Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 221
Filter
Add more filters

Publication year range
1.
Immunity ; 51(2): 285-297.e5, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31272808

ABSTRACT

Interactions with the microbiota influence many aspects of immunity, including immune cell development, differentiation, and function. Here, we examined the impact of the microbiota on CD8+ T cell memory. Antigen-activated CD8+ T cells transferred into germ-free mice failed to transition into long-lived memory cells and had transcriptional impairments in core genes associated with oxidative metabolism. The microbiota-derived short-chain fatty acid (SCFA) butyrate promoted cellular metabolism, enhanced memory potential of activated CD8+ T cells, and SCFAs were required for optimal recall responses upon antigen re-encounter. Mechanistic experiments revealed that butyrate uncoupled the tricarboxylic acid cycle from glycolytic input in CD8+ T cells, which allowed preferential fueling of oxidative phosphorylation through sustained glutamine utilization and fatty acid catabolism. Our findings reveal a role for the microbiota in promoting CD8+ T cell long-term survival as memory cells and suggest that microbial metabolites guide the metabolic rewiring of activated CD8+ T cells to enable this transition.


Subject(s)
Butyrates/metabolism , CD8-Positive T-Lymphocytes/immunology , Fatty Acids, Volatile/metabolism , Immunologic Memory , Microbiota/immunology , Adoptive Transfer , Animals , Antigens/immunology , Cell Differentiation , Cells, Cultured , Glycolysis , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction
2.
PLoS Pathog ; 20(9): e1012484, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39241090

ABSTRACT

Glycophosphatidylinositol (GPI) anchors are the predominant glycoconjugate in Plasmodium parasites, enabling modified proteins to associate with biological membranes. GPI biosynthesis commences with donation of a mannose residue held by dolichol-phosphate at the endoplasmic reticulum membrane. In Plasmodium dolichols are derived from isoprenoid precursors synthesised in the Plasmodium apicoplast, a relict plastid organelle of prokaryotic origin. We found that treatment of Plasmodium parasites with apicoplast inhibitors decreases the synthesis of isoprenoid and GPI intermediates resulting in GPI-anchored proteins becoming untethered from their normal membrane association. Even when other isoprenoids were chemically rescued, GPI depletion led to an arrest in schizont stage parasites, which had defects in segmentation and egress. In those daughter parasites (merozoites) that did form, proteins that would normally be GPI-anchored were mislocalised, and when these merozoites were artificially released they were able to attach to but not invade new red blood cells. Our data provides further evidence for the importance of GPI biosynthesis during the asexual cycle of P. falciparum, and indicates that GPI biosynthesis, and by extension egress and invasion, is dependent on isoprenoids synthesised in the apicoplast.


Subject(s)
Apicoplasts , Glycosylphosphatidylinositols , Plasmodium falciparum , Terpenes , Plasmodium falciparum/metabolism , Apicoplasts/metabolism , Glycosylphosphatidylinositols/metabolism , Glycosylphosphatidylinositols/biosynthesis , Terpenes/metabolism , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Erythrocytes/parasitology , Erythrocytes/metabolism , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/metabolism , Animals , Merozoites/metabolism
3.
PLoS Pathog ; 20(8): e1012440, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39207937

ABSTRACT

Reconstructing the evolutionary origins of Mycobacterium tuberculosis, the causative agent of human tuberculosis, has helped identify bacterial factors that have led to the tubercle bacillus becoming such a formidable human pathogen. Here we report the discovery and detailed characterization of an exceedingly slow growing mycobacterium that is closely related to M. tuberculosis for which we have proposed the species name Mycobacterium spongiae sp. nov., (strain ID: FSD4b-SM). The bacterium was isolated from a marine sponge, taken from the waters of the Great Barrier Reef in Queensland, Australia. Comparative genomics revealed that, after the opportunistic human pathogen Mycobacterium decipiens, M. spongiae is the most closely related species to the M. tuberculosis complex reported to date, with 80% shared average nucleotide identity and extensive conservation of key M. tuberculosis virulence factors, including intact ESX secretion systems and associated effectors. Proteomic and lipidomic analyses showed that these conserved systems are functional in FSD4b-SM, but that it also produces cell wall lipids not previously reported in mycobacteria. We investigated the virulence potential of FSD4b-SM in mice and found that, while the bacteria persist in lungs for 56 days after intranasal infection, no overt pathology was detected. The similarities with M. tuberculosis, together with its lack of virulence, motivated us to investigate the potential of FSD4b-SM as a vaccine strain and as a genetic donor of the ESX-1 genetic locus to improve BCG immunogenicity. However, neither of these approaches resulted in superior protection against M. tuberculosis challenge compared to BCG vaccination alone. The discovery of M. spongiae adds to our understanding of the emergence of the M. tuberculosis complex and it will be another useful resource to refine our understanding of the factors that shaped the evolution and pathogenesis of M. tuberculosis.


Subject(s)
Porifera , Animals , Mice , Virulence , Porifera/microbiology , Mycobacterium tuberculosis/pathogenicity , Mycobacterium tuberculosis/genetics , Tuberculosis/microbiology , Virulence Factors/genetics , Female , Biological Evolution , Humans , Phylogeny , Mycobacterium/pathogenicity , Mycobacterium/genetics
4.
PLoS Pathog ; 19(7): e1011006, 2023 07.
Article in English | MEDLINE | ID: mdl-37523385

ABSTRACT

A key element of Plasmodium biology and pathogenesis is the trafficking of ~10% of the parasite proteome into the host red blood cell (RBC) it infects. To cross the parasite-encasing parasitophorous vacuole membrane, exported proteins utilise a channel-forming protein complex termed the Plasmodium translocon of exported proteins (PTEX). PTEX is obligatory for parasite survival, both in vitro and in vivo, suggesting that at least some exported proteins have essential metabolic functions. However, to date only one essential PTEX-dependent process, the new permeability pathways, has been described. To identify other essential PTEX-dependant proteins/processes, we conditionally knocked down the expression of one of its core components, PTEX150, and examined which pathways were affected. Surprisingly, the food vacuole mediated process of haemoglobin (Hb) digestion was substantially perturbed by PTEX150 knockdown. Using a range of transgenic parasite lines and approaches, we show that two major Hb proteases; falcipain 2a and plasmepsin II, interact with PTEX core components, implicating the translocon in the trafficking of Hb proteases. We propose a model where these proteases are translocated into the PV via PTEX in order to reach the cytostome, located at the parasite periphery, prior to food vacuole entry. This work offers a second mechanistic explanation for why PTEX function is essential for growth of the parasite within its host RBC.


Subject(s)
Parasites , Plasmodium falciparum , Animals , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Vacuoles/metabolism , Protein Transport , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Erythrocytes/parasitology , Parasites/metabolism , Peptide Hydrolases/metabolism
5.
Mol Cell ; 67(3): 457-470.e5, 2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28712726

ABSTRACT

Acylglycerol kinase (AGK) is a mitochondrial lipid kinase that catalyzes the phosphorylation of monoacylglycerol and diacylglycerol to lysophosphatidic acid and phosphatidic acid, respectively. Mutations in AGK cause Sengers syndrome, which is characterized by congenital cataracts, hypertrophic cardiomyopathy, skeletal myopathy, exercise intolerance, and lactic acidosis. Here we identified AGK as a subunit of the mitochondrial TIM22 protein import complex. We show that AGK functions in a kinase-independent manner to maintain the integrity of the TIM22 complex, where it facilitates the import and assembly of mitochondrial carrier proteins. Mitochondria isolated from Sengers syndrome patient cells and tissues show a destabilized TIM22 complex and defects in the biogenesis of carrier substrates. Consistent with this phenotype, we observe perturbations in the tricarboxylic acid (TCA) cycle in cells lacking AGK. Our identification of AGK as a bona fide subunit of TIM22 provides an exciting and unexpected link between mitochondrial protein import and Sengers syndrome.


Subject(s)
Cardiomyopathies/enzymology , Cataract/enzymology , Mitochondria/enzymology , Mitochondrial Membrane Transport Proteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Cardiomyopathies/genetics , Cataract/genetics , Citric Acid Cycle , Genetic Predisposition to Disease , HEK293 Cells , HeLa Cells , Humans , Mitochondrial Membrane Transport Proteins/genetics , Multiprotein Complexes , Mutation , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Stability , Protein Transport , Transfection
6.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35074914

ABSTRACT

Catabolism of sulfoquinovose (SQ; 6-deoxy-6-sulfoglucose), the ubiquitous sulfosugar produced by photosynthetic organisms, is an important component of the biogeochemical carbon and sulfur cycles. Here, we describe a pathway for SQ degradation that involves oxidative desulfurization to release sulfite and enable utilization of the entire carbon skeleton of the sugar to support the growth of the plant pathogen Agrobacterium tumefaciens SQ or its glycoside sulfoquinovosyl glycerol are imported into the cell by an ATP-binding cassette transporter system with an associated SQ binding protein. A sulfoquinovosidase hydrolyzes the SQ glycoside and the liberated SQ is acted on by a flavin mononucleotide-dependent sulfoquinovose monooxygenase, in concert with an NADH-dependent flavin reductase, to release sulfite and 6-oxo-glucose. An NAD(P)H-dependent oxidoreductase reduces the 6-oxo-glucose to glucose, enabling entry into primary metabolic pathways. Structural and biochemical studies provide detailed insights into the recognition of key metabolites by proteins in this pathway. Bioinformatic analyses reveal that the sulfoquinovose monooxygenase pathway is distributed across Alpha- and Betaproteobacteria and is especially prevalent within the Rhizobiales order. This strategy for SQ catabolism is distinct from previously described pathways because it enables the complete utilization of all carbons within SQ by a single organism with concomitant production of inorganic sulfite.


Subject(s)
Bacteria/metabolism , Bacterial Physiological Phenomena , Metabolic Networks and Pathways , Methylglucosides/metabolism , Oxidative Stress , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Carbohydrate Metabolism , Gene Expression Regulation, Bacterial , Models, Biological , Models, Molecular , Protein Binding , Protein Conformation , Structure-Activity Relationship , Sulfur/metabolism
7.
EMBO J ; 39(21): e105111, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32945574

ABSTRACT

Elevated ribosome biogenesis in oncogene-driven cancers is commonly targeted by DNA-damaging cytotoxic drugs. Our previous first-in-human trial of CX-5461, a novel, less genotoxic agent that specifically inhibits ribosome biogenesis via suppression of RNA polymerase I (Pol I) transcription, revealed single-agent efficacy in refractory blood cancers. Despite this clinical response, patients were not cured. In parallel, we demonstrated a marked improvement in the in vivo efficacy of CX-5461 in combination with PI3K/AKT/mTORC1 pathway inhibitors. Here, we reveal the molecular basis for this improved efficacy observed in vivo, which is associated with specific suppression of translation of mRNAs encoding regulators of cellular metabolism. Importantly, acquired resistance to this cotreatment is driven by translational rewiring that results in dysregulated cellular metabolism and induction of a cAMP-dependent pathway critical for the survival of blood cancers including lymphoma and acute myeloid leukemia. Our studies thus identify key molecular mechanisms underpinning the response of blood cancers to selective inhibition of ribosome biogenesis and define metabolic vulnerabilities that will facilitate the rational design of more effective regimens for Pol I-directed therapies.


Subject(s)
Neoplasms/metabolism , Protein Biosynthesis/genetics , Protein Biosynthesis/physiology , Ribosomes/metabolism , Transcription, Genetic/drug effects , Animals , Antineoplastic Agents/pharmacology , Benzothiazoles/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm , Guanine Nucleotide Exchange Factors/metabolism , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Naphthyridines/pharmacology , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Biosynthesis/drug effects , Protein Kinase Inhibitors , RNA Polymerase I/metabolism , RNA, Messenger/metabolism , RNA, Ribosomal , Ribosomes/drug effects , Transcriptome
8.
J Biol Chem ; 298(1): 101468, 2022 01.
Article in English | MEDLINE | ID: mdl-34896149

ABSTRACT

Apicomplexan parasites, such as Toxoplasma gondii, are unusual in that each cell contains a single apicoplast, a plastid-like organelle that compartmentalizes enzymes involved in the essential 2C-methyl-D-erythritol 4-phosphate pathway of isoprenoid biosynthesis. The last two enzymatic steps in this organellar pathway require electrons from a redox carrier. However, the small iron-sulfur cluster-containing protein ferredoxin, a likely candidate for this function, has not been investigated in this context. We show here that inducible knockdown of T. gondii ferredoxin results in progressive inhibition of growth and eventual parasite death. Surprisingly, this phenotype is not accompanied by ultrastructural changes in the apicoplast or overall cell morphology. The knockdown of ferredoxin was instead associated with a dramatic decrease in cellular levels of the last two metabolites in isoprenoid biosynthesis, 1-hydroxy-2-methyl-2-(E)- butenyl-4-pyrophosphate, and isomeric dimethylallyl pyrophosphate/isopentenyl pyrophosphate. Ferredoxin depletion was also observed to impair gliding motility, consistent with isoprenoid metabolites being important for dolichol biosynthesis, protein prenylation, and modification of other proteins involved in motility. Significantly, pharmacological inhibition of isoprenoid synthesis of the host cell exacerbated the impact of ferredoxin depletion on parasite replication, suggesting that the slow onset of parasite death after ferredoxin depletion is because of isoprenoid scavenging from the host cell and leading to partial compensation of the depleted parasite metabolites upon ferredoxin knockdown. Overall, these findings show that ferredoxin has an essential physiological function as an electron donor for the 2C-methyl-D-erythritol 4-phosphate pathway and is a potential drug target for apicomplexan parasites.


Subject(s)
Apicoplasts , Ferredoxins , Iron-Sulfur Proteins , Protozoan Proteins , Toxoplasma , Apicoplasts/genetics , Apicoplasts/metabolism , Biosynthetic Pathways , Diphosphates/metabolism , Electrons , Erythritol/analogs & derivatives , Erythritol/metabolism , Ferredoxins/genetics , Ferredoxins/metabolism , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Sugar Phosphates/metabolism , Terpenes/metabolism , Toxoplasma/genetics , Toxoplasma/metabolism
9.
Neurobiol Dis ; 176: 105933, 2023 01.
Article in English | MEDLINE | ID: mdl-36436748

ABSTRACT

In Huntington's disease (HD), a key pathological feature includes the development of inclusion-bodies of fragments of the mutant huntingtin protein in the neurons of the striatum and hippocampus. To examine the molecular changes associated with inclusion-body formation, we applied MALDI-mass spectrometry imaging and deuterium pulse labelling to determine lipid levels and synthesis rates in the hippocampus of a transgenic mouse model of HD (R6/1 line). The R6/1 HD mice lacked inclusions in the hippocampus at 6 weeks of age (pre-symptomatic), whereas inclusions were pervasive by 16 weeks of age (symptomatic). Hippocampal subfields (CA1, CA3 and DG), which formed the highest density of inclusion formation in the mouse brain showed a reduction in the relative abundance of neuron-enriched lipids that have roles in neurotransmission, synaptic plasticity, neurogenesis, and ER-stress protection. Lipids involved in the adaptive response to ER stress (phosphatidylinositol, phosphatidic acid, and ganglioside classes) displayed increased rates of synthesis in HD mice relative to WT mice at all the ages examined, including prior to the formation of the inclusion bodies. Our findings, therefore, support a role for ER stress occurring pre-symptomatically and potentially contributing to pathological mechanisms underlying HD.


Subject(s)
Huntington Disease , Mice , Animals , Mice, Transgenic , Huntington Disease/metabolism , Neurons/metabolism , Hippocampus/metabolism , Disease Models, Animal , Lipids , Huntingtin Protein/genetics , Huntingtin Protein/metabolism
10.
PLoS Pathog ; 17(8): e1009835, 2021 08.
Article in English | MEDLINE | ID: mdl-34432856

ABSTRACT

Intracellular parasites of the phylum Apicomplexa are dependent on the scavenging of essential amino acids from their hosts. We previously identified a large family of apicomplexan-specific plasma membrane-localized amino acid transporters, the ApiATs, and showed that the Toxoplasma gondii transporter TgApiAT1 functions in the selective uptake of arginine. TgApiAT1 is essential for parasite virulence, but dispensable for parasite growth in medium containing high concentrations of arginine, indicating the presence of at least one other arginine transporter. Here we identify TgApiAT6-1 as the second arginine transporter. Using a combination of parasite assays and heterologous characterisation of TgApiAT6-1 in Xenopus laevis oocytes, we demonstrate that TgApiAT6-1 is a general cationic amino acid transporter that mediates both the high-affinity uptake of lysine and the low-affinity uptake of arginine. TgApiAT6-1 is the primary lysine transporter in the disease-causing tachyzoite stage of T. gondii and is essential for parasite proliferation. We demonstrate that the uptake of cationic amino acids by TgApiAT6-1 is 'trans-stimulated' by cationic and neutral amino acids and is likely promoted by an inwardly negative membrane potential. These findings demonstrate that T. gondii has evolved overlapping transport mechanisms for the uptake of essential cationic amino acids, and we draw together our findings into a comprehensive model that highlights the finely-tuned, regulated processes that mediate cationic amino acid scavenging by these intracellular parasites.


Subject(s)
Amino Acid Transport Systems, Basic/metabolism , Amino Acids, Essential/metabolism , Fibroblasts/metabolism , Oocytes/metabolism , Protozoan Proteins/metabolism , Toxoplasmosis/metabolism , Amino Acid Transport Systems, Basic/genetics , Animals , Arginine/metabolism , Biological Transport , Fibroblasts/parasitology , Humans , Lysine/metabolism , Oocytes/parasitology , Protozoan Proteins/genetics , Toxoplasma/physiology , Toxoplasmosis/parasitology , Xenopus laevis
11.
Appl Environ Microbiol ; 89(2): e0201622, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36728421

ABSTRACT

Sulfoquinovose (SQ) is a major metabolite in the global sulfur cycle produced by nearly all photosynthetic organisms. One of the major pathways involved in the catabolism of SQ in bacteria such as Escherichia coli is a variant of the glycolytic Embden-Meyerhof-Parnas (EMP) pathway termed the sulfoglycolytic EMP (sulfo-EMP) pathway, which leads to the consumption of three of the six carbons of SQ and the excretion of 2,3-dihydroxypropanesulfonate (DHPS). Comparative metabolite profiling of aerobically glucose (Glc)-grown and SQ-grown E. coli cells was undertaken to identify the metabolic consequences of the switch from glycolysis to sulfoglycolysis. Sulfoglycolysis was associated with the diversion of triose phosphates (triose-P) to synthesize sugar phosphates (gluconeogenesis) and an unexpected accumulation of trehalose and glycogen storage carbohydrates. Sulfoglycolysis was also associated with global changes in central carbon metabolism, as indicated by the changes in the levels of intermediates in the tricarboxylic acid (TCA) cycle, the pentose phosphate pathway (PPP), polyamine metabolism, pyrimidine metabolism, and many amino acid metabolic pathways. Upon entry into stationary phase and the depletion of SQ, E. coli cells utilize their glycogen, indicating a reversal of metabolic fluxes to allow glycolytic metabolism. IMPORTANCE The sulfosugar sulfoquinovose is estimated to be produced on a scale of 10 billion metric tons per annum, making it a major organosulfur species in the biosulfur cycle. The microbial degradation of sulfoquinovose through sulfoglycolysis allows the utilization of its carbon content and contributes to the biomineralization of its sulfur. However, the metabolic consequences of microbial growth on sulfoquinovose are unclear. We use metabolomics to identify the metabolic adaptations that Escherichia coli undergoes when grown on sulfoquinovose versus glucose. This revealed the increased flux into storage carbohydrates through gluconeogenesis and the reduced flux of carbon into the TCA cycle and downstream metabolism. These changes are relieved upon entry into stationary phase and reversion to glycolytic metabolism. This work provides new insights into the metabolic consequences of microbial growth on an abundant sulfosugar.


Subject(s)
Carbon , Escherichia coli , Carbon/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Glycolysis , Glucose/metabolism , Glycogen/metabolism , Trioses/metabolism , Sulfur/metabolism
12.
Proc Natl Acad Sci U S A ; 117(39): 24475-24483, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32913052

ABSTRACT

Wolbachia-infected mosquitoes are refractory to flavivirus infections, but the role of lipids in Wolbachia-mediated virus blocking remains to be elucidated. Here, we use liquid chromatography mass spectrometry to provide a comprehensive picture of the lipidome of Aedes aegypti (Aag2) cells infected with Wolbachia only, either dengue or Zika virus only, and Wolbachia-infected Aag2 cells superinfected with either dengue or Zika virus. This approach identifies a class of lipids, acyl-carnitines, as being down-regulated during Wolbachia infection. Furthermore, treatment with an acyl-carnitine inhibitor assigns a crucial role for acyl-carnitines in the replication of dengue and Zika viruses. In contrast, depletion of acyl-carnitines increases Wolbachia density while addition of commercially available acyl-carnitines impairs Wolbachia production. Finally, we show an increase in flavivirus infection of Wolbachia-infected cells with the addition of acyl-carnitines. This study uncovers a previously unknown role for acyl-carnitines in this tripartite interaction that suggests an important and broad mechanism that underpins Wolbachia-mediated pathogen blocking.


Subject(s)
Aedes/microbiology , Aedes/virology , Carnitine/metabolism , Wolbachia/physiology , Zika Virus/physiology , Aedes/chemistry , Aedes/metabolism , Animals , Carnitine/chemistry , Female , Mosquito Vectors/chemistry , Mosquito Vectors/metabolism , Mosquito Vectors/microbiology , Mosquito Vectors/virology
13.
Mol Syst Biol ; 17(4): e10023, 2021 04.
Article in English | MEDLINE | ID: mdl-33821563

ABSTRACT

The malaria parasite, Plasmodium falciparum, proliferates rapidly in human erythrocytes by actively scavenging multiple carbon sources and essential nutrients from its host cell. However, a global overview of the metabolic capacity of intraerythrocytic stages is missing. Using multiplex 13 C-labelling coupled with untargeted mass spectrometry and unsupervised isotopologue grouping, we have generated a draft metabolome of P. falciparum and its host erythrocyte consisting of 911 and 577 metabolites, respectively, corresponding to 41% of metabolites and over 70% of the metabolic reaction predicted from the parasite genome. An additional 89 metabolites and 92 reactions were identified that were not predicted from genomic reconstructions, with the largest group being associated with metabolite damage-repair systems. Validation of the draft metabolome revealed four previously uncharacterised enzymes which impact isoprenoid biosynthesis, lipid homeostasis and mitochondrial metabolism and are necessary for parasite development and proliferation. This study defines the metabolic fate of multiple carbon sources in P. falciparum, and highlights the activity of metabolite repair pathways in these rapidly growing parasite stages, opening new avenues for drug discovery.


Subject(s)
Isotope Labeling , Metabolic Networks and Pathways , Metabolomics , Parasites/metabolism , Plasmodium falciparum/metabolism , Animals , Electron Transport , Erythrocytes/parasitology , Glycine Hydroxymethyltransferase/metabolism , Hemoglobins/metabolism , Humans , Metabolic Flux Analysis , Metabolome , Mitochondria/metabolism , Parasites/growth & development , Phosphoprotein Phosphatases/metabolism , Plasmodium falciparum/growth & development , Protozoan Proteins/metabolism , Serine/metabolism , Terpenes/metabolism , Trophozoites/metabolism
14.
PLoS Biol ; 17(7): e3000376, 2019 07.
Article in English | MEDLINE | ID: mdl-31318858

ABSTRACT

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.


Subject(s)
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
15.
Proc Natl Acad Sci U S A ; 116(9): 3722-3727, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30808758

ABSTRACT

Staphylococcus aureus is a notorious human bacterial pathogen with considerable capacity to develop antibiotic resistance. We have observed that human infections caused by highly drug-resistant S. aureus are more prolonged, complicated, and difficult to eradicate. Here we describe a metabolic adaptation strategy used by clinical S. aureus strains that leads to resistance to the last-line antibiotic, daptomycin, and simultaneously affects host innate immunity. This response was characterized by a change in anionic membrane phospholipid composition induced by point mutations in the phospholipid biosynthesis gene, cls2, encoding cardiolipin synthase. Single cls2 point mutations were sufficient for daptomycin resistance, antibiotic treatment failure, and persistent infection. These phenotypes were mediated by enhanced cardiolipin biosynthesis, leading to increased bacterial membrane cardiolipin and reduced phosphatidylglycerol. The changes in membrane phospholipid profile led to modifications in membrane structure that impaired daptomycin penetration and membrane disruption. The cls2 point mutations also allowed S. aureus to evade neutrophil chemotaxis, mediated by the reduction in bacterial membrane phosphatidylglycerol, a previously undescribed bacterial-driven chemoattractant. Together, these data illustrate a metabolic strategy used by S. aureus to circumvent antibiotic and immune attack and provide crucial insights into membrane-based therapeutic targeting of this troublesome pathogen.


Subject(s)
Drug Resistance, Bacterial/genetics , Membrane Proteins/genetics , Methicillin-Resistant Staphylococcus aureus/genetics , Staphylococcal Infections/genetics , Transferases (Other Substituted Phosphate Groups)/genetics , Anti-Bacterial Agents/pharmacology , Daptomycin/pharmacology , Drug Resistance, Bacterial/immunology , Gene Expression Regulation, Bacterial/drug effects , Host-Pathogen Interactions/immunology , Humans , Immune Evasion/genetics , Immune Evasion/immunology , Membrane Proteins/metabolism , Methicillin-Resistant Staphylococcus aureus/immunology , Methicillin-Resistant Staphylococcus aureus/metabolism , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Microbial Sensitivity Tests , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Transferases (Other Substituted Phosphate Groups)/metabolism
16.
J Biol Chem ; 295(18): 6108-6119, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32217691

ABSTRACT

Pathogenic bacteria of the genera Mycobacterium and Corynebacterium cause severe human diseases such as tuberculosis (Mycobacterium tuberculosis) and diphtheria (Corynebacterium diphtheriae). The cells of these species are surrounded by protective cell walls rich in long-chain mycolic acids. These fatty acids are conjugated to the disaccharide trehalose on the cytoplasmic side of the bacterial cell membrane. They are then transported across the membrane to the periplasm where they act as donors for other reactions. We have previously shown that transient acetylation of the glycolipid trehalose monohydroxycorynomycolate (hTMCM) enables its efficient transport to the periplasm in Corynebacterium glutamicum and that acetylation is mediated by the membrane protein TmaT. Here, we show that a putative methyltransferase, encoded at the same genetic locus as TmaT, is also required for optimal hTMCM transport. Deletion of the C. glutamicum gene NCgl2764 (Rv0224c in M. tuberculosis) abolished acetyltrehalose monocorynomycolate (AcTMCM) synthesis, leading to accumulation of hTMCM in the inner membrane and delaying its conversion to trehalose dihydroxycorynomycolate (h2TDCM). Complementation with NCgl2764 normalized turnover of hTMCM to h2TDCM. In contrast, complementation with NCgl2764 derivatives mutated at residues essential for methyltransferase activity failed to rectify the defect, suggesting that NCgl2764/Rv0224c encodes a methyltransferase, designated here as MtrP. Comprehensive analyses of the individual mtrP and tmaT mutants and of a double mutant revealed strikingly similar changes across several lipid classes compared with WT bacteria. These findings indicate that both MtrP and TmaT have nonredundant roles in regulating AcTMCM synthesis, revealing additional complexity in the regulation of trehalose mycolate transport in the Corynebacterineae.


Subject(s)
Cell Membrane/metabolism , Corynebacterium glutamicum/cytology , Corynebacterium glutamicum/enzymology , Methyltransferases/metabolism , Mycolic Acids/chemistry , Trehalose/chemistry , Trehalose/metabolism , Biological Transport , Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/metabolism , Methyltransferases/genetics , Mutation , Mycobacterium tuberculosis/genetics , Sequence Homology, Nucleic Acid
17.
Brief Bioinform ; 20(2): 384-389, 2019 03 22.
Article in English | MEDLINE | ID: mdl-29106479

ABSTRACT

EMBL Australia Bioinformatics Resource (EMBL-ABR) is a developing national research infrastructure, providing bioinformatics resources and support to life science and biomedical researchers in Australia. EMBL-ABR comprises 10 geographically distributed national nodes with one coordinating hub, with current funding provided through Bioplatforms Australia and the University of Melbourne for its initial 2-year development phase. The EMBL-ABR mission is to: (1) increase Australia's capacity in bioinformatics and data sciences; (2) contribute to the development of training in bioinformatics skills; (3) showcase Australian data sets at an international level and (4) enable engagement in international programs. The activities of EMBL-ABR are focussed in six key areas, aligning with comparable international initiatives such as ELIXIR, CyVerse and NIH Commons. These key areas-Tools, Data, Standards, Platforms, Compute and Training-are described in this article.


Subject(s)
Biological Science Disciplines , Biomedical Research , Computational Biology/education , Computational Biology/methods , Data Curation/methods , Australia , Humans
18.
PLoS Pathog ; 15(2): e1007577, 2019 02.
Article in English | MEDLINE | ID: mdl-30742695

ABSTRACT

Apicomplexan parasites are auxotrophic for a range of amino acids which must be salvaged from their host cells, either through direct uptake or degradation of host proteins. Here, we describe a family of plasma membrane-localized amino acid transporters, termed the Apicomplexan Amino acid Transporters (ApiATs), that are ubiquitous in apicomplexan parasites. Functional characterization of the ApiATs of Toxoplasma gondii indicate that several of these transporters are important for intracellular growth of the tachyzoite stage of the parasite, which is responsible for acute infections. We demonstrate that the ApiAT protein TgApiAT5-3 is an exchanger for aromatic and large neutral amino acids, with particular importance for L-tyrosine scavenging and amino acid homeostasis, and that TgApiAT5-3 is critical for parasite virulence. Our data indicate that T. gondii expresses additional proteins involved in the uptake of aromatic amino acids, and we present a model for the uptake and homeostasis of these amino acids. Our findings identify a family of amino acid transporters in apicomplexans, and highlight the importance of amino acid scavenging for the biology of this important phylum of intracellular parasites.


Subject(s)
Amino Acid Transport Systems/metabolism , Toxoplasma/metabolism , Tyrosine/physiology , Animals , Apicomplexa/metabolism , Biological Transport , Host-Parasite Interactions , Ion Transport , Parasites , Protozoan Proteins , Tyrosine/metabolism
19.
PLoS Biol ; 16(3): e2004328, 2018 03.
Article in English | MEDLINE | ID: mdl-29529020

ABSTRACT

Within the human host, the malaria parasite Plasmodium falciparum is exposed to multiple selection pressures. The host environment changes dramatically in severe malaria, but the extent to which the parasite responds to-or is selected by-this environment remains unclear. From previous studies, the parasites that cause severe malaria appear to increase expression of a restricted but poorly defined subset of the PfEMP1 variant, surface antigens. PfEMP1s are major targets of protective immunity. Here, we used RNA sequencing (RNAseq) to analyse gene expression in 44 parasite isolates that caused severe and uncomplicated malaria in Papuan patients. The transcriptomes of 19 parasite isolates associated with severe malaria indicated that these parasites had decreased glycolysis without activation of compensatory pathways; altered chromatin structure and probably transcriptional regulation through decreased histone methylation; reduced surface expression of PfEMP1; and down-regulated expression of multiple chaperone proteins. Our RNAseq also identified novel associations between disease severity and PfEMP1 transcripts, domains, and smaller sequence segments and also confirmed all previously reported associations between expressed PfEMP1 sequences and severe disease. These findings will inform efforts to identify vaccine targets for severe malaria and also indicate how parasites adapt to-or are selected by-the host environment in severe malaria.


Subject(s)
Antigens, Protozoan/genetics , Antigens, Surface/genetics , Malaria/parasitology , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Transcriptome , Gene Expression Regulation , Humans , Malaria/pathology , Plasmodium falciparum/isolation & purification , Plasmodium falciparum/metabolism , Sequence Analysis, RNA
20.
PLoS Biol ; 16(9): e2005642, 2018 09.
Article in English | MEDLINE | ID: mdl-30208022

ABSTRACT

The phylum Apicomplexa comprises a group of obligate intracellular parasites that alternate between intracellular replicating stages and actively motile extracellular forms that move through tissue. Parasite cytosolic Ca2+ signalling activates motility, but how this is switched off after invasion is complete to allow for replication to begin is not understood. Here, we show that the cyclic adenosine monophosphate (cAMP)-dependent protein kinase A catalytic subunit 1 (PKAc1) of Toxoplasma is responsible for suppression of Ca2+ signalling upon host cell invasion. We demonstrate that PKAc1 is sequestered to the parasite periphery by dual acylation of PKA regulatory subunit 1 (PKAr1). Upon genetic depletion of PKAc1 we show that newly invaded parasites exit host cells shortly thereafter, in a perforin-like protein 1 (PLP-1)-dependent fashion. Furthermore, we demonstrate that loss of PKAc1 prevents rapid down-regulation of cytosolic [Ca2+] levels shortly after invasion. We also provide evidence that loss of PKAc1 sensitises parasites to cyclic GMP (cGMP)-induced Ca2+ signalling, thus demonstrating a functional link between cAMP and these other signalling modalities. Together, this work provides a new paradigm in understanding how Toxoplasma and related apicomplexan parasites regulate infectivity.


Subject(s)
Calcium Signaling , Cyclic AMP-Dependent Protein Kinases/metabolism , Toxoplasma/enzymology , Acylation , Animals , Calcium/metabolism , Cyclic AMP/metabolism , Cytosol/metabolism , Fibroblasts/parasitology , Host-Parasite Interactions , Humans , Life Cycle Stages , Mice , Parasites/enzymology , Parasites/growth & development , Protein Subunits/metabolism , Protozoan Proteins , Signal Transduction , Toxoplasma/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL