Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
mSphere ; : e0010924, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38578105

ABSTRACT

The two species that account for most cases of Acinetobacter-associated bacteremia in the United Kingdom are Acinetobacter lwoffii, often a commensal but also an emerging pathogen, and Acinetobacter baumannii, a well-known antibiotic-resistant species. While these species both cause similar types of human infection and occupy the same niche, A. lwoffii (unlike A. baumannii) has thus far remained susceptible to antibiotics. Comparatively little is known about the biology of A. lwoffii, and this is the largest study on it conducted to date, providing valuable insights into its behaviour and potential threat to human health. This study aimed to explain the antibiotic susceptibility, virulence, and fundamental biological differences between these two species. The relative susceptibility of A. lwoffii was explained as it encoded fewer antibiotic resistance and efflux pump genes than A. baumannii (9 and 30, respectively). While both species had markers of horizontal gene transfer, A. lwoffii encoded more DNA defense systems and harbored a far more restricted range of plasmids. Furthermore, A. lwoffii displayed a reduced ability to select for antibiotic resistance mutations, form biofilm, and infect both in vivo and in in vitro models of infection. This study suggests that the emerging pathogen A. lwoffii has remained susceptible to antibiotics because mechanisms exist to make it highly selective about the DNA it acquires, and we hypothesize that the fact that it only harbors a single RND system restricts the ability to select for resistance mutations. This provides valuable insights into how development of resistance can be constrained in Gram-negative bacteria. IMPORTANCE: Acinetobacter lwoffii is often a harmless commensal but is also an emerging pathogen and is the most common cause of Acinetobacter-derived bloodstream infections in England and Wales. In contrast to the well-studied and often highly drug-resistant A. baumannii, A. lwoffii has remained susceptible to antibiotics. This study explains why this organism has not evolved resistance to antibiotics. These new insights are important to understand why and how some species develop antibiotic resistance, while others do not, and could inform future novel treatment strategies.

2.
mSphere ; 8(6): e0051123, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-37975677

ABSTRACT

IMPORTANCE: Toxoplasma gondii (Tg) is a ubiquitous parasitic pathogen, infecting about one-third of the global population. Tg is controlled in immunocompetent people by mechanisms that are not fully understood. Tg infection drives the production of the inflammatory cytokine interferon gamma (IFNγ), which upregulates intracellular anti-pathogen defense pathways. In this study, we describe host proteins p97/VCP, UBXD1, and ANKRD13A that control Tg at the parasitophorous vacuole (PV) in IFNγ-stimulated endothelial cells. p97/VCP is an ATPase that interacts with a network of cofactors and is active in a wide range of ubiquitin-dependent cellular processes. We demonstrate that PV ubiquitination is a pre-requisite for recruitment of these host defense proteins, and their deposition directs Tg PVs to acidification in endothelial cells. We show that p97/VCP universally targets PVs in human cells and restricts Tg in different human cell types. Overall, these findings reveal new players of intracellular host defense of a vacuolated pathogen.


Subject(s)
Parasites , Toxoplasma , Animals , Humans , Toxoplasma/metabolism , Interferons/metabolism , Vacuoles/metabolism , Endothelial Cells , Interferon-gamma , Valosin Containing Protein/metabolism
3.
Science ; 382(6666): eadg2253, 2023 10 06.
Article in English | MEDLINE | ID: mdl-37797010

ABSTRACT

Disruption of cellular activities by pathogen virulence factors can trigger innate immune responses. Interferon-γ (IFN-γ)-inducible antimicrobial factors, such as the guanylate binding proteins (GBPs), promote cell-intrinsic defense by attacking intracellular pathogens and by inducing programmed cell death. Working in human macrophages, we discovered that GBP1 expression in the absence of IFN-γ killed the cells and induced Golgi fragmentation. IFN-γ exposure improved macrophage survival through the activity of the kinase PIM1. PIM1 phosphorylated GBP1, leading to its sequestration by 14-3-3σ, which thereby prevented GBP1 membrane association. During Toxoplasma gondii infection, the virulence protein TgIST interfered with IFN-γ signaling and depleted PIM1, thereby increasing GBP1 activity. Although infected cells can restrain pathogens in a GBP1-dependent manner, this mechanism can protect uninfected bystander cells. Thus, PIM1 can provide a bait for pathogen virulence factors, guarding the integrity of IFN-γ signaling.


Subject(s)
GTP-Binding Proteins , Host-Pathogen Interactions , Immunity, Innate , Interferon-gamma , Proto-Oncogene Proteins c-pim-1 , Toxoplasma , Toxoplasmosis , Humans , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Interferon-gamma/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Toxoplasmosis/immunology , Virulence Factors/metabolism , Macrophages/immunology , 14-3-3 Proteins/metabolism , Host-Pathogen Interactions/immunology
4.
Curr Opin Immunol ; 84: 102373, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37536111

ABSTRACT

Cell-intrinsic defense is an essential part of the immune response against intracellular pathogens regulated by cytokine-induced proteins and pathways. One of the most upregulated families of proteins in this defense system are the guanylate-binding proteins (GBPs), large GTPases of the dynamin family, induced in response to interferon gamma. Human GBPs (hGBPs) exert their antimicrobial activity through detection of pathogen-associated molecular patterns and/or damage-associated molecular patterns to execute control mechanisms directed at the pathogen itself as well as the vacuolar compartments in which it resides. Consequently, hGBPs are also inducers of canonical and noncanonical inflammasome responses leading to host cell death. The mechanisms are both cell-type and pathogen-dependent with hGBP1 acting as a pioneer sensor for intracellular invaders. This review focuses on the most recent functional roles of hGBPs in pathways of pathogen detection, destruction, and host cell death induction.

5.
Pathog Dis ; 79(9)2022 01 07.
Article in English | MEDLINE | ID: mdl-34931666

ABSTRACT

Human guanylate binding proteins (GBPs) are key players of interferon-gamma (IFNγ)-induced cell intrinsic defense mechanisms targeting intracellular pathogens. In this study, we combine the well-established Toxoplasmagondii infection model with three in vitro macrophage culture systems to delineate the contribution of individual GBP family members to control this apicomplexan parasite. Use of high-throughput imaging assays and genome engineering allowed us to define a role for GBP1, 2 and 5 in parasite infection control. While GBP1 performs a pathogen-proximal, parasiticidal and growth-restricting function through accumulation at the parasitophorous vacuole of intracellular Toxoplasma, GBP2 and GBP5 perform a pathogen-distal, growth-restricting role. We further find that mutants of the GTPase or isoprenylation site of GBP1/2/5 affect their normal function in Toxoplasma control by leading to mis-localization of the proteins.


Subject(s)
GTP-Binding Proteins/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/parasitology , Toxoplasma/immunology , Toxoplasmosis/immunology , Toxoplasmosis/metabolism , Toxoplasmosis/parasitology , Biomarkers , Disease Susceptibility , Host-Parasite Interactions , Humans
6.
Cell Microbiol ; 23(7): e13349, 2021 07.
Article in English | MEDLINE | ID: mdl-33930228

ABSTRACT

To study the dynamics of infection processes, it is common to manually enumerate imaging-based infection assays. However, manual counting of events from imaging data is biased, error-prone and a laborious task. We recently presented HRMAn (Host Response to Microbe Analysis), an automated image analysis program using state-of-the-art machine learning and artificial intelligence algorithms to analyse pathogen growth and host defence behaviour. With HRMAn, we can quantify intracellular infection by pathogens such as Toxoplasma gondii and Salmonella in a variety of cell types in an unbiased and highly reproducible manner, measuring multiple parameters including pathogen growth, pathogen killing and activation of host cell defences. Since HRMAn is based on the KNIME Analytics platform, it can easily be adapted to work with other pathogens and produce more readouts from quantitative imaging data. Here we showcase improvements to HRMAn resulting in the release of HRMAn 2.0 and new applications of HRMAn 2.0 for the analysis of host-pathogen interactions using the established pathogen T. gondii and further extend it for use with the bacterial pathogen Chlamydia trachomatis and the fungal pathogen Cryptococcus neoformans.


Subject(s)
Chlamydia Infections/diagnostic imaging , Cryptococcosis/diagnostic imaging , Host-Pathogen Interactions , Image Processing, Computer-Assisted/methods , Toxoplasmosis/diagnostic imaging , Artificial Intelligence , Cell Line, Tumor , Humans
7.
mSphere ; 5(5)2020 09 09.
Article in English | MEDLINE | ID: mdl-32907956

ABSTRACT

The use of deep neural networks (DNNs) for analysis of complex biomedical images shows great promise but is hampered by a lack of large verified data sets for rapid network evolution. Here, we present a novel strategy, termed "mimicry embedding," for rapid application of neural network architecture-based analysis of pathogen imaging data sets. Embedding of a novel host-pathogen data set, such that it mimics a verified data set, enables efficient deep learning using high expressive capacity architectures and seamless architecture switching. We applied this strategy across various microbiological phenotypes, from superresolved viruses to in vitro and in vivo parasitic infections. We demonstrate that mimicry embedding enables efficient and accurate analysis of two- and three-dimensional microscopy data sets. The results suggest that transfer learning from pretrained network data may be a powerful general strategy for analysis of heterogeneous pathogen fluorescence imaging data sets.IMPORTANCE In biology, the use of deep neural networks (DNNs) for analysis of pathogen infection is hampered by a lack of large verified data sets needed for rapid network evolution. Artificial neural networks detect handwritten digits with high precision thanks to large data sets, such as MNIST, that allow nearly unlimited training. Here, we developed a novel strategy we call mimicry embedding, which allows artificial intelligence (AI)-based analysis of variable pathogen-host data sets. We show that deep learning can be used to detect and classify single pathogens based on small differences.


Subject(s)
Deep Learning , Host-Pathogen Interactions , Image Processing, Computer-Assisted/methods , Neural Networks, Computer , Animals , Artificial Intelligence , Microscopy/methods , Toxoplasma/pathogenicity , Vaccinia virus/pathogenicity , Zebrafish
8.
Cell Rep ; 32(6): 108008, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32783936

ABSTRACT

Interferon-inducible guanylate-binding proteins (GBPs) promote cell-intrinsic defense through host cell death. GBPs target pathogens and pathogen-containing vacuoles and promote membrane disruption for release of microbial molecules that activate inflammasomes. GBP1 mediates pyroptosis or atypical apoptosis of Salmonella Typhimurium (STm)- or Toxoplasma gondii (Tg)- infected human macrophages, respectively. The pathogen-proximal detection-mechanisms of GBP1 remain poorly understood, as humans lack functional immunity-related GTPases (IRGs) that assist murine Gbps. Here, we establish that GBP1 promotes the lysis of Tg-containing vacuoles and parasite plasma membranes, releasing Tg-DNA. In contrast, we show GBP1 targets cytosolic STm and recruits caspase-4 to the bacterial surface for its activation by lipopolysaccharide (LPS), but does not contribute to bacterial vacuole escape. Caspase-1 cleaves and inactivates GBP1, and a cleavage-deficient GBP1D192E mutant increases caspase-4-driven pyroptosis due to the absence of feedback inhibition. Our studies elucidate microbe-specific roles of GBP1 in infection detection and its triggering of the assembly of divergent caspase signaling platforms.


Subject(s)
Caspases/immunology , GTP-Binding Proteins/immunology , Salmonella typhimurium/immunology , Toxoplasma/immunology , Cell Death/immunology , HEK293 Cells , Humans , Inflammasomes/immunology , Interferon-gamma/pharmacology , Ligands , Salmonella Infections/immunology , Salmonella Infections/microbiology , THP-1 Cells , Toxoplasma/genetics , Toxoplasmosis/immunology , Toxoplasmosis/microbiology , Vacuoles/immunology
9.
Methods Mol Biol ; 2071: 411-433, 2020.
Article in English | MEDLINE | ID: mdl-31758464

ABSTRACT

Research on Toxoplasma gondii and its interplay with the host is often performed using fluorescence microscopy-based imaging experiments combined with manual quantification of acquired images. We present here an accurate and unbiased quantification method for host-pathogen interactions. We describe how to plan experiments and prepare, stain and image infected specimens and analyze them with the program HRMAn (Host Response to Microbe Analysis). HRMAn is a high-content image analysis method based on KNIME Analytics Platform. Users of this guide will be able to perform infection studies in high-throughput volume and to a greater level of detail. Relying on cutting edge machine learning algorithms, HRMAn can be trained and tailored to many experimental settings and questions.


Subject(s)
Toxoplasma/pathogenicity , Algorithms , Artificial Intelligence , Host-Pathogen Interactions , Machine Learning , Microscopy, Fluorescence/methods
10.
PLoS Pathog ; 15(12): e1008097, 2019 12.
Article in English | MEDLINE | ID: mdl-31830133
11.
Wellcome Open Res ; 4: 124, 2019.
Article in English | MEDLINE | ID: mdl-31544161

ABSTRACT

Background: Infections cause the production of inflammatory cytokines such as Interferon gamma (IFNγ). IFNγ in turn prompts the upregulation of a range of host defence proteins including members of the family of guanylate binding proteins (Gbps). In humans and mice alike, GBPs restrict the intracellular replication of invasive microbes and promote inflammation. To study the physiological functions of Gbp family members, the most commonly chosen in vivo models are mice harbouring loss-of-function mutations in either individual Gbp genes or the entire Gbp gene cluster on mouse chromosome 3. Individual Gbp deletion strains differ in their design, as some strains exist on a pure C57BL/6 genetic background, while other strains contain a 129-derived genetic interval encompassing the Gbp gene cluster on an otherwise C57BL/6 genetic background. Methods: To determine whether the presence of 129 alleles of paralogous Gbps could influence the phenotypes of 129-congenic Gbp-deficient strains, we studied the expression of Gbps in both C57BL/6J and 129/Sv mice following in vivo stimulation with adjuvants and after infection with either Toxoplasma gondii or Shigella flexneri. Results: We show that C57BL/6J relative to 129/Sv mice display moderately elevated expression of Gbp2, but more prominently, are also defective for Gbp2b (formerly Gbp1) mRNA induction upon immune priming. Notably, Toxoplasma infections induce robust Gbp2b protein expression in both strains of mice, suggestive of a Toxoplasma-activated mechanism driving Gbp2b protein translation. We further find that the higher expression of Gbp2b mRNA in 129/Sv mice correlates with a gene duplication event at the Gbp2b locus resulting in two copies of the Gbp2b gene on the haploid genome of the 129/Sv strain. Conclusions: Our findings demonstrate functional differences between 129 and C57BL/6 Gbp alleles which need to be considered in the design and interpretation of studies utilizing mouse models, particularly for phenotypes influenced by Gbp2 or Gbp2b expression.

12.
EMBO J ; 38(13): e100926, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31268602

ABSTRACT

The guanylate binding protein (GBP) family of interferon-inducible GTPases promotes antimicrobial immunity and cell death. During bacterial infection, multiple mouse Gbps, human GBP2, and GBP5 support the activation of caspase-1-containing inflammasome complexes or caspase-4 which trigger pyroptosis. Whether GBPs regulate other forms of cell death is not known. The apicomplexan parasite Toxoplasma gondii causes macrophage death through unidentified mechanisms. Here we report that Toxoplasma-induced death of human macrophages requires GBP1 and its ability to target Toxoplasma parasitophorous vacuoles through its GTPase activity and prenylation. Mechanistically, GBP1 promoted Toxoplasma detection by AIM2, which induced GSDMD-independent, ASC-, and caspase-8-dependent apoptosis. Identical molecular determinants targeted GBP1 to Salmonella-containing vacuoles. GBP1 facilitated caspase-4 recruitment to Salmonella leading to its enhanced activation and pyroptosis. Notably, GBP1 could be bypassed by the delivery of Toxoplasma DNA or bacterial LPS into the cytosol, pointing to its role in liberating microbial molecules. GBP1 thus acts as a gatekeeper of cell death pathways, which respond specifically to infecting microbes. Our findings expand the immune roles of human GBPs in regulating not only pyroptosis, but also apoptosis.


Subject(s)
GTP-Binding Proteins/metabolism , Macrophages/parasitology , Toxoplasma/pathogenicity , Toxoplasmosis/metabolism , Caspases, Initiator/metabolism , DNA-Binding Proteins/metabolism , Humans , Macrophages/metabolism , Protein Prenylation , Pyroptosis , THP-1 Cells , Toxoplasmosis/parasitology
13.
Elife ; 82019 02 12.
Article in English | MEDLINE | ID: mdl-30744806

ABSTRACT

For image-based infection biology, accurate unbiased quantification of host-pathogen interactions is essential, yet often performed manually or using limited enumeration employing simple image analysis algorithms based on image segmentation. Host protein recruitment to pathogens is often refractory to accurate automated assessment due to its heterogeneous nature. An intuitive intelligent image analysis program to assess host protein recruitment within general cellular pathogen defense is lacking. We present HRMAn (Host Response to Microbe Analysis), an open-source image analysis platform based on machine learning algorithms and deep learning. We show that HRMAn has the capacity to learn phenotypes from the data, without relying on researcher-based assumptions. Using Toxoplasma gondii and Salmonella enterica Typhimurium we demonstrate HRMAn's capacity to recognize, classify and quantify pathogen killing, replication and cellular defense responses. HRMAn thus presents the only intelligent solution operating at human capacity suitable for both single image and high content image analysis. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).


Subject(s)
Artificial Intelligence , Host-Pathogen Interactions , Image Processing, Computer-Assisted/methods , Microscopy, Fluorescence/methods , Salmonella typhimurium/growth & development , Single-Cell Analysis/methods , Toxoplasma/growth & development , HeLa Cells , Humans , Workflow
14.
Sci Rep ; 7(1): 5209, 2017 07 12.
Article in English | MEDLINE | ID: mdl-28701773

ABSTRACT

Interferon gamma (IFNγ) is the major proinflammatory cytokine conferring resistance to the intracellular vacuolar pathogen Toxoplasma gondii by inducing the destruction of the parasitophorous vacuole (PV). We previously identified TRIM21 as an IFNγ-driven E3 ubiquitin ligase mediating the deposition of ubiquitin around pathogen inclusions. Here, we show that TRIM21 knockout mice were highly susceptible to Toxoplasma infection, exhibiting decreased levels of serum inflammatory cytokines and higher parasite burden in the peritoneum and brain. We demonstrate that IFNγ drives recruitment of TRIM21 to GBP1-positive Toxoplasma vacuoles, leading to Lys63-linked ubiquitination of the vacuole and restriction of parasite early replication without interfering with vacuolar disruption. As seen in vivo, TRIM21 impacted the secretion of inflammatory cytokines. This study identifies TRIM21 as a previously unknown modulator of Toxoplasma gondii resistance in vivo thereby extending host innate immune recognition of eukaryotic pathogens to include E3 ubiquitin ligases.


Subject(s)
Fibroblasts/parasitology , GTP-Binding Proteins/metabolism , Host-Parasite Interactions/immunology , Macrophages/parasitology , Ribonucleoproteins/physiology , Toxoplasmosis/parasitology , Vacuoles/parasitology , Animals , Autophagy , Cytokines/metabolism , Female , Fibroblasts/immunology , Fibroblasts/metabolism , Interferon-gamma/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Toxoplasma/pathogenicity , Toxoplasmosis/immunology , Toxoplasmosis/metabolism , Ubiquitin/metabolism , Ubiquitination , Vacuoles/immunology , Vacuoles/metabolism , Virulence Factors/metabolism
15.
Trends Parasitol ; 33(6): 473-488, 2017 06.
Article in English | MEDLINE | ID: mdl-28330745

ABSTRACT

The parasitophorous vacuole is a unique replicative niche for apicomplexan parasites, including Toxoplasma gondii. Derived from host plasma membrane, the vacuole is rendered nonfusogenic with the host endolysosomal system. Toxoplasma secretes numerous proteins to modify the forming vacuole, enable nutrient uptake, and set up mechanisms of host subversion. Here we describe the pathways of host-parasite interaction at the parasitophorous vacuole employed by Toxoplasma and host, leading to the intricate balance of host defence versus parasite survival.


Subject(s)
Host-Parasite Interactions/physiology , Toxoplasma/immunology , Animals , Humans , Toxoplasma/cytology , Vacuoles/immunology
16.
PLoS Pathog ; 12(11): e1006027, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27875583

ABSTRACT

Toxoplasma gondii is the most common protozoan parasitic infection in man. Gamma interferon (IFNγ) activates haematopoietic and non-haematopoietic cells to kill the parasite and mediate host resistance. IFNγ-driven host resistance pathways and parasitic virulence factors are well described in mice, but a detailed understanding of pathways that kill Toxoplasma in human cells is lacking. Here we show, that contrary to the widely held belief that the Toxoplasma vacuole is non-fusogenic, in an immune-stimulated environment, the vacuole of type II Toxoplasma in human cells is able to fuse with the host endo-lysosomal machinery leading to parasite death by acidification. Similar to murine cells, we find that type II, but not type I Toxoplasma vacuoles are targeted by K63-linked ubiquitin in an IFNγ-dependent manner in non-haematopoetic primary-like human endothelial cells. Host defence proteins p62 and NDP52 are subsequently recruited to the type II vacuole in distinct, overlapping microdomains with a loss of IFNγ-dependent restriction in p62 knocked down cells. Autophagy proteins Atg16L1, GABARAP and LC3B are recruited to <10% of parasite vacuoles and show no parasite strain preference, which is consistent with inhibition and enhancement of autophagy showing no effect on parasite replication. We demonstrate that this differs from HeLa human epithelial cells, where type II Toxoplasma are restricted by non-canonical autophagy leading to growth stunting that is independent of lysosomal acidification. In contrast to mouse cells, human vacuoles do not break. In HUVEC, the ubiquitinated vacuoles are targeted for destruction in acidified LAMP1-positive endo-lysosomal compartments. Consequently, parasite death can be prevented by inhibiting host ubiquitination and endosomal acidification. Thus, K63-linked ubiquitin recognition leading to vacuolar endo-lysosomal fusion and acidification is an important, novel virulence-driven Toxoplasma human host defence pathway.


Subject(s)
Host-Parasite Interactions/immunology , Interferon-gamma/immunology , Lysosomes/immunology , Toxoplasmosis/immunology , Ubiquitination/immunology , Flow Cytometry , Humans , Immunoblotting , Lysine/metabolism , Lysosomes/metabolism , Lysosomes/parasitology , Microscopy, Fluorescence , Toxoplasma/immunology , Toxoplasma/metabolism , Toxoplasmosis/metabolism , Vacuoles/immunology , Vacuoles/metabolism , Vacuoles/parasitology
17.
Cell Microbiol ; 18(8): 1056-64, 2016 08.
Article in English | MEDLINE | ID: mdl-26874079

ABSTRACT

Guanylate binding proteins (GBPs) are a family of large interferon-inducible GTPases that are transcriptionally upregulated upon infection with intracellular pathogens. Murine GBPs (mGBPs) including mGBP1 and 2 localize to and disrupt pathogen-containing vacuoles (PVs) resulting in the cell-autonomous clearing or innate immune detection of PV-resident pathogens. Human GBPs (hGBPs) are known to exert antiviral host defense and activate the NLRP3 inflammasome, but it is unclear whether hGBPs can directly recognize and control intravacuolar pathogens. Here, we report that endogenous or ectopically expressed hGBP1 fails to associate with PVs formed in human cells by the bacterial pathogens Chlamydia trachomatis or Salmonella typhimurium or the protozoan pathogen Toxoplasma gondii. While we find that hGBP1 expression has no discernible effect on intracellular replication of C. trachomatis and S. typhimurium, we observed enhanced early Toxoplasma replication in CRISPR hGBP1-deleted human epithelial cells. We thus identified a novel role for hGBP1 in cell-autonomous immunity that is independent of PV translocation, as observed for mGBPs. This study highlights fundamental differences between human and murine GBPs and underlines the need to study the functions of GBPs at cellular locations away from PVs.


Subject(s)
GTP-Binding Proteins/metabolism , Toxoplasma/physiology , Vacuoles/metabolism , A549 Cells , Chlamydia trachomatis/physiology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/parasitology , Humans , Protein Transport , Salmonella typhimurium/physiology , Vacuoles/parasitology
18.
Antimicrob Agents Chemother ; 58(10): 6032-43, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25070106

ABSTRACT

PfCDPK1 is a Plasmodium falciparum calcium-dependent protein kinase, which has been identified as a potential target for novel antimalarial chemotherapeutics. In order to further investigate the role of PfCDPK1, we established a high-throughput in vitro biochemical assay and used it to screen a library of over 35,000 small molecules. Five chemical series of inhibitors were initially identified from the screen, from which series 1 and 2 were selected for chemical optimization. Indicative of their mechanism of action, enzyme inhibition by these compounds was found to be sensitive to both the ATP concentration and substitution of the amino acid residue present at the "gatekeeper" position at the ATP-binding site of the enzyme. Medicinal chemistry efforts led to a series of PfCDPK1 inhibitors with 50% inhibitory concentrations (IC50s) below 10 nM against PfCDPK1 in a biochemical assay and 50% effective concentrations (EC50s) less than 100 nM for inhibition of parasite growth in vitro. Potent inhibition was combined with acceptable absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties and equipotent inhibition of Plasmodium vivax CDPK1. However, we were unable to correlate biochemical inhibition with parasite growth inhibition for this series overall. Inhibition of Plasmodium berghei CDPK1 correlated well with PfCDPK1 inhibition, enabling progression of a set of compounds to in vivo evaluation in the P. berghei rodent model for malaria. These chemical series have potential for further development as inhibitors of CDPK1.


Subject(s)
Antimalarials/pharmacology , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Protozoan Proteins/metabolism , Animals , Antimalarials/therapeutic use , Malaria/drug therapy , Mice , Plasmodium berghei/drug effects , Plasmodium berghei/pathogenicity , Plasmodium falciparum/pathogenicity , Plasmodium vivax/drug effects , Plasmodium vivax/pathogenicity , Protein Kinase Inhibitors/therapeutic use , Protozoan Proteins/antagonists & inhibitors
19.
J Med Chem ; 57(8): 3570-87, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24689770

ABSTRACT

A structure-guided design approach using a homology model of Plasmodium falciparum calcium-dependent protein kinase 1 (PfCDPK1) was used to improve the potency of a series of imidazopyridazine inhibitors as potential antimalarial agents. This resulted in high affinity compounds with PfCDPK1 enzyme IC50 values less than 10 nM and in vitro P. falciparum antiparasite EC50 values down to 12 nM, although these compounds did not have suitable ADME properties to show in vivo efficacy in a mouse model. Structural modifications designed to address the ADME issues, in particular permeability, were initially accompanied by losses in antiparasite potency, but further optimization allowed a good balance in the compound profile to be achieved. Upon testing in vivo in a murine model of efficacy against malaria, high levels of compound exposure relative to their in vitro activities were achieved, and the modest efficacy that resulted raises questions about the level of effect that is achievable through the targeting of PfCDPK1.


Subject(s)
Antimalarials/chemical synthesis , Plasmodium falciparum/drug effects , Protein Kinase Inhibitors/chemical synthesis , Protozoan Proteins/antagonists & inhibitors , Pyridazines/chemical synthesis , Animals , Antimalarials/pharmacology , Mice , Plasmodium falciparum/enzymology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/chemistry , Protozoan Proteins/chemistry , Pyridazines/pharmacology , Structure-Activity Relationship
20.
Nat Chem ; 6(2): 112-21, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24451586

ABSTRACT

Malaria is an infectious disease caused by parasites of the genus Plasmodium, which leads to approximately one million deaths per annum worldwide. Chemical validation of new antimalarial targets is urgently required in view of rising resistance to current drugs. One such putative target is the enzyme N-myristoyltransferase, which catalyses the attachment of the fatty acid myristate to protein substrates (N-myristoylation). Here, we report an integrated chemical biology approach to explore protein myristoylation in the major human parasite P. falciparum, combining chemical proteomic tools for identification of the myristoylated and glycosylphosphatidylinositol-anchored proteome with selective small-molecule N-myristoyltransferase inhibitors. We demonstrate that N-myristoyltransferase is an essential and chemically tractable target in malaria parasites both in vitro and in vivo, and show that selective inhibition of N-myristoylation leads to catastrophic and irreversible failure to assemble the inner membrane complex, a critical subcellular organelle in the parasite life cycle. Our studies provide the basis for the development of new antimalarials targeting N-myristoyltransferase.


Subject(s)
Acyltransferases/antagonists & inhibitors , Antimalarials/chemistry , Enzyme Inhibitors/chemistry , Acyl Coenzyme A/chemistry , Acyl Coenzyme A/metabolism , Acyltransferases/genetics , Acyltransferases/metabolism , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use , Binding Sites , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Cell Cycle Proteins/metabolism , Crystallography, X-Ray , Cycloaddition Reaction , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Malaria/drug therapy , Malaria/parasitology , Molecular Docking Simulation , Plasmodium falciparum/drug effects , Plasmodium vivax/drug effects , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...