 PMCID: PMC10795524 DOI: 10.7717/peerj.16595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/14/2023] [Indexed: 01/22/2024] Open
Abstract
Background Plasmodium falciparum possesses a cobalamin-dependent methionine synthase (MS). MS is putatively encoded by the PF3D7_1233700 gene, which is orthologous and syntenic in Plasmodium. However, its vulnerability as an antimalarial target has not been assessed. Methods We edited the PF3D7_1233700 and PF3D7_0417200 (dihydrofolate reductase-thymidylate synthase, DHFR-TS) genes and obtained transgenic P. falciparum parasites expressing epitope-tagged target proteins under the control of the glmS ribozyme. Conditional loss-of-function mutants were obtained by treating transgenic parasites with glucosamine. Results DHFR-TS, but not MS mutants showed a significant proliferation defect over 96 h, suggesting that P. falciparum MS is not a vulnerable antimalarial target.
Collapse
Affiliation(s)
- Nirut Leela
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Bangkok, Thailand
| | - Parichat Prommana
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Sumalee Kamchonwongpaisan
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Tana Taechalertpaisarn
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Bangkok, Thailand
| | - Philip J. Shaw
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, Thailand
| |
Collapse
|
18
|
Shunmugam S, Quansah N, Flammersfeld A, Islam MM, Sassmannshausen J, Bennink S, Yamaryo-Botté Y, Pradel G, Botté CY. The patatin-like phospholipase PfPNPLA2 is involved in the mitochondrial degradation of phosphatidylglycerol during Plasmodium falciparum blood stage development. Front Cell Infect Microbiol 2023; 13:997245. [PMID: 38089812 PMCID: PMC10711835 DOI: 10.3389/fcimb.2023.997245] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/09/2023] [Indexed: 12/18/2023] Open
Abstract
Plasmodium falciparum is an Apicomplexa responsible for human malaria, a major disease causing more than ½ million deaths every year, against which there is no fully efficient vaccine. The current rapid emergence of drug resistances emphasizes the need to identify novel drug targets. Increasing evidences show that lipid synthesis and trafficking are essential for parasite survival and pathogenesis, and that these pathways represent potential points of attack. Large amounts of phospholipids are needed for the generation of membrane compartments for newly divided parasites in the host cell. Parasite membrane homeostasis is achieved by an essential combination of parasite de novo lipid synthesis/recycling and massive host lipid scavenging. Latest data suggest that the mobilization and channeling of lipid resources is key for asexual parasite survival within the host red blood cell, but the molecular actors allowing lipid acquisition are poorly characterized. Enzymes remodeling lipids such as phospholipases are likely involved in these mechanisms. P. falciparum possesses an unusually large set of phospholipases, whose functions are largely unknown. Here we focused on the putative patatin-like phospholipase PfPNPLA2, for which we generated an glmS-inducible knockdown line and investigated its role during blood stages malaria. Disruption of the mitochondrial PfPNPLA2 in the asexual blood stages affected mitochondrial morphology and further induced a significant defect in parasite replication and survival, in particular under low host lipid availability. Lipidomic analyses revealed that PfPNPLA2 specifically degrades the parasite membrane lipid phosphatidylglycerol to generate lysobisphosphatidic acid. PfPNPLA2 knockdown further resulted in an increased host lipid scavenging accumulating in the form of storage lipids and free fatty acids. These results suggest that PfPNPLA2 is involved in the recycling of parasite phosphatidylglycerol to sustain optimal intraerythrocytic development when the host resources are scarce. This work strengthens our understanding of the complex lipid homeostasis pathways to acquire lipids and allow asexual parasite survival.
Collapse
Affiliation(s)
- Serena Shunmugam
- Apicolipid Team, Institute for Avanced Biosciences, Centre National pour la Recherche Scientifique (CNRS) UMR5309, Institut National de la Santé et de la Recherche Médicale, Université Grenoble Alpes, Grenoble, France
| | - Nyamekye Quansah
- Apicolipid Team, Institute for Avanced Biosciences, Centre National pour la Recherche Scientifique (CNRS) UMR5309, Institut National de la Santé et de la Recherche Médicale, Université Grenoble Alpes, Grenoble, France
| | - Ansgar Flammersfeld
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Md Muzahidul Islam
- International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Juliane Sassmannshausen
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Sandra Bennink
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Yoshiki Yamaryo-Botté
- Apicolipid Team, Institute for Avanced Biosciences, Centre National pour la Recherche Scientifique (CNRS) UMR5309, Institut National de la Santé et de la Recherche Médicale, Université Grenoble Alpes, Grenoble, France
| | - Gabriele Pradel
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Cyrille Y. Botté
- Apicolipid Team, Institute for Avanced Biosciences, Centre National pour la Recherche Scientifique (CNRS) UMR5309, Institut National de la Santé et de la Recherche Médicale, Université Grenoble Alpes, Grenoble, France
| |
Collapse
|
19
|
Narwal M, Jain S, Rathore S, Mohmmed A. Plasmodium falciparum OPA3-like protein (PfOPA3) is essential for maintenance of mitochondrial homeostasis and parasite proliferation. FASEB J 2023; 37:e23235. [PMID: 37819580 DOI: 10.1096/fj.202201386rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/01/2023] [Accepted: 09/20/2023] [Indexed: 10/13/2023]
Abstract
Metabolic pathways and proteins responsible for maintaining mitochondrial dynamics and homeostasis in the Plasmodium parasite, the causative agent of malaria, remain to be elucidated. Here, we identified and functionally characterized a novel OPA3-like domain-containing protein in P. falciparum (PfOPA3). We show that PfOPA3 is expressed in the intraerythrocytic stages of the parasite and localizes to the mitochondria. Inducible knock-down of PfOPA3 using GlmS ribozyme hindered the normal intraerythrocytic cycle of the parasites; specifically, PfOPA3-iKD disrupted parasite development as well as parasite division and segregation at schizont stages, which resulted in a drastic reduction in the number of merozoites progenies. Parasites lacking PfOPA3 show severe defects in the development of functional mitochondria; the mitochondria showed reduced activity of mtETC but not ATP synthesis, as evidenced by reduced activity of complex III of the mtETC, and increased sensitivity for drugs targeting DHODH as well as complex III, but not to the drugs targeting complex V. Further, PfOPA3 downregulation leads to reduction in the level of mitochondrial proton transport uncoupling protein (PfUCP) to compensate reduced activity of complex III and maintain proton efflux across the inner membrane. The reduced activity of DHODH, which is responsible for pyrimidine biosynthesis required for nuclear DNA synthesis, resulted in a significant reduction in parasite nuclear division and generation of progeny. In conclusion, we show that PfOPA3 is essential for the functioning of mtETC and homeostasis required for the development of functional mitochondria as well as for parasite segregation, and thus PfOPA3 is crucial for parasite survival during blood stages.
Collapse
Affiliation(s)
- Monika Narwal
- Parasite Cell Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Shilpi Jain
- Parasite Cell Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Sumit Rathore
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Asif Mohmmed
- Parasite Cell Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
20
|
Lima C, Verdaguer IB, Wunderlich G, Katzin AM, Crabb BS, Gilson PR, Azevedo MF. Conditional expression of NanoLuc luciferase through a multimodular system offers rapid detection of antimalarial drug activity. Exp Parasitol 2023; 254:108620. [PMID: 37716462 DOI: 10.1016/j.exppara.2023.108620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023]
Abstract
Conditional gene expression is a powerful tool to investigate putative vaccine and drug targets, especially in a haploid organism such as Plasmodium falciparum. Inducible systems based on regulation of either transcription, translation, protein or mRNA stability, among others, allow switching on an off the expression of any desired gene causing specific gain or loss of function phenotypes. However, those systems can be cumbersome involving the construction of large plasmids and generation of multiple transgenic parasite lines. In addition, the dynamic range of regulation achieved is not predictable for each individual gene and can be insufficient to generate detectable phenotypes when the genes of interest are silenced. Here, we combined up to three distinct inducible systems to regulate the expression of a single gene. Expression of the reporter NanoLuc luciferase was regulated over 40-fold, which correlates to the regulation achieved by each individual system multiplied by each other. We applied the conditionally expressed NanoLuc to evaluate the effect of fast-acting antimalarials such as chloroquine and artesunate as well as of slower-acting ones such as atovaquone. The conditionally expressed reporter allowed faster and more reliable detection of toxicity to the parasite, which correlated to the expected action of each compound. Bioluminescence achieved by the expression of this inducible highly sensitive reporter is therefore a promising tool to investigate the temporal effect of potential new antimalarials. This single plasmid combination system might also prove useful to achieve sufficient regulation of genes of interest to produce loss-of-function phenotypes.
Collapse
Affiliation(s)
- Caroline Lima
- Federal University of Sao Paulo, Santos, Sao Paulo, Brazil
| | - Ignasi B Verdaguer
- Departamento de Parasitologia, Instituto de Ciência Biomédicas, Universidade de São Paulo, Avenida Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-900, Brazil
| | - Gerhard Wunderlich
- Departamento de Parasitologia, Instituto de Ciência Biomédicas, Universidade de São Paulo, Avenida Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-900, Brazil
| | - Alejandro M Katzin
- Departamento de Parasitologia, Instituto de Ciência Biomédicas, Universidade de São Paulo, Avenida Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-900, Brazil
| | - Brendan S Crabb
- Burnet Institute, Melbourne, VIC, 3004, Australia; University of Melbourne, VIC, 3052, Australia; Monash University, Melbourne, VIC, 3004, Australia
| | - Paul R Gilson
- Burnet Institute, Melbourne, VIC, 3004, Australia; University of Melbourne, VIC, 3052, Australia
| | | |
Collapse
|
21
|
Rosa C, Singh P, Chen P, Sinha A, Claës A, Preiser PR, Dedon PC, Baumgarten S, Scherf A, Bryant JM. Cohesin contributes to transcriptional repression of stage-specific genes in the human malaria parasite. EMBO Rep 2023; 24:e57090. [PMID: 37592911 PMCID: PMC10561359 DOI: 10.15252/embr.202357090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 08/19/2023] Open
Abstract
The complex life cycle of the human malaria parasite, Plasmodium falciparum, is driven by specific transcriptional programs, but it is unclear how most genes are activated or silenced at specific times. There is an association between transcription and spatial organization; however, the molecular mechanisms behind genome organization are unclear. While P. falciparum lacks key genome-organizing proteins found in metazoans, it has all core components of the cohesin complex. To investigate the role of cohesin in P. falciparum, we functionally characterize the cohesin subunit Structural Maintenance of Chromosomes protein 3 (SMC3). SMC3 knockdown during early stages of the intraerythrocytic developmental cycle (IDC) upregulates a subset of genes involved in erythrocyte egress and invasion, which are normally expressed at later stages. ChIP-seq analyses reveal that during the IDC, SMC3 enrichment at the promoter regions of these genes inversely correlates with gene expression and chromatin accessibility. These data suggest that SMC3 binding contributes to the repression of specific genes until their appropriate time of expression, revealing a new mode of stage-specific gene repression in P. falciparum.
Collapse
Affiliation(s)
- Catarina Rosa
- Institut Pasteur, Université Paris Cité, INSERM U1201, CNRS EMR9195, Biology of Host‐Parasite Interactions UnitParisFrance
- Sorbonne Université, Collège Doctoral Complexité du Vivant ED515ParisFrance
| | - Parul Singh
- Institut Pasteur, Université Paris Cité, INSERM U1201, CNRS EMR9195, Biology of Host‐Parasite Interactions UnitParisFrance
| | - Patty Chen
- Institut Pasteur, Université Paris Cité, INSERM U1201, CNRS EMR9195, Biology of Host‐Parasite Interactions UnitParisFrance
| | - Ameya Sinha
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore‐MIT Alliance for Research and TechnologySingaporeSingapore
| | - Aurélie Claës
- Institut Pasteur, Université Paris Cité, INSERM U1201, CNRS EMR9195, Biology of Host‐Parasite Interactions UnitParisFrance
| | - Peter R Preiser
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore‐MIT Alliance for Research and TechnologySingaporeSingapore
| | - Peter C Dedon
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore‐MIT Alliance for Research and TechnologySingaporeSingapore
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| | | | - Artur Scherf
- Institut Pasteur, Université Paris Cité, INSERM U1201, CNRS EMR9195, Biology of Host‐Parasite Interactions UnitParisFrance
| | - Jessica M Bryant
- Institut Pasteur, Université Paris Cité, INSERM U1201, CNRS EMR9195, Biology of Host‐Parasite Interactions UnitParisFrance
| |
Collapse
|
22
|
McHugh E, Bulloch MS, Batinovic S, Patrick CJ, Sarna DK, Ralph SA. Nonsense-mediated decay machinery in Plasmodium falciparum is inefficient and non-essential. mSphere 2023; 8:e0023323. [PMID: 37366629 PMCID: PMC10449492 DOI: 10.1128/msphere.00233-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 05/15/2023] [Indexed: 06/28/2023] Open
Abstract
Nonsense-mediated decay (NMD) is a conserved mRNA quality control process that eliminates transcripts bearing a premature termination codon. In addition to its role in removing erroneous transcripts, NMD is involved in post-transcriptional regulation of gene expression via programmed intron retention in metazoans. The apicomplexan parasite Plasmodium falciparum shows relatively high levels of intron retention, but it is unclear whether these variant transcripts are functional targets of NMD. In this study, we use CRISPR-Cas9 to disrupt and epitope-tag the P. falciparum orthologs of two core NMD components: PfUPF1 (PF3D7_1005500) and PfUPF2 (PF3D7_0925800). We localize both PfUPF1 and PfUPF2 to puncta within the parasite cytoplasm and show that these proteins interact with each other and other mRNA-binding proteins. Using RNA-seq, we find that although these core NMD orthologs are expressed and interact in P. falciparum, they are not required for degradation of nonsense transcripts. Furthermore, our work suggests that the majority of intron retention in P. falciparum has no functional role and that NMD is not required for parasite growth ex vivo. IMPORTANCE In many organisms, the process of destroying nonsense transcripts is dependent on a small set of highly conserved proteins. We show that in the malaria parasite, these proteins do not impact the abundance of nonsense transcripts. Furthermore, we demonstrate efficient CRISPR-Cas9 editing of the malaria parasite using commercial Cas9 nuclease and synthetic guide RNA, streamlining genomic modifications in this genetically intractable organism.
Collapse
Affiliation(s)
- Emma McHugh
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Michaela S. Bulloch
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Steven Batinovic
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Cameron J. Patrick
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Victoria, Australia
| | - Drishti K. Sarna
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Stuart A. Ralph
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
23
|
Fang J, Wang J, Wang Y, Liu X, Chen B, Zou W. Ribo-On and Ribo-Off tools using a self-cleaving ribozyme allow manipulation of endogenous gene expression in C. elegans. Commun Biol 2023; 6:816. [PMID: 37542105 PMCID: PMC10403566 DOI: 10.1038/s42003-023-05184-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 07/26/2023] [Indexed: 08/06/2023] Open
Abstract
Investigating gene function relies on the efficient manipulation of endogenous gene expression. Currently, a limited number of tools are available to robustly manipulate endogenous gene expression between "on" and "off" states. In this study, we insert a 63 bp coding sequence of T3H38 ribozyme into the 3' untranslated region (UTR) of C. elegans endogenous genes using the CRISPR/Cas9 technology, which reduces the endogenous gene expression to a nearly undetectable level and generated loss-of-function phenotypes similar to that of the genetic null animals. To achieve conditional knockout, a cassette of loxP-flanked transcriptional termination signal and ribozyme is inserted into the 3' UTR of endogenous genes, which eliminates gene expression spatially or temporally via the controllable expression of the Cre recombinase. Conditional endogenous gene turn-on can be achieved by either injecting morpholino, which blocks the ribozyme self-cleavage activity or using the Cre recombinase to remove the loxP-flanked ribozyme. Together, our results demonstrate that these ribozyme-based tools can efficiently manipulate endogenous gene expression both in space and time and expand the toolkit for studying the functions of endogenous genes.
Collapse
Affiliation(s)
- Jie Fang
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, 322000, Yiwu, China
- Institute of Translational Medicine, Zhejiang University, 310058, Hangzhou, China
- Department of Cell Biology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Jie Wang
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, 322000, Yiwu, China
- Institute of Translational Medicine, Zhejiang University, 310058, Hangzhou, China
| | - Yuzhi Wang
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, 322000, Yiwu, China
- Institute of Translational Medicine, Zhejiang University, 310058, Hangzhou, China
| | - Xiaofan Liu
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, 322000, Yiwu, China
- Institute of Translational Medicine, Zhejiang University, 310058, Hangzhou, China
| | - Baohui Chen
- Department of Cell Biology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China.
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, China.
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China.
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China.
| | - Wei Zou
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, 322000, Yiwu, China.
- Institute of Translational Medicine, Zhejiang University, 310058, Hangzhou, China.
| |
Collapse
|
24
|
Ghosh S, Kundu R, Chandana M, Das R, Anand A, Beura S, Bobde RC, Jain V, Prabhu SR, Behera PK, Mohanty AK, Chakrapani M, Satyamoorthy K, Suryawanshi AR, Dixit A, Padmanaban G, Nagaraj VA. Distinct evolution of type I glutamine synthetase in Plasmodium and its species-specific requirement. Nat Commun 2023; 14:4216. [PMID: 37452051 PMCID: PMC10349072 DOI: 10.1038/s41467-023-39670-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/22/2023] [Indexed: 07/18/2023] Open
Abstract
Malaria parasite lacks canonical pathways for amino acid biosynthesis and depends primarily on hemoglobin degradation and extracellular resources for amino acids. Interestingly, a putative gene for glutamine synthetase (GS) is retained despite glutamine being an abundant amino acid in human and mosquito hosts. Here we show Plasmodium GS has evolved as a unique type I enzyme with distinct structural and regulatory properties to adapt to the asexual niche. Methionine sulfoximine (MSO) and phosphinothricin (PPT) inhibit parasite GS activity. GS is localized to the parasite cytosol and abundantly expressed in all the life cycle stages. Parasite GS displays species-specific requirement in Plasmodium falciparum (Pf) having asparagine-rich proteome. Targeting PfGS affects asparagine levels and inhibits protein synthesis through eIF2α phosphorylation leading to parasite death. Exposure of artemisinin-resistant Pf parasites to MSO and PPT inhibits the emergence of viable parasites upon artemisinin treatment.
Collapse
Affiliation(s)
- Sourav Ghosh
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, 121001, Haryana, India
| | - Rajib Kundu
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, 121001, Haryana, India
| | - Manjunatha Chandana
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, 751024, Odisha, India
| | - Rahul Das
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, 121001, Haryana, India
| | - Aditya Anand
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, 121001, Haryana, India
| | - Subhashree Beura
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
| | - Ruchir Chandrakant Bobde
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, 121001, Haryana, India
| | - Vishal Jain
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
| | - Sowmya Ramakant Prabhu
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | | | - Akshaya Kumar Mohanty
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- Ispat General Hospital, Sector 19, Rourkela, 769005, Odisha, India
| | - Mahabala Chakrapani
- Department of Medicine, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | | | - Anshuman Dixit
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
| | - Govindarajan Padmanaban
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, Karnataka, India
| | | |
Collapse
|
25
|
Harris CT, Tong X, Campelo R, Marreiros IM, Vanheer LN, Nahiyaan N, Zuzarte-Luís VA, Deitsch KW, Mota MM, Rhee KY, Kafsack BFC. Sexual differentiation in human malaria parasites is regulated by competition between phospholipid metabolism and histone methylation. Nat Microbiol 2023; 8:1280-1292. [PMID: 37277533 PMCID: PMC11163918 DOI: 10.1038/s41564-023-01396-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 04/25/2023] [Indexed: 06/07/2023]
Abstract
For Plasmodium falciparum, the most widespread and virulent malaria parasite that infects humans, persistence depends on continuous asexual replication in red blood cells, while transmission to their mosquito vector requires asexual blood-stage parasites to differentiate into non-replicating gametocytes. This decision is controlled by stochastic derepression of a heterochromatin-silenced locus encoding AP2-G, the master transcription factor of sexual differentiation. The frequency of ap2-g derepression was shown to be responsive to extracellular phospholipid precursors but the mechanism linking these metabolites to epigenetic regulation of ap2-g was unknown. Through a combination of molecular genetics, metabolomics and chromatin profiling, we show that this response is mediated by metabolic competition for the methyl donor S-adenosylmethionine between histone methyltransferases and phosphoethanolamine methyltransferase, a critical enzyme in the parasite's pathway for de novo phosphatidylcholine synthesis. When phosphatidylcholine precursors are scarce, increased consumption of SAM for de novo phosphatidylcholine synthesis impairs maintenance of the histone methylation responsible for silencing ap2-g, increasing the frequency of derepression and sexual differentiation. This provides a key mechanistic link that explains how LysoPC and choline availability can alter the chromatin status of the ap2-g locus controlling sexual differentiation.
Collapse
Affiliation(s)
- Chantal T Harris
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
- Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Xinran Tong
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
- BCMB Allied Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Riward Campelo
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Inês M Marreiros
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina Universidade de Lisboa, Lisbon, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Leen N Vanheer
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Navid Nahiyaan
- Division of Infectious Diseases, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Vanessa A Zuzarte-Luís
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina Universidade de Lisboa, Lisbon, Portugal
| | - Kirk W Deitsch
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Maria M Mota
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina Universidade de Lisboa, Lisbon, Portugal
| | - Kyu Y Rhee
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
- Division of Infectious Diseases, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Björn F C Kafsack
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
26
|
Jonsdottir TK, Elsworth B, Cobbold S, Gabriela M, Ploeger E, Parkyn Schneider M, Charnaud SC, Dans MG, McConville M, Bullen HE, Crabb BS, Gilson PR. PTEX helps efficiently traffic haemoglobinases to the food vacuole in Plasmodium falciparum. PLoS Pathog 2023; 19:e1011006. [PMID: 37523385 PMCID: PMC10414648 DOI: 10.1371/journal.ppat.1011006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 08/10/2023] [Accepted: 07/16/2023] [Indexed: 08/02/2023] Open
Abstract
A key element of Plasmodium biology and pathogenesis is the trafficking of ~10% of the parasite proteome into the host red blood cell (RBC) it infects. To cross the parasite-encasing parasitophorous vacuole membrane, exported proteins utilise a channel-forming protein complex termed the Plasmodium translocon of exported proteins (PTEX). PTEX is obligatory for parasite survival, both in vitro and in vivo, suggesting that at least some exported proteins have essential metabolic functions. However, to date only one essential PTEX-dependent process, the new permeability pathways, has been described. To identify other essential PTEX-dependant proteins/processes, we conditionally knocked down the expression of one of its core components, PTEX150, and examined which pathways were affected. Surprisingly, the food vacuole mediated process of haemoglobin (Hb) digestion was substantially perturbed by PTEX150 knockdown. Using a range of transgenic parasite lines and approaches, we show that two major Hb proteases; falcipain 2a and plasmepsin II, interact with PTEX core components, implicating the translocon in the trafficking of Hb proteases. We propose a model where these proteases are translocated into the PV via PTEX in order to reach the cytostome, located at the parasite periphery, prior to food vacuole entry. This work offers a second mechanistic explanation for why PTEX function is essential for growth of the parasite within its host RBC.
Collapse
Affiliation(s)
- Thorey K. Jonsdottir
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
- Department of Immunology and Microbiology, University of Melbourne, Melbourne, Australia
| | - Brendan Elsworth
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
| | - Simon Cobbold
- Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Melbourne, Australia
| | - Mikha Gabriela
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
- School of Medicine, Deakin University, Geelong, Australia
| | - Ellen Ploeger
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
| | | | - Sarah C. Charnaud
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
| | - Madeline G. Dans
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
| | - Malcolm McConville
- Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Melbourne, Australia
| | - Hayley E. Bullen
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
- Department of Immunology and Microbiology, University of Melbourne, Melbourne, Australia
| | - Brendan S. Crabb
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
- Department of Immunology and Microbiology, University of Melbourne, Melbourne, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, Australia
| | - Paul R. Gilson
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
- Department of Immunology and Microbiology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
27
|
He L, Qiu Y, Pang G, Li S, Wang J, Feng Y, Chen L, Zhu L, Liu Y, Cui L, Cao Y, Zhu X. Plasmodium falciparum GAP40 Plays an Essential Role in Merozoite Invasion and Gametocytogenesis. Microbiol Spectr 2023; 11:e0143423. [PMID: 37249423 PMCID: PMC10269477 DOI: 10.1128/spectrum.01434-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/03/2023] [Indexed: 05/31/2023] Open
Abstract
Cyclic invasion of red blood cells (RBCs) by Plasmodium merozoites is associated with the symptoms and pathology of malaria. Merozoite invasion is powered actively and rapidly by a parasite actomyosin motor called the glideosome. The ability of the glideosome to generate force to support merozoite entry into the host RBCs is thought to rely on its stable anchoring within the inner membrane complex (IMC) through membrane-resident proteins, such as GAP50 and GAP40. Using a conditional knockdown (KD) approach, we determined that PfGAP40 was required for asexual blood-stage replication. PfGAP40 is not needed for merozoite egress from host RBCs or for the attachment of merozoites to new RBCs. PfGAP40 coprecipitates with PfGAP45 and PfGAP50. During merozoite invasion, PfGAP40 is associated strongly with stabilizing the expression levels of PfGAP45 and PfGAP50 in the schizont stage. Although PfGAP40 KD did not influence IMC integrity, it impaired the maturation of gametocytes. In addition, PfGAP40 is phosphorylated, and mutations that block phosphorylation of PfGAP40 at the C-terminal serine residues S370, S372, S376, S405, S409, S420, and S445 reduced merozoite invasion efficiency. Overall, our findings implicate PfGAP40 as an important regulator for the gliding activity of merozoites and suggest that phosphorylation is required for PfGAP40 function. IMPORTANCE Red blood cell invasion is central to the pathogenesis of the malaria parasite, and the parasite proteins involved in this process are potential therapeutic targets. Gliding motility powers merozoite invasion and is driven by a unique molecular motor termed the glideosome. The glideosome is stably anchored to the parasite inner membrane complex (IMC) through membrane-resident proteins. In the present study, we demonstrate the importance of an IMC-resident glideosome component, PfGAP40, that plays a critical role in stabilizing the expression levels of glideosome components in the schizont stage. We determined that phosphorylation of PfGAP40 at C-terminal residues is required for efficient merozoite invasion.
Collapse
Affiliation(s)
- Lu He
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Yue Qiu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Geping Pang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Siqi Li
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Jingjing Wang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Yonghui Feng
- Department of Laboratory Medicine, the First Hospital of China Medical University, Shenyang, Liaoning, China
- National Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning, China
| | - Lumeng Chen
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Liying Zhu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Yinjie Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Liwang Cui
- College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Xiaotong Zhu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
28
|
Dobrescu I, Hammam E, Dziekan JM, Claës A, Halby L, Preiser P, Bozdech Z, Arimondo PB, Scherf A, Nardella F. Plasmodium falciparum Eukaryotic Translation Initiation Factor 3 is Stabilized by Quinazoline-Quinoline Bisubstrate Inhibitors. ACS Infect Dis 2023; 9:1257-1266. [PMID: 37216290 PMCID: PMC10262199 DOI: 10.1021/acsinfecdis.3c00127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Indexed: 05/24/2023]
Abstract
Malaria drug resistance is hampering the fight against the deadliest parasitic disease affecting over 200 million people worldwide. We recently developed quinoline-quinazoline-based inhibitors (as compound 70) as promising new antimalarials. Here, we aimed to investigate their mode of action by using thermal proteome profiling (TPP). The eukaryotic translation initiation factor 3 (EIF3i) subunit I was identified as the main target protein stabilized by compound 70 in Plasmodium falciparum. This protein has never been characterized in malaria parasites. P. falciparum parasite lines were generated expressing either a HA tag or an inducible knockdown of the PfEIF3i gene to further characterize the target protein. PfEIF3i was stabilized in the presence of compound 70 in a cellular thermal shift Western blot assay, pointing that PfEIF3i indeed interacts with quinoline-quinazoline-based inhibitors. In addition, PfEIF3i-inducible knockdown blocks intra-erythrocytic development in the trophozoite stage, indicating that it has a vital function. We show that PfEIF3i is mostly expressed in late intra-erythrocytic stages and localizes in the cytoplasm. Previous mass spectrometry reports show that PfEIF3i is expressed in all parasite life cycle stages. Further studies will explore the potential of PfEIF3i as a target for the design of new antimalarial drugs active all along the life cycle of the parasite.
Collapse
Affiliation(s)
- Irina Dobrescu
- Unité
Biology of Host-Parasite Interactions, Department of Parasites and
Insect Vectors, Institut Pasteur, Université
de Paris-Cité, CNRS EMR 9195, INSERM Unit U1201, 25-28 Rue Du Dr Roux, Paris 75015, France
| | - Elie Hammam
- Unité
Biology of Host-Parasite Interactions, Department of Parasites and
Insect Vectors, Institut Pasteur, Université
de Paris-Cité, CNRS EMR 9195, INSERM Unit U1201, 25-28 Rue Du Dr Roux, Paris 75015, France
| | - Jerzy M. Dziekan
- School
of Biological Sciences, Nanyang Technological
University, Singapore 639798, Singapore
| | - Aurélie Claës
- Unité
Biology of Host-Parasite Interactions, Department of Parasites and
Insect Vectors, Institut Pasteur, Université
de Paris-Cité, CNRS EMR 9195, INSERM Unit U1201, 25-28 Rue Du Dr Roux, Paris 75015, France
| | - Ludovic Halby
- Epigenetic
Chemical Biology, Department of Structural Biology and Chemistry,
Institut Pasteur, Université de Paris-Cité,
UMR n3523 Chem4Life, CNRS, 28 Rue Du Dr Roux, Paris 75015, France
| | - Peter Preiser
- School
of Biological Sciences, Nanyang Technological
University, Singapore 639798, Singapore
| | - Zbynek Bozdech
- School
of Biological Sciences, Nanyang Technological
University, Singapore 639798, Singapore
| | - Paola B. Arimondo
- Epigenetic
Chemical Biology, Department of Structural Biology and Chemistry,
Institut Pasteur, Université de Paris-Cité,
UMR n3523 Chem4Life, CNRS, 28 Rue Du Dr Roux, Paris 75015, France
| | - Artur Scherf
- Unité
Biology of Host-Parasite Interactions, Department of Parasites and
Insect Vectors, Institut Pasteur, Université
de Paris-Cité, CNRS EMR 9195, INSERM Unit U1201, 25-28 Rue Du Dr Roux, Paris 75015, France
| | - Flore Nardella
- Unité
Biology of Host-Parasite Interactions, Department of Parasites and
Insect Vectors, Institut Pasteur, Université
de Paris-Cité, CNRS EMR 9195, INSERM Unit U1201, 25-28 Rue Du Dr Roux, Paris 75015, France
| |
Collapse
|
29
|
Najeh S, Zandi K, Kharma N, Perreault J. Computational design and experimental verification of pseudoknotted ribozymes. RNA (NEW YORK, N.Y.) 2023; 29:764-776. [PMID: 36868786 PMCID: PMC10187678 DOI: 10.1261/rna.079148.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/27/2022] [Indexed: 05/18/2023]
Abstract
The design of new RNA sequences that retain the function of a model RNA structure is a challenge in bioinformatics because of the structural complexity of these molecules. RNA can fold into its secondary and tertiary structures by forming stem-loops and pseudoknots. A pseudoknot is a set of base pairs between a region within a stem-loop and nucleotides outside of this stem-loop; this motif is very important for numerous functional structures. It is important for any computational design algorithm to take into account these interactions to give a reliable result for any structures that include pseudoknots. In our study, we experimentally validated synthetic ribozymes designed by Enzymer, which implements algorithms allowing for the design of pseudoknots. Enzymer is a program that uses an inverse folding approach to design pseudoknotted RNAs; we used it in this study to design two types of ribozymes. The ribozymes tested were the hammerhead and the glmS, which have a self-cleaving activity that allows them to liberate the new RNA genome copy during rolling-circle replication or to control the expression of the downstream genes, respectively. We demonstrated the efficiency of Enzymer by showing that the pseudoknotted hammerhead and glmS ribozymes sequences it designed were extensively modified compared to wild-type sequences and were still active.
Collapse
Affiliation(s)
- Sabrine Najeh
- INRS - Institut Armand-Frappier, Laval, QC H7V 1B7, Canada
| | - Kasra Zandi
- Software Engineering and Computer Science Department, Concordia University, Montreal, Quebec H3G 1M8, Canada
| | - Nawwaf Kharma
- Electrical and Computer Engineering Department, Concordia University, Montreal, Quebec H3G 1M8, Canada
| | | |
Collapse
|
30
|
Wenz C, Simon CS, Romão TP, Stürmer VS, Machado M, Klages N, Klemmer A, Voß Y, Ganter M, Brochet M, Guizetti J. An Sfi1-like centrin-interacting centriolar plaque protein affects nuclear microtubule homeostasis. PLoS Pathog 2023; 19:e1011325. [PMID: 37130129 PMCID: PMC10180636 DOI: 10.1371/journal.ppat.1011325] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 05/12/2023] [Accepted: 03/28/2023] [Indexed: 05/03/2023] Open
Abstract
Malaria-causing parasites achieve rapid proliferation in human blood through multiple rounds of asynchronous nuclear division followed by daughter cell formation. Nuclear divisions critically depend on the centriolar plaque, which organizes intranuclear spindle microtubules. The centriolar plaque consists of an extranuclear compartment, which is connected via a nuclear pore-like structure to a chromatin-free intranuclear compartment. Composition and function of this non-canonical centrosome remain largely elusive. Centrins, which reside in the extranuclear part, are among the very few centrosomal proteins conserved in Plasmodium falciparum. Here we identify a novel centrin-interacting centriolar plaque protein. Conditional knock down of this Sfi1-like protein (PfSlp) caused a growth delay in blood stages, which correlated with a reduced number of daughter cells. Surprisingly, intranuclear tubulin abundance was significantly increased, which raises the hypothesis that the centriolar plaque might be implicated in regulating tubulin levels. Disruption of tubulin homeostasis caused excess microtubules and aberrant mitotic spindles. Time-lapse microscopy revealed that this prevented or delayed mitotic spindle extension but did not significantly interfere with DNA replication. Our study thereby identifies a novel extranuclear centriolar plaque factor and establishes a functional link to the intranuclear compartment of this divergent eukaryotic centrosome.
Collapse
Affiliation(s)
- Christoph Wenz
- Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | | | | | | | - Marta Machado
- Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Graduate Program in Areas of Basic and Applied Biology, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Natacha Klages
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Anja Klemmer
- Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Yannik Voß
- Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Markus Ganter
- Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Mathieu Brochet
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Julien Guizetti
- Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
31
|
Schneider V, Visone J, Harris C, Florini F, Hadjimichael E, Zhang X, Gross M, Rhee K, Ben Mamoun C, Kafsack B, Deitsch K. The human malaria parasite Plasmodium falciparum can sense environmental changes and respond by antigenic switching. Proc Natl Acad Sci U S A 2023; 120:e2302152120. [PMID: 37068249 PMCID: PMC10151525 DOI: 10.1073/pnas.2302152120] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/20/2023] [Indexed: 04/19/2023] Open
Abstract
The primary antigenic and virulence determinant of the human malaria parasite Plasmodium falciparum is a variant surface protein called PfEMP1. Different forms of PfEMP1 are encoded by a multicopy gene family called var, and switching between active genes enables the parasites to evade the antibody response of their human hosts. var gene switching is key for the maintenance of chronic infections; however, what controls switching is unknown, although it has been suggested to occur at a constant frequency with little or no environmental influence. var gene transcription is controlled epigenetically through the activity of histone methyltransferases (HMTs). Studies in model systems have shown that metabolism and epigenetic control of gene expression are linked through the availability of intracellular S-adenosylmethionine (SAM), the principal methyl donor in biological methylation modifications, which can fluctuate based on nutrient availability. To determine whether environmental conditions and changes in metabolism can influence var gene expression, P. falciparum was cultured in media with altered concentrations of nutrients involved in SAM metabolism. We found that conditions that influence lipid metabolism induce var gene switching, indicating that parasites can respond to changes in their environment by altering var gene expression patterns. Genetic modifications that directly modified expression of the enzymes that control SAM levels similarly led to profound changes in var gene expression, confirming that changes in SAM availability modulate var gene switching. These observations directly challenge the paradigm that antigenic variation in P. falciparum follows an intrinsic, programed switching rate, which operates independently of any external stimuli.
Collapse
Affiliation(s)
- Victoria M. Schneider
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, Ithaca, NY14853
- Laboratory of Chemical Biology and Microbial Pathogenesis, Rockefeller University, New York, NY 10065
| | - Joseph E. Visone
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, Ithaca, NY14853
| | - Chantal T. Harris
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, Ithaca, NY14853
| | - Francesca Florini
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, Ithaca, NY14853
| | - Evi Hadjimichael
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, Ithaca, NY14853
| | - Xu Zhang
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, Ithaca, NY14853
| | - Mackensie R. Gross
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, Ithaca, NY14853
| | - Kyu Y. Rhee
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, Ithaca, NY14853
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, Cornell University, Ithaca, NY14853
| | - Choukri Ben Mamoun
- Section of Infectious Disease, Department of Microbial Pathogenesis, Yale School of Medicine, Yale University New Haven, CT 06510
| | - Björn F. C. Kafsack
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, Ithaca, NY14853
| | - Kirk W. Deitsch
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, Ithaca, NY14853
| |
Collapse
|
32
|
Sheokand PK, Yamaryo-Botté Y, Narwal M, Arnold CS, Thakur V, Islam MM, Banday MM, Asad M, Botté CY, Mohmmed A. A Plasmodium falciparum lysophospholipase regulates host fatty acid flux via parasite lipid storage to enable controlled asexual schizogony. Cell Rep 2023; 42:112251. [PMID: 37015228 DOI: 10.1016/j.celrep.2023.112251] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 11/04/2022] [Accepted: 02/24/2023] [Indexed: 04/05/2023] Open
Abstract
Phospholipid metabolism is crucial for membrane biogenesis and homeostasis of Plasmodium falciparum. To generate such phospholipids, the parasite extensively scavenges, recycles, and reassembles host lipids. P. falciparum possesses an unusually large number of lysophospholipases, whose roles and importance remain to be elucidated. Here, we functionally characterize one P. falciparum lysophospholipase, PfLPL3, to reveal its key role in parasite propagation during asexual blood stages. PfLPL3 displays a dynamic localization throughout asexual stages, mainly localizing in the host-parasite interface. Inducible knockdown of PfLPL3 disrupts parasite development from trophozoites to schizont, inducing a drastic reduction in merozoite progenies. Detailed lipidomic analyses show that PfLPL3 generates fatty acids from scavenged host lipids to generate neutral lipids. These are then timely mobilized to allow schizogony and merozoite formation. We then identify inhibitors of PfLPL3 from Medicine for Malaria Venture (MMV) with potent antimalarial activity, which could also serve as pertinent chemical tools to study parasite lipid synthesis.
Collapse
Affiliation(s)
- Pradeep Kumar Sheokand
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India
| | - Yoshiki Yamaryo-Botté
- ApicoLipid Team, Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, Grenoble, France
| | - Monika Narwal
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India
| | - Christophe-Sébastien Arnold
- ApicoLipid Team, Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, Grenoble, France
| | - Vandana Thakur
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India
| | - Md Muzahidul Islam
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India
| | - Mudassir M Banday
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India
| | - Mohd Asad
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India
| | - Cyrille Y Botté
- ApicoLipid Team, Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, Grenoble, France.
| | - Asif Mohmmed
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India.
| |
Collapse
|
33
|
Elsworth B, Keroack C, Rezvani Y, Paul A, Barazorda K, Tennessen J, Sack S, Moreira C, Gubbels MJ, Meyers M, Zarringhalam K, Duraisingh M. Babesia divergens egress from host cells is orchestrated by essential and druggable kinases and proteases. RESEARCH SQUARE 2023:rs.3.rs-2553721. [PMID: 36909484 PMCID: PMC10002801 DOI: 10.21203/rs.3.rs-2553721/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Apicomplexan egress from host cells is fundamental to the spread of infection and is poorly characterized in Babesia spp., parasites of veterinary importance and emerging zoonoses. Through the use of video microscopy, transcriptomics and chemical genetics, we have implicated signaling, proteases and gliding motility as key drivers of egress by Babesia divergens. We developed reverse genetics to perform a knockdown screen of putative mediators of egress, identifying kinases and proteases involved in distinct steps of egress (ASP3, PKG and CDPK4) and invasion (ASP2, ASP3 and PKG). Inhibition of egress leads to continued intracellular replication, indicating exit from the replication cycle is uncoupled from egress. Chemical genetics validated PKG, ASP2 and ASP3 as druggable targets in Babesia spp. All taken together, egress in B. divergens more closely resembles T. gondii than the more evolutionarily-related Plasmodium spp. We have established a molecular framework for biological and translational studies of B. divergens egress.
Collapse
|
34
|
Wichers-Misterek JS, Binder AM, Mesén-Ramírez P, Dorner LP, Safavi S, Fuchs G, Lenz TL, Bachmann A, Wilson D, Frischknecht F, Gilberger TW. A Microtubule-Associated Protein Is Essential for Malaria Parasite Transmission. mBio 2023; 14:e0331822. [PMID: 36625655 PMCID: PMC9973338 DOI: 10.1128/mbio.03318-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 01/11/2023] Open
Abstract
Mature gametocytes of Plasmodium falciparum display a banana (falciform) shape conferred by a complex array of subpellicular microtubules (SPMT) associated with the inner membrane complex (IMC). Microtubule-associated proteins (MAPs) define MT populations and modulate interaction with pellicular components. Several MAPs have been identified in Toxoplasma gondii, and homologues can be found in the genomes of Plasmodium species, but the function of these proteins for asexual and sexual development of malaria parasites is still unknown. Here, we identified a novel subpellicular MAP, termed SPM3, that is conserved within the genus Plasmodium, especially within the subgenus Laverania, but absent in other Apicomplexa. Conditional knockdown and targeted gene disruption of Pfspm3 in Plasmodium falciparum cause severe morphological defects during gametocytogenesis, leading to round, nonfalciform gametocytes with an aberrant SPMT pattern. In contrast, Pbspm3 knockout in Plasmodium berghei, a species with round gametocytes, caused no defect in gametocytogenesis, but sporozoites displayed an aberrant motility and a dramatic defect in invasion of salivary glands, leading to a decreased efficiency in transmission. Electron microscopy revealed a dissociation of the SPMT from the IMC in Pbspm3 knockout parasites, suggesting a function of SPM3 in anchoring MTs to the IMC. Overall, our results highlight SPM3 as a pellicular component with essential functions for malaria parasite transmission. IMPORTANCE A key structural feature driving the transition between different life cycle stages of the malaria parasite is the unique three-membrane pellicle, consisting of the parasite plasma membrane (PPM) and a double membrane structure underlying the PPM termed the inner membrane complex (IMC). Additionally, there are numerous linearly arranged intramembranous particles (IMPs) linked to the IMC, which likely link the IMC to the subpellicular microtubule cytoskeleton. Here, we identified, localized, and characterized a novel subpellicular microtubule-associated protein unique to the genus Plasmodium. The knockout of this protein in the human-pathogenic species P. falciparum resulted in malformed gametocytes and aberrant microtubules. We confirmed the microtubule association in the P. berghei rodent malaria homologue and show that its knockout results in a perturbed microtubule architecture, aberrant sporozoite motility, and decreased transmission efficiency.
Collapse
Affiliation(s)
- Jan Stephan Wichers-Misterek
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
| | - Annika M. Binder
- Integrative Parasitology, Department of Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Paolo Mesén-Ramírez
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
| | - Lilian Patrick Dorner
- Integrative Parasitology, Department of Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Soraya Safavi
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
| | - Gwendolin Fuchs
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
| | - Tobias L. Lenz
- Biology Department, University of Hamburg, Hamburg, Germany
- Research Unit for Evolutionary Immunogenomics, Department of Biology, University of Hamburg, Hamburg, Germany
| | - Anna Bachmann
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Borstel-Lübeck-Riems, Hamburg, Germany
| | - Danny Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
- Burnet Institute, Melbourne, Victoria, Australia
- Institute for Photonics and Advanced Sensing, University of Adelaide, Adelaide, South Australia, Australia
| | - Friedrich Frischknecht
- Integrative Parasitology, Department of Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
- German Center for Infection Research, Partner Site Heidelberg, Heidelberg, Germany
| | - Tim-Wolf Gilberger
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
| |
Collapse
|
35
|
A Plasmodium falciparum RING Finger E3 Ubiquitin Ligase Modifies the Roles of PfMDR1 and PfCRT in Parasite Drug Responses. Antimicrob Agents Chemother 2023; 67:e0082122. [PMID: 36625569 PMCID: PMC9933707 DOI: 10.1128/aac.00821-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Protein ubiquitination is an important posttranslational regulation mechanism that mediates Plasmodium development and modifies parasite responses to antimalarial drugs. Although mutations in several parasite ubiquitination enzymes have been linked to increased drug tolerance, the molecular mechanisms by which ubiquitination pathways mediate these parasite responses remain largely unknown. Here, we investigate the roles of a Plasmodium falciparum ring finger ubiquitin ligase (PfRFUL) in parasite development and in responses to antimalarial drugs. We engineered a transgenic parasite having the Pfrful gene tagged with an HA-2A-NeoR-glmS sequence to knockdown (KD) Pfrful expression using glucosamine (GlcN). A Western blot analysis of the proteins from GlcN-treated pSLI-HA-NeoR-glmS-tagged (PfRFULg) parasites, relative to their wild-type (Dd2) controls, showed changes in the ubiquitination of numerous proteins. PfRFUL KD rendered the parasites more sensitive to multiple antimalarial drugs, including mefloquine, piperaquine, amodiaquine, and dihydroartemisinin. PfRFUL KD also decreased the protein level of the P. falciparum multiple drug resistance 1 protein (PfMDR1) and altered the ratio of two bands of the P. falciparum chloroquine resistance transporter (PfCRT), suggesting contributions to the changed drug responses by the altered ubiquitination of these two molecules. The inhibition of proteasomal protein degradation by epoxomicin increased the PfRFUL level, suggesting the degradation of PfRFUL by the proteasome pathways, whereas the inhibition of E3 ubiquitin ligase activities by JNJ26854165 reduced the PfRFUL level. This study reveals the potential mechanisms of PfRFUL in modifying the expression of drug transporters and their roles in parasite drug responses. PfRFUL could be a potential target for antimalarial drug development.
Collapse
|
36
|
Quansah E, Chen Y, Yang S, Wang J, Sun D, Zhao Y, Chen M, Yu L, Zhang C. CRISPR-Cas13 in malaria parasite: Diagnosis and prospective gene function identification. Front Microbiol 2023; 14:1076947. [PMID: 36760507 PMCID: PMC9905151 DOI: 10.3389/fmicb.2023.1076947] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/03/2023] [Indexed: 01/26/2023] Open
Abstract
Malaria caused by Plasmodium is still a serious public health problem. Genomic editing is essential to understand parasite biology, elucidate mechanical pathways, uncover gene functions, identify novel therapeutic targets, and develop clinical diagnostic tools. Recent advances have seen the development of genomic diagnostic technologies and the emergence of genetic manipulation toolbox comprising a host of several systems for editing the genome of Plasmodium at the DNA, RNA, and protein level. Genomic manipulation at the RNA level is critical as it allows for the functional characterization of several transcripts. Of notice, some developed artificial RNA genome editing tools hinge on the endogenous RNA interference system of Plasmodium. However, Plasmodium lacks a robust RNAi machinery, hampering the progress of these editing tools. CRISPR-Cas13, which belongs to the VI type of the CRISPR system, can specifically bind and cut RNA under the guidance of crRNA, with no or minimal permanent genetic scar on genes. This review summarizes CRISPR-Cas13 system from its discovery, classification, principle of action, and diagnostic platforms. Further, it discusses the application prospects of Cas13-based systems in Plasmodium and highlights its advantages and drawbacks.
Collapse
Affiliation(s)
- Elvis Quansah
- Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Key Laboratory of Zoonoses, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yihuan Chen
- The Second Clinical Medical College, Anhui Medical University, Hefei, China
| | - Shijie Yang
- The Second Clinical Medical College, Anhui Medical University, Hefei, China
| | - Junyan Wang
- The Second Clinical Medical College, Anhui Medical University, Hefei, China
| | - Danhong Sun
- The Second Clinical Medical College, Anhui Medical University, Hefei, China
| | - Yangxi Zhao
- The First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Ming Chen
- The Second Clinical Medical College, Anhui Medical University, Hefei, China
| | - Li Yu
- Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Key Laboratory of Zoonoses, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China,*Correspondence: Li Yu, ✉
| | - Chao Zhang
- Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Key Laboratory of Zoonoses, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China,Chao Zhang, ✉
| |
Collapse
|
37
|
Hussain T, Linera-Gonzalez J, Beck JM, Fierro MA, Mair GR, Smith RC, Beck JR. The PTEX Pore Component EXP2 Is Important for Intrahepatic Development during the Plasmodium Liver Stage. mBio 2022; 13:e0309622. [PMID: 36445080 PMCID: PMC9765067 DOI: 10.1128/mbio.03096-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 12/02/2022] Open
Abstract
During vertebrate infection, obligate intracellular malaria parasites develop within a parasitophorous vacuole, which constitutes the interface between the parasite and its hepatocyte or erythrocyte host cells. To traverse this barrier, Plasmodium spp. utilize a dual-function pore formed by EXP2 for nutrient transport and, in the context of the PTEX translocon, effector protein export across the vacuole membrane. While critical to blood-stage survival, less is known about EXP2/PTEX function in the liver stage, although major differences in the export mechanism are suggested by absence of the PTEX unfoldase HSP101 in the intrahepatic vacuole. Here, we employed the glucosamine-activated glmS ribozyme to study the role of EXP2 during Plasmodium berghei liver-stage development in hepatoma cells. Insertion of the glmS sequence into the exp2 3' untranslated region (UTR) enabled glucosamine-dependent depletion of EXP2 after hepatocyte invasion, allowing separation of EXP2 function during intrahepatic development from a recently reported role in hepatocyte invasion. Postinvasion EXP2 knockdown reduced parasite size and largely abolished expression of the mid- to late-liver-stage marker LISP2. As an orthogonal approach to monitor development, EXP2-glmS parasites and controls were engineered to express nanoluciferase. Activation of glmS after invasion substantially decreased luminescence in hepatoma monolayers and in culture supernatants at later time points corresponding to merosome detachment, which marks the culmination of liver-stage development. Collectively, our findings extend the utility of the glmS ribozyme to study protein function in the liver stage and reveal that EXP2 is important for intrahepatic parasite development, indicating that PTEX components also function at the hepatocyte-parasite interface. IMPORTANCE After the mosquito bite that initiates a Plasmodium infection, parasites first travel to the liver and develop in hepatocytes. This liver stage is asymptomatic but necessary for the parasite to transition to the merozoite form, which infects red blood cells and causes malaria. To take over their host cells, avoid immune defenses, and fuel their growth, these obligately intracellular parasites must import nutrients and export effector proteins across a vacuole membrane in which they reside. In the blood stage, these processes depend on a translocon called PTEX, but it is unclear if PTEX also functions during the liver stage. Here, we adapted the glmS ribozyme to control expression of EXP2, the membrane pore component of PTEX, during the liver stage of the rodent malaria parasite Plasmodium berghei. Our results show that EXP2 is important for intracellular development in the hepatocyte, revealing that PTEX components are also functionally important during liver-stage infection.
Collapse
Affiliation(s)
- Tahir Hussain
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| | | | - John M. Beck
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, Iowa, USA
| | - Manuel A. Fierro
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| | - Gunnar R. Mair
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| | - Ryan C. Smith
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, Iowa, USA
| | - Josh R. Beck
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, Iowa, USA
| |
Collapse
|
38
|
Su XZ, Wu J, Xu F, Pattaradilokrat S. Genetic mapping of determinants in drug resistance, virulence, disease susceptibility, and interaction of host-rodent malaria parasites. Parasitol Int 2022; 91:102637. [PMID: 35926693 PMCID: PMC9452477 DOI: 10.1016/j.parint.2022.102637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 12/31/2022]
Abstract
Genetic mapping has been widely employed to search for genes linked to phenotypes/traits of interest. Because of the ease of maintaining rodent malaria parasites in laboratory mice, many genetic crosses of rodent malaria parasites have been performed to map the parasite genes contributing to malaria parasite development, drug resistance, host immune response, and disease pathogenesis. Drs. Richard Carter, David Walliker, and colleagues at the University of Edinburgh, UK, were the pioneers in developing the systems for genetic mapping of malaria parasite traits, including characterization of genetic markers to follow the inheritance and recombination of parasite chromosomes and performing the first genetic cross using rodent malaria parasites. Additionally, many genetic crosses of inbred mice have been performed to link mouse chromosomal loci to the susceptibility to malaria parasite infections. In this chapter, we review and discuss past and recent advances in genetic marker development, performing genetic crosses, and genetic mapping of both parasite and host genes. Genetic mappings using models of rodent malaria parasites and inbred mice have contributed greatly to our understanding of malaria, including parasite development within their hosts, mechanism of drug resistance, and host-parasite interaction.
Collapse
Affiliation(s)
- Xin-Zhuan Su
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA.
| | - Jian Wu
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Fangzheng Xu
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
39
|
Russell TJ, De Silva EK, Crowley VM, Shaw-Saliba K, Dube N, Josling G, Pasaje CFA, Kouskoumvekaki I, Panagiotou G, Niles JC, Jacobs-Lorena M, Denise Okafor C, Gamo FJ, Llinás M. Inhibitors of ApiAP2 protein DNA binding exhibit multistage activity against Plasmodium parasites. PLoS Pathog 2022; 18:e1010887. [PMID: 36223427 PMCID: PMC9591056 DOI: 10.1371/journal.ppat.1010887] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 10/24/2022] [Accepted: 09/17/2022] [Indexed: 11/06/2022] Open
Abstract
Plasmodium parasites are reliant on the Apicomplexan AP2 (ApiAP2) transcription factor family to regulate gene expression programs. AP2 DNA binding domains have no homologs in the human or mosquito host genomes, making them potential antimalarial drug targets. Using an in-silico screen to dock thousands of small molecules into the crystal structure of the AP2-EXP (Pf3D7_1466400) AP2 domain (PDB:3IGM), we identified putative AP2-EXP interacting compounds. Four compounds were found to block DNA binding by AP2-EXP and at least one additional ApiAP2 protein. Our top ApiAP2 competitor compound perturbs the transcriptome of P. falciparum trophozoites and results in a decrease in abundance of log2 fold change > 2 for 50% (46/93) of AP2-EXP target genes. Additionally, two ApiAP2 competitor compounds have multi-stage anti-Plasmodium activity against blood and mosquito stage parasites. In summary, we describe a novel set of antimalarial compounds that interact with AP2 DNA binding domains. These compounds may be used for future chemical genetic interrogation of ApiAP2 proteins or serve as starting points for a new class of antimalarial therapeutics.
Collapse
Affiliation(s)
- Timothy James Russell
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, Pennsylvania, United States of America
- Huck Institutes Center for Eukaryotic Gene Regulation (CEGR), Pennsylvania State University, State College, Pennsylvania, United States of America
- Huck Institutes Center for Malaria Research (CMaR), Pennsylvania State University, State College, Pennsylvania, United States of America
- Huck Institutes Center for Infectious Disease Dynamics, Pennsylvania State University, State College, Pennsylvania, United States of America
| | - Erandi K. De Silva
- Lewis-Singler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
| | - Valerie M. Crowley
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, Pennsylvania, United States of America
| | - Kathryn Shaw-Saliba
- Department of Molecular Biology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Namita Dube
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, Pennsylvania, United States of America
| | - Gabrielle Josling
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, Pennsylvania, United States of America
- Huck Institutes Center for Malaria Research (CMaR), Pennsylvania State University, State College, Pennsylvania, United States of America
- Huck Institutes Center for Infectious Disease Dynamics, Pennsylvania State University, State College, Pennsylvania, United States of America
| | - Charisse Flerida A. Pasaje
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Irene Kouskoumvekaki
- Department of Systems Biology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Gianni Panagiotou
- Systems Biology and Bioinformatics, Leibniz Institute for Natural Products Research and Infection Biology, Hans Knöll Institute, Jena, Germany
- Department of Medicine, the University of Hong Kong, Hong Kong SAR, China
| | - Jacquin C. Niles
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Marcelo Jacobs-Lorena
- Department of Molecular Biology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - C. Denise Okafor
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, Pennsylvania, United States of America
- Department of Chemistry, Pennsylvania State University, State College, Pennsylvania, United States of America
| | | | - Manuel Llinás
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, Pennsylvania, United States of America
- Huck Institutes Center for Eukaryotic Gene Regulation (CEGR), Pennsylvania State University, State College, Pennsylvania, United States of America
- Huck Institutes Center for Malaria Research (CMaR), Pennsylvania State University, State College, Pennsylvania, United States of America
- Huck Institutes Center for Infectious Disease Dynamics, Pennsylvania State University, State College, Pennsylvania, United States of America
- Department of Chemistry, Pennsylvania State University, State College, Pennsylvania, United States of America
| |
Collapse
|
40
|
Edgar RCS, Siddiqui G, Hjerrild K, Malcolm TR, Vinh NB, Webb CT, Holmes C, MacRaild CA, Chernih HC, Suen WW, Counihan NA, Creek DJ, Scammells PJ, McGowan S, de Koning-Ward TF. Genetic and chemical validation of Plasmodium falciparum aminopeptidase PfA-M17 as a drug target in the hemoglobin digestion pathway. eLife 2022; 11:e80813. [PMID: 36097817 PMCID: PMC9470162 DOI: 10.7554/elife.80813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/02/2022] [Indexed: 11/18/2022] Open
Abstract
Plasmodium falciparum, the causative agent of malaria, remains a global health threat as parasites continue to develop resistance to antimalarial drugs used throughout the world. Accordingly, drugs with novel modes of action are desperately required to combat malaria. P. falciparum parasites infect human red blood cells where they digest the host's main protein constituent, hemoglobin. Leucine aminopeptidase PfA-M17 is one of several aminopeptidases that have been implicated in the last step of this digestive pathway. Here, we use both reverse genetics and a compound specifically designed to inhibit the activity of PfA-M17 to show that PfA-M17 is essential for P. falciparum survival as it provides parasites with free amino acids for growth, many of which are highly likely to originate from hemoglobin. We further show that loss of PfA-M17 results in parasites exhibiting multiple digestive vacuoles at the trophozoite stage. In contrast to other hemoglobin-degrading proteases that have overlapping redundant functions, we validate PfA-M17 as a potential novel drug target.
Collapse
Affiliation(s)
- Rebecca CS Edgar
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Ghizal Siddiqui
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | | | - Tess R Malcolm
- Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityMelbourneAustralia
| | - Natalie B Vinh
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Chaille T Webb
- Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityMelbourneAustralia
| | - Clare Holmes
- CSIRO Australian Centre for Disease PreparednessGeelongAustralia
| | - Christopher A MacRaild
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Hope C Chernih
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Willy W Suen
- CSIRO Australian Centre for Disease PreparednessGeelongAustralia
| | - Natalie A Counihan
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Darren J Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Peter J Scammells
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Sheena McGowan
- Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityMelbourneAustralia
| | - Tania F de Koning-Ward
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| |
Collapse
|
41
|
Sanchez CP, Manson EDT, Moliner Cubel S, Mandel L, Weidt SK, Barrett MP, Lanzer M. The Knock-Down of the Chloroquine Resistance Transporter PfCRT Is Linked to Oligopeptide Handling in Plasmodium falciparum. Microbiol Spectr 2022; 10:e0110122. [PMID: 35867395 PMCID: PMC9431119 DOI: 10.1128/spectrum.01101-22] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 06/27/2022] [Indexed: 11/20/2022] Open
Abstract
The chloroquine resistance transporter, PfCRT, is an essential factor during intraerythrocytic development of the human malaria parasite Plasmodium falciparum. PfCRT resides at the digestive vacuole of the parasite, where hemoglobin taken up by the parasite from its host cell is degraded. PfCRT can acquire several mutations that render PfCRT a drug transporting system expelling compounds targeting hemoglobin degradation from the digestive vacuole. The non-drug related function of PfCRT is less clear, although a recent study has suggested a role in oligopeptide transport based on studies conducted in a heterologous expression system. The uncertainty about the natural function of PfCRT is partly due to a lack of a null mutant and a dearth of functional assays in the parasite. Here, we report on the generation of a conditional PfCRT knock-down mutant in P. falciparum. The mutant accumulated oligopeptides 2 to at least 8 residues in length under knock-down conditions, as shown by comparative global metabolomics. The accumulated oligopeptides were structurally diverse, had an isoelectric point between 4.0 and 5.4 and were electrically neutral or carried a single charge at the digestive vacuolar pH of 5.2. Fluorescently labeled dipeptides and live cell imaging identified the digestive vacuole as the compartment where oligopeptides accumulated. Our findings suggest a function of PfCRT in oligopeptide transport across the digestive vacuolar membrane in P. falciparum and associated with it a role in nutrient acquisition and the maintenance of the colloid osmotic balance. IMPORTANCE The chloroquine resistance transporter, PfCRT, is important for the survival of the human malaria parasite Plasmodium falciparum. It increases the tolerance to many antimalarial drugs, and it is essential for the development of the parasite within red blood cells. While we understand the role of PfCRT in drug resistance in ever increasing detail, the non-drug resistance functions are still debated. Identifying the natural substrate of PfCRT has been hampered by a paucity of functional assays to test putative substrates in the parasite system and the absence of a parasite mutant deficient for the PfCRT encoding gene. By generating a conditional PfCRT knock-down mutant, together with comparative metabolomics and uptake studies using fluorescently labeled oligopeptides, we could show that PfCRT is an oligopeptide transporter. The oligopeptides were structurally diverse and were electrically neutral or carried a single charge. Our data support a function of PfCRT in oligopeptide transport.
Collapse
Affiliation(s)
- Cecilia P. Sanchez
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | | | - Sonia Moliner Cubel
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | | | - Stefan K. Weidt
- Glasgow Polyomics, University of Glasgow, Wolfson Wohl Cancer Research Centre, Glasgow, United Kingdom
| | - Michael P. Barrett
- Glasgow Polyomics, University of Glasgow, Wolfson Wohl Cancer Research Centre, Glasgow, United Kingdom
- The Wellcome Centre for Integrative Parasitology, Institute for Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Michael Lanzer
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| |
Collapse
|
42
|
A nuclear redox sensor modulates gene activation and var switching in Plasmodium falciparum. Proc Natl Acad Sci U S A 2022; 119:e2201247119. [PMID: 35939693 PMCID: PMC9388093 DOI: 10.1073/pnas.2201247119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The virulence of Plasmodium falciparum, which causes the deadliest form of human malaria, is attributed to its ability to evade the human immune response. These parasites "choose" to express a single variant from a repertoire of surface antigens called PfEMP1, which are placed on the surface of the infected red cell. Immune evasion is achieved by switches in expression between var genes, each encoding a different PfEMP1 variant. While the mechanisms that regulate mutually exclusive expression of var genes are still elusive, antisense long-noncoding RNAs (lncRNAs) transcribed from the intron of the active var gene were implicated in the "choice" of the single active var gene. Here, we show that this lncRNA colocalizes with the site of var mRNA transcription and is anchored to the var locus via DNA:RNA interactions. We define the var lncRNA interactome and identify a redox sensor, P. falciparum thioredoxin peroxidase I (PfTPx-1), as one of the proteins associated with the var antisense lncRNA. We show that PfTPx-1 localizes to a nuclear subcompartment associated with active transcription on the nuclear periphery, in ring-stage parasite, when var transcription occurs. In addition, PfTPx-1 colocalizes with S-adenosylmethionine synthetase (PfSAMS) in the nucleus, and its overexpression leads to activation of var2csa, similar to overexpression of PfSAMS. Furthermore, we show that PfTPx-1 knockdown alters the var switch rate as well as activation of additional gene subsets. Taken together, our data indicate that nuclear PfTPx-1 plays a role in gene activation possibly by providing a redox-controlled nuclear microenvironment ideal for active transcription.
Collapse
|
43
|
The Plasmodium falciparum Nuclear Protein Phosphatase NIF4 Is Required for Efficient Merozoite Invasion and Regulates Artemisinin Sensitivity. mBio 2022; 13:e0189722. [PMID: 35938722 PMCID: PMC9426563 DOI: 10.1128/mbio.01897-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Artemisinin resistance in Plasmodium falciparum has been associated with a mutation in the NLI-interacting factor-like phosphatase PfNIF4, in addition to the mutations in the Kelch13 protein as the major determinant. We found that PfNIF4 was predominantly expressed at the schizont stage and localized in the nuclei of the parasite. To elucidate the functions of PfNIF4 in P. falciparum, we performed PfNIF4 knockdown (KD) using the inducible ribozyme system. PfNIF4 KD attenuated merozoite invasion and affected gametocytogenesis. PfNIF4 KD parasites also showed significantly increased in vitro susceptibility to artemisinins. Transcriptomic and proteomic analysis revealed that PfNIF4 KD led to the downregulation of gene categories involved in invasion and artemisinin resistance (e.g., mitochondrial function, membrane, and Kelch13 interactome) at the trophozoite and/or schizont stage. Consistent with PfNIF4 being a protein phosphatase, PfNIF4 KD resulted in an overall upregulation of the phosphoproteome of infected erythrocytes. Quantitative phosphoproteomic profiling identified a set of PfNIF4-regulated phosphoproteins with functional similarity to FCP1 substrates, particularly proteins involved in chromatin organization and transcriptional regulation. Specifically, we observed increased phosphorylation of Ser2/5 of the tandem repeats in the C-terminal domain (CTD) of RNA polymerase II (RNAPII) upon PfNIF4 KD. Furthermore, using the TurboID-based proteomic approach, we identified that PfNIF4 interacted with the RNAPII components, AP2-domain transcription factors, and chromatin-modifiers and binders. These findings suggest that PfNIF4 may act as the RNAPII CTD phosphatase, regulating the expression of general and parasite-specific cellular pathways during the blood-stage development.
Collapse
|
44
|
Roling L, Flammersfeld A, Pradel G, Bennink S. The WD40-Protein PfWLP1 Ensures Stability of the PfCCp-Based Adhesion Protein Complex in Plasmodium falciparum Gametocytes. Front Cell Infect Microbiol 2022; 12:942364. [PMID: 35923798 PMCID: PMC9339629 DOI: 10.3389/fcimb.2022.942364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Members of the WD40-repeat protein family can be found in all eukaryotic proteomes where they usually serve as interaction platforms for the assembly of large protein complexes and are therefore essential for the integrity of these complexes. In the malaria parasite Plasmodium falciparum, the WD40-repeat protein PfWLP1 has been shown to interact with members of distinct adhesion protein complexes in the asexual blood stages and gametocyte stages. In this study, we demonstrate that the presence of PfWLP1 is crucial for both the stability of these gametocyte-specific adhesion complexes as well as for gametocyte maturation and gametogenesis. Using reverse genetics, we generated a PfWLP1-knockdown parasite line for functional characterization of the protein. Knockdown of PfWLP1 resulted in a slight reduction of gametocyte numbers and significantly the impaired ability of the gametocytes to exflagellate. PfWLP1-knockdown further led to reduced protein levels of the Limulus coagulation factor C-like (LCCL)-domain proteins PfCCp1 and PfCCp2, which are key components of the adhesion complexes. These findings suggest that the interaction of PfWLP1 with members of the PfCCp-based adhesion complex ensures complex stability and thereby contributes to gametocyte viability and exflagellation.
Collapse
|
45
|
Musabyimana JP, Distler U, Sassmannshausen J, Berks C, Manti J, Bennink S, Blaschke L, Burda PC, Flammersfeld A, Tenzer S, Ngwa CJ, Pradel G. Plasmodium falciparum S-Adenosylmethionine Synthetase Is Essential for Parasite Survival through a Complex Interaction Network with Cytoplasmic and Nuclear Proteins. Microorganisms 2022; 10:1419. [PMID: 35889137 PMCID: PMC9320499 DOI: 10.3390/microorganisms10071419] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/01/2022] [Accepted: 07/11/2022] [Indexed: 11/16/2022] Open
Abstract
S-adenosylmethionine synthetase (SAMS) is a key enzyme for the synthesis of the lone methyl donor S-adenosyl methionine (SAM), which is involved in transmethylation reactions and hence required for cellular processes such as DNA, RNA, and histone methylation, but also polyamine biosynthesis and proteostasis. In the human malaria parasite Plasmodium falciparum, PfSAMS is encoded by a single gene and has been suggested to be crucial for malaria pathogenesis and transmission; however, to date, PfSAMS has not been fully characterized. To gain deeper insight into the function of PfSAMS, we generated a conditional gene knockdown (KD) using the glmS ribozyme system. We show that PfSAMS localizes to the cytoplasm and the nucleus of blood-stage parasites. PfSAMS-KD results in reduced histone methylation and leads to impaired intraerythrocytic growth and gametocyte development. To further determine the interaction network of PfSAMS, we performed a proximity-dependent biotin identification analysis. We identified a complex network of 1114 proteins involved in biological processes such as cell cycle control and DNA replication, or transcription, but also in phosphatidylcholine and polyamine biosynthesis and proteasome regulation. Our findings highlight the diverse roles of PfSAMS during intraerythrocytic growth and sexual stage development and emphasize that PfSAMS is a potential drug target.
Collapse
Affiliation(s)
- Jean Pierre Musabyimana
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (J.P.M.); (J.S.); (C.B.); (J.M.); (S.B.); (L.B.); (A.F.); (C.J.N.)
| | - Ute Distler
- Proteomics Core Facility, Institute of Immunology, University Medical Center of the Johannes-Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (U.D.); (S.T.)
| | - Juliane Sassmannshausen
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (J.P.M.); (J.S.); (C.B.); (J.M.); (S.B.); (L.B.); (A.F.); (C.J.N.)
| | - Christina Berks
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (J.P.M.); (J.S.); (C.B.); (J.M.); (S.B.); (L.B.); (A.F.); (C.J.N.)
| | - Janice Manti
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (J.P.M.); (J.S.); (C.B.); (J.M.); (S.B.); (L.B.); (A.F.); (C.J.N.)
| | - Sandra Bennink
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (J.P.M.); (J.S.); (C.B.); (J.M.); (S.B.); (L.B.); (A.F.); (C.J.N.)
| | - Lea Blaschke
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (J.P.M.); (J.S.); (C.B.); (J.M.); (S.B.); (L.B.); (A.F.); (C.J.N.)
| | - Paul-Christian Burda
- Centre for Structural Systems Biology (CSSB) c/o DESY, Bernhard Nocht Institute, University of Hamburg, Notkestraße 85, Building 15, 22607 Hamburg, Germany;
| | - Ansgar Flammersfeld
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (J.P.M.); (J.S.); (C.B.); (J.M.); (S.B.); (L.B.); (A.F.); (C.J.N.)
| | - Stefan Tenzer
- Proteomics Core Facility, Institute of Immunology, University Medical Center of the Johannes-Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (U.D.); (S.T.)
| | - Che Julius Ngwa
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (J.P.M.); (J.S.); (C.B.); (J.M.); (S.B.); (L.B.); (A.F.); (C.J.N.)
| | - Gabriele Pradel
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (J.P.M.); (J.S.); (C.B.); (J.M.); (S.B.); (L.B.); (A.F.); (C.J.N.)
| |
Collapse
|
46
|
Liang X, Boonhok R, Siddiqui FA, Xiao B, Li X, Qin J, Min H, Jiang L, Cui L, Miao J. A Leak-Free Inducible CRISPRi/a System for Gene Functional Studies in Plasmodium falciparum. Microbiol Spectr 2022; 10:e0278221. [PMID: 35510853 PMCID: PMC9241666 DOI: 10.1128/spectrum.02782-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 04/18/2022] [Indexed: 12/16/2022] Open
Abstract
By fusing catalytically dead Cas9 (dCas9) to active domains of histone deacetylase (Sir2a) or acetyltransferase (GCN5), this CRISPR interference/activation (CRISPRi/a) system allows gene regulation at the transcriptional level without causing permanent changes in the parasite genome. However, the constitutive expression of dCas9 poses a challenge for studying essential genes, which may lead to adaptive changes in the parasite, masking the true phenotypes. Here, we developed a leak-free inducible CRISPRi/a system by integrating the DiCre/loxP regulon to allow the expression of dCas9-GCN5/-Sir2a upon transient induction with rapamycin, which allows convenient transcriptional regulation of a gene of interest by introducing a guide RNA targeting its transcription start region. Using eight genes that are either silent or expressed from low to high levels during asexual erythrocytic development, we evaluated the robustness and versatility of this system in the asexual parasites. For most genes analyzed, this inducible CRISPRi/a system led to 1.5- to 3-fold up-or downregulation of the target genes at the mRNA level. Alteration in the expression of PfK13 and PfMYST resulted in altered sensitivities to artemisinin. For autophagy-related protein 18, an essential gene related to artemisinin resistance, a >2-fold up- or downregulation was obtained by inducible CRISPRi/a, leading to growth retardation. For the master regulator of gametocytogenesis, PfAP2-G, a >10-fold increase of the PfAP2-G transcripts was obtained by CRISPRa, resulting in >4-fold higher gametocytemia in the induced parasites. Additionally, inducible CRISPRi/a could also regulate gene expression in gametocytes. This inducible epigenetic regulation system offers a fast way of studying gene functions in Plasmodium falciparum. IMPORTANCE Understanding the fundamental biology of malaria parasites through functional genetic/genomic studies is critical for identifying novel targets for antimalarial development. Conditional knockout/knockdown systems are required to study essential genes in the haploid blood stages of the parasite. In this study, we developed an inducible CRISPRi/a system via the integration of DiCre/loxP. We evaluated the robustness and versatility of this system by activating or repressing eight selected genes and achieved up- and downregulation of the targeted genes located in both the euchromatin and heterochromatin regions. This system offers the malaria research community another tool for functional genetic studies.
Collapse
Affiliation(s)
- Xiaoying Liang
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Rachasak Boonhok
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Faiza Amber Siddiqui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Bo Xiao
- Unit of Human Parasite Molecular and Cell Biology, Key Laboratory of Molecular Virology and Immunology, Pasteur Institute of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Xiaolian Li
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Junling Qin
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Hui Min
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Lubin Jiang
- Unit of Human Parasite Molecular and Cell Biology, Key Laboratory of Molecular Virology and Immunology, Pasteur Institute of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Jun Miao
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
47
|
Wichers JS, Mesén-Ramírez P, Fuchs G, Yu-Strzelczyk J, Stäcker J, von Thien H, Alder A, Henshall I, Liffner B, Nagel G, Löw C, Wilson D, Spielmann T, Gao S, Gilberger TW, Bachmann A, Strauss J. PMRT1, a Plasmodium-Specific Parasite Plasma Membrane Transporter, Is Essential for Asexual and Sexual Blood Stage Development. mBio 2022; 13:e0062322. [PMID: 35404116 PMCID: PMC9040750 DOI: 10.1128/mbio.00623-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 12/12/2022] Open
Abstract
Membrane transport proteins perform crucial roles in cell physiology. The obligate intracellular parasite Plasmodium falciparum, an agent of human malaria, relies on membrane transport proteins for the uptake of nutrients from the host, disposal of metabolic waste, exchange of metabolites between organelles, and generation and maintenance of transmembrane electrochemical gradients for its growth and replication within human erythrocytes. Despite their importance for Plasmodium cellular physiology, the functional roles of a number of membrane transport proteins remain unclear, which is particularly true for orphan membrane transporters that have no or limited sequence homology to transporter proteins in other evolutionary lineages. Therefore, in the current study, we applied endogenous tagging, targeted gene disruption, conditional knockdown, and knockout approaches to investigate the subcellular localization and essentiality of six membrane transporters during intraerythrocytic development of P. falciparum parasites. They are localized at different subcellular structures-the food vacuole, the apicoplast, and the parasite plasma membrane-and four out of the six membrane transporters are essential during asexual development. Additionally, the plasma membrane resident transporter 1 (PMRT1; PF3D7_1135300), a unique Plasmodium-specific plasma membrane transporter, was shown to be essential for gametocytogenesis and functionally conserved within the genus Plasmodium. Overall, we reveal the importance of four orphan transporters to blood stage P. falciparum development, which have diverse intracellular localizations and putative functions. IMPORTANCE Plasmodium falciparum-infected erythrocytes possess multiple compartments with designated membranes. Transporter proteins embedded in these membranes not only facilitate movement of nutrients, metabolites, and other molecules between these compartments, but also are common therapeutic targets and can confer antimalarial drug resistance. Orphan membrane transporters in P. falciparum without sequence homology to transporters in other evolutionary lineages and divergent from host transporters may constitute attractive targets for novel intervention approaches. Here, we localized six of these putative transporters at different subcellular compartments and probed their importance during asexual parasite growth by using reverse genetic approaches. In total, only two candidates turned out to be dispensable for the parasite, highlighting four candidates as putative targets for therapeutic interventions. This study reveals the importance of several orphan transporters to blood stage P. falciparum development.
Collapse
Affiliation(s)
- Jan Stephan Wichers
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
| | | | - Gwendolin Fuchs
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
| | - Jing Yu-Strzelczyk
- Institute of Physiology, Department of Neurophysiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Jan Stäcker
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Heidrun von Thien
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Arne Alder
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
| | - Isabelle Henshall
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Benjamin Liffner
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Georg Nagel
- Institute of Physiology, Department of Neurophysiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Christian Löw
- Centre for Structural Systems Biology, Hamburg, Germany
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| | - Danny Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia
- Burnet Institute, Melbourne, Victoria, Australia
| | - Tobias Spielmann
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Shiqiang Gao
- Institute of Physiology, Department of Neurophysiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Tim-Wolf Gilberger
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
| | - Anna Bachmann
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
| | - Jan Strauss
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| |
Collapse
|
48
|
Quinn JE, Jeninga MD, Limm K, Pareek K, Meißgeier T, Bachmann A, Duffy MF, Petter M. The Putative Bromodomain Protein PfBDP7 of the Human Malaria Parasite Plasmodium Falciparum Cooperates With PfBDP1 in the Silencing of Variant Surface Antigen Expression. Front Cell Dev Biol 2022; 10:816558. [PMID: 35493110 PMCID: PMC9039026 DOI: 10.3389/fcell.2022.816558] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/18/2022] [Indexed: 01/08/2023] Open
Abstract
Epigenetic regulation is a critical mechanism in controlling virulence, differentiation, and survival of the human malaria parasite Plasmodium (P.) falciparum. Bromodomain proteins contribute to this process by binding to acetylated lysine residues of histones and thereby targeting the gene regulatory machinery to gene promoters. A protein complex containing the P. falciparum bromodomain proteins (PfBDP) 1 and PfBDP2 (BDP1/BDP2 core complex) was previously shown to play an essential role for the correct transcription of invasion related genes. Here, we performed a functional characterization of a third component of this complex, which we dubbed PfBDP7, because structural modelling predicted a typical bromodomain fold. We confirmed that PfBDP7 is a nuclear protein that interacts with PfBDP1 at invasion gene promoters in mature schizont stage parasites and contributes to their transcription. Although partial depletion of PfBDP7 showed no significant effect on parasite viability, conditional knock down of either PfBDP7 or PfBDP1 resulted in the de-repression of variant surface antigens (VSA), which are important pathogenicity factors. This de-repression was evident both on mRNA and protein level. To understand the underlying mechanism, we mapped the genome wide binding sites of PfBDP7 by ChIPseq and showed that in early schizonts, PfBDP7 and PfBDP1 are commonly enriched in heterochromatic regions across the gene body of all VSA families, including genes coding for PfEMP1, RIFIN, STEVOR, and PfMC-2TM. This suggests that PfBDP7 and PfBDP1 contribute to the silencing of VSAs by associating with heterochromatin. In conclusion, we identified PfBDP7 as a chromatin binding protein that is a constitutive part of the P. falciparum BDP1/BDP2 core complex and established PfBDP1 and PfBDP7 as novel players in the silencing of heterochromatin regulated virulence gene families of the malaria parasite P. falciparum.
Collapse
Affiliation(s)
- Jennifer E. Quinn
- Mikrobiologisches Institut—Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Myriam D. Jeninga
- Mikrobiologisches Institut—Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Katharina Limm
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Kapil Pareek
- Mikrobiologisches Institut—Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Tina Meißgeier
- Mikrobiologisches Institut—Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Bachmann
- Department of Cellular Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Centre for Structural Systems Biology (CSSB), Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
| | - Michael F. Duffy
- Department of Microbiology and Immunology, The University of Melbourne, Bio21 Institute, Parkville, VIC, Australia
| | - Michaela Petter
- Mikrobiologisches Institut—Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Department of Medicine, The University of Melbourne, Royal Melbourne Hospital, Parkville, VIC, Australia
- *Correspondence: Michaela Petter,
| |
Collapse
|
49
|
Bullen HE, Sanders PR, Dans MG, Jonsdottir TK, Riglar DT, Looker O, Palmer CS, Kouskousis B, Charnaud SC, Triglia T, Gabriela M, Schneider MP, Chan J, de Koning‐Ward TF, Baum J, Kazura JW, Beeson JG, Cowman AF, Gilson PR, Crabb BS. The
Plasmodium falciparum
parasitophorous vacuole protein
P113
interacts with the parasite protein export machinery and maintains normal vacuole architecture. Mol Microbiol 2022; 117:1245-1262. [PMID: 35403274 PMCID: PMC9544671 DOI: 10.1111/mmi.14904] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Hayley E. Bullen
- Burnet Institute 85 Commercial Road Prahran Australia
- Department of Microbiology and Immunology University of Melbourne Parkville Australia
| | | | - Madeline G. Dans
- Burnet Institute 85 Commercial Road Prahran Australia
- School of Medicine Deakin University Waurn Ponds Victoria Australia
- Walter and Eliza Hall Institute 1G Royal Parade Parkville Australia
| | - Thorey K. Jonsdottir
- Burnet Institute 85 Commercial Road Prahran Australia
- Department of Microbiology and Immunology University of Melbourne Parkville Australia
| | - David T. Riglar
- Walter and Eliza Hall Institute 1G Royal Parade Parkville Australia
- Department of Medical Biology University of Melbourne Parkville Australia
- Imperial College London Department of Infectious Diseases, South Kensington Campus, SW72AZ UK
| | - Oliver Looker
- Burnet Institute 85 Commercial Road Prahran Australia
| | - Catherine S. Palmer
- Burnet Institute 85 Commercial Road Prahran Australia
- Bio21, 30 Road Parkville Flemington Australia
| | | | - Sarah C. Charnaud
- Burnet Institute 85 Commercial Road Prahran Australia
- WHO Geneva Switzerland
| | - Tony Triglia
- Walter and Eliza Hall Institute 1G Royal Parade Parkville Australia
- Department of Medical Biology University of Melbourne Parkville Australia
| | - Mikha Gabriela
- Burnet Institute 85 Commercial Road Prahran Australia
- School of Medicine Deakin University Waurn Ponds Victoria Australia
- Walter and Eliza Hall Institute 1G Royal Parade Parkville Australia
| | | | - Jo‐Anne Chan
- Burnet Institute 85 Commercial Road Prahran Australia
- Department of Medicine University of Melbourne Parkville Victoria Australia
| | | | - Jake Baum
- Walter and Eliza Hall Institute 1G Royal Parade Parkville Australia
- Department of Medical Biology University of Melbourne Parkville Australia
- Imperial College London Department of Infectious Diseases, South Kensington Campus, SW72AZ UK
| | - James W. Kazura
- Centre for Global Health and Diseases Case Western Reserve University Cleveland Ohio USA
| | - James G. Beeson
- Burnet Institute 85 Commercial Road Prahran Australia
- Department of Medicine University of Melbourne Parkville Victoria Australia
- Department of Immunology, Central clinical school Monash University Melbourne Victoria Australia
| | - Alan F. Cowman
- Walter and Eliza Hall Institute 1G Royal Parade Parkville Australia
- Department of Medical Biology University of Melbourne Parkville Australia
| | - Paul R. Gilson
- Burnet Institute 85 Commercial Road Prahran Australia
- Department of Microbiology and Immunology University of Melbourne Parkville Australia
| | - Brendan S. Crabb
- Burnet Institute 85 Commercial Road Prahran Australia
- Department of Microbiology and Immunology University of Melbourne Parkville Australia
| |
Collapse
|
50
|
Aroonsri A, Wongsombat C, Shaw P, Franke S, Przyborski J, Kaiser A. Investigation of an Allosteric Deoxyhypusine Synthase Inhibitor in P. falciparum. Molecules 2022; 27:molecules27082463. [PMID: 35458660 PMCID: PMC9030622 DOI: 10.3390/molecules27082463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 11/30/2022] Open
Abstract
The treatment of a variety of protozoal infections, in particular those causing disabling human diseases, is still hampered by a lack of drugs or increasing resistance to registered drugs. However, in recent years, remarkable progress has been achieved to combat neglected tropical diseases by sequencing the parasites’ genomes or the validation of new targets in the parasites by novel genetic manipulation techniques, leading to loss of function. The novel amino acid hypusine is a posttranslational modification (PTM) that occurs in eukaryotic initiation factor 5A (EIF5A) at a specific lysine residue. This modification occurs by two steps catalyzed by deoxyhypusine synthase (dhs) and deoxyhypusine hydroxylase (DOHH) enzymes. dhs from Plasmodium has been validated as a druggable target by small molecules and reverse genetics. Recently, the synthesis of a series of human dhs inhibitors led to 6-bromo-N-(1H-indol-4yl)-1-benzothiophene-2-carboxamide, a potent allosteric inhibitor with an IC50 value of 0.062 µM. We investigated this allosteric dhs inhibitor in Plasmodium. In vitro P. falciparum growth assays showed weak inhibition activity, with IC50 values of 46.1 µM for the Dd2 strain and 51.5 µM for the 3D7 strain, respectively. The antimalarial activity could not be attributed to the targeting of the Pfdhs gene, as shown by chemogenomic profiling with transgenically modified P. falciparum lines. Moreover, in dose-dependent enzymatic assays with purified recombinant P. falciparum dhs protein, only 45% inhibition was observed at an inhibitor dose of 0.4 µM. These data are in agreement with a homology-modeled Pfdhs, suggesting significant structural differences in the allosteric site between the human and parasite enzymes. Virtual screening of the allosteric database identified candidate ligand binding to novel binding pockets identified in P. falciparum dhs, which might foster the development of parasite-specific inhibitors.
Collapse
Affiliation(s)
- Aiyada Aroonsri
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Khlong Luang 12120, Thailand; (A.A.); (C.W.); (P.S.)
| | - Chayaphat Wongsombat
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Khlong Luang 12120, Thailand; (A.A.); (C.W.); (P.S.)
| | - Philip Shaw
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Khlong Luang 12120, Thailand; (A.A.); (C.W.); (P.S.)
| | - Siegrid Franke
- Interdisziplinäres Forschungszentrum IFZ, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany; (S.F.); (J.P.)
| | - Jude Przyborski
- Interdisziplinäres Forschungszentrum IFZ, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany; (S.F.); (J.P.)
| | - Annette Kaiser
- Medical Research Centre, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany
- Correspondence:
| |
Collapse
|