Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 63
1.
Biomolecules ; 11(9)2021 09 07.
Article En | MEDLINE | ID: mdl-34572535

Calcineurin (CaN) is present in all eukaryotic cells, including intracellular trypanosomatid parasites such as Trypanosoma cruzi (Tc) and Leishmania spp. (Lspp). In this study, we performed an in silico analysis of the CaN subunits, comparing them with the human (Hs) and looking their structure, post-translational mechanisms, subcellular distribution, interactors, and secretion potential. The differences in the structure of the domains suggest the existence of regulatory mechanisms and differential activity between these protozoa. Regulatory subunits are partially conserved, showing differences in their Ca2+-binding domains and myristoylation potential compared with human CaN. The subcellular distribution reveals that the catalytic subunits TcCaNA1, TcCaNA2, LsppCaNA1, LsppCaNA1_var, and LsppCaNA2 associate preferentially with the plasma membrane compared with the cytoplasmic location of HsCaNAα. For regulatory subunits, HsCaNB-1 and LsppCaNB associate preferentially with the nucleus and cytoplasm, and TcCaNB with chloroplast and cytoplasm. Calpain cleavage sites on CaNA suggest differential processing. CaNA and CaNB of these trypanosomatids have the potential to be secreted and could play a role in remote communication. Therefore, this background can be used to develop new drugs for protozoan pathogens that cause neglected disease.


Calcineurin/metabolism , Computer Simulation , Intracellular Space/parasitology , Leishmania/pathogenicity , Protozoan Proteins/metabolism , Trypanosoma cruzi/pathogenicity , Amino Acid Sequence , Calcineurin/chemistry , Calpain/metabolism , Conserved Sequence , Humans , Immunophilins/metabolism , Immunosuppressive Agents/pharmacology , Myristic Acid/metabolism , Phosphorylation , Protein Domains , Protein Subunits/metabolism , Protozoan Proteins/chemistry , Subcellular Fractions/metabolism
2.
Int J Mol Sci ; 22(12)2021 Jun 17.
Article En | MEDLINE | ID: mdl-34204285

Pathogenic intracellular bacteria, parasites and viruses have evolved sophisticated mechanisms to manipulate mammalian host cells to serve as niches for persistence and proliferation. The intracellular lifestyles of pathogens involve the manipulation of membrane-bound organellar compartments of host cells. In this review, we described how normal structural organization and cellular functions of endosomes, endoplasmic reticulum, Golgi apparatus, mitochondria, or lipid droplets are targeted by microbial virulence mechanisms. We focus on the specific interactions of Salmonella, Legionella pneumophila, Rickettsia rickettsii, Chlamydia spp. and Mycobacterium tuberculosis representing intracellular bacterial pathogens, and of Plasmodium spp. and Toxoplasma gondii representing intracellular parasites. The replication strategies of various viruses, i.e., Influenza A virus, Poliovirus, Brome mosaic virus, Epstein-Barr Virus, Hepatitis C virus, severe acute respiratory syndrome virus (SARS), Dengue virus, Zika virus, and others are presented with focus on the specific manipulation of the organelle compartments. We compare the specific features of intracellular lifestyle and replication cycles, and highlight the communalities in mechanisms of manipulation deployed.


Host-Pathogen Interactions , Organelles/metabolism , Animals , Biological Transport , Biomarkers , Energy Metabolism , Host-Parasite Interactions , Humans , Intracellular Space/metabolism , Intracellular Space/microbiology , Intracellular Space/parasitology , Intracellular Space/virology , Organelles/microbiology , Organelles/parasitology , Organelles/ultrastructure , Phagocytosis
3.
Front Immunol ; 12: 662944, 2021.
Article En | MEDLINE | ID: mdl-33959131

Extracellular vesicles (EVs) have garnered significant interest in recent years due to their contributions to cell-to-cell communication and disease processes. EVs are composed of a complex profile of bioactive molecules, which include lipids, nucleic acids, metabolites, and proteins. Although the biogenesis of EVs released by cells under various normal and abnormal conditions has been well-studied, there is incomplete knowledge about how infection influences EV biogenesis. EVs from infected cells contain specific molecules of both host and pathogen origin that may contribute to pathogenesis and the elicitation of the host immune response. Intracellular pathogens exhibit diverse lifestyles that undoubtedly dictate the mechanisms by which their molecules enter the cell's exosome biogenesis schemes. We will discuss the current understanding of the mechanisms used during infection to traffic molecules from their vacuolar niche to host EVs by selected intravacuolar pathogens. We initially review general exosome biogenesis schemes and then discuss what is known about EV biogenesis in Mycobacterium, Plasmodium, Toxoplasma, and Leishmania infections, which are pathogens that reside within membrane delimited compartments in phagocytes at some time in their life cycle within mammalian hosts. The review includes discussion of the need for further studies into the biogenesis of EVs to better understand the contributions of these vesicles to host-pathogen interactions, and to uncover potential therapeutic targets to control these pathogens.


Extracellular Vesicles/metabolism , Host-Pathogen Interactions/immunology , Virulence Factors/metabolism , Animals , Biological Transport , Cell Communication , Exosomes , Extracellular Vesicles/microbiology , Extracellular Vesicles/parasitology , Gene Expression Regulation , Host-Parasite Interactions , Host-Pathogen Interactions/genetics , Humans , Intracellular Space/immunology , Intracellular Space/metabolism , Intracellular Space/microbiology , Intracellular Space/parasitology , Protein Processing, Post-Translational , Signal Transduction
4.
Cell Calcium ; 94: 102337, 2021 03.
Article En | MEDLINE | ID: mdl-33524795

Toxoplasma gondii is an obligate intracellular parasite and replicates inside a parasitophorous vacuole (PV) within the host cell. The membrane of the PV (PVM) contains pores that permits for equilibration of ions and small molecules between the host cytosol and the PV lumen. Ca2+ signaling is universal and both T. gondii and its mammalian host cell utilize Ca2+ signals to stimulate diverse cellular functions. Egress of T. gondii from host cells is an essential step for the infection cycle of T. gondii, and a cytosolic Ca2+ increase initiates a Ca2+ signaling cascade that culminates in the stimulation of motility and egress. In this work we demonstrate that intracellular T. gondii tachyzoites are able to take up Ca2+ from the host cytoplasm during host cell signaling events. Both intracellular and extracellular Ca2+ sources are important in reaching a threshold of parasite cytosolic Ca2+ needed for successful egress. Two peaks of Ca2+ were observed in egressing single parasites with the second peak resulting from Ca2+ entry. We patched infected host cells to allow the delivery of precise concentrations of Ca2+ for the stimulation of motility and egress. Using this approach of patching infected host cells, allowed us to determine that increasing the host cytosolic Ca2+ to a specific concentration can trigger egress, which is further accelerated by diminishing the concentration of potassium (K+).


Calcium Signaling , Host-Pathogen Interactions , Potassium/metabolism , Toxoplasma/metabolism , Animals , Calcium/metabolism , Cell Membrane/metabolism , Cytosol/metabolism , HeLa Cells , Humans , Intracellular Space/parasitology , Models, Biological , Parasites/metabolism
5.
Chem Biol Interact ; 332: 109296, 2020 Dec 01.
Article En | MEDLINE | ID: mdl-33096056

Leishmaniasis is a parasitic neglected tropical disease and result in a broad spectrum of clinical manifestations, ranging from a single ulceration to a progressive and fatal visceral disease. Comprising a limited and highly toxic therapeutic arsenal, new treatments are urgently needed. Targeting delivery of drugs has been a promising approach for visceral leishmaniasis (VL). Phosphatidylserine-liposomes have demonstrated superior efficacy in VL, targeting intracellular parasites in host cells through macrophage scavenger receptors. In this work, we investigated the in vitro and in vivo efficacy of the antihelminthic drug nitazoxanide in a nanoliposomal formulation against Leishmania (L.) infantum. Physicochemical parameters of liposomes containing nitazoxanide (NTZ-LP) were determined by dynamic light scattering and small angle X-ray scattering. The efficacy of the formulation was verified in an intracellular amastigote model and in an experimental hamster model. Our findings showed that NTZ-LP was able to eliminate the amastigotes inside the host cell with an IC50 value of 16 µM. NTZ-LP was labelled a fluorescent probe and by spectrofluorimetry, we observed that the infected macrophages internalized similar levels of the drug to the uninfected cells. The confocal microscopy images confirmed the uptake and demonstrated a diffuse distribution of the NTZ-LP in the cytoplasm of Leishmania-infected macrophages, with the vesicles in a closer proximity to the parasites. For the in vivo efficacy, the liposomal NTZ-LP was administrated intraperitoneally to Leishmania-infected hamsters for 10 consecutive days at 2 mg/kg/day. By qPCR we demonstrated a reduction of the parasite burden by 82% and 50% in the liver (p < 0.05) and spleen (p < 0.05), respectively. NTZ (non-liposomal) was administered at 100 mg/kg/day per oral (p.o.) for the same period, but demonstrated no efficacy. This liposomal formulation ensured a targeting delivery of NTZ to the intracellular parasites, resulting in an good efficacy at a low dose in animals, and it may represent a new candidate therapy for VL.


Intracellular Space/parasitology , Leishmania infantum/drug effects , Liposomes/chemistry , Nanoparticles/chemistry , Phosphatidylserines/metabolism , Thiazoles/pharmacology , Animals , Antiprotozoal Agents/pharmacology , Dynamic Light Scattering , Female , Inhibitory Concentration 50 , Macrophages/drug effects , Macrophages/parasitology , Male , Mesocricetus , Mice, Inbred BALB C , Nitro Compounds , Scattering, Small Angle , Static Electricity , X-Ray Diffraction
6.
Chem Biol Interact ; 330: 109165, 2020 Oct 01.
Article En | MEDLINE | ID: mdl-32771326

The effect of N-geranyl-ethane-1,2-diamine dihydochloride (GIB24), a synthetic diamine, was assayed against different developmental forms of the parasitic protozoan Trypanosoma cruzi (strain Dm28c). The compound was effective against culture epimastigote forms (IC50/24h = 5.64 µM; SI = 16.4) and intracellular amastigotes (IC50/24h = 12.89 µM; SI = 7.18), as detected by the MTT methodology and by cell counting, respectively. Incubation of epimastigotes for 6h with 6 µM GIB24 (IC50/24h value) resulted in significant dissipation of the mitochondrial membrane potential, prior to permeabilization of the plasma membrane. Rounded epimastigotes with cell size reduction were observed by scanning electron microscopy. These morpho-physiological changes induced by GIB24 suggest an incidental death process. Treatment of infected Vero cells did not prevent the intracellular amastigotes from completing the intracellular cycle. However, there was a decrease in the number of released parasites, increasing the ratio amastigotes/trypomastigotes. Proteomic analysis of 15 µM GIB24 resistant epimastigotes indicated that the compound acts mainly on mitochondrial components involved in the Krebs cycle and in maintaining the oxidative homeostasis of the parasites. Our data suggest that GIB24 is active against the main morphological forms of T. cruzi.


Diamines/pharmacology , Drug Resistance , Intracellular Space/drug effects , Proteomics , Terpenes/chemistry , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/growth & development , Animals , Chlorocebus aethiops , Diamines/chemistry , Intracellular Space/parasitology , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/metabolism , Vero Cells
7.
J Am Soc Mass Spectrom ; 31(9): 1815-1824, 2020 Sep 02.
Article En | MEDLINE | ID: mdl-32830963

The obligate intracellular apicomplexan parasites Toxoplasma gondii and Besnoitia besnoiti are important causes of disease in both humans and cattle. To date, effective specific treatments are lacking for both infections. To counteract severe symptoms leading to, e.g., disabilities and even abortion in the case of human toxoplasmosis and bovine besnoitiosis, novel targets are required for development of drugs and vaccines. A promising emerging technique for molecular characterization of organisms is high-resolution atmospheric-pressure scanning microprobe matrix-assisted laser desorption/ionization (AP-SMALDI) mass spectrometry imaging (MSI) which enables semiquantitative visualization of metabolite distributions. MSI was here used to trace and characterize lipid metabolites in primary bovine umbilical vein endothelial cells (BUVECs) upon infection with tachyzoites, an early and pathogenic fast-replicating life stage of T. gondii and B. besnoiti. A cell bulk, derived from noninfected controls and parasite-infected cell pellets, was analyzed by AP-SMALDI MSI in technical and biological triplicates. Multivariate statistical analysis including hierarchical clustering and principle component analysis revealed infection-specific metabolites in both positive- and negative-ion mode, identified by combining database search and LC-MS2 experiments. MSI analyses of host cell monolayers were conducted at 5 µm lateral resolution, allowing single apicomplexan-infected cells to be allocated. This is the first mass spectrometry imaging study on intracellular T. gondii and B. besnoiti infections and the first detailed metabolomic characterization of B. besnoiti tachyzoites. MSI was used here as an efficient tool to discriminate infected from noninfected cells at the single-cell level in vitro.


Coccidiosis , Intracellular Space/parasitology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Toxoplasmosis, Animal , Animals , Cattle , Cells, Cultured , Coccidiosis/diagnostic imaging , Coccidiosis/parasitology , Endothelial Cells/cytology , Endothelial Cells/parasitology , Molecular Imaging , Sarcocystidae/pathogenicity , Single-Cell Analysis , Toxoplasma/pathogenicity , Toxoplasmosis, Animal/diagnostic imaging , Toxoplasmosis, Animal/parasitology
8.
Cell Rep ; 30(11): 3778-3792.e9, 2020 03 17.
Article En | MEDLINE | ID: mdl-32187549

Apicomplexan parasites are unicellular eukaryotic pathogens that must obtain and combine lipids from both host cell scavenging and de novo synthesis to maintain parasite propagation and survival within their human host. Major questions on the role and regulation of each lipid source upon fluctuating host nutritional conditions remain unanswered. Characterization of an apicoplast acyltransferase, TgATS2, shows that the apicoplast provides (lyso)phosphatidic acid, required for the recruitment of a critical dynamin (TgDrpC) during parasite cytokinesis. Disruption of TgATS2 also leads parasites to shift metabolic lipid acquisition from de novo synthesis toward host scavenging. We show that both lipid scavenging and de novo synthesis pathways in wild-type parasites exhibit major metabolic and cellular plasticity upon sensing host lipid-deprived environments through concomitant (1) upregulation of de novo fatty acid synthesis capacities in the apicoplast and (2) parasite-driven host remodeling to generate multi-membrane-bound structures from host organelles that are imported toward the parasite.


Adaptation, Physiological , Apicoplasts/metabolism , Cell Division , Host-Parasite Interactions , Lipid Metabolism , Parasites/metabolism , Toxoplasma/metabolism , Toxoplasma/physiology , Acyltransferases/metabolism , Animals , Cell Membrane/metabolism , Cytokinesis , Fatty Acid Synthases/metabolism , Fatty Acids/biosynthesis , Gene Deletion , Humans , Intracellular Space/parasitology , Life Cycle Stages , Lipidomics , Male , Models, Biological , Multivesicular Bodies/metabolism , Multivesicular Bodies/ultrastructure , Mutation/genetics , Nutrients , Parasites/growth & development , Parasites/physiology , Parasites/ultrastructure , Protozoan Proteins/metabolism , Toxoplasma/growth & development , Toxoplasma/ultrastructure
9.
Molecules ; 24(17)2019 Aug 30.
Article En | MEDLINE | ID: mdl-31480402

The current chemotherapy of Chagas disease needs to be urgently improved. With this aim, a series of 16 hybrids of Cinchona alkaloids and bile acids were prepared by functionalization at position C-2 of the quinoline nucleus by a radical attack of a norcholane substituent via a Barton-Zard decarboxylation reaction. The antitrypanosomal activity of the hybrids was tested on different stages and strains of T. cruzi. In particular, eight out of 16 hybrids presented an IC50 ≤1 µg/mL against trypomastigotes of the CL Brener strain and/or a selectivity index higher than 10. These promising hybrids yielded similar results when tested on trypomastigotes from the RA strain of T. cruzi (discrete typing unit-DTU-VI). Surprisingly, trypomastigotes of the Y strain (DTU II) were more resistant to benznidazole and to most of the hybrids than those of the CL Brener and RA strains. However, the peracetylated and non-acetylated forms of the cinchonine/chenodeoxycholic bile acid conjugate 4f and 5f were the most trypanocidal hybrids against Y strain trypomastigotes, with IC50 values of 0.5 and 0.65 µg/mL, respectively. More importantly, promising results were observed in invasion assays using the Y strain, where hybrids 5f and 4f induced a significant reduction in intracellular amastigotes and on the release of trypomastigotes from infected cells.


Antiparasitic Agents/pharmacology , Bile Acids and Salts/pharmacology , Cinchona Alkaloids/pharmacology , Trypanosoma cruzi/drug effects , Animals , Chlorocebus aethiops , Inhibitory Concentration 50 , Intracellular Space/parasitology , Rats , Vero Cells
10.
PLoS Biol ; 17(7): e3000376, 2019 07.
Article En | MEDLINE | ID: mdl-31318858

Apicomplexan parasites possess a plastid organelle called the apicoplast. Inhibitors that selectively target apicoplast housekeeping functions, including DNA replication and protein translation, are lethal for the parasite, and several (doxycycline, clindamycin, and azithromycin) are in clinical use as antimalarials. A major limitation of such drugs is that treated parasites only arrest one intraerythrocytic development cycle (approximately 48 hours) after treatment commences, a phenotype known as the 'delayed death' effect. The molecular basis of delayed death is a long-standing mystery in parasitology, and establishing the mechanism would aid rational clinical implementation of apicoplast-targeted drugs. Parasites undergoing delayed death transmit defective apicoplasts to their daughter cells and cannot produce the sole, blood-stage essential metabolic product of the apicoplast: the isoprenoid precursor isopentenyl-pyrophosphate. How the isoprenoid precursor depletion kills the parasite remains unknown. We investigated the requirements for the range of isoprenoids in the human malaria parasite Plasmodium falciparum and characterised the molecular and morphological phenotype of parasites experiencing delayed death. Metabolomic profiling reveals disruption of digestive vacuole function in the absence of apicoplast derived isoprenoids. Three-dimensional electron microscopy reveals digestive vacuole fragmentation and the accumulation of cytostomal invaginations, characteristics common in digestive vacuole disruption. We show that digestive vacuole disruption results from a defect in the trafficking of vesicles to the digestive vacuole. The loss of prenylation of vesicular trafficking proteins abrogates their membrane attachment and function and prevents the parasite from feeding. Our data show that the proximate cause of delayed death is an interruption of protein prenylation and consequent cellular trafficking defects.


Apicoplasts/metabolism , Intracellular Space/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Animals , Antimalarials/pharmacology , Cell Death/drug effects , Hemiterpenes/metabolism , Hemiterpenes/pharmacology , Humans , Intracellular Space/drug effects , Intracellular Space/parasitology , Malaria, Falciparum/parasitology , Metabolomics/methods , Organophosphorus Compounds/metabolism , Organophosphorus Compounds/pharmacology , Plasmodium falciparum/drug effects , Plasmodium falciparum/physiology , Protein Prenylation/drug effects , Protein Transport/drug effects , Vacuoles/drug effects , Vacuoles/metabolism , Vacuoles/parasitology
11.
J Cell Sci ; 132(13)2019 07 01.
Article En | MEDLINE | ID: mdl-31182647

Centrins are EF-hand containing proteins ubiquitously found in eukaryotes and are key components of centrioles/basal bodies as well as certain contractile fibers. We previously identified three centrins in the human parasite Toxoplasma gondii, all of which localized to the centrioles. However, one of them, T. gondii (Tg) Centrin2 (CEN2), is also targeted to structures at the apical and basal ends of the parasite, as well as to annuli at the base of the apical cap of the membrane cortex. The role(s) that CEN2 play in these locations were unknown. Here, we report the functional characterization of CEN2 using a conditional knockdown method that combines transcriptional and protein stability control. The knockdown resulted in an ordered loss of CEN2 from its four compartments, due to differences in incorporation kinetics and structural inheritance over successive generations. This was correlated with a major invasion deficiency at early stages of CEN2 knockdown, and replication defects at later stages. These results indicate that CEN2 is incorporated into multiple cytoskeletal structures to serve distinct functions that are required for parasite survival.


Intracellular Space/parasitology , Parasites/growth & development , Parasites/metabolism , Protozoan Proteins/metabolism , Toxoplasma/growth & development , Toxoplasma/metabolism , Amino Acid Sequence , Animals , Down-Regulation , Green Fluorescent Proteins/metabolism , Humans , Life Cycle Stages , Male , Mutation/genetics , Phylogeny , Protein Biosynthesis , Protozoan Proteins/chemistry , Transcription, Genetic
12.
Curr Opin Immunol ; 60: 111-116, 2019 10.
Article En | MEDLINE | ID: mdl-31229914

Hypoxia-inducible transcription factor-1α (HIF-1α) was originally identified as a master regulator of cellular responses to hypoxia. More recently, HIF-1α has emerged as a critical regulator of immune cell function that couples shifts in cellular metabolism to cell type-specific transcriptional outputs. Activation of macrophages with inflammatory stimuli leads to induction of the metabolic program aerobic glycolysis and to HIF-1α stabilization, which reinforce one another in a positive feedback loop that helps drive macrophage activation. This activation of aerobic glycolysis and HIF-1α is important both for production of inflammatory cytokines, such as IL-1ß, and for cell intrinsic control of infection. Here, we review the importance of HIF-1α for control of bacterial, fungal, and protozoan intracellular pathogens, highlighting recent findings that reveal mechanisms by which HIF-1α is activated during infection and how HIF-1α coordinates antimicrobial responses of macrophages.


Biomarkers , Disease Resistance , Host-Pathogen Interactions , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Animals , Disease Resistance/genetics , Disease Resistance/immunology , Energy Metabolism , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunity, Innate , Intracellular Space/immunology , Intracellular Space/metabolism , Intracellular Space/microbiology , Intracellular Space/parasitology , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Macrophages/parasitology , Phagocytosis/immunology , Signal Transduction , Transcription Factors/metabolism
13.
Int J Biol Macromol ; 121: 498-507, 2019 Jan.
Article En | MEDLINE | ID: mdl-30316767

Trypanothione based redox metabolism is unique to the Trypanosomatida family. Despite extensive studies on redox metabolism of Leishmania parasites, a prominent question of why Leishmania adopt this unique redox pathway remains elusive. We have episomally expressed human glutathione reductase (HuGR) in Leishmania donovani (LdGR+) and investigated its effect. LdGR+ strain has slower growth compared to the wild type (Ld) indicating decreased survival ability of the strain. Further, LdGR+ strain showed enhanced accumulation of intracellular reactive oxygen species (ROS) and more sensitivity to the anti-leishmanial drug, Miltefosine, inferring increased stress level. In contrast, the expression analyses of genes specific to redox metabolism were increased significantly in LdGR+ strain compared to wild type. Lower infectivity index of the LdGR+ strain substantiated the above findings and indicated that the expression of HuGR reduces the stress tolerance ability of the parasite. From molecular docking studies with HuGR, it was observed that oxidized trypanothione (TS2) binds much better than oxidized glutathione (GS2). These results also give us hints that the parasite is losing infectivity potential due to an overall increase in intracellular stress caused with the expression of HuGR, showcasing a possible role of evolutionary pressure on the Leishmania parasites posed by HuGR.


Evolution, Molecular , Gene Expression Regulation, Enzymologic , Glutathione Reductase/metabolism , Leishmania donovani/metabolism , Plasmids/metabolism , Stress, Physiological , Cell Line , Humans , Intracellular Space/metabolism , Intracellular Space/parasitology , Leishmania donovani/enzymology , Leishmania donovani/physiology , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/metabolism
14.
Int J Mol Sci ; 19(12)2018 Dec 19.
Article En | MEDLINE | ID: mdl-30572624

Microsporidia are fungi-like parasites that have the smallest known eukaryotic genome, and for that reason they are used as a model to study the phenomenon of genome decay in parasitic forms of life. Similar to other intracellular parasites that reproduce asexually in an environment with alleviated natural selection, Microsporidia experience continuous genome decay that is driven by Muller's ratchet-an evolutionary process of irreversible accumulation of deleterious mutations that lead to gene loss and the miniaturization of cellular components. Particularly, Microsporidia have remarkably small ribosomes in which the rRNA is reduced to the minimal enzymatic core. In this study, we analyzed microsporidian ribosomes to study an apparent impact of Muller's ratchet on structure of RNA and protein molecules in parasitic forms of life. Through mass spectrometry of microsporidian proteome and analysis of microsporidian genomes, we found that massive rRNA reduction in microsporidian ribosomes appears to annihilate the binding sites for ribosomal proteins eL8, eL27, and eS31, suggesting that these proteins are no longer bound to the ribosome in microsporidian species. We then provided an evidence that protein eS31 is retained in Microsporidia due to its non-ribosomal function in ubiquitin biogenesis. Our study illustrates that, while Microsporidia carry the same set of ribosomal proteins as non-parasitic eukaryotes, some ribosomal proteins are no longer participating in protein synthesis in Microsporidia and they are preserved from genome decay by having extra-ribosomal functions. More generally, our study shows that many components of parasitic cells, which are identified by automated annotation of pathogenic genomes, may lack part of their biological functions due to continuous genome decay.


Intracellular Space/parasitology , Microsporidia/metabolism , Parasites/metabolism , Ribosomes/metabolism , Amino Acid Sequence , Animals , Binding Sites , Biological Evolution , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , RNA, Ribosomal/metabolism , Ribosomal Proteins/metabolism
15.
Parasit Vectors ; 11(1): 521, 2018 Sep 20.
Article En | MEDLINE | ID: mdl-30236162

BACKGROUND: Trypanosoma cruzi invades and replicates inside mammalian cells, which can lead to chronic Chagas disease in humans. Trypanosoma copemani infects Australian marsupials and recent investigations indicate it may be able to invade mammalian cells in vitro, similar to T. cruzi. Here, T. cruzi 10R26 strain (TcIIa) and two strains of T. copemani [genotype 1 (G1) and genotype 2 (G2)] were incubated with marsupial cells in vitro. Live-cell time-lapse and fluorescent microscopy, combined with high-resolution microscopy (transmission and scanning electron microscopy) were used to investigate surface interactions between parasites and mammalian cells. RESULTS: The number of parasites invading cells was significantly higher in T. cruzi compared to either genotype of T. copemani, between which there was no significant difference. While capable of cellular invasion, T. copemani did not multiply in host cells in vitro as there was no increase in intracellular amastigotes over time and no release of new trypomastigotes from host cells, as observed in T. cruzi. Exposure of host cells to G2 trypomastigotes resulted in increased host cell membrane permeability within 24 h of infection, and host cell death/blebbing was also observed. G2 parasites also became embedded in the host cell membrane. CONCLUSIONS: Trypanosoma copemani is unlikely to have an obligate intracellular life-cycle like T. cruzi. However, T. copemani adversely affects cell health in vitro and should be investigated in vivo in infected host tissues to better understand this host-parasite relationship. Future research should focus on increasing understanding of the T. copemani life history and the genetic, physiological and ecological differences between different genotypes.


Host-Parasite Interactions , Trypanosoma/physiology , Trypanosomiasis/parasitology , Animals , Australia , Cell Death , Chagas Disease/parasitology , Genotype , Humans , Intracellular Space/parasitology , Life Cycle Stages , Marsupialia , Microscopy, Electron, Scanning/veterinary , Microscopy, Electron, Transmission/veterinary , Species Specificity , Time-Lapse Imaging/veterinary , Trypanosoma/genetics , Trypanosoma/growth & development , Trypanosoma/ultrastructure , Trypanosoma cruzi/genetics , Trypanosoma cruzi/growth & development , Trypanosoma cruzi/physiology , Trypanosoma cruzi/ultrastructure
16.
Cell Microbiol ; 20(10): e12867, 2018 10.
Article En | MEDLINE | ID: mdl-29895095

Valosin-containing protein (VCP)/p97/Cdc48 is one of the best-characterised type II cytosolic AAA+ ATPases most known for their role in ubiquitin-dependent protein quality control. Here, we provide functional insights into the role of the Leishmania VCP/p97 homologue (LiVCP) in the parasite intracellular development. We demonstrate that although LiVCP is an essential gene, Leishmania infantum promastigotes can grow with less VCP. In contrast, growth of axenic and intracellular amastigotes is dramatically affected upon decreased LiVCP levels in heterozygous and temperature sensitive (ts) LiVCP mutants or the expression of dominant negative mutants known to specifically target the second conserved VCP ATPase domain, a major contributor of the VCP overall ATPase activity. Interestingly, these VCP mutants are also unable to survive heat stress, and a ts VCP mutant is defective in amastigote growth. Consistent with LiVCP's essential function in amastigotes, LiVCP messenger ribonucleic acid undergoes 3'Untranslated Region (UTR)-mediated developmental regulation, resulting in higher VCP expression in amastigotes. Furthermore, we show that parasite mutant lines expressing lower VCP levels or dominant negative VCP forms exhibit high accumulation of polyubiquitinated proteins and increased sensitivity to proteotoxic stress, supporting the ubiquitin-selective chaperone function of LiVCP. Together, these results emphasise the crucial role LiVCP plays under heat stress and during the parasite intracellular development.


Intracellular Space/parasitology , Leishmania infantum/growth & development , Valosin Containing Protein/metabolism , 3' Untranslated Regions/genetics , Base Sequence/genetics , Endoplasmic Reticulum/metabolism , Gene Expression Regulation/genetics , Germ-Free Life/physiology , Heat-Shock Response/physiology , Leishmania infantum/genetics , Molecular Chaperones/metabolism , Protein Domains/genetics , RNA, Messenger/genetics , Ubiquitin/metabolism , Ubiquitination , Valosin Containing Protein/genetics
17.
Biophys J ; 114(7): 1695-1706, 2018 04 10.
Article En | MEDLINE | ID: mdl-29642038

Severe malaria is primarily caused by Plasmodium falciparum parasites during their asexual reproduction cycle within red blood cells. One of the least understood stages in this cycle is the brief preinvasion period during which merozoite-red cell contacts lead to apical alignment of the merozoite in readiness for penetration, a stage of major relevance in the control of invasion efficiency. Red blood cell deformations associated with this process were suggested to be active plasma membrane responses mediated by transients of elevated intracellular calcium. Few studies have addressed this hypothesis because of technical challenges, and the results remained inconclusive. Here, Fluo-4 was used as a fluorescent calcium indicator with optimized protocols to investigate the distribution of the dye in red blood cell populations used as P. falciparum invasion targets in egress-invasion assays. Preinvasion dynamics was observed simultaneously under bright-field and fluorescence microscopy by recording egress-invasion events. All the egress-invasion sequences showed red blood cell deformations of varied intensities during the preinvasion period and the echinocytic changes that follow during invasion. Intraerythrocytic calcium signals were absent throughout this interval in over half the records and totally absent during the preinvasion period, regardless of deformation strength. When present, calcium signals were of a punctate modality, initiated within merozoites already poised for invasion. These results argue against a role of elevated intracellular calcium during the preinvasion stage. We suggest an alternative mechanism of merozoite-induced preinvasion deformations based on passive red cell responses to transient agonist-receptor interactions associated with the formation of adhesive coat filaments.


Calcium/metabolism , Intracellular Space/metabolism , Intracellular Space/parasitology , Plasmodium falciparum/physiology , Aniline Compounds/metabolism , Erythrocytes/cytology , Erythrocytes/parasitology , Formaldehyde/pharmacology , Humans , Plasmodium falciparum/drug effects , Pyruvic Acid/pharmacology , Xanthenes/metabolism
18.
mSphere ; 3(2)2018 04 25.
Article En | MEDLINE | ID: mdl-29669882

Metabolomics is increasingly popular for the study of pathogens. For the malaria parasite Plasmodium falciparum, both targeted and untargeted metabolomics have improved our understanding of pathogenesis, host-parasite interactions, and antimalarial drug treatment and resistance. However, purification and analysis procedures for performing metabolomics on intracellular pathogens have not been explored. Here, we purified in vitro-grown ring-stage intraerythrocytic P. falciparum parasites for untargeted metabolomics studies; the small size of this developmental stage amplifies the challenges associated with metabolomics studies as the ratio between host and parasite biomass is maximized. Following metabolite identification and data preprocessing, we explored multiple confounding factors that influence data interpretation, including host contamination and normalization approaches (including double-stranded DNA, total protein, and parasite numbers). We conclude that normalization parameters have large effects on differential abundance analysis and recommend the thoughtful selection of these parameters. However, normalization does not remove the contribution from the parasite's extracellular environment (culture media and host erythrocyte). In fact, we found that extraparasite material is as influential on the metabolome as treatment with a potent antimalarial drug with known metabolic effects (artemisinin). Because of this influence, we could not detect significant changes associated with drug treatment. Instead, we identified metabolites predictive of host and medium contamination that could be used to assess sample purification. Our analysis provides the first quantitative exploration of the effects of these factors on metabolomics data analysis; these findings provide a basis for development of improved experimental and analytical methods for future metabolomics studies of intracellular organisms.IMPORTANCE Molecular characterization of pathogens such as the malaria parasite can lead to improved biological understanding and novel treatment strategies. However, the distinctive biology of the Plasmodium parasite, including its repetitive genome and the requirement for growth within a host cell, hinders progress toward these goals. Untargeted metabolomics is a promising approach to learn about pathogen biology. By measuring many small molecules in the parasite at once, we gain a better understanding of important pathways that contribute to the parasite's response to perturbations such as drug treatment. Although increasingly popular, approaches for intracellular parasite metabolomics and subsequent analysis are not well explored. The findings presented in this report emphasize the critical need for improvements in these areas to limit misinterpretation due to host metabolites and to standardize biological interpretation. Such improvements will aid both basic biological investigations and clinical efforts to understand important pathogens.


Erythrocytes/parasitology , Intracellular Space/parasitology , Metabolome , Plasmodium falciparum/metabolism , Animals , Antimalarials/pharmacology , Artemisinins/pharmacology , Culture Media/chemistry , Genome, Protozoan , Host-Parasite Interactions , Malaria, Falciparum/metabolism , Mass Spectrometry , Metabolomics , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
19.
Semin Immunol ; 38: 15-23, 2018 08.
Article En | MEDLINE | ID: mdl-29551246

Interleukin 32 (IL-32) is an intracellular cytokine produced by immune and non immune cells after different stimuli. It contributes to inflammation and control of intracellular pathogens mainly by inducing proinflammatory cytokines and microbicidal molecules. Evidence is rising showing that IL-32 can be considered an endogenous danger signal after tissue injury, amplifying the inflammatory process and acquired immune responses. It seems to be a master regulator of intracellular infectious diseases. In this review, first the general properties of IL-32 are described followed by its role in the immunopathogenesis of inflammatory and infectious diseases. Roles of IL-32 in the control of infectious diseases caused by intracellular pathogens are reported, and later a focus on IL-32 in leishmaniases, diseases caused by an intracellular protozoan, is presented.


Inflammation Mediators/immunology , Interleukins/immunology , Intracellular Space/immunology , Leishmania/immunology , Leishmaniasis/immunology , Signal Transduction/immunology , Animals , Cytokines/immunology , Cytokines/metabolism , Host-Parasite Interactions/immunology , Humans , Inflammation Mediators/metabolism , Interleukins/metabolism , Intracellular Space/parasitology , Leishmania/physiology , Leishmaniasis/metabolism , Leishmaniasis/parasitology
20.
Eur J Med Chem ; 143: 1312-1324, 2018 Jan 01.
Article En | MEDLINE | ID: mdl-29126735

Screening of a designed collection of mono-substituted amino-1H-phenalen-1-ones against promastigote forms of L. donovani and L. amazonensis, identified seven compounds with anti-leishmanial activities comparable or better than the commonly prescribed anti-leishmanial drug, miltefosine. Structure-activity analysis revealed that appendages containing a basic tertiary nitrogen were favored, and that the position of the appendage also affected their potency. Like miltefosine, several of these active compounds significantly reduced the mitochondrial membrane potential in promastigotes. Further studies in amastigotes of L. amazonensis revealed that compounds 14, 15 and 33 were more active and more selective than miltefosine, with sub-micromolar potencies and selectivity indices >100.


Antiprotozoal Agents/chemical synthesis , Antiprotozoal Agents/pharmacology , Drug Design , Leishmania donovani/drug effects , Phenalenes/chemical synthesis , Phenalenes/pharmacology , Antiprotozoal Agents/chemistry , Chemistry Techniques, Synthetic , Intracellular Space/drug effects , Intracellular Space/parasitology , Leishmania donovani/physiology , Membrane Potential, Mitochondrial/drug effects , Phenalenes/chemistry
...