Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Mol Microbiol ; 115(5): 860-869, 2021 05.
Article in English | MEDLINE | ID: mdl-33565178

ABSTRACT

Theileria are tick-transmitted parasites that cause often fatal leuko-proliferative diseases in cattle called tropical theileriosis (T. annulata) and East Coast fever (T. parva). However, upon treatment with anti-theilerial drug-transformed leukocytes die of apoptosis indicating that Theileria-induced transformation is reversible making infected leukocytes a powerful example of how intracellular parasites interact with their hosts. Theileria-transformed leukocytes disseminate throughout infected cattle causing a cancer-like disease and here, we discuss how cytokines, noncoding RNAs and oncometabolites can contribute to the transformed phenotype and disease pathology.


Subject(s)
Cattle Diseases/physiopathology , Leukocytes/parasitology , Theileria/physiology , Theileriasis/physiopathology , Animals , Cattle , Cattle Diseases/genetics , Cattle Diseases/immunology , Cattle Diseases/parasitology , Cytokines/genetics , Cytokines/immunology , Host-Parasite Interactions , Leukocytes/immunology , Theileria/genetics , Theileriasis/genetics , Theileriasis/immunology , Theileriasis/parasitology
2.
PLoS Pathog ; 16(5): e1008106, 2020 05.
Article in English | MEDLINE | ID: mdl-32463830

ABSTRACT

Toxoplasma gondii possesses an armada of secreted virulent factors that enable parasite invasion and survival into host cells. These factors are contained in specific secretory organelles, the rhoptries, micronemes and dense granules that release their content upon host cell recognition. Dense granules are secreted in a constitutive manner during parasite replication and play a crucial role in modulating host metabolic and immune responses. While the molecular mechanisms triggering rhoptry and microneme release upon host cell adhesion have been well studied, constitutive secretion remains a poorly explored aspect of T. gondii vesicular trafficking. Here, we investigated the role of the small GTPase Rab11A, a known regulator of exocytosis in eukaryotic cells. Our data revealed an essential role of Rab11A in promoting the cytoskeleton driven transport of dense granules and the release of their content into the vacuolar space. Rab11A also regulates transmembrane protein trafficking and localization during parasite replication, indicating a broader role of Rab11A in cargo exocytosis at the plasma membrane. Moreover, we found that Rab11A also regulates extracellular parasite motility and adhesion to host cells. In line with these findings, MIC2 secretion was altered in Rab11A-defective parasites, which also exhibited severe morphological defects. Strikingly, by live imaging we observed a polarized accumulation of Rab11A-positive vesicles and dense granules at the apical pole of extracellular motile and invading parasites suggesting that apically polarized Rab11A-dependent delivery of cargo regulates early secretory events during parasite entry into host cells.


Subject(s)
Transport Vesicles/metabolism , Vacuoles/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Cell Adhesion , Cell Line , Cell Membrane/metabolism , Cytoskeleton/metabolism , Host-Parasite Interactions/physiology , Humans , Membrane Proteins/metabolism , Microtubules/metabolism , Parasites/metabolism , Protein Transport , Protozoan Proteins , Toxoplasma/metabolism , Toxoplasmosis/metabolism , rab GTP-Binding Proteins/physiology
3.
PLoS Pathog ; 16(6): e1008640, 2020 06.
Article in English | MEDLINE | ID: mdl-32569299

ABSTRACT

Ubiquitylation is a common post translational modification of eukaryotic proteins and in the human malaria parasite, Plasmodium falciparum (Pf) overall ubiquitylation increases in the transition from intracellular schizont to extracellular merozoite stages in the asexual blood stage cycle. Here, we identify specific ubiquitylation sites of protein substrates in three intraerythrocytic parasite stages and extracellular merozoites; a total of 1464 sites in 546 proteins were identified (data available via ProteomeXchange with identifier PXD014998). 469 ubiquitylated proteins were identified in merozoites compared with only 160 in the preceding intracellular schizont stage, suggesting a large increase in protein ubiquitylation associated with merozoite maturation. Following merozoite invasion of erythrocytes, few ubiquitylated proteins were detected in the first intracellular ring stage but as parasites matured through trophozoite to schizont stages the apparent extent of ubiquitylation increased. We identified commonly used ubiquitylation motifs and groups of ubiquitylated proteins in specific areas of cellular function, for example merozoite pellicle proteins involved in erythrocyte invasion, exported proteins, and histones. To investigate the importance of ubiquitylation we screened ubiquitin pathway inhibitors in a parasite growth assay and identified the ubiquitin activating enzyme (UBA1 or E1) inhibitor MLN7243 (TAK-243) to be particularly effective. This small molecule was shown to be a potent inhibitor of recombinant PfUBA1, and a structural homology model of MLN7243 bound to the parasite enzyme highlights avenues for the development of P. falciparum specific inhibitors. We created a genetically modified parasite with a rapamycin-inducible functional deletion of uba1; addition of either MLN7243 or rapamycin to the recombinant parasite line resulted in the same phenotype, with parasite development blocked at the schizont stage. Nuclear division and formation of intracellular structures was interrupted. These results indicate that the intracellular target of MLN7243 is UBA1, and this activity is essential for the final differentiation of schizonts to merozoites.


Subject(s)
Merozoites/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Ubiquitin/metabolism , Ubiquitination , Humans , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Ubiquitin/genetics
4.
Biol Cell ; 113(4): 220-233, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33314227

ABSTRACT

Theileria parasites are classified in the phylum Apicomplexa that includes several genera of medical and veterinary importance such as Plasmodium, Babesia, Toxoplasma and Cryptosporidium. These protozoans have evolved subtle ways to reshape their intracellular niche for their own benefit and Theileria is no exception. This tick transmitted microorganism is unique among all eukaryotes in that its intracellular schizont stage is able to transform its mammalian host leukocytes into an immortalised highly disseminating cell that phenocopies tumour cells. Here, we describe what is known about secreted Theileria-encoded host cell manipulators.


Subject(s)
Apicomplexa , Leukocytes , Theileria , Animals , Antigens, Protozoan , Apicomplexa/immunology , Apicomplexa/metabolism , Cell Transformation, Neoplastic , Host-Parasite Interactions/immunology , Host-Parasite Interactions/physiology , Leukocytes/parasitology , Leukocytes/pathology , Mammals/parasitology , Theileria/immunology , Theileria/metabolism
5.
Cell Microbiol ; 22(12): e13255, 2020 12.
Article in English | MEDLINE | ID: mdl-32830401

ABSTRACT

Theileria annulata is a tick-transmitted apicomplexan parasite that infects and transforms bovine leukocytes into disseminating tumours that cause a disease called tropical theileriosis. Using comparative transcriptomics we identified genes transcriptionally perturbed during Theileria-induced leukocyte transformation. Dataset comparisons highlighted a small set of genes associated with Theileria-transformed leukocyte dissemination. The roles of Granzyme A (GZMA) and RAS guanyl-releasing protein 1 (RASGRP1) were verified by CRISPR/Cas9-mediated knockdown. Knocking down expression of GZMA and RASGRP1 in attenuated macrophages led to a regain in their dissemination in Rag2/γC mice confirming their role as dissemination suppressors in vivo. We further evaluated the roles of GZMA and RASGRP1 in human B lymphomas by comparing the transcriptome of 934 human cancer cell lines to that of Theileria-transformed bovine host cells. We confirmed dampened dissemination potential of human B lymphomas that overexpress GZMA and RASGRP1. Our results provide evidence that GZMA and RASGRP1 have a novel tumour suppressor function in both T. annulata-infected bovine host leukocytes and in human B lymphomas.


Subject(s)
DNA-Binding Proteins/genetics , Genes, Tumor Suppressor/physiology , Granzymes/genetics , Guanine Nucleotide Exchange Factors/genetics , Leukocytes/parasitology , Lymphoma, B-Cell/genetics , Macrophages/parasitology , Theileria annulata/genetics , Animals , Cattle , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Lymphoma, B-Cell/parasitology , Mice , Theileria annulata/pathogenicity
6.
PLoS Pathog ; 14(3): e1006942, 2018 03.
Article in English | MEDLINE | ID: mdl-29570727

ABSTRACT

Theileria annulata is an apicomplexan parasite that infects and transforms bovine macrophages that disseminate throughout the animal causing a leukaemia-like disease called tropical theileriosis. Using deep RNAseq of T. annulata-infected B cells and macrophages we identify a set of microRNAs induced by infection, whose expression diminishes upon loss of the hyper-disseminating phenotype of virulent transformed macrophages. We describe how infection-induced upregulation of miR-126-5p ablates JIP-2 expression to release cytosolic JNK to translocate to the nucleus and trans-activate AP-1-driven transcription of mmp9 to promote tumour dissemination. In non-disseminating attenuated macrophages miR-126-5p levels drop, JIP-2 levels increase, JNK1 is retained in the cytosol leading to decreased c-Jun phosphorylation and dampened AP-1-driven mmp9 transcription. We show that variation in miR-126-5p levels depends on the tyrosine phosphorylation status of AGO2 that is regulated by Grb2-recruitment of PTP1B. In attenuated macrophages Grb2 levels drop resulting in less PTP1B recruitment, greater AGO2 phosphorylation, less miR-126-5p associated with AGO2 and a consequent rise in JIP-2 levels. Changes in miR-126-5p levels therefore, underpin both the virulent hyper-dissemination and the attenuated dissemination of T. annulata-infected macrophages.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , MAP Kinase Kinase 4/metabolism , Macrophages/microbiology , MicroRNAs/genetics , Theileriasis/microbiology , Transcription Factor AP-1/metabolism , Virulence/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Cattle , Cells, Cultured , MAP Kinase Kinase 4/genetics , Macrophages/metabolism , Theileria annulata/pathogenicity , Theileriasis/genetics , Theileriasis/metabolism , Transcription Factor AP-1/genetics
7.
Cell Microbiol ; 21(3): e12973, 2019 03.
Article in English | MEDLINE | ID: mdl-30412643

ABSTRACT

Constitutive c-Jun N-terminal kinase (JNK) activity characterizes bovine T and B cells infected with Theileria parva, and B cells and macrophages infected with Theileria annulata. Here, we show that T. annulata infection of macrophages manipulates JNK activation by recruiting JNK2 and not JNK1 to the parasite surface, whereas JNK1 is found predominantly in the host cell nucleus. At the parasite's surface, JNK2 forms a complex with p104, a GPI-(GlycosylPhosphatidylInositol)-anchor T. annulata plasma membrane protein. Sequestration of JNK2 depended on Protein Kinase-A (PKA)-mediated phosphorylation of a JNK-binding motif common to T. parva and a cell penetrating peptide harbouring the conserved p104 JNK-binding motif competitively ablated binding, whereupon liberated JNK2 became ubiquitinated and degraded. Cytosolic sequestration of JNK2 suppressed small mitochondrial ARF-mediated autophagy, whereas it sustained nuclear JNK1 levels, c-Jun phosphorylation, and matrigel traversal. Therefore, T. annulata sequestration of JNK2 contributes to both survival and dissemination of Theileria-transformed macrophages.


Subject(s)
Host-Pathogen Interactions , Immune Evasion , Macrophages/parasitology , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Protozoan Proteins/metabolism , Theileria annulata/growth & development , Animals , Macrophages/immunology , Mitogen-Activated Protein Kinase 8/metabolism , Models, Theoretical , Protein Binding , Theileria annulata/metabolism , Theileriasis/parasitology , Theileriasis/pathology
8.
PLoS Pathog ; 13(4): e1006331, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28430827

ABSTRACT

Toxoplasma gondii possesses a highly polarized secretory system, which efficiently assembles de novo micronemes and rhoptries during parasite replication. These apical secretory organelles release their contents into host cells promoting parasite invasion and survival. Using a CreLox-based inducible knock-out strategy and the ddFKBP over-expression system, we unraveled novel functions of the clathrin adaptor complex TgAP1. First, our data indicate that AP1 in T. gondii likely functions as a conserved heterotetrameric complex composed of the four subunits γ, ß, µ1, σ1 and interacts with known regulators of clathrin-mediated vesicular budding such as the unique ENTH-domain containing protein, which we named Epsin-like protein (TgEpsL). Disruption of the µ1 subunit resulted in the mis-sorting of microneme proteins at the level of the Trans-Golgi-Network (TGN). Furthermore, we demonstrated that TgAP1 regulates rhoptry biogenesis by activating rhoptry protein exit from the TGN, but also participates in the post-Golgi maturation process of preROP compartments into apically anchored club-shaped mature organelles. For this latter activity, our data indicate a specific functional relationship between TgAP1 and the Rab5A-positive endosome-like compartment. In addition, we unraveled an original role for TgAP1 in the regulation of parasite division. APµ1-depleted parasites undergo normal daughter cell budding and basal complex assembly but fail to segregate at the end of cytokinesis.


Subject(s)
Adaptor Protein Complex 1/metabolism , Protozoan Proteins/metabolism , Toxoplasma/metabolism , Adaptor Protein Complex 1/genetics , Animals , Cell Division , Clathrin/genetics , Clathrin/metabolism , Cytokinesis , Endosomes/metabolism , Gene Expression , Gene Knockout Techniques , Golgi Apparatus/metabolism , Mass Spectrometry , Models, Biological , Organelles/metabolism , Protein Transport , Protozoan Proteins/genetics , Toxoplasma/genetics , Toxoplasma/ultrastructure , trans-Golgi Network/metabolism
9.
Bioorg Med Chem ; 26(6): 1127-1134, 2018 03 15.
Article in English | MEDLINE | ID: mdl-28917447

ABSTRACT

One powerful application of cell penetrating peptides is the delivery into cells of molecules that function as specific competitors or inhibitors of protein-protein interactions. Ablating defined protein-protein interactions is a refined way to explore their contribution to a particular cellular phenotype in a given disease context. Cell-penetrating peptides can be synthetically constrained through various chemical modifications that stabilize a given structural fold with the potential to improve competitive binding to specific targets. Theileria-transformed leukocytes display high PKA activity, but PKA is an enzyme that plays key roles in multiple cellular processes; consequently genetic ablation of kinase activity gives rise to a myriad of confounding phenotypes. By contrast, ablation of a specific kinase-substrate interaction has the potential to give more refined information and we illustrate this here by describing how surgically ablating PKA interactions with BAD gives precise information on the type of glycolysis performed by Theileria-transformed leukocytes. In addition, we provide two other examples of how ablating specific protein-protein interactions in Theileria-infected leukocytes leads to precise phenotypes and argue that constrained penetrating peptides have great therapeutic potential to combat infectious diseases in general.


Subject(s)
Cell-Penetrating Peptides/metabolism , Theileria/pathogenicity , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/chemistry , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , GRB2 Adaptor Protein/chemistry , GRB2 Adaptor Protein/metabolism , Hexokinase/chemistry , Hexokinase/metabolism , Humans , Leukocytes/cytology , Leukocytes/metabolism , Leukocytes/parasitology , Oxidative Phosphorylation , Protein Interaction Maps
10.
J Biol Chem ; 291(28): 14732-46, 2016 Jul 08.
Article in English | MEDLINE | ID: mdl-27226564

ABSTRACT

Differential functions of Rab5 isoforms in endocytosis are not well characterized. Here, we cloned, expressed, and characterized Rab5a and Rab5b from Leishmania and found that both of them are localized in the early endosome. To understand the role of LdRab5 isoforms in different modes of endocytosis in Leishmania, we generated transgenic parasites overexpressing LdRab5a, LdRab5b, or their dominant-positive (LdRab5a:Q93L and LdRab5b:Q80L) or dominant-negative mutants (LdRab5a:N146I and LdRab5b:N133I). Using LdRab5a or its mutants overexpressing parasites, we found that LdRab5a specifically regulates the fluid-phase endocytosis of horseradish peroxidase and also specifically induced the transport of dextran-Texas Red to the lysosomes. In contrast, cells overexpressing LdRab5b or its mutants showed that LdRab5b explicitly controls receptor-mediated endocytosis of hemoglobin, and overexpression of LdRab5b:WT enhanced the transport of internalized Hb to the lysosomes in comparison with control cells. To unequivocally demonstrate the role of Rab5 isoforms in endocytosis in Leishmania, we tried to generate null-mutants of LdRab5a and LdRab5b parasites, but both were lethal indicating their essential functions in parasites. Therefore, we used heterozygous LdRab5a(+/-) and LdRab5b(+/-) cells. LdRab5a(+/-) Leishmania showed 50% inhibition of HRP uptake, but hemoglobin endocytosis was uninterrupted. In contrast, about 50% inhibition of Hb endocytosis was observed in LdRab5b(+/-) cells without any significant effect on HRP uptake. Finally, we tried to identify putative LdRab5a and LdRab5b effectors. We found that LdRab5b interacts with clathrin heavy chain and hemoglobin receptor. However, LdRab5a failed to interact with the clathrin heavy chain, and interaction with hemoglobin receptor was significantly less. Thus, our results showed that LdRab5a and LdRab5b differentially regulate fluid phase and receptor-mediated endocytosis in Leishmania.


Subject(s)
Endocytosis/physiology , Leishmania donovani/metabolism , Protein Isoforms/physiology , rab5 GTP-Binding Proteins/physiology , Amino Acid Sequence , Animals , Mutation , Sequence Homology, Amino Acid , rab5 GTP-Binding Proteins/chemistry , rab5 GTP-Binding Proteins/genetics
11.
PLoS Pathog ; 11(5): e1004815, 2015 May.
Article in English | MEDLINE | ID: mdl-25951195

ABSTRACT

Blocking Plasmodium falciparum transmission to mosquitoes has been designated a strategic objective in the global agenda of malaria elimination. Transmission is ensured by gametocyte-infected erythrocytes (GIE) that sequester in the bone marrow and at maturation are released into peripheral blood from where they are taken up during a mosquito blood meal. Release into the blood circulation is accompanied by an increase in GIE deformability that allows them to pass through the spleen. Here, we used a microsphere matrix to mimic splenic filtration and investigated the role of cAMP-signalling in regulating GIE deformability. We demonstrated that mature GIE deformability is dependent on reduced cAMP-signalling and on increased phosphodiesterase expression in stage V gametocytes, and that parasite cAMP-dependent kinase activity contributes to the stiffness of immature gametocytes. Importantly, pharmacological agents that raise cAMP levels in transmissible stage V gametocytes render them less deformable and hence less likely to circulate through the spleen. Therefore, phosphodiesterase inhibitors that raise cAMP levels in P. falciparum infected erythrocytes, such as sildenafil, represent new candidate drugs to block transmission of malaria parasites.


Subject(s)
Erythrocyte Deformability/physiology , Erythrocytes/parasitology , Malaria, Falciparum/parasitology , Plasmodium falciparum/physiology , Signal Transduction , Animals , Culicidae , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Malaria, Falciparum/transmission
12.
Biochim Biophys Acta ; 1854(10 Pt B): 1575-85, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25847873

ABSTRACT

Many cellular functions in eukaryotic pathogens are mediated by the cyclic nucleotide binding (CNB) domain, which senses second messengers such as cyclic AMP and cyclic GMP. Although CNB domain-containing proteins have been identified in many pathogenic organisms, an incomplete understanding of how CNB domains in pathogens differ from other eukaryotic hosts has hindered the development of selective inhibitors for CNB domains associated with infectious diseases. Here, we identify and classify CNB domain-containing proteins in eukaryotic genomes to understand the evolutionary basis for CNB domain functional divergence in pathogens. We identify 359 CNB domain-containing proteins in 31 pathogenic organisms and classify them into distinct subfamilies based on sequence similarity within the CNB domain as well as functional domains associated with the CNB domain. Our study reveals novel subfamilies with pathogen-specific variations in the phosphate-binding cassette. Analyzing these variations in light of existing structural and functional data provides new insights into ligand specificity and promiscuity and clues for drug design. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases.


Subject(s)
Evolution, Molecular , Host-Pathogen Interactions/genetics , Protein Kinases/genetics , Protein Structure, Tertiary/genetics , Cyclic AMP/chemistry , Cyclic AMP/genetics , Cyclic GMP/chemistry , Cyclic GMP/genetics , Drug Design , Genome , Humans , Infections/genetics , Infections/pathology , Phylogeny , Protein Binding , Protein Kinases/chemistry , Protein Structure, Tertiary/drug effects , Signal Transduction
13.
Blood Cells Mol Dis ; 58: 35-44, 2016 May.
Article in English | MEDLINE | ID: mdl-27067487

ABSTRACT

The phosphorylation status of red blood cell proteins is strongly altered during the infection by the malaria parasite Plasmodium falciparum. We identify the key phosphorylation events that occur in the erythrocyte membrane and cytoskeleton during infection, by a comparative analysis of global phospho-proteome screens between infected (obtained at schizont stage) and uninfected RBCs. The meta-analysis of reported mass spectrometry studies revealed a novel compendium of 495 phosphorylation sites in 182 human proteins with regulatory roles in red cell morphology and stability, with about 25% of these sites specific to infected cells. A phosphorylation motif analysis detected 7 unique motifs that were largely mapped to kinase consensus sequences of casein kinase II and of protein kinase A/protein kinase C. This analysis highlighted prominent roles for PKA/PKC involving 78 phosphorylation sites. We then compared the phosphorylation status of PKA (PKC) specific sites in adducin, dematin, Band 3 and GLUT-1 in uninfected RBC stimulated or not by cAMP to their phosphorylation status in iRBC. We showed cAMP-induced phosphorylation of adducin S59 by immunoblotting and we were able to demonstrate parasite-induced phosphorylation for adducin S726, Band 3 and GLUT-1, corroborating the protein phosphorylation status in our erythrocyte phosphorylation site compendium.


Subject(s)
Erythrocytes/parasitology , Malaria, Falciparum/metabolism , Plasmodium falciparum/physiology , Proteome/metabolism , Amino Acid Sequence , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/analysis , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Cytoskeleton/parasitology , Erythrocytes/chemistry , Erythrocytes/metabolism , Glucose Transporter Type 1/analysis , Glucose Transporter Type 1/metabolism , Humans , Phosphorylation , Proteome/analysis
14.
PLoS Pathog ; 10(12): e1004520, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25522250

ABSTRACT

All pathogenesis and death associated with Plasmodium falciparum malaria is due to parasite-infected erythrocytes. Invasion of erythrocytes by P. falciparum merozoites requires specific interactions between host receptors and parasite ligands that are localized in apical organelles called micronemes. Here, we identify cAMP as a key regulator that triggers the timely secretion of microneme proteins enabling receptor-engagement and invasion. We demonstrate that exposure of merozoites to a low K+ environment, typical of blood plasma, activates a bicarbonate-sensitive cytoplasmic adenylyl cyclase to raise cytosolic cAMP levels and activate protein kinase A, which regulates microneme secretion. We also show that cAMP regulates merozoite cytosolic Ca2+ levels via induction of an Epac pathway and demonstrate that increases in both cAMP and Ca2+ are essential to trigger microneme secretion. Our identification of the different elements in cAMP-dependent signaling pathways that regulate microneme secretion during invasion provides novel targets to inhibit blood stage parasite growth and prevent malaria.


Subject(s)
Cyclic AMP/physiology , Erythrocytes/parasitology , Malaria, Falciparum/physiopathology , Merozoites/growth & development , Plasmodium falciparum/pathogenicity , Calcium/physiology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/physiology , Erythrocytes/drug effects , Erythrocytes/pathology , Humans , Hydrogen-Ion Concentration , Merozoites/physiology , Potassium/pharmacology , Signal Transduction/physiology
15.
Cell Microbiol ; 17(4): 467-72, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25620534

ABSTRACT

Within 2 h of infection by Theileria annulata sporozoites, bovine macrophages display a two- to fourfold increase in transcription of hypoxia inducible factor (HIF-1α). Twenty hours post-invasion sporozoites develop into multi-nucleated macroschizonts that transform the infected macrophage into an immortalized, permanently proliferating, hyper-invasive and disease-causing leukaemia-like cell. Once immortalized Theileria-infected leukocytes can be propagated as cell lines and even though cultivated under normoxic conditions, both infected B cells and macrophages display sustained activation of HIF-1α. Attenuated macrophages used as live vaccines against tropical theileriosis also display HIF-1α activation even though they have lost their tumorigenic phenotype. Here, we review data that ascribes HIF-1α activation to the proliferation status of the infected leukocyte and discuss the possibility that Theileria may have lost its ability to render its host macrophage virulent due to continuous parasite replication in a high Reactive Oxygen Species (ROS) environment. We propose a model where uninfected macrophages have low levels of H2 O2 output, whereas virulent-infected macrophages produce high amounts of H2 O2 . Further increase in H2 O2 output leads to dampening of infected macrophage virulence, a characteristic of disease-resistant macrophages. At the same time exposure to H2 O2 sustains HIF-1α that induces the switch from mitochondrial oxidative phosphorylation to Warburg glycolysis, a metabolic shift that underpins uncontrolled infected macrophage proliferation. We propose that as macroschizonts develop into merozoites and infected macrophage proliferation arrests, HIF-1α levels will decrease and glycolysis will switch back from Warburg to oxidative glycolysis. As Theileria infection transforms its host leukocyte into an aggressive leukaemic-like cell, we propose that manipulating ROS levels, HIF-1α induction and oxidative over Warburg glycolysis could contribute to improved disease control. Finally, as excess amounts of H2 O2 drive virulent Theileria-infected macrophages towards attenuation it highlights how infection-induced pathology and redox balance are intimately linked.


Subject(s)
Glycolysis , Host-Pathogen Interactions , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Leukocytes/metabolism , Leukocytes/parasitology , Theileria annulata/growth & development , Animals , Cattle , Cell Proliferation , Hydrogen Peroxide/metabolism , Models, Biological
16.
Malar J ; 15: 248, 2016 Apr 29.
Article in English | MEDLINE | ID: mdl-27129434

ABSTRACT

BACKGROUND: The asexual intra-erythrocytic multiplication of the malaria parasite Plasmodium falciparum is regulated by various molecular mechanisms. In eukaryotic cells, protein kinases are known to play key roles in cell cycle regulation and signaling pathways. The activity of cAMP-dependent protein kinase (PKA) depends on A-kinase anchoring proteins (AKAPs) through protein interactions. While several components of the cAMP dependent pathway-including the PKA catalytic and regulatory subunits-have been characterized in P. falciparum, whether AKAPs are involved in this pathway remains unclear. Here, PfAKAL, an open reading frame of a potential AKAP-like protein in the P. falciparum genome was identified, and its protein partners and putative cellular functions characterized. METHODS: The expression of PfAKAL throughout the erythrocytic cycle of the 3D7 strain was assessed by RT-qPCR and the presence of the corresponding protein by immunofluorescence assays. In order to study physical interactions between PfAKAL and other proteins, pull down experiments were performed using a recombinant PfAKAL protein and parasite protein extracts, or with recombinant proteins. These interactions were also tested by combining biochemical and proteomic approaches. As phosphorylation could be involved in the regulation of protein complexes, both PfAKAL and Pf14-3-3I phosphorylation was studied using a radiolabel kinase activity assay. Finally, to identify a potential function of the protein, PfAKAL sequence was aligned and structurally modeled, revealing a conserved nucleotide-binding pocket; confirmed by qualitative nucleotide binding experiments. RESULTS: PfAKAL is the first AKAP-like protein in P. falciparum to be identified, and shares 23 % sequence identity with the central domain of human AKAP18δ. PfAKAL is expressed in mature asexual stages, merozoites and gametocytes. In spite of homology to AKAP18, biochemical and immunochemical analyses demonstrated that PfAKAL does not interact directly with the P. falciparum PKA regulatory subunit (PfPKA-R), but instead binds and colocalizes with Pf14-3-3I, which in turn interacts with PfPKA-R. In vivo, these different interactions could be regulated by phosphorylation, as PfPKA-R and Pf14-3-3I, but not PfAKAL, are phosphorylated in vitro by PKA. Interestingly, PfAKAL binds nucleotides such as AMP and cAMP, suggesting that this protein may be involved in the AMP-activated protein kinase (AMPK) pathway, or associated with phosphodiesterase activities. CONCLUSION: PfAKAL is an atypical AKAP that shares common features with human AKAP18, such as nucleotides binding. The interaction of PfAKAL with PfPKA-R could be indirectly mediated through a join interaction with Pf14-3-3I. Therefore, PfPKA localization could not depend on PfAKAL, but rather involves other partners.


Subject(s)
A Kinase Anchor Proteins/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Plasmodium falciparum/enzymology , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , A Kinase Anchor Proteins/chemistry , A Kinase Anchor Proteins/metabolism , Amino Acid Sequence , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Signal Transduction
17.
Proteomics ; 15(15): 2716-29, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25886026

ABSTRACT

Pathology of the most lethal form of malaria is caused by Plasmodium falciparum asexual blood stages and initiated by merozoite invasion of erythrocytes. We present a phosphoproteome analysis of extracellular merozoites revealing 1765 unique phosphorylation sites including 785 sites not previously detected in schizonts. All MS data have been deposited in the ProteomeXchange with identifier PXD001684 (http://proteomecentral.proteomexchange.org/dataset/PXD001684). The observed differential phosphorylation between extra and intraerythrocytic life-cycle stages was confirmed using both phospho-site and phospho-motif specific antibodies and is consistent with the core motif [K/R]xx[pS/pT] being highly represented in merozoite phosphoproteins. Comparative bioinformatic analyses highlighted protein sets and pathways with established roles in invasion. Within the merozoite phosphoprotein interaction network a subnetwork of 119 proteins with potential roles in cellular movement and invasion was identified and suggested that it is coregulated by a further small subnetwork of protein kinase A (PKA), two calcium-dependent protein kinases (CDPKs), a phosphatidyl inositol kinase (PI3K), and a GCN2-like elF2-kinase with a predicted role in translational arrest and associated changes in the ubquitinome. To test this notion experimentally, we examined the overall ubiquitination level in intracellular schizonts versus extracellular merozoites and found it highly upregulated in merozoites. We propose that alterations in the phosphoproteome and ubiquitinome reflect a starvation-induced translational arrest as intracellular schizonts transform into extracellular merozoites.


Subject(s)
Erythrocytes/parasitology , Merozoites/metabolism , Plasmodium falciparum/metabolism , Proteome/metabolism , Protozoan Proteins/metabolism , Schizonts/metabolism , Extracellular Space/parasitology , Host-Parasite Interactions , Humans , Merozoites/growth & development , Phosphorylation , Plasmodium falciparum/cytology , Plasmodium falciparum/physiology , Schizonts/growth & development
18.
Infect Immun ; 83(5): 1869-80, 2015 May.
Article in English | MEDLINE | ID: mdl-25690101

ABSTRACT

Transforming growth factor beta (TGF-ß) is a pleiotropic cytokine known to regulate cell growth, differentiation, and motility and is a potent modulator of immune function. TGF-ß consequently plays a central role in carcinogenesis, and a dampened TGF-ß2 response by Theileria annulata-infected monocytes/macrophages underpins disease resistance to tropical theileriosis. Here, we show that concomitant with the loss of TGF-ß2 production, there is ablated expression of COX2 and EP4, which leads to a drop in cyclic AMP (cAMP) levels and, consequently, reduced activation of protein kinase A (PKA) and EPAC. This ablated phenotype can be rescued in attenuated macrophages by the addition of exogenous TGF-ß2, which reactivates the expression of COX2 and EP4 while repressing that of protein kinase inhibitor gamma (PKIG) to the levels in virulent macrophages. TGF-ß2 therefore promotes the adhesion and invasiveness of virulent macrophages by modulating COX2, EP4, and PKIG transcription to initiate a prostaglandin E2 (PGE2)-driven autostimulatory loop that augments PKA and EPAC activities. A virulence phenotype stemming from the double activation of PKA and EPAC is the induction of a CREB-mediated transcriptional program and the upregulation of JAM-L- and integrin 4αß1-mediated adhesion of Theileria-infected macrophages.


Subject(s)
Cyclooxygenase 2/biosynthesis , Dinoprostone/metabolism , Macrophages/parasitology , Receptors, Prostaglandin E, EP4 Subtype/biosynthesis , Theileria annulata/physiology , Transcription, Genetic , Transforming Growth Factor beta2/metabolism , Animals , Cell Adhesion , Gene Expression Regulation , Host-Pathogen Interactions
19.
PLoS Pathog ; 9(3): e1003213, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23505371

ABSTRACT

The basic organisation of the endomembrane system is conserved in all eukaryotes and comparative genome analyses provides compelling evidence that the endomembrane system of the last common eukaryotic ancestor (LCEA) is complex with many genes required for regulated traffic being present. Although apicomplexan parasites, causative agents of severe human and animal diseases, appear to have only a basic set of trafficking factors such as Rab-GTPases, they evolved unique secretory organelles (micronemes, rhoptries and dense granules) that are sequentially secreted during invasion of the host cell. In order to define the secretory pathway of apicomplexans, we performed an overexpression screen of Rabs in Toxoplasma gondii and identified Rab5A and Rab5C as important regulators of traffic to micronemes and rhoptries. Intriguingly, we found that not all microneme proteins traffic depends on functional Rab5A and Rab5C, indicating the existence of redundant microneme targeting pathways. Using two-colour super-resolution stimulated emission depletion (STED) we verified distinct localisations of independent microneme proteins and demonstrate that micronemal organelles are organised in distinct subsets or subcompartments. Our results suggest that apicomplexan parasites modify classical regulators of the endocytic system to carryout essential parasite-specific roles in the biogenesis of their unique secretory organelles.


Subject(s)
Protozoan Proteins/metabolism , Toxoplasma/metabolism , rab5 GTP-Binding Proteins/metabolism , Cell Line , Fibroblasts/parasitology , Humans , Organelles/metabolism , Protein Transport , Protozoan Proteins/genetics , Secretory Pathway
20.
Cell Microbiol ; 16(2): 269-79, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24112286

ABSTRACT

Theileria annulata infects predominantly macrophages, and to a lesser extent B cells, and causes a widespread disease of cattle called tropical theileriosis. Disease-causing infected macrophages are aggressively invasive, but this virulence trait can be attenuated by long-term culture. Attenuated macrophages are used as live vaccines against tropical theileriosis and via their characterization one gains insights into what host cell trait is altered concomitant with loss of virulence. We established that sporozoite infection of monocytes rapidly induces hif1-α transcription and that constitutive induction of HIF-1α in transformed leukocytes is parasite-dependent. In both infected macrophages and B cells induction of HIF-1α activates transcription of its target genes that drive host cells to perform Warburg-like glycolysis. We propose that Theileria-infected leukocytes maintain a HIF-1α-driven transcriptional programme typical of Warburg glycolysis in order to reduce as much as possible host cell H2 O2 type oxidative stress. However, in attenuated macrophages H2O2 production increases and HIF-1α levels consequently remained high, even though adhesion and aggressive invasiveness diminished. This indicates that Theileria infection generates a host leukocytes hypoxic response that if not properly controlled leads to loss of virulence.


Subject(s)
Hydrogen Peroxide/metabolism , Monocytes/immunology , Monocytes/parasitology , Oxidative Stress , Theileria annulata/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cattle , Cells, Cultured , Glycolysis , Monocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL