Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(13): 11401-11420, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38918002

ABSTRACT

Structure-activity relationship studies of 2,8-disubstituted-1,5-naphthyridines, previously reported as potent inhibitors of Plasmodium falciparum (Pf) phosphatidylinositol-4-kinase ß (PI4K), identified 1,5-naphthyridines with basic groups at 8-position, which retained Plasmodium PI4K inhibitory activity but switched primary mode of action to the host hemoglobin degradation pathway through inhibition of hemozoin formation. These compounds showed minimal off-target inhibitory activity against the human phosphoinositide kinases and MINK1 and MAP4K kinases, which were associated with the teratogenicity and testicular toxicity observed in rats for the PfPI4K inhibitor clinical candidate MMV390048. A representative compound from the series retained activity against field isolates and lab-raised drug-resistant strains of Pf. It was efficacious in the humanized NSG mouse malaria infection model at a single oral dose of 32 mg/kg. This compound was nonteratogenic in the zebrafish embryo model of teratogenicity and has a low predicted human dose, indicating that this series has the potential to deliver a preclinical candidate for malaria.


Subject(s)
1-Phosphatidylinositol 4-Kinase , Antimalarials , Hemeproteins , Naphthyridines , Plasmodium falciparum , Zebrafish , Plasmodium falciparum/drug effects , Animals , Naphthyridines/pharmacology , Naphthyridines/chemistry , Naphthyridines/chemical synthesis , Naphthyridines/therapeutic use , Antimalarials/pharmacology , Antimalarials/chemistry , Antimalarials/chemical synthesis , 1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , 1-Phosphatidylinositol 4-Kinase/metabolism , Humans , Structure-Activity Relationship , Hemeproteins/antagonists & inhibitors , Hemeproteins/metabolism , Mice , Rats , Malaria, Falciparum/drug therapy , Male , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis
2.
Nat Commun ; 15(1): 937, 2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38297033

ABSTRACT

Malaria poses an enormous threat to human health. With ever increasing resistance to currently deployed drugs, breakthrough compounds with novel mechanisms of action are urgently needed. Here, we explore pyrimidine-based sulfonamides as a new low molecular weight inhibitor class with drug-like physical parameters and a synthetically accessible scaffold. We show that the exemplar, OSM-S-106, has potent activity against parasite cultures, low mammalian cell toxicity and low propensity for resistance development. In vitro evolution of resistance using a slow ramp-up approach pointed to the Plasmodium falciparum cytoplasmic asparaginyl-tRNA synthetase (PfAsnRS) as the target, consistent with our finding that OSM-S-106 inhibits protein translation and activates the amino acid starvation response. Targeted mass spectrometry confirms that OSM-S-106 is a pro-inhibitor and that inhibition of PfAsnRS occurs via enzyme-mediated production of an Asn-OSM-S-106 adduct. Human AsnRS is much less susceptible to this reaction hijacking mechanism. X-ray crystallographic studies of human AsnRS in complex with inhibitor adducts and docking of pro-inhibitors into a model of Asn-tRNA-bound PfAsnRS provide insights into the structure-activity relationship and the selectivity mechanism.


Subject(s)
Antimalarials , Aspartate-tRNA Ligase , Animals , Humans , Plasmodium falciparum/genetics , Asparagine/metabolism , Aspartate-tRNA Ligase/genetics , RNA, Transfer, Amino Acyl/metabolism , Antimalarials/pharmacology , Mammals/genetics
3.
Cell Chem Biol ; 31(2): 312-325.e9, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-37995692

ABSTRACT

Our previous study identified 52 antiplasmodial peptaibols isolated from fungi. To understand their antiplasmodial mechanism of action, we conducted phenotypic assays, assessed the in vitro evolution of resistance, and performed a transcriptome analysis of the most potent peptaibol, HZ NPDG-I. HZ NPDG-I and 2 additional peptaibols were compared for their killing action and stage dependency, each showing a loss of digestive vacuole (DV) content via ultrastructural analysis. HZ NPDG-I demonstrated a stepwise increase in DV pH, impaired DV membrane permeability, and the ability to form ion channels upon reconstitution in planar membranes. This compound showed no signs of cross resistance to targets of current clinical candidates, and 3 independent lines evolved to resist HZ NPDG-I acquired nonsynonymous changes in the P. falciparum multidrug resistance transporter, pfmdr1. Conditional knockdown of PfMDR1 showed varying effects to other peptaibol analogs, suggesting differing sensitivity.


Subject(s)
Antimalarials , Malaria, Falciparum , Humans , Peptaibols/metabolism , Peptaibols/pharmacology , Antimalarials/pharmacology , Membrane Transport Proteins , Cell Membrane Permeability
4.
EMBO J ; 42(23): e113155, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37886905

ABSTRACT

Apicomplexan parasites discharge specialized organelles called rhoptries upon host cell contact to mediate invasion. The events that drive rhoptry discharge are poorly understood, yet essential to sustain the apicomplexan parasitic life cycle. Rhoptry discharge appears to depend on proteins secreted from another set of organelles called micronemes, which vary in function from allowing host cell binding to facilitation of gliding motility. Here we examine the function of the microneme protein CLAMP, which we previously found to be necessary for Toxoplasma gondii host cell invasion, and demonstrate its essential role in rhoptry discharge. CLAMP forms a distinct complex with two other microneme proteins, the invasion-associated SPATR, and a previously uncharacterized protein we name CLAMP-linked invasion protein (CLIP). CLAMP deficiency does not impact parasite adhesion or microneme protein secretion; however, knockdown of any member of the CLAMP complex affects rhoptry discharge. Phylogenetic analysis suggests orthologs of the essential complex components, CLAMP and CLIP, are ubiquitous across apicomplexans. SPATR appears to act as an accessory factor in Toxoplasma, but despite incomplete conservation is also essential for invasion during Plasmodium falciparum blood stages. Together, our results reveal a new protein complex that mediates rhoptry discharge following host-cell contact.


Subject(s)
Toxoplasma , Toxoplasma/metabolism , Microneme , Protozoan Proteins/metabolism , Phylogeny , Organelles/metabolism
5.
Res Sq ; 2023 Jul 27.
Article in English | MEDLINE | ID: mdl-37546892

ABSTRACT

Malaria poses an enormous threat to human health. With ever increasing resistance to currently deployed drugs, breakthrough compounds with novel mechanisms of action are urgently needed. Here, we explore pyrimidine-based sulfonamides as a new low molecular weight inhibitor class with drug-like physical parameters and a synthetically accessible scaffold. We show that the exemplar, OSM-S-106, has potent activity against parasite cultures, low mammalian cell toxicity and low propensity for resistance development. In vitro evolution of resistance using a slow ramp-up approach pointed to the Plasmodium falciparum cytoplasmic asparaginyl tRNA synthetase (PfAsnRS) as the target, consistent with our finding that OSM-S-106 inhibits protein translation and activates the amino acid starvation response. Targeted mass spectrometry confirms that OSM-S-106 is a pro-inhibitor and that inhibition of PfAsnRS occurs via enzyme-mediated production of an Asn-OSM-S-106 adduct. Human AsnRS is much less susceptible to this reaction hijacking mechanism. X-ray crystallographic studies of human AsnRS in complex with inhibitor adducts and docking of pro-inhibitors into a model of Asn-tRNA-bound PfAsnRS provide insights into the structure activity relationship and the selectivity mechanism.

6.
Br J Pharmacol ; 180(15): 1899-1929, 2023 08.
Article in English | MEDLINE | ID: mdl-37197802

ABSTRACT

Antimalarial drug discovery has until recently been driven by high-throughput phenotypic cellular screening, allowing millions of compounds to be assayed and delivering clinical drug candidates. In this review, we will focus on target-based approaches, describing recent advances in our understanding of druggable targets in the malaria parasite. Targeting multiple stages of the Plasmodium lifecycle, rather than just the clinically symptomatic asexual blood stage, has become a requirement for new antimalarial medicines, and we link pharmacological data clearly to the parasite stages to which it applies. Finally, we highlight the IUPHAR/MMV Guide to MALARIA PHARMACOLOGY, a web resource developed for the malaria research community that provides open and optimized access to published data on malaria pharmacology.


Subject(s)
Antimalarials , Malaria , Humans , Malaria/drug therapy , Antimalarials/pharmacology , Antimalarials/therapeutic use , Drug Discovery , High-Throughput Screening Assays
7.
Cell Chem Biol ; 30(5): 470-485.e6, 2023 05 18.
Article in English | MEDLINE | ID: mdl-36963402

ABSTRACT

The Plasmodium falciparum proteasome constitutes a promising antimalarial target, with multiple chemotypes potently and selectively inhibiting parasite proliferation and synergizing with the first-line artemisinin drugs, including against artemisinin-resistant parasites. We compared resistance profiles of vinyl sulfone, epoxyketone, macrocyclic peptide, and asparagine ethylenediamine inhibitors and report that the vinyl sulfones were potent even against mutant parasites resistant to other proteasome inhibitors and did not readily select for resistance, particularly WLL that displays covalent and irreversible binding to the catalytic ß2 and ß5 proteasome subunits. We also observed instances of collateral hypersensitivity, whereby resistance to one inhibitor could sensitize parasites to distinct chemotypes. Proteasome selectivity was confirmed using CRISPR/Cas9-edited mutant and conditional knockdown parasites. Molecular modeling of proteasome mutations suggested spatial contraction of the ß5 P1 binding pocket, compromising compound binding. Dual targeting of P. falciparum proteasome subunits using covalent inhibitors provides a potential strategy for restoring artemisinin activity and combating the spread of drug-resistant malaria.


Subject(s)
Antimalarials , Artemisinins , Malaria, Falciparum , Plasmodium , Humans , Antimalarials/pharmacology , Antimalarials/chemistry , Proteasome Endopeptidase Complex/metabolism , Plasmodium/metabolism , Artemisinins/chemistry , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/chemistry
8.
Nat Commun ; 14(1): 1455, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36927839

ABSTRACT

Identifying how small molecules act to kill malaria parasites can lead to new "chemically validated" targets. By pressuring Plasmodium falciparum asexual blood stage parasites with three novel structurally-unrelated antimalarial compounds (MMV665924, MMV019719 and MMV897615), and performing whole-genome sequence analysis on resistant parasite lines, we identify multiple mutations in the P. falciparum acyl-CoA synthetase (ACS) genes PfACS10 (PF3D7_0525100, M300I, A268D/V, F427L) and PfACS11 (PF3D7_1238800, F387V, D648Y, and E668K). Allelic replacement and thermal proteome profiling validates PfACS10 as a target of these compounds. We demonstrate that this protein is essential for parasite growth by conditional knockdown and observe increased compound susceptibility upon reduced expression. Inhibition of PfACS10 leads to a reduction in triacylglycerols and a buildup of its lipid precursors, providing key insights into its function. Analysis of the PfACS11 gene and its mutations point to a role in mediating resistance via decreased protein stability.


Subject(s)
Antimalarials , Malaria, Falciparum , Humans , Plasmodium falciparum/metabolism , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Antimalarials/pharmacology , Antimalarials/therapeutic use , Mutation , Ligases/metabolism
9.
Sci Transl Med ; 14(667): eabo7219, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36260689

ABSTRACT

Compounds acting on multiple targets are critical to combating antimalarial drug resistance. Here, we report that the human "mammalian target of rapamycin" (mTOR) inhibitor sapanisertib has potent prophylactic liver stage activity, in vitro and in vivo asexual blood stage (ABS) activity, and transmission-blocking activity against the protozoan parasite Plasmodium spp. Chemoproteomics studies revealed multiple potential Plasmodium kinase targets, and potent inhibition of Plasmodium phosphatidylinositol 4-kinase type III beta (PI4Kß) and cyclic guanosine monophosphate-dependent protein kinase (PKG) was confirmed in vitro. Conditional knockdown of PI4Kß in ABS cultures modulated parasite sensitivity to sapanisertib, and laboratory-generated P. falciparum sapanisertib resistance was mediated by mutations in PI4Kß. Parasite metabolomic perturbation profiles associated with sapanisertib and other known PI4Kß and/or PKG inhibitors revealed similarities and differences between chemotypes, potentially caused by sapanisertib targeting multiple parasite kinases. The multistage activity of sapanisertib and its in vivo antimalarial efficacy, coupled with potent inhibition of at least two promising drug targets, provides an opportunity to reposition this pyrazolopyrimidine for malaria.


Subject(s)
Antimalarials , Plasmodium , Animals , Humans , Antimalarials/pharmacology , Antimalarials/therapeutic use , Plasmodium falciparum , MTOR Inhibitors , 1-Phosphatidylinositol 4-Kinase , Guanosine Monophosphate , Life Cycle Stages , TOR Serine-Threonine Kinases , Sirolimus , Mammals
10.
PLoS Pathog ; 18(10): e1010887, 2022 10.
Article in English | MEDLINE | ID: mdl-36223427

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.


Subject(s)
Antimalarials , DNA-Binding Proteins , Plasmodium , Humans , Antimalarials/pharmacology , Antimalarials/metabolism , DNA/metabolism , Plasmodium/drug effects , Plasmodium/genetics , Protozoan Proteins/metabolism , DNA-Binding Proteins/metabolism
11.
Nat Commun ; 13(1): 4067, 2022 07 13.
Article in English | MEDLINE | ID: mdl-35831417

ABSTRACT

Plasmodium falciparum has developed extensive mechanisms to evade host immune clearance. Currently, most of our understanding is based on in vitro studies of individual parasite variant surface antigens and how this relates to the processes in vivo is not well-understood. Here, we have used a humanized mouse model to identify parasite factors important for in vivo growth. We show that upregulation of the specific PfEMP1, VAR2CSA, provides the parasite with protection from macrophage phagocytosis and clearance in the humanized mice. Furthermore, parasites adapted to thrive in the humanized mice show reduced NK cell-mediated killing through interaction with the immune inhibitory receptor, LILRB1. Taken together, these findings reveal new insights into the molecular and cellular mechanisms that the parasite utilizes to coordinate immune escape in vivo. Identification and targeting of these specific parasite variant surface antigens crucial for immune evasion provides a unique approach for therapy.


Subject(s)
Malaria, Falciparum , Plasmodium falciparum , Animals , Antigens, Protozoan , Antigens, Surface/metabolism , Erythrocytes/parasitology , Malaria, Falciparum/parasitology , Mice , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism
12.
Trends Parasitol ; 38(9): 722-723, 2022 09.
Article in English | MEDLINE | ID: mdl-35871979

ABSTRACT

The recent study by Campelo Morillo et al. has shown that one of the small number of non-ApiAP2 DNA-binding proteins in the Plasmodium falciparum genome acts as a transcription factor in the gametocytogenesis cascade and is responsible for the gametocyte's distinctive morphology.


Subject(s)
Plasmodium falciparum , Transcription Factors , Gene Expression Regulation , Plasmodium falciparum/genetics , Transcription Factors/genetics
13.
Science ; 376(6597): 1074-1079, 2022 06 03.
Article in English | MEDLINE | ID: mdl-35653481

ABSTRACT

Aminoacyl transfer RNA (tRNA) synthetases (aaRSs) are attractive drug targets, and we present class I and II aaRSs as previously unrecognized targets for adenosine 5'-monophosphate-mimicking nucleoside sulfamates. The target enzyme catalyzes the formation of an inhibitory amino acid-sulfamate conjugate through a reaction-hijacking mechanism. We identified adenosine 5'-sulfamate as a broad-specificity compound that hijacks a range of aaRSs and ML901 as a specific reagent a specific reagent that hijacks a single aaRS in the malaria parasite Plasmodium falciparum, namely tyrosine RS (PfYRS). ML901 exerts whole-life-cycle-killing activity with low nanomolar potency and single-dose efficacy in a mouse model of malaria. X-ray crystallographic studies of plasmodium and human YRSs reveal differential flexibility of a loop over the catalytic site that underpins differential susceptibility to reaction hijacking by ML901.


Subject(s)
Antimalarials , Malaria, Falciparum , Molecular Targeted Therapy , Plasmodium falciparum , Protein Biosynthesis , Protozoan Proteins , Tyrosine-tRNA Ligase , Adenosine/analogs & derivatives , Animals , Antimalarials/chemistry , Antimalarials/pharmacology , Antimalarials/therapeutic use , Crystallography, X-Ray , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Mice , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Protein Biosynthesis/drug effects , Protein Conformation , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Sulfonic Acids/chemistry , Tyrosine-tRNA Ligase/chemistry , Tyrosine-tRNA Ligase/metabolism
14.
J Anim Sci ; 100(6)2022 Jun 01.
Article in English | MEDLINE | ID: mdl-35579995

ABSTRACT

In pigs, the gut microbiota composition plays a major role in the process of digestion, but is influenced by many external factors, especially diet. To be used in breeding applications, genotype by diet interactions on microbiota composition have to be quantified, as well as their impact on genetic covariances with feed efficiency (FE) and digestive efficiency (DE) traits. This study aimed at determining the impact of an alternative diet on variance components of microbiota traits (genera and alpha diversity indices) and estimating genetic correlations between microbiota and efficiency traits for pigs fed a conventional (CO) or a high-fiber (HF) diet. Fecal microbes of 812 full-siblings fed a CO diet and 752 pigs fed the HF diet were characterized at 16 weeks of age by sequencing the V3-V4 region of the 16S rRNA gene. A total of 231 genera were identified. Digestibility coefficients of nitrogen, organic matter, and energy were predicted analyzing the same fecal samples with near infrared spectrometry. Daily feed intake, feed conversion ratio, residual feed intake and average daily gain (ADG) were also recorded. The 71 genera present in more than 20% of individuals were retained for genetic analyses. Heritability (h²) of microbiota traits were similar between diets (from null to 0.38 ±â€…0.12 in the CO diet and to 0.39 ±â€…0.12 in the HF diet). Only three out of the 24 genera and two alpha diversity indices with significant h² in both diets had genetic correlations across diets significantly different from 0.99 (P < 0.05), indicating limited genetic by diet interactions for these traits. When both diets were analyzed jointly, 59 genera had h² significantly different from zero. Based on the genetic correlations between these genera and ADG, FE, and DE traits, three groups of genera could be identified. A group of 29 genera had abundances favorably correlated with DE and FE traits, 14 genera were unfavorably correlated with DE traits, and the last group of 16 genera had abundances with correlations close to zero with production traits. However, genera abundances favorably correlated with DE and FE traits were unfavorably correlated with ADG, and vice versa. Alpha diversity indices had correlation patterns similar to the first group. In the end, genetic by diet interactions on gut microbiota composition of growing pigs were limited in this study. Based on this study, microbiota-based traits could be used as proxies to improve FE and DE in growing pigs.


The link between the composition of the gut microbiota, i.e the composition of microorganisms in the gut, in pigs and their feed efficiency, i.e. their ability to utilize nutrients, as well as their ability to digest were studied from a genetic point of view. A family structure of 1,564 pigs were studied and fed with two different diets. One of the full-sib was fed a conventional diet used in breeding farms and the other one an alternative diet containing raw materials, less expensive but with a higher content of dietary fibers more difficult to digest. This study has shown that some microbiota microorganisms were genetically correlated with feed and digestive efficiency performances, positively or negatively, depending on the microorganisms. In addition, the diversity of microorganisms in the animal's gut was favorably correlated with the feed and digestive performances studied. Therefore, there is a genetic link between these performances and the composition of the animal's gut microbiota. Thus, a potential genetic selection on some intestinal microorganisms or diversity of microorganisms would allow to improve these performances, and in particular when pigs are fed with diet more difficult to digest.


Subject(s)
Gastrointestinal Microbiome , Animal Feed/analysis , Animals , Diet/veterinary , Feces , RNA, Ribosomal, 16S/genetics , Swine/genetics
15.
Nat Commun ; 13(1): 2158, 2022 04 20.
Article in English | MEDLINE | ID: mdl-35444200

ABSTRACT

Drug resistance and a dire lack of transmission-blocking antimalarials hamper malaria elimination. Here, we present the pantothenamide MMV693183 as a first-in-class acetyl-CoA synthetase (AcAS) inhibitor to enter preclinical development. Our studies demonstrate attractive drug-like properties and in vivo efficacy in a humanized mouse model of Plasmodium falciparum infection. The compound shows single digit nanomolar in vitro activity against P. falciparum and P. vivax clinical isolates, and potently blocks P. falciparum transmission to Anopheles mosquitoes. Genetic and biochemical studies identify AcAS as the target of the MMV693183-derived antimetabolite, CoA-MMV693183. Pharmacokinetic-pharmacodynamic modelling predict that a single 30 mg oral dose is sufficient to cure a malaria infection in humans. Toxicology studies in rats indicate a > 30-fold safety margin in relation to the predicted human efficacious exposure. In conclusion, MMV693183 represents a promising candidate for further (pre)clinical development with a novel mode of action for treatment of malaria and blocking transmission.


Subject(s)
Antimalarials , Folic Acid Antagonists , Malaria, Falciparum , Malaria, Vivax , Malaria , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use , Malaria/drug therapy , Malaria, Falciparum/drug therapy , Malaria, Vivax/drug therapy , Mice , Pantothenic Acid/analogs & derivatives , Plasmodium falciparum/genetics , Rats
16.
Nat Commun ; 13(1): 1275, 2022 03 11.
Article in English | MEDLINE | ID: mdl-35277503

ABSTRACT

The RAP (RNA-binding domain abundant in Apicomplexans) protein family has been identified in various organisms. Despite expansion of this protein family in apicomplexan parasites, their main biological functions remain unknown. In this study, we use inducible knockdown studies in the human malaria parasite, Plasmodium falciparum, to show that two RAP proteins, PF3D7_0105200 (PfRAP01) and PF3D7_1470600 (PfRAP21), are essential for parasite survival and localize to the mitochondrion. Using transcriptomics, metabolomics, and proteomics profiling experiments, we further demonstrate that these RAP proteins are involved in mitochondrial RNA metabolism. Using high-throughput sequencing of RNA isolated by crosslinking immunoprecipitation (eCLIP-seq), we validate that PfRAP01 and PfRAP21 are true RNA-binding proteins and interact specifically with mitochondrial rRNAs. Finally, mitochondrial enrichment experiments followed by deep sequencing of small RNAs demonstrate that PfRAP21 controls mitochondrial rRNA expression. Collectively, our results establish the role of these RAP proteins in mitoribosome activity and contribute to further understanding this protein family in malaria parasites.


Subject(s)
Malaria, Falciparum , Mitochondrial Ribosomes , Plasmodium falciparum , Protozoan Proteins , RNA-Binding Proteins , Genomics , Humans , Malaria, Falciparum/parasitology , Mitochondrial Ribosomes/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
17.
CRISPR J ; 5(1): 155-164, 2022 02.
Article in English | MEDLINE | ID: mdl-35191751

ABSTRACT

Functional characterization of the multitude of poorly described proteins in the human malarial pathogen, Plasmodium falciparum, requires tools to enable genome-scale perturbation studies. Here, we present GeneTargeter (genetargeter.mit.edu), a software tool for automating the design of homology-directed repair donor vectors to achieve gene knockouts, conditional knockdowns, and epitope tagging of P. falciparum genes. We demonstrate GeneTargeter-facilitated genome-scale design of six different types of knockout and conditional knockdown constructs for the P. falciparum genome and validate the computational design process experimentally with successful donor vector assembly and transfection. The software's modular nature accommodates arbitrary destination vectors and allows customizable designs that extend the genome manipulation outcomes attainable in Plasmodium and other organisms.


Subject(s)
Malaria, Falciparum , Parasites , Animals , CRISPR-Cas Systems/genetics , Gene Editing , Humans , Malaria, Falciparum/genetics , Malaria, Falciparum/parasitology , Parasites/genetics , Plasmodium falciparum/genetics
18.
Animal ; 16(2): 100447, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35074648

ABSTRACT

Digestive efficiency traits are promising selection criteria to improve feed efficiency in pigs. However, the genetic relationships between digestive efficiency and sow reproductive traits are mostly unknown and need to be estimated. In this study, reproductive traits were available for 61 601 litters recorded on 21 719 Large White purebred sows. The traits were comprised of the number of born alive (NBA) and the number of weaned piglets (NWP), the number of stillbirths (NSB) and piglet mortality during suckling (PM). For a subset of 32518 litters, the mean (MBW) and CV of piglet birth weights (CVBW) were deduced from individual piglet weights as well as the proportion of piglets weighing less than 1 kg (PPL1K). Growth and feed efficiency traits were available for 4 643 Large White male pigs related to sows with reproductive performances. They comprised average daily gain (ADG), daily feed intake (DFI) and feed conversion ratio (FCR). A subset of 1 391 pigs had predictions for digestibility coefficients (DC) of energy, organic matter and nitrogen obtained by analysing faecal samples with near-infrared spectrometry. Estimated heritabilities were low for NBA, NSB, NWP and PM (0.08 ± 0.01 to 0.11 ± 0.01) and low to moderate for litter weight characteristics (0.14 ± 0.02 to 0.38 ± 0.01). Heritability estimates were moderate to high for ADG, DFI and FCR (0.37 ± 0.04 to 0.54 ± 0.05) and moderate for DC traits (0.26 ± 0.06 to 0.38 ± 0.07). Genetic correlations were low between ADG, or alternatively FCR, and reproductive traits. They were significantly different from zero with MBW (0.19 ± 0.06 with ADG and -0.15 ± 0.06 with FCR) and PPL1K (-0.19 ± 0.07 with ADG and 0.18 ± 0.07 with FCR). All genetic correlations between DFI and reproductive traits were low and not significantly different from zero. Genetic correlations between DC traits and NBA were significantly different from zero for DC of organic matter and energy (<-0.25 ± 0.11). DC traits were moderately correlated with MBW (>0.30 ± 0.11), CVBW (<-0.36 ± 0.11) and PPL1K (<-0.37 ± 0.11) at the genetic level. Genetic correlations between DC traits and PM were significantly negative and hence favourable (<-0.38 ± 0.12). Finally, genetic correlations between DC traits and NWP were close to zero. These results suggested that sows closely related to growing pigs with the best digestive efficiency would produce heavier and more homogeneous piglets, with slightly smaller litter sizes at birth but better survival. Hence, there is usable genetic variation in DC that could be exploited to define new selection strategies in maternal lines aiming at improving not only feed efficiency but also piglet survival.


Subject(s)
Lactation , Reproduction , Animals , Eating , Female , Lactation/genetics , Litter Size/genetics , Male , Pregnancy , Reproduction/genetics , Swine/genetics , Weaning
19.
Cell Chem Biol ; 29(2): 191-201.e8, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34348113

ABSTRACT

We identify the Plasmodium falciparum acetyl-coenzyme A synthetase (PfAcAS) as a druggable target, using genetic and chemical validation. In vitro evolution of resistance with two antiplasmodial drug-like compounds (MMV019721 and MMV084978) selects for mutations in PfAcAS. Metabolic profiling of compound-treated parasites reveals changes in acetyl-CoA levels for both compounds. Genome editing confirms that mutations in PfAcAS are sufficient to confer resistance. Knockdown studies demonstrate that PfAcAS is essential for asexual growth, and partial knockdown induces hypersensitivity to both compounds. In vitro biochemical assays using recombinantly expressed PfAcAS validates that MMV019721 and MMV084978 directly inhibit the enzyme by preventing CoA and acetate binding, respectively. Immunolocalization studies reveal that PfAcAS is primarily localized to the nucleus. Functional studies demonstrate inhibition of histone acetylation in compound-treated wild-type, but not in resistant parasites. Our findings identify and validate PfAcAS as an essential, druggable target involved in the epigenetic regulation of gene expression.


Subject(s)
Acetate-CoA Ligase/antagonists & inhibitors , Antimalarials/pharmacology , Enzyme Inhibitors/pharmacology , Malaria/drug therapy , Plasmodium falciparum/drug effects , Acetate-CoA Ligase/metabolism , Antimalarials/chemistry , Enzyme Inhibitors/chemistry , Humans , Malaria/metabolism , Models, Molecular , Molecular Structure , Parasitic Sensitivity Tests , Plasmodium falciparum/enzymology
20.
Cell Chem Biol ; 29(5): 824-839.e6, 2022 05 19.
Article in English | MEDLINE | ID: mdl-34233174

ABSTRACT

Widespread Plasmodium falciparum resistance to first-line antimalarials underscores the vital need to develop compounds with novel modes of action and identify new druggable targets. Here, we profile five compounds that potently inhibit P. falciparum asexual blood stages. Resistance selection studies with three carboxamide-containing compounds, confirmed by gene editing and conditional knockdowns, identify point mutations in the parasite transporter ABCI3 as the primary mediator of resistance. Selection studies with imidazopyridine or quinoline-carboxamide compounds also yield changes in ABCI3, this time through gene amplification. Imidazopyridine mode of action is attributed to inhibition of heme detoxification, as evidenced by cellular accumulation and heme fractionation assays. For the copy-number variation-selecting imidazopyridine and quinoline-carboxamide compounds, we find that resistance, manifesting as a biphasic concentration-response curve, can independently be mediated by mutations in the chloroquine resistance transporter PfCRT. These studies reveal the interconnectedness of P. falciparum transporters in overcoming drug pressure in different parasite strains.


Subject(s)
Antimalarials , Folic Acid Antagonists , Malaria, Falciparum , Parasites , Quinolines , ATP-Binding Cassette Transporters/genetics , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use , Heme , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Membrane Transport Proteins/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Quinolines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...