268 results on '"McConville MJ"'
Search Results
2. Fatty acid-induced hepatocyte phospholipidome changes and their potential roles in NAFLD
- Author
-
PENG, KY, MELLETT, NA, KAMMOUN, HL, FEBBRAIO, MA, BARLOW, CK, MCCONVILLE, MJ, and MEIKLE, PJ
- Published
- 2015
3. Post-translational processng of avariant surface protein of Giardia
- Author
-
Köhler, P, Papanastasiou, P, McConville, MJ, Hülsmeier, A, Ralton, J, and Hiltpold, A
- Published
- 1998
- Full Text
- View/download PDF
4. MLKL deficiency elevates testosterone production in male mice independently of necroptotic functions.
- Author
-
Chiou S, Cawthorne W, Soerianto T, Hofferek V, Patel KM, Garnish SE, Tovey Crutchfield EC, Hall C, Hildebrand JM, McConville MJ, Lawlor KE, Hawkins ED, Samson AL, and Murphy JM
- Subjects
- Animals, Male, Mice, Mice, Inbred C57BL, Macrophages metabolism, Spermatozoa metabolism, Testosterone metabolism, Necroptosis, Protein Kinases metabolism, Testis metabolism
- Abstract
Mixed lineage kinase domain-like (MLKL) is a pseudokinase, best known for its role as the terminal effector of the necroptotic cell death pathway. MLKL-mediated necroptosis has long been linked to various age-related pathologies including neurodegeneration, atherosclerosis and male reproductive decline, however many of these attributions remain controversial. Here, we investigated the role of MLKL and necroptosis in the adult mouse testis: an organ divided into sperm-producing seminiferous tubules and the surrounding testosterone-producing interstitium. We find that sperm-producing cells within seminiferous tubules lack expression of key necroptotic mediators and thus are resistant to a pro-necroptotic challenge. By comparison, coordinated expression of the necroptotic pathway occurs in the testicular interstitium, rendering cells within this compartment, especially the lysozyme-positive macrophages, vulnerable to necroptotic cell death. We also uncover a non-necroptotic role for MLKL in regulating testosterone levels. Thus, MLKL serves two roles in the mouse testes - one involving the canonical response of macrophages to necroptotic insult, and the other a non-canonical function in male reproductive hormone control., Competing Interests: Competing interests: KMP, JMH, KEL, ALS, and JMM contribute to or have contributed to a project developing necroptosis inhibitors in collaboration with Anaxis Pharma. The other authors declare no competing interests. Ethics declaration: All experiments were approved by the WEHI Animal Ethics Committee following the Prevention of Cruelty to Animals Act (1996) and the Australian National Health and Medical Research Council Code of Practice for the Care and Use of Animals for Scientific Purposes (1997)., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
5. Apicoplast-derived isoprenoids are essential for biosynthesis of GPI protein anchors, and consequently for egress and invasion in Plasmodium falciparum.
- Author
-
Bulloch MS, Huynh LK, Kennedy K, Ralton JE, McConville MJ, and Ralph SA
- Subjects
- Protozoan Proteins metabolism, Protozoan Proteins genetics, Erythrocytes parasitology, Erythrocytes metabolism, Humans, Malaria, Falciparum parasitology, Malaria, Falciparum metabolism, Animals, Merozoites metabolism, Plasmodium falciparum metabolism, Apicoplasts metabolism, Glycosylphosphatidylinositols metabolism, Glycosylphosphatidylinositols biosynthesis, Terpenes metabolism
- 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., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2024 Bulloch et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
6. Accurate detection of acute sleep deprivation using a metabolomic biomarker-A machine learning approach.
- Author
-
Jeppe K, Ftouni S, Nijagal B, Grant LK, Lockley SW, Rajaratnam SMW, Phillips AJK, McConville MJ, Tull D, and Anderson C
- Subjects
- Humans, Wakefulness, Metabolomics, Machine Learning, Sleep Deprivation metabolism, Sleep
- Abstract
Sleep deprivation enhances risk for serious injury and fatality on the roads and in workplaces. To facilitate future management of these risks through advanced detection, we developed and validated a metabolomic biomarker of sleep deprivation in healthy, young participants, across three experiments. Bi-hourly plasma samples from 2 × 40-hour extended wake protocols (for train/test models) and 1 × 40-hour protocol with an 8-hour overnight sleep interval were analyzed by untargeted liquid chromatography-mass spectrometry. Using a knowledge-based machine learning approach, five consistently important variables were used to build predictive models. Sleep deprivation (24 to 38 hours awake) was predicted accurately in classification models [versus well-rested (0 to 16 hours)] (accuracy = 94.7%/AUC 99.2%, 79.3%/AUC 89.1%) and to a lesser extent in regression ( R
2 = 86.1 and 47.8%) models for within- and between-participant models, respectively. Metabolites were identified for replicability/future deployment. This approach for detecting acute sleep deprivation offers potential to reduce accidents through "fitness for duty" or "post-accident analysis" assessments.- Published
- 2024
- Full Text
- View/download PDF
7. Matrix Selection for the Visualization of Small Molecules and Lipids in Brain Tumors Using Untargeted MALDI-TOF Mass Spectrometry Imaging.
- Author
-
Lu T, Freytag L, Narayana VK, Moore Z, Oliver SJ, Valkovic A, Nijagal B, Peterson AL, de Souza DP, McConville MJ, Whittle JR, Best SA, and Freytag S
- Abstract
Matrix-assisted laser desorption/ionization mass spectrometry imaging allows for the study of metabolic activity in the tumor microenvironment of brain cancers. The detectable metabolites within these tumors are contingent upon the choice of matrix, deposition technique, and polarity setting. In this study, we compared the performance of three different matrices, two deposition techniques, and the use of positive and negative polarity in two different brain cancer types and across two species. Optimal combinations were confirmed by a comparative analysis of lipid and small-molecule abundance by using liquid chromatography-mass spectrometry and RNA sequencing to assess differential metabolites and enzymes between normal and tumor regions. Our findings indicate that in the tumor-bearing brain, the recrystallized α-cyano-4-hydroxycinnamic acid matrix with positive polarity offered superior performance for both detected metabolites and consistency with other techniques. Beyond these implications for brain cancer, our work establishes a workflow to identify optimal matrices for spatial metabolomics studies.
- Published
- 2023
- Full Text
- View/download PDF
8. Integrative omics identifies conserved and pathogen-specific responses of sepsis-causing bacteria.
- Author
-
Mu A, Klare WP, Baines SL, Ignatius Pang CN, Guérillot R, Harbison-Price N, Keller N, Wilksch J, Nhu NTK, Phan MD, Keller B, Nijagal B, Tull D, Dayalan S, Chua HHC, Skoneczny D, Koval J, Hachani A, Shah AD, Neha N, Jadhav S, Partridge SR, Cork AJ, Peters K, Bertolla O, Brouwer S, Hancock SJ, Álvarez-Fraga L, De Oliveira DMP, Forde B, Dale A, Mujchariyakul W, Walsh CJ, Monk I, Fitzgerald A, Lum M, Correa-Ospina C, Roy Chowdhury P, Parton RG, De Voss J, Beckett J, Monty F, McKinnon J, Song X, Stephen JR, Everest M, Bellgard MI, Tinning M, Leeming M, Hocking D, Jebeli L, Wang N, Ben Zakour N, Yasar SA, Vecchiarelli S, Russell T, Zaw T, Chen T, Teng D, Kassir Z, Lithgow T, Jenney A, Cole JN, Nizet V, Sorrell TC, Peleg AY, Paterson DL, Beatson SA, Wu J, Molloy MP, Syme AE, Goode RJA, Hunter AA, Bowland G, West NP, Wilkins MR, Djordjevic SP, Davies MR, Seemann T, Howden BP, Pascovici D, Tyagi S, Schittenhelm RB, De Souza DP, McConville MJ, Iredell JR, Cordwell SJ, Strugnell RA, Stinear TP, Schembri MA, and Walker MJ
- Subjects
- Humans, Anti-Bacterial Agents therapeutic use, Proteomics, Bacteria, Escherichia coli, Klebsiella, Microbial Sensitivity Tests, Sepsis microbiology, Staphylococcal Infections
- Abstract
Even in the setting of optimal resuscitation in high-income countries severe sepsis and septic shock have a mortality of 20-40%, with antibiotic resistance dramatically increasing this mortality risk. To develop a reference dataset enabling the identification of common bacterial targets for therapeutic intervention, we applied a standardized genomic, transcriptomic, proteomic and metabolomic technological framework to multiple clinical isolates of four sepsis-causing pathogens: Escherichia coli, Klebsiella pneumoniae species complex, Staphylococcus aureus and Streptococcus pyogenes. Exposure to human serum generated a sepsis molecular signature containing global increases in fatty acid and lipid biosynthesis and metabolism, consistent with cell envelope remodelling and nutrient adaptation for osmoprotection. In addition, acquisition of cholesterol was identified across the bacterial species. This detailed reference dataset has been established as an open resource to support discovery and translational research., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
9. Remodeling of Carbon Metabolism during Sulfoglycolysis in Escherichia coli.
- Author
-
Mui JW, De Souza DP, Saunders EC, McConville MJ, and Williams SJ
- Subjects
- Glycolysis, Glucose metabolism, Glycogen metabolism, Trioses metabolism, Sulfur metabolism, Carbon metabolism, Escherichia coli genetics, Escherichia coli metabolism
- 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.
- Published
- 2023
- Full Text
- View/download PDF
10. Longitudinal spatial mapping of lipid metabolites reveals pre-symptomatic changes in the hippocampi of Huntington's disease transgenic mice.
- Author
-
Farzana F, McConville MJ, Renoir T, Li S, Nie S, Tran H, Hannan AJ, Hatters DM, and Boughton BA
- Subjects
- Mice, Animals, Mice, Transgenic, Neurons metabolism, Hippocampus metabolism, Disease Models, Animal, Lipids, Huntingtin Protein genetics, Huntingtin Protein metabolism, Huntington Disease metabolism
- 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., Competing Interests: Declaration of Competing Interest The authors declare no competing financial interests., (Copyright © 2022. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
11. Relationship of circulating Plasmodium falciparum lifecycle stage to circulating parasitemia and total parasite biomass.
- Author
-
Duffy MF, Tonkin-Hill GQ, Trianty L, Noviyanti R, Nguyen HHT, Rambhatla JS, McConville MJ, Rogerson SJ, Brown GV, Price RN, Anstey NM, Day KP, and Papenfuss AT
- Subjects
- Animals, Biomass, Life Cycle Stages, Parasitemia diagnosis, Parasitemia parasitology, Plasmodium falciparum, Malaria, Falciparum diagnosis, Malaria, Falciparum parasitology, Parasites
- Published
- 2022
- Full Text
- View/download PDF
12. In-cell DNP NMR reveals multiple targeting effect of antimicrobial peptide.
- Author
-
Separovic F, Hofferek V, Duff AP, McConville MJ, and Sani MA
- Abstract
Dynamic nuclear polarization NMR spectroscopy was used to investigate the effect of the antimicrobial peptide (AMP) maculatin 1.1 on E. coli cells. The enhanced
15 N NMR signals from nucleic acids, proteins and lipids identified a number of unanticipated physiological responses to peptide stress, revealing that membrane-active AMPs can have a multi-target impact on E. coli cells. DNP-enhanced15 N-observed31 P-dephased REDOR NMR allowed monitoring how Mac1 induced DNA condensation and prevented intermolecular salt bridges between the main E. coli lipid phosphatidylethanolamine (PE) molecules. The latter was supported by similar results obtained using E. coli PE lipid systems. Overall, the ability to monitor the action of antimicrobial peptides in situ will provide greater insight into their mode of action., Competing Interests: The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (© 2022 The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
13. Microbial Metabolites in the Maturation and Activation of Dendritic Cells and Their Relevance for Respiratory Immunity.
- Author
-
Wilson KR, Gressier E, McConville MJ, and Bedoui S
- Subjects
- Cell Differentiation, Humans, Lung, T-Lymphocytes, Dendritic Cells, Respiratory Tract Infections metabolism
- Abstract
The respiratory tract is a gateway for viruses and bacteria from the external environment to invade the human body. Critical to the protection against these invaders are dendritic cells (DCs) - a group of highly specialized myeloid cells that monitors the lung microenvironment and relays contextual and antigenic information to T cells. Following the recognition of danger signals and/or pathogen molecular associated patterns in the lungs, DCs undergo activation. This process arms DCs with the unique ability to induce the proliferation and differentiation of T cells responding to matching antigen in complex with MHC molecules. Depending on how DCs interact with T cells, the ensuing T cell response can be tolerogenic or immunogenic and as such, the susceptibility and severity of respiratory infections is influenced by the signals DCs receive, integrate, and then convey to T cells. It is becoming increasingly clear that these facets of DC biology are heavily influenced by the cellular components and metabolites produced by the lung and gut microbiota. In this review, we discuss the roles of different DC subsets in respiratory infections and outline how microbial metabolites impact the development, propensity for activation and subsequent activation of DCs. In particular, we highlight these concepts in the context of respiratory immunity., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Wilson, Gressier, McConville and Bedoui.)
- Published
- 2022
- Full Text
- View/download PDF
14. Tetraspanin CD82 restrains phagocyte migration but supports macrophage activation.
- Author
-
McGowan ENS, Wong O, Jones E, Nguyen J, Wee J, Demaria MC, Deliyanti D, Johnson CJ, Hickey MJ, McConville MJ, Wilkinson-Berka JL, Wright MD, and Binger KJ
- Abstract
Phagocytes migrate into tissues to combat infection and maintain tissue homeostasis. As dysregulated phagocyte migration and function can lead to inflammation or susceptibility to infection, identifying molecules that control these processes is critical. Here, we show that the tetraspanin CD82 restrains the migration of neutrophils and macrophages into tissues. Cd82
-/- phagocytes exhibited excessive migration during in vivo models of peritoneal inflammation, superfusion of CXCL1, retinopathy of prematurity, and infection with the protozoan parasite L. mexicana . However, with the latter, while Cd82-/- macrophages infiltrated infection sites at higher proportions, cutaneous L. mexicana lesions were larger and persisted, indicating a failure to control infection. Analyses of in vitro bone-marrow-derived macrophages showed CD82 deficiency altered cellular morphology, and impaired gene expression and metabolism in response to anti-inflammatory activation. Altogether, this work reveals an important role for CD82 in restraining phagocyte infiltration and mediating their differentiation in response to stimulatory cues., Competing Interests: The authors declare no competing interests., (© 2022 The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
15. Type I interferon antagonism of the JMJD3-IRF4 pathway modulates macrophage activation and polarization.
- Author
-
Ming-Chin Lee K, Achuthan AA, De Souza DP, Lupancu TJ, Binger KJ, Lee MKS, Xu Y, McConville MJ, de Weerd NA, Dragoljevic D, Hertzog PJ, Murphy AJ, Hamilton JA, and Fleetwood AJ
- Subjects
- Granulocyte-Macrophage Colony-Stimulating Factor genetics, Ketoglutaric Acids metabolism, Ketoglutaric Acids pharmacology, Succinic Acid, Interferon Type I, Macrophage Activation
- Abstract
Metabolic adaptations can directly influence the scope and scale of macrophage activation and polarization. Here we explore the impact of type I interferon (IFNβ) on macrophage metabolism and its broader impact on cytokine signaling pathways. We find that IFNβ simultaneously increased the expression of immune-responsive gene 1 and itaconate production while inhibiting isocitrate dehydrogenase activity and restricting α-ketoglutarate accumulation. IFNβ also increased the flux of glutamine-derived carbon into the tricarboxylic acid cycle to boost succinate levels. Combined, we identify that IFNβ controls the cellular α-ketoglutarate/succinate ratio. We show that by lowering the α-ketoglutarate/succinate ratio, IFNβ potently blocks the JMJD3-IRF4-dependent pathway in GM-CSF and IL-4 activated macrophages. The suppressive effects of IFNβ on JMJD3-IRF4-dependent responses, including M2 polarization and GM-CSF-induced inflammatory pain, were reversed by supplementation with α-ketoglutarate. These results reveal that IFNβ modulates macrophage activation and polarization through control of the cellular α-ketoglutarate/succinate ratio., Competing Interests: Declaration of interests The authors declare no conflict of interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
16. YAP regulates an SGK1/mTORC1/SREBP-dependent lipogenic program to support proliferation and tissue growth.
- Author
-
Vaidyanathan S, Salmi TM, Sathiqu RM, McConville MJ, Cox AG, and Brown KK
- Subjects
- Cell Proliferation, Mechanistic Target of Rapamycin Complex 1 metabolism, Sterol Regulatory Element Binding Protein 1 genetics, Sterol Regulatory Element Binding Protein 1 metabolism, Lipogenesis genetics, Sterol Regulatory Element Binding Proteins genetics, Sterol Regulatory Element Binding Proteins metabolism
- Abstract
The coordinated regulation of growth control and metabolic pathways is required to meet the energetic and biosynthetic demands associated with proliferation. Emerging evidence suggests that the Hippo pathway effector Yes-associated protein 1 (YAP) reprograms cellular metabolism to meet the anabolic demands of growth, although the mechanisms involved are poorly understood. Here, we demonstrate that YAP co-opts the sterol regulatory element-binding protein (SREBP)-dependent lipogenic program to facilitate proliferation and tissue growth. Mechanistically, YAP stimulates de novo lipogenesis via mechanistic target of rapamcyin (mTOR) complex 1 (mTORC1) signaling and subsequent activation of SREBP. Importantly, YAP-dependent regulation of serum- and glucocorticoid-regulated kinase 1 (SGK1) is required to activate mTORC1/SREBP and stimulate de novo lipogenesis. We also find that the SREBP target genes fatty acid synthase (FASN) and stearoyl-CoA desaturase (SCD) are conditionally required to support YAP-dependent proliferation and tissue growth. These studies reveal that de novo lipogenesis is a metabolic vulnerability that can be targeted to disrupt YAP-dependent proliferation and tissue growth., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
17. Oxidative desulfurization pathway for complete catabolism of sulfoquinovose by bacteria.
- Author
-
Sharma M, Lingford JP, Petricevic M, Snow AJD, Zhang Y, Järvå MA, Mui JW, Scott NE, Saunders EC, Mao R, Epa R, da Silva BM, Pires DEV, Ascher DB, McConville MJ, Davies GJ, Williams SJ, and Goddard-Borger ED
- Subjects
- 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, Bacteria metabolism, Bacterial Physiological Phenomena, Metabolic Networks and Pathways, Methylglucosides metabolism, Oxidative Stress
- 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., Competing Interests: The authors declare no competing interest., (Copyright © 2022 the Author(s). Published by PNAS.)
- Published
- 2022
- Full Text
- View/download PDF
18. Toxoplasma gondii apicoplast-resident ferredoxin is an essential electron transfer protein for the MEP isoprenoid-biosynthetic pathway.
- Author
-
Henkel S, Frohnecke N, Maus D, McConville MJ, Laue M, Blume M, and Seeber F
- Subjects
- Biosynthetic Pathways, Diphosphates metabolism, Electrons, Erythritol analogs & derivatives, Erythritol metabolism, Sugar Phosphates metabolism, Terpenes metabolism, Apicoplasts genetics, Apicoplasts metabolism, Ferredoxins genetics, Ferredoxins metabolism, Iron-Sulfur Proteins genetics, Iron-Sulfur Proteins metabolism, Protozoan Proteins genetics, Protozoan Proteins metabolism, Toxoplasma genetics, Toxoplasma metabolism
- 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., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
19. MmpA, a Conserved Membrane Protein Required for Efficient Surface Transport of Trehalose Lipids in Corynebacterineae.
- Author
-
Cashmore TJ, Klatt S, Brammananth R, Rainczuk AK, Crellin PK, McConville MJ, and Coppel RL
- Subjects
- Acetylation, Acetyltransferases metabolism, Bacterial Proteins genetics, Bacterial Proteins metabolism, Cell Wall chemistry, Cytoplasm metabolism, Gene Deletion, Lipidomics, Methyltransferases metabolism, Trehalose chemistry, Corynebacterium glutamicum metabolism, Membrane Proteins genetics, Membrane Proteins metabolism, Trehalose biosynthesis
- Abstract
Cell walls of bacteria of the genera Mycobacterium and Corynebacterium contain high levels of (coryno)mycolic acids. These very long chain fatty acids are synthesized on the cytoplasmic leaflet of the inner membrane (IM) prior to conjugation to the disaccharide, trehalose, and transport to the periplasm. Recent studies on Corynebacterium glutamicum have shown that acetylation of trehalose monohydroxycorynomycolate (hTMCM) promotes its transport across the inner membrane. Acetylation is mediated by the membrane acetyltransferase, TmaT, and is dependent on the presence of a putative methyltransferase, MtrP. Here, we identify a third protein that is required for the acetylation and membrane transport of hTMCM. Deletion of the C. glutamicum gene NCgl2761 ( Rv0226c in Mycobacterium tuberculosis ) abolished synthesis of acetylated hTMCM (AcTMCM), resulting in an accumulation of hTMCM in the inner membrane and reduced synthesis of trehalose dihydroxycorynomycolate (h2TDCM), a major outer membrane glycolipid. Complementation with the NCgl2761 gene, designated here as mmpA , restored the hTMCM:h2TDCM ratio. Comprehensive lipidomic analysis of the Δ tmaT , Δ mtrP and Δ mmpA mutants revealed strikingly similar global changes in overall membrane lipid composition. Our findings suggest that the acetylation and membrane transport of hTMCM is regulated by multiple proteins: MmpA, MtrP and TmaT, and that defects in this process lead to global, potentially compensatory changes in the composition of inner and outer membranes.
- Published
- 2021
- Full Text
- View/download PDF
20. The Redox Homeostasis of Skeletal Muscle Cells Regulates Stage Differentiation of Toxoplasma gondii .
- Author
-
Rahman MT, Swierzy IJ, Downie B, Salinas G, Blume M, McConville MJ, and Lüder CGK
- Subjects
- Animals, Cell Differentiation, Homeostasis, Humans, Mice, Muscle Fibers, Skeletal, Oxidation-Reduction, Persistent Infection, Toxoplasma
- Abstract
Toxoplasma gondii is an obligatory intracellular parasite that causes persistent infections in birds and mammals including ~30% of the world's human population. Differentiation from proliferative and metabolically active tachyzoites to largely dormant bradyzoites initiates the chronic phase of infection and occurs predominantly in brain and muscle tissues. Here we used murine skeletal muscle cells (SkMCs) to decipher host cellular factors that favor T. gondii bradyzoite formation in terminally differentiated and syncytial myotubes, but not in proliferating myoblast precursors. Genome-wide transcriptome analyses of T. gondii -infected SkMCs and non-infected controls identified ~6,500 genes which were differentially expressed (DEGs) in myotubes compared to myoblasts, largely irrespective of infection. On the other hand, genes related to central carbohydrate metabolism, to redox homeostasis, and to the Nrf2-dependent stress response pathway were enriched in both infected myoblast precursors and myotubes. Stable isotope-resolved metabolite profiling indicated increased fluxes into the oxidative branch of the pentose phosphate pathway (OxPPP) in infected myoblasts and into the TCA cycle in infected myotubes. High OxPPP activity in infected myoblasts was associated with increased NADPH/NADP
+ ratio while myotubes exhibited higher ROS levels and lower expression of anti-oxidants and detoxification enzymes. Pharmacological reduction of ROS levels in SkMCs inhibited bradyzoite differentiation, while increased ROS induced bradyzoite formation. Thus, we identified a novel host cell-dependent mechanism that triggers stage conversion of T. gondii into persistent tissue cysts in its natural host cell type., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Rahman, Swierzy, Downie, Salinas, Blume, McConville and Lüder.)- Published
- 2021
- Full Text
- View/download PDF
21. Evolution and function of carbohydrate reserve biosynthesis in parasitic protists.
- Author
-
Ralton JE, Sernee MF, and McConville MJ
- Subjects
- Animals, Carbohydrates, Eukaryota, Life Cycle Stages, Leishmania, Parasites metabolism
- Abstract
Nearly all eukaryotic cells synthesize carbohydrate reserves, such as glycogen, starch, or low-molecular-weight oligosaccharides. However, a number of parasitic protists have lost this capacity while others have lost, and subsequently evolved, entirely new pathways. Recent studies suggest that retention, loss, or acquisition of these pathways in different protists is intimately linked to their lifestyle. In particular, parasites with carbohydrate reserves often establish long-lived chronic infections and/or produce environmental cysts, whereas loss of these pathways is associated with parasites that have highly proliferative and metabolically active life-cycle stages. The evolution of mannogen biosynthesis in Leishmania and related parasites indicates that these pathways have played a role in defining the host range and niches occupied by some protists., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
22. Metabolic stringent response in intracellular stages of Leishmania.
- Author
-
Saunders EC, Sernee MF, Ralton JE, and McConville MJ
- Subjects
- Animals, Fatty Acids, Macrophages, Phagosomes, Leishmania genetics, Parasites
- Abstract
Leishmania are unusual in being able to survive long-term in the mature phagolysosome compartment of macrophages and other phagocytic cells in their mammalian hosts. Key to their survival in this niche, Leishmania amastigotes switch to a slow growth state and activate a stringent metabolic response. The stringent metabolic response may be triggered by multiple stresses and is associated with decreased metabolic fluxes, restricted use of sugars and fatty acids as carbon sources and increased dependence on metabolic homeostasis pathways. Heterogeneity in expression of the Leishmania stringent response occurs in vivo reflects temporal and spatial heterogeneity in lesion tissues and includes non-dividing dormant stages. This response underpins the capacity of these parasites to maintain long-term chronic infections and survive drug treatments., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
23. Coordinated action of multiple transporters in the acquisition of essential cationic amino acids by the intracellular parasite Toxoplasma gondii.
- Author
-
Fairweather SJ, Rajendran E, Blume M, Javed K, Steinhöfel B, McConville MJ, Kirk K, Bröer S, and van Dooren GG
- Subjects
- 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, Amino Acid Transport Systems, Basic metabolism, Amino Acids, Essential metabolism, Fibroblasts metabolism, Oocytes metabolism, Protozoan Proteins metabolism, Toxoplasmosis metabolism
- 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., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
24. Malaria thriving on steroids.
- Author
-
McConville MJ and Engwerda CR
- Published
- 2021
- Full Text
- View/download PDF
25. Identification of Metabolically Quiescent Leishmania mexicana Parasites in Peripheral and Cured Dermal Granulomas Using Stable Isotope Tracing Imaging Mass Spectrometry.
- Author
-
Kloehn J, Boughton BA, Saunders EC, O'Callaghan S, Binger KJ, and McConville MJ
- Subjects
- Animals, Disease Models, Animal, Female, Isotope Labeling, Leishmaniasis, Cutaneous pathology, Macrophages parasitology, Mice, Mice, Inbred BALB C, Muscles parasitology, Muscles pathology, Skin parasitology, Deuterium Oxide, Granuloma parasitology, Host-Parasite Interactions, Image Processing, Computer-Assisted methods, Leishmania mexicana metabolism, Leishmaniasis, Cutaneous parasitology, Mass Spectrometry methods, Skin pathology
- Abstract
Leishmania are sandfly-transmitted protists that induce granulomatous lesions in their mammalian host. Although infected host cells in these tissues can exist in different activation states, the extent to which intracellular parasites stages also exist in different growth or physiological states remains poorly defined. Here, we have mapped the spatial distribution of metabolically quiescent and active subpopulations of Leishmania mexicana in dermal granulomas in susceptible BALB/c mice, using in vivo heavy water labeling and ultra high-resolution imaging mass spectrometry. Quantitation of the rate of turnover of parasite and host-specific lipids at high spatial resolution, suggested that the granuloma core comprised mixed populations of metabolically active and quiescent parasites. Unexpectedly, a significant population of metabolically quiescent parasites was also identified in the surrounding collagen-rich, dermal mesothelium. Mesothelium-like tissues harboring quiescent parasites progressively replaced macrophage-rich granuloma tissues following treatment with the first-line drug, miltefosine. In contrast to the granulomatous tissue, neither the mesothelium nor newly deposited tissue sequestered miltefosine. These studies suggest that the presence of quiescent parasites in acute granulomatous tissues, together with the lack of miltefosine accumulation in cured lesion tissue, may contribute to drug failure and nonsterile cure. IMPORTANCE Many microbial pathogens switch between different growth and physiological states in vivo in order to adapt to local nutrient levels and host microbicidal responses. Heterogeneity in microbial growth and metabolism may also contribute to nongenetic mechanisms of drug resistance and drug failure. In this study, we have developed a new approach for measuring spatial heterogeneity in microbial metabolism in vivo using a combination of heavy water (
2 H2 O) labeling and imaging mass spectrometry. Using this approach, we show that lesions contain a patchwork of metabolically distinct parasite populations, while the underlying dermal tissues contain a large population of metabolically quiescent parasites. Quiescent parasites also dominate drug-depleted tissues in healed animals, providing an explanation for failure of some first line drugs to completely eradicate parasites. This approach is broadly applicable to study the metabolic and growth dynamics in other host-pathogen interactions., (Copyright © 2021 Kloehn et al.)- Published
- 2021
- Full Text
- View/download PDF
26. Non-canonical metabolic pathways in the malaria parasite detected by isotope-tracing metabolomics.
- Author
-
Cobbold SA, V Tutor M, Frasse P, McHugh E, Karnthaler M, Creek DJ, Odom John A, Tilley L, Ralph SA, and McConville MJ
- Subjects
- 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, Isotope Labeling, Metabolic Networks and Pathways, Metabolomics, Parasites metabolism, Plasmodium falciparum metabolism
- 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., (© 2021 The Authors. Published under the terms of the CC BY 4.0 license.)- Published
- 2021
- Full Text
- View/download PDF
27. The Key Glycolytic Enzyme Phosphofructokinase Is Involved in Resistance to Antiplasmodial Glycosides.
- Author
-
Fisher GM, Cobbold SA, Jezewski A, Carpenter EF, Arnold M, Cowell AN, Tjhin ET, Saliba KJ, Skinner-Adams TS, Lee MCS, Odom John A, Winzeler EA, McConville MJ, Poulsen SA, and Andrews KT
- Subjects
- Alleles, Antimalarials chemistry, Dose-Response Relationship, Drug, Drug Resistance, Erythrocytes metabolism, Erythrocytes parasitology, Glycolysis, Glycosides chemistry, Metabolomics methods, Models, Molecular, Molecular Structure, Parasitic Sensitivity Tests, Phosphofructokinases genetics, Plasmodium falciparum genetics, Polymorphism, Single Nucleotide, Protein Conformation, Structure-Activity Relationship, Antimalarials pharmacology, Glycosides pharmacology, Phosphofructokinases metabolism, Plasmodium falciparum drug effects, Plasmodium falciparum enzymology
- Abstract
Plasmodium parasites rely heavily on glycolysis for ATP production and for precursors for essential anabolic pathways, such as the methylerythritol phosphate (MEP) pathway. Here, we show that mutations in the Plasmodium falciparum glycolytic enzyme, phosphofructokinase ( Pf PFK9), are associated with in vitro resistance to a primary sulfonamide glycoside (PS-3). Flux through the upper glycolysis pathway was significantly reduced in PS-3-resistant parasites, which was associated with reduced ATP levels but increased flux into the pentose phosphate pathway. PS-3 may directly or indirectly target enzymes in these pathways, as PS-3-treated parasites had elevated levels of glycolytic and tricarboxylic acid (TCA) cycle intermediates. PS-3 resistance also led to reduced MEP pathway intermediates, and PS-3-resistant parasites were hypersensitive to the MEP pathway inhibitor, fosmidomycin. Overall, this study suggests that PS-3 disrupts core pathways in central carbon metabolism, which is compensated for by mutations in Pf PFK9, highlighting a novel metabolic drug resistance mechanism in P. falciparum IMPORTANCE Malaria, caused by Plasmodium parasites, continues to be a devastating global health issue, causing 405,000 deaths and 228 million cases in 2018. Understanding key metabolic processes in malaria parasites is critical to the development of new drugs to combat this major infectious disease. The Plasmodium glycolytic pathway is essential to the malaria parasite, providing energy for growth and replication and supplying important biomolecules for other essential Plasmodium anabolic pathways. Despite this overreliance on glycolysis, no current drugs target glycolysis, and there is a paucity of information on critical glycolysis targets. Our work addresses this unmet need, providing new mechanistic insights into this key pathway., (Copyright © 2020 Fisher et al.)
- Published
- 2020
- Full Text
- View/download PDF
28. Reprogrammed mRNA translation drives resistance to therapeutic targeting of ribosome biogenesis.
- Author
-
Kusnadi EP, Trigos AS, Cullinane C, Goode DL, Larsson O, Devlin JR, Chan KT, De Souza DP, McConville MJ, McArthur GA, Thomas G, Sanij E, Poortinga G, Hannan RD, Hannan KM, Kang J, and Pearson RB
- Subjects
- 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, Neoplasms metabolism, Protein Biosynthesis genetics, Protein Biosynthesis physiology, Ribosomes metabolism, Transcription, Genetic drug effects
- 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., (© 2020 The Authors. Published under the terms of the CC BY NC ND 4.0 license.)
- Published
- 2020
- Full Text
- View/download PDF
29. Immunometabolism of Leishmania granulomas.
- Author
-
Saunders EC and McConville MJ
- Subjects
- Humans, Signal Transduction, Cellular Reprogramming, Granuloma parasitology, Leishmania metabolism, Macrophages immunology, Macrophages metabolism
- Abstract
Leishmania are parasitic protists that cause a spectrum of diseases in humans characterized by the formation of granulomatous lesions in the skin or other tissues, such as liver and spleen. The extent to which Leishmania granulomas constrain or promote parasite growth is critically dependent on the host T-helper type 1/T-helper type 2 immune response and the localized functional polarization of infected and noninfected macrophages toward a classically (M1) or alternatively (M2) activated phenotype. Recent studies have shown that metabolic reprograming of M1 and M2 macrophages underpins the capacity of these cells to act as permissive or nonpermissive host reservoirs, respectively. In this review, we highlight the metabolic requirements of Leishmania amastigotes and the evidence that these parasites induce and/or exploit metabolic reprogramming of macrophage metabolism. We also focus on recent studies highlighting the role of key macrophage metabolic signaling pathways, such as mechanistic target of rapamycin, adenosine monophosphate-activated protein kinase and peroxisome proliferator receptor gamma in regulating the pathological progression of Leishmania granulomas. These studies highlight the intimate connectivity between Leishmania and host cell metabolism, the need to investigate these interactions in vivo and the potential to exploit host cell metabolic signaling pathways in developing new host-directed therapies., (© 2020 Australian and New Zealand Society for Immunology Inc.)
- Published
- 2020
- Full Text
- View/download PDF
30. Modulation of acyl-carnitines, the broad mechanism behind Wolbachia -mediated inhibition of medically important flaviviruses in Aedes aegypti .
- Author
-
Manokaran G, Flores HA, Dickson CT, Narayana VK, Kanojia K, Dayalan S, Tull D, McConville MJ, Mackenzie JM, and Simmons CP
- Subjects
- Aedes chemistry, Aedes metabolism, Animals, Carnitine chemistry, Female, Mosquito Vectors chemistry, Mosquito Vectors metabolism, Mosquito Vectors microbiology, Mosquito Vectors virology, Aedes microbiology, Aedes virology, Carnitine metabolism, Wolbachia physiology, Zika Virus physiology
- 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., Competing Interests: The authors declare no competing interest., (Copyright © 2020 the Author(s). Published by PNAS.)
- Published
- 2020
- Full Text
- View/download PDF
31. Microbe-Metabolite Associations Linked to the Rebounding Murine Gut Microbiome Postcolonization with Vancomycin-Resistant Enterococcus faecium.
- Author
-
Mu A, Carter GP, Li L, Isles NS, Vrbanac AF, Morton JT, Jarmusch AK, De Souza DP, Narayana VK, Kanojia K, Nijagal B, McConville MJ, Knight R, Howden BP, and Stinear TP
- Abstract
Vancomycin-resistant Enterococcus faecium (VREfm) is an emerging antibiotic-resistant pathogen. Strain-level investigations are beginning to reveal the molecular mechanisms used by VREfm to colonize regions of the human bowel. However, the role of commensal bacteria during VREfm colonization, in particular following antibiotic treatment, remains largely unknown. We employed amplicon 16S rRNA gene sequencing and metabolomics in a murine model system to try and investigate functional roles of the gut microbiome during VREfm colonization. First-order taxonomic shifts between Bacteroidetes and Tenericutes within the gut microbial community composition were detected both in response to pretreatment using ceftriaxone and to subsequent VREfm challenge. Using neural networking approaches to find cooccurrence profiles of bacteria and metabolites, we detected key metabolome features associated with butyric acid during and after VREfm colonization. These metabolite features were associated with Bacteroides , indicative of a transition toward a preantibiotic naive microbiome. This study shows the impacts of antibiotics on the gut ecosystem and the progression of the microbiome in response to colonization with VREfm. Our results offer insights toward identifying potential nonantibiotic alternatives to eliminate VREfm through metabolic reengineering to preferentially select for Bacteroides IMPORTANCE This study demonstrates the importance and power of linking bacterial composition profiling with metabolomics to find the interactions between commensal gut bacteria and a specific pathogen. Knowledge from this research will inform gut microbiome engineering strategies, with the aim of translating observations from animal models to human-relevant therapeutic applications., (Copyright © 2020 Mu et al.)
- Published
- 2020
- Full Text
- View/download PDF
32. The natural function of the malaria parasite's chloroquine resistance transporter.
- Author
-
Shafik SH, Cobbold SA, Barkat K, Richards SN, Lancaster NS, Llinás M, Hogg SJ, Summers RL, McConville MJ, and Martin RE
- Subjects
- Animals, Biological Transport, Active, Drug Resistance genetics, Female, Host-Parasite Interactions genetics, Host-Parasite Interactions physiology, Humans, Malaria, Falciparum drug therapy, Malaria, Falciparum metabolism, Malaria, Falciparum parasitology, Membrane Transport Proteins genetics, Models, Biological, Mutant Proteins genetics, Mutant Proteins metabolism, Oligopeptides metabolism, Oocytes metabolism, Plasmodium falciparum genetics, Protein Transport, Protozoan Proteins genetics, Xenopus laevis, Antimalarials pharmacology, Chloroquine pharmacology, Membrane Transport Proteins metabolism, Plasmodium falciparum drug effects, Plasmodium falciparum metabolism, Protozoan Proteins metabolism
- Abstract
The Plasmodium falciparum chloroquine resistance transporter (PfCRT) is a key contributor to multidrug resistance and is also essential for the survival of the malaria parasite, yet its natural function remains unresolved. We identify host-derived peptides of 4-11 residues, varying in both charge and composition, as the substrates of PfCRT in vitro and in situ, and show that PfCRT does not mediate the non-specific transport of other metabolites and/or ions. We find that drug-resistance-conferring mutations reduce both the peptide transport capacity and substrate range of PfCRT, explaining the impaired fitness of drug-resistant parasites. Our results indicate that PfCRT transports peptides from the lumen of the parasite's digestive vacuole to the cytosol, thereby providing a source of amino acids for parasite metabolism and preventing osmotic stress of this organelle. The resolution of PfCRT's native substrates will aid the development of drugs that target PfCRT and/or restore the efficacy of existing antimalarials.
- Published
- 2020
- Full Text
- View/download PDF
33. A Sulfoglycolytic Entner-Doudoroff Pathway in Rhizobium leguminosarum bv. trifolii SRDI565.
- Author
-
Li J, Epa R, Scott NE, Skoneczny D, Sharma M, Snow AJD, Lingford JP, Goddard-Borger ED, Davies GJ, McConville MJ, and Williams SJ
- Subjects
- Proteomics, Bacterial Proteins metabolism, Glycerol metabolism, Methylglucosides metabolism, Proteome metabolism, Rhizobium leguminosarum metabolism
- Abstract
Rhizobia are nitrogen-fixing bacteria that engage in symbiotic relationships with plant hosts but can also persist as free-living bacteria in the soil and rhizosphere. Here, we show that free-living Rhizobium leguminosarum SRDI565 can grow on the sulfosugar sulfoquinovose (SQ) or the related glycoside SQ-glycerol using a sulfoglycolytic Entner-Doudoroff (sulfo-ED) pathway, resulting in production of sulfolactate (SL) as the major metabolic end product. Comparative proteomics supports the involvement of a sulfo-ED operon encoding an ABC transporter, sulfo-ED enzymes, and an SL exporter. Consistent with an oligotrophic lifestyle, proteomics data revealed little change in expression of the sulfo-ED proteins during growth on SQ versus mannitol, a result confirmed through biochemical assay of sulfoquinovosidase activity in cell lysates. Metabolomics analysis showed that growth on SQ involves gluconeogenesis to satisfy metabolic requirements for glucose-6-phosphate and fructose-6-phosphate. Metabolomics analysis also revealed the unexpected production of small amounts of sulfofructose and 2,3-dihydroxypropanesulfonate, which are proposed to arise from promiscuous activities of the glycolytic enzyme phosphoglucose isomerase and a nonspecific aldehyde reductase, respectively. The discovery of a rhizobium isolate with the ability to degrade SQ builds our knowledge of how these important symbiotic bacteria persist within soil. IMPORTANCE Sulfonate sulfur is a major form of organic sulfur in soils but requires biomineralization before it can be utilized by plants. Very little is known about the biochemical processes used to mobilize sulfonate sulfur. We show that a rhizobial isolate from soil, Rhizobium leguminosarum SRDI565, possesses the ability to degrade the abundant phototroph-derived carbohydrate sulfonate SQ through a sulfoglycolytic Entner-Doudoroff pathway. Proteomics and metabolomics demonstrated the utilization of this pathway during growth on SQ and provided evidence for gluconeogenesis. Unexpectedly, off-cycle sulfoglycolytic species were also detected, pointing to the complexity of metabolic processes within cells under conditions of sulfoglycolysis. Thus, rhizobial metabolism of the abundant sulfosugar SQ may contribute to persistence of the bacteria in the soil and to mobilization of sulfur in the pedosphere., (Copyright © 2020 American Society for Microbiology.)
- Published
- 2020
- Full Text
- View/download PDF
34. Publisher Correction: Metabolic characteristics of CD8 + T cell subsets in young and aged individuals are not predictive of functionality.
- Author
-
Quinn KM, Hussain T, Kraus F, Formosa LE, Lam WK, Dagley MJ, Saunders EC, Assmus LM, Wynne-Jones E, Loh L, van de Sandt CE, Cooper L, Good-Jacobson KL, Kedzierska K, Mackay LK, McConville MJ, Ramm G, Ryan MT, and La Gruta NL
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
- Full Text
- View/download PDF
35. Metabolic characteristics of CD8 + T cell subsets in young and aged individuals are not predictive of functionality.
- Author
-
Quinn KM, Hussain T, Kraus F, Formosa LE, Lam WK, Dagley MJ, Saunders EC, Assmus LM, Wynne-Jones E, Loh L, van de Sandt CE, Cooper L, Good-Jacobson KL, Kedzierska K, Mackay LK, McConville MJ, Ramm G, Ryan MT, and La Gruta NL
- Subjects
- Adult, Aged, Animals, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes ultrastructure, Cell Differentiation immunology, Cell Proliferation, Disease Models, Animal, Female, Humans, Influenza A virus immunology, Influenza, Human blood, Influenza, Human immunology, Influenza, Human virology, Male, Mice, Microscopy, Electron, Transmission, Mitochondria metabolism, Mitochondria ultrastructure, T-Lymphocyte Subsets cytology, T-Lymphocyte Subsets metabolism, T-Lymphocyte Subsets ultrastructure, Young Adult, Aging immunology, CD8-Positive T-Lymphocytes immunology, Immunologic Memory, T-Lymphocyte Subsets immunology
- Abstract
Virtual memory T (T
VM ) cells are antigen-naïve CD8+ T cells that exist in a semi-differentiated state and exhibit marked proliferative dysfunction in advanced age. High spare respiratory capacity (SRC) has been proposed as a defining metabolic characteristic of antigen-experienced memory T (TMEM ) cells, facilitating rapid functionality and survival. Given the semi-differentiated state of TVM cells and their altered functionality with age, here we investigate TVM cell metabolism and its association with longevity and functionality. Elevated SRC is a feature of TVM , but not TMEM , cells and it increases with age in both subsets. The elevated SRC observed in aged mouse TVM cells and human CD8+ T cells from older individuals is associated with a heightened sensitivity to IL-15. We conclude that elevated SRC is a feature of TVM , but not TMEM , cells, is driven by physiological levels of IL-15, and is not indicative of enhanced functionality in CD8+ T cells.- Published
- 2020
- Full Text
- View/download PDF
36. Leishmania Encodes a Bacterium-like 2,4-Dienoyl-Coenzyme A Reductase That Is Required for Fatty Acid β-Oxidation and Intracellular Parasite Survival.
- Author
-
Semini G, Paape D, Blume M, Sernee MF, Peres-Alonso D, Calvignac-Spencer S, Döllinger J, Jehle S, Saunders E, McConville MJ, and Aebischer T
- Subjects
- Amino Acid Sequence, Animals, Fatty Acid Desaturases genetics, Female, Leishmania major growth & development, Leishmania mexicana genetics, Macrophages parasitology, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Oxidation-Reduction, Phylogeny, Fatty Acid Desaturases metabolism, Fatty Acids metabolism, Leishmania major enzymology, Leishmania major genetics
- Abstract
Leishmania spp. are protozoan parasites that cause a spectrum of important diseases in humans. These parasites develop as extracellular promastigotes in the digestive tract of their insect vectors and as obligate intracellular amastigotes that infect macrophages and other phagocytic cells in their vertebrate hosts. Promastigote-to-amastigote differentiation is associated with marked changes in metabolism, including the upregulation of enzymes involved in fatty acid β-oxidation, which may reflect adaptation to the intracellular niche. Here, we have investigated the function of one of these enzymes, a putative 2,4-dienoyl-coenzyme A (CoA) reductase (DECR), which is specifically required for the β-oxidation of polyunsaturated fatty acids. The Leishmania DECR shows close homology to bacterial DECR proteins, suggesting that it was acquired by lateral gene transfer. It is present in other trypanosomatids that have obligate intracellular stages (i.e., Trypanosoma cruzi and Angomonas ) but is absent from dixenous parasites with an exclusively extracellular lifestyle (i.e., Trypanosoma brucei ). A DECR-green fluorescent protein (GFP) fusion protein was localized to the mitochondrion in both promastigote and amastigote stages, and the levels of expression increased in the latter stages. A Leishmania major Δ decr null mutant was unable to catabolize unsaturated fatty acids and accumulated the intermediate 2,4-decadienoyl-CoA, confirming DECR's role in β-oxidation. Strikingly, the L. major Δ decr mutant was unable to survive in macrophages and was avirulent in BALB/c mice. These findings suggest that β-oxidation of polyunsaturated fatty acids is essential for intracellular parasite survival and that the bacterial origin of key enzymes in this pathway could be exploited in developing new therapies. IMPORTANCE The Trypanosomatidae are protozoan parasites that infect insects, plants, and animals and have evolved complex monoxenous (single host) and dixenous (two hosts) lifestyles. A number of species of Trypanosomatidae, including Leishmania spp., have evolved the capacity to survive within intracellular niches in vertebrate hosts. The adaptations, metabolic and other, that are associated with development of intracellular lifestyles remain poorly defined. We show that genomes of Leishmania and Trypanosomatidae that can survive intracellularly encode a 2,4-dienoyl-CoA reductase that is involved in catabolism of a subclass of fatty acids. The trypanosomatid enzyme shows closest similarity to the corresponding bacterial enzymes and is located in the mitochondrion and essential for intracellular growth of Leishmania The findings suggest that acquisition of this gene by lateral gene transfer from bacteria by ancestral monoxenous Trypanosomatidae likely contributed to the development of a dixenous lifestyle of these parasites., (Copyright © 2020 Semini et al.)
- Published
- 2020
- Full Text
- View/download PDF
37. EirA Is a Novel Protein Essential for Intracellular Replication of Coxiella burnetii.
- Author
-
Kuba M, Neha N, Newton P, Lee YW, Bennett-Wood V, Hachani A, De Souza DP, Nijagal B, Dayalan S, Tull D, McConville MJ, Sansom FM, and Newton HJ
- Subjects
- Bacterial Proteins metabolism, Cell Membrane, Humans, Metabolome, Metabolomics methods, Microbial Viability, Models, Biological, Mutation, Protein Transport, Vacuoles microbiology, Virulence genetics, Virulence Factors genetics, Bacterial Proteins genetics, Coxiella burnetii physiology, Host-Pathogen Interactions genetics, Q Fever microbiology
- Abstract
The zoonotic bacterial pathogen Coxiella burnetii is the causative agent of Q fever, a febrile illness which can cause a serious chronic infection. C. burnetii is a unique intracellular bacterium which replicates within host lysosome-derived vacuoles. The ability of C. burnetii to replicate within this normally hostile compartment is dependent on the activity of the Dot/Icm type 4B secretion system. In a previous study, a transposon mutagenesis screen suggested that the disruption of the gene encoding the novel protein CBU2072 rendered C. burnetii incapable of intracellular replication. This protein, subsequently named EirA ( e ssential for i ntracellular r eplication A), is indispensable for intracellular replication and virulence, as demonstrated by infection of human cell lines and in vivo infection of Galleria mellonella The putative N-terminal signal peptide is essential for protein function but is not required for localization of EirA to the bacterial inner membrane compartment and axenic culture supernatant. In the absence of EirA, C. burnetii remains viable but nonreplicative within the host phagolysosome, as coinfection with C. burnetii expressing native EirA rescues the replicative defect in the mutant strain. In addition, while the bacterial ultrastructure appears to be intact, there is an altered metabolic profile shift in the absence of EirA, suggesting that EirA may impact overall metabolism. Most strikingly, in the absence of EirA, Dot/Icm effector translocation was inhibited even when EirA-deficient C. burnetii replicated in the wild type (WT)-supported Coxiella containing vacuoles. EirA may therefore have a novel role in the control of Dot/Icm activity and represent an important new therapeutic target., (Copyright © 2020 American Society for Microbiology.)
- Published
- 2020
- Full Text
- View/download PDF
38. Unique properties of a subset of human pluripotent stem cells with high capacity for self-renewal.
- Author
-
Lau KX, Mason EA, Kie J, De Souza DP, Kloehn J, Tull D, McConville MJ, Keniry A, Beck T, Blewitt ME, Ritchie ME, Naik SH, Zalcenstein D, Korn O, Su S, Romero IG, Spruce C, Baker CL, McGarr TC, Wells CA, and Pera MF
- Subjects
- Animals, Cell Differentiation, Chromatin metabolism, DNA Methylation, Epigenome, Flow Cytometry, Fluorescent Antibody Technique, Indirect, G1 Phase, Germ Layers metabolism, Glycolysis, Humans, MAP Kinase Signaling System, Metabolomics, Mice, Mitochondria metabolism, Oxidative Phosphorylation, RNA-Seq, Signal Transduction, Embryonic Stem Cells cytology, Germ Layers cytology, Pluripotent Stem Cells cytology
- Abstract
Archetypal human pluripotent stem cells (hPSC) are widely considered to be equivalent in developmental status to mouse epiblast stem cells, which correspond to pluripotent cells at a late post-implantation stage of embryogenesis. Heterogeneity within hPSC cultures complicates this interspecies comparison. Here we show that a subpopulation of archetypal hPSC enriched for high self-renewal capacity (ESR) has distinct properties relative to the bulk of the population, including a cell cycle with a very low G1 fraction and a metabolomic profile that reflects a combination of oxidative phosphorylation and glycolysis. ESR cells are pluripotent and capable of differentiation into primordial germ cell-like cells. Global DNA methylation levels in the ESR subpopulation are lower than those in mouse epiblast stem cells. Chromatin accessibility analysis revealed a unique set of open chromatin sites in ESR cells. RNA-seq at the subpopulation and single cell levels shows that, unlike mouse epiblast stem cells, the ESR subset of hPSC displays no lineage priming, and that it can be clearly distinguished from gastrulating and extraembryonic cell populations in the primate embryo. ESR hPSC correspond to an earlier stage of post-implantation development than mouse epiblast stem cells.
- Published
- 2020
- Full Text
- View/download PDF
39. MtrP, a putative methyltransferase in Corynebacteria, is required for optimal membrane transport of trehalose mycolates.
- Author
-
Rainczuk AK, Klatt S, Yamaryo-Botté Y, Brammananth R, McConville MJ, Coppel RL, and Crellin PK
- Subjects
- Biological Transport, Corynebacterium glutamicum genetics, Corynebacterium glutamicum metabolism, Methyltransferases genetics, Mutation, Mycobacterium tuberculosis genetics, Sequence Homology, Nucleic Acid, Cell Membrane metabolism, Corynebacterium glutamicum cytology, Corynebacterium glutamicum enzymology, Methyltransferases metabolism, Mycolic Acids chemistry, Trehalose chemistry, Trehalose metabolism
- 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., (© 2020 Rainczuk et al.)
- Published
- 2020
- Full Text
- View/download PDF
40. Correction: Function of hTim8a in complex IV assembly in neuronal cells provides insight into pathomechanism underlying Mohr-Tranebjærg syndrome.
- Author
-
Kang Y, Anderson AJ, Jackson TD, Palmer CS, De Souza DP, Fujihara KM, Stait T, Frazier AE, Clemons NJ, Tull D, Thorburn DR, McConville MJ, Ryan MT, Stroud DA, and Stojanovski D
- Published
- 2020
- Full Text
- View/download PDF
41. Metabolomics Provide Sensitive Insights into the Impacts of Low Level Environmental Contamination on Fish Health-A Pilot Study.
- Author
-
Long SM, Tull DL, De Souza DP, Kouremenos KA, Dayalan S, McConville MJ, Hassell KL, Pettigrove VJ, and Gagnon MM
- Abstract
This exploratory study aims to investigate the health of sand flathead ( Platycephalus bassensis ) sampled from five sites in Port Phillip Bay, Australia using gas chromatography-mass spectrometry (GC-MS) metabolomics approaches. Three of the sites were the recipients of industrial, agricultural, and urban run-off and were considered urban sites, while the remaining two sites were remote from contaminant inputs, and hence classed as rural sites. Morphological parameters as well as polar and free fatty acid metabolites were used to investigate inter-site differences in fish health. Significant differences in liver somatic index (LSI) and metabolite abundance were observed between the urban and rural sites. Differences included higher LSI, an increased abundance of amino acids and energy metabolites, and reduced abundance of free fatty acids at the urban sites compared to the rural sites. These differences might be related to the additional energy requirements needed to cope with low-level contaminant exposure through energy demanding processes such as detoxification and antioxidant responses as well as differences in diet between the sites. In this study, we demonstrate that metabolomics approaches can offer a greater level of sensitivity compared to traditional parameters such as physiological parameters or biochemical markers of fish health, most of which showed no or little inter-site differences in the present study. Moreover, the metabolite responses are more informative than traditional biomarkers in terms of biological significance as disturbances in specific metabolic pathways can be identified.
- Published
- 2020
- Full Text
- View/download PDF
42. Analysis of the Physiological and Metabolic State of Leishmania Using Heavy Water Labeling.
- Author
-
Kloehn J and McConville MJ
- Subjects
- Animals, DNA, Protozoan analysis, DNA, Protozoan chemistry, DNA, Protozoan metabolism, Disease Models, Animal, Female, Gas Chromatography-Mass Spectrometry methods, Humans, Leishmania mexicana isolation & purification, Leishmaniasis, Cutaneous immunology, Leishmaniasis, Cutaneous parasitology, Leishmaniasis, Cutaneous pathology, Life Cycle Stages physiology, Metabolomics methods, Mice, Polysaccharides analysis, Polysaccharides chemistry, Polysaccharides metabolism, Protozoan Proteins analysis, Protozoan Proteins chemistry, Protozoan Proteins metabolism, RNA, Protozoan analysis, RNA, Protozoan chemistry, RNA, Protozoan metabolism, Skin parasitology, Deuterium Oxide chemistry, Isotope Labeling methods, Leishmania mexicana metabolism, Leishmaniasis, Cutaneous diagnosis
- Abstract
This protocol describes the use of heavy water (
2 H2 O) labeling to determine the growth rate and metabolic state of Leishmania parasites in culture and in infected animals. In vitro labeling studies are undertaken by cultivating defined parasite developmental stages in standard medium supplemented with 5%2 H2 O, resulting in the incorporation of deuterium (2 H) into a range of metabolic precursors used in macromolecule (DNA, RNA, protein, lipid, and glycan) synthesis. The rate of turnover of different parasite macromolecules can subsequently be determined by analysis of deuterium enrichment in the different constituents of these macromolecules by gas chromatography-mass spectrometry (GC-MS). To measure the growth rate and physiological state of parasite stages in lesion tissue, infected mice were provided with 9%2 H2 O in their drinking water for various periods of time and2 H-enrichment in the macromolecular constituents of isolated lesion-derived parasite stages determined by GC-MS. This protocol provides quantitative information on key cellular processes, such as replication (DNA turnover), transcription (RNA turnover), translation (protein turnover), membrane biogenesis (lipid turnover), and central carbon metabolism (glycan turnover) that define the growth state and phenome of different parasite stages in vitro and in vivo. This approach can be used to assess the impact of host immune responses on parasite growth and physiology (using different Leishmania strains/species, mouse lines), characterize different parasite populations during chronic and acute infections, and assess parasite responses to drug treatments. It is also broadly applicable to other microbial pathogens.- Published
- 2020
- Full Text
- View/download PDF
43. Metabolomic Analysis of Toxoplasma gondii Tachyzoites.
- Author
-
King EFB, Cobbold SA, Uboldi AD, Tonkin CJ, and McConville MJ
- Subjects
- Animals, Chromatography, Liquid, Fibroblasts parasitology, Foreskin cytology, Gas Chromatography-Mass Spectrometry, Humans, Male, Mass Spectrometry, Metabolomics, Software, Toxoplasmosis, Toxoplasma metabolism, Toxoplasma pathogenicity
- Abstract
This protocol describes the use of
13 C-stable isotope labeling, combined with metabolite profiling, to investigate the metabolism of the tachyzoite stage of the protozoan parasite Toxoplasma gondii. T. gondii tachyzoites can infect any nucleated cell in their vertebrate (including human) hosts, and utilize a range of carbon sources that freely permeate across the limiting membrane of the specialized vacuole within which they proliferate. Methods for cultivating tachyzoites in human foreskin fibroblasts and metabolically labeling intracellular and naturally egressed tachyzoites with a range of13 C-labeled carbon sources are described. Parasites are harvested and purified from host metabolites, with rapid metabolic quenching and13 C-enrichment in intracellular polar metabolites quantified by gas chromatography-mass spectrometry (GC-MS) and liquid chromatography-mass spectrometry (LC-MS). The mass isotopomer distribution of key metabolites is determined using DExSI software. This method can be used to measure perturbations in parasite metabolism induced by drug inhibition or genetic manipulation of enzyme levels and is broadly applicable to other cultured or intracellular parasite stages.- Published
- 2020
- Full Text
- View/download PDF
44. The Metabolite Repair Enzyme Phosphoglycolate Phosphatase Regulates Central Carbon Metabolism and Fosmidomycin Sensitivity in Plasmodium falciparum.
- Author
-
Dumont L, Richardson MB, van der Peet P, Marapana DS, Triglia T, Dixon MWA, Cowman AF, Williams SJ, Tilley L, McConville MJ, and Cobbold SA
- Subjects
- Fosfomycin pharmacology, Glycolysis drug effects, Humans, Lactates pharmacology, Malaria, Falciparum drug therapy, Malaria, Falciparum metabolism, Sugar Acids pharmacology, Antimalarials pharmacology, Carbon metabolism, Drug Resistance drug effects, Fosfomycin analogs & derivatives, Phosphoric Monoester Hydrolases metabolism, Plasmodium falciparum drug effects, Plasmodium falciparum metabolism
- Abstract
Members of the haloacid dehalogenase (HAD) family of metabolite phosphatases play an important role in regulating multiple pathways in Plasmodium falciparum central carbon metabolism. We show that the P. falciparum HAD protein, phosphoglycolate phosphatase (PGP), regulates glycolysis and pentose pathway flux in asexual blood stages via detoxifying the damaged metabolite 4-phosphoerythronate (4-PE). Disruption of the P. falciparum pgp gene caused accumulation of two previously uncharacterized metabolites, 2-phospholactate and 4-PE. 4-PE is a putative side product of the glycolytic enzyme, glyceraldehyde-3-phosphate dehydrogenase, and its accumulation inhibits the pentose phosphate pathway enzyme, 6-phosphogluconate dehydrogenase (6-PGD). Inhibition of 6-PGD by 4-PE leads to an unexpected feedback response that includes increased flux into the pentose phosphate pathway as a result of partial inhibition of upper glycolysis, with concomitant increased sensitivity to antimalarials that target pathways downstream of glycolysis. These results highlight the role of metabolite detoxification in regulating central carbon metabolism and drug sensitivity of the malaria parasite. IMPORTANCE The malaria parasite has a voracious appetite, requiring large amounts of glucose and nutrients for its rapid growth and proliferation inside human red blood cells. The host cell is resource rich, but this is a double-edged sword; nutrient excess can lead to undesirable metabolic reactions and harmful by-products. Here, we demonstrate that the parasite possesses a metabolite repair enzyme (PGP) that suppresses harmful metabolic by-products (via substrate dephosphorylation) and allows the parasite to maintain central carbon metabolism. Loss of PGP leads to the accumulation of two damaged metabolites and causes a domino effect of metabolic dysregulation. Accumulation of one damaged metabolite inhibits an essential enzyme in the pentose phosphate pathway, leading to substrate accumulation and secondary inhibition of glycolysis. This work highlights how the parasite coordinates metabolic flux by eliminating harmful metabolic by-products to ensure rapid proliferation in its resource-rich niche., (Copyright © 2019 Dumont et al.)
- Published
- 2019
- Full Text
- View/download PDF
45. The multifunctional enzyme S-adenosylhomocysteine/methylthioadenosine nucleosidase is a key metabolic enzyme in the virulence of Salmonella enterica var Typhimurium.
- Author
-
Husna AU, Wang N, Wilksch JJ, Newton HJ, Hocking DM, Hay ID, Cobbold SA, Davies MR, McConville MJ, Lithgow T, and Strugnell RA
- Subjects
- Animals, Bacterial Proteins genetics, Female, Gene Expression Regulation, Bacterial, Humans, Male, Mice, Mice, Inbred C57BL, Multifunctional Enzymes genetics, Multifunctional Enzymes metabolism, N-Glycosyl Hydrolases genetics, Purine-Nucleoside Phosphorylase genetics, S-Adenosylhomocysteine metabolism, Salmonella typhimurium genetics, Virulence, Bacterial Proteins metabolism, N-Glycosyl Hydrolases metabolism, Purine-Nucleoside Phosphorylase metabolism, Salmonella Infections microbiology, Salmonella typhimurium enzymology, Salmonella typhimurium pathogenicity
- Abstract
Key physiological differences between bacterial and mammalian metabolism provide opportunities for the development of novel antimicrobials. We examined the role of the multifunctional enzyme S-adenosylhomocysteine/Methylthioadenosine (SAH/MTA) nucleosidase (Pfs) in the virulence of S. enterica var Typhimurium (S. Typhimurium) in mice, using a defined Pfs deletion mutant (i.e. Δpfs). Pfs was essential for growth of S. Typhimurium in M9 minimal medium, in tissue cultured cells, and in mice. Studies to resolve which of the three known functions of Pfs were key to murine virulence suggested that downstream production of autoinducer-2, spermidine and methylthioribose were non-essential for Salmonella virulence in a highly sensitive murine model. Mass spectrometry revealed the accumulation of SAH in S. Typhimurium Δpfs and complementation of the Pfs mutant with the specific SAH hydrolase from Legionella pneumophila reduced SAH levels, fully restored growth ex vivo and the virulence of S. Typhimurium Δpfs for mice. The data suggest that Pfs may be a legitimate target for antimicrobial development, and that the key role of Pfs in bacterial virulence may be in reducing the toxic accumulation of SAH which, in turn, suppresses an undefined methyltransferase., (© 2019 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2019
- Full Text
- View/download PDF
46. Decreased K13 Abundance Reduces Hemoglobin Catabolism and Proteotoxic Stress, Underpinning Artemisinin Resistance.
- Author
-
Yang T, Yeoh LM, Tutor MV, Dixon MW, McMillan PJ, Xie SC, Bridgford JL, Gillett DL, Duffy MF, Ralph SA, McConville MJ, Tilley L, and Cobbold SA
- Subjects
- Humans, Mutation, Artemisinins metabolism, Hemoglobins genetics, Malaria, Falciparum drug therapy, Plasmodium falciparum genetics
- Abstract
Increased tolerance of Plasmodium falciparum to front-line artemisinin antimalarials (ARTs) is associated with mutations in Kelch13 (K13), although the precise role of K13 remains unclear. Here, we show that K13 mutations result in decreased expression of this protein, while mislocalization of K13 mimics resistance-conferring mutations, pinpointing partial loss of function of K13 as the relevant molecular event. K13-GFP is associated with ∼170 nm diameter doughnut-shaped structures at the parasite periphery, consistent with the location and dimensions of cytostomes. Moreover, the hemoglobin-peptide profile of ring-stage parasites is reduced when K13 is mislocalized. We developed a pulse-SILAC approach to quantify protein turnover and observe less disruption to protein turnover following ART exposure when K13 is mislocalized. Our findings suggest that K13 regulates digestive vacuole biogenesis and the uptake/degradation of hemoglobin and that ART resistance is mediated by a decrease in heme-dependent drug activation, less proteotoxicity, and increased survival of parasite ring stages., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
47. Function of hTim8a in complex IV assembly in neuronal cells provides insight into pathomechanism underlying Mohr-Tranebjærg syndrome.
- Author
-
Kang Y, Anderson AJ, Jackson TD, Palmer CS, De Souza DP, Fujihara KM, Stait T, Frazier AE, Clemons NJ, Tull D, Thorburn DR, McConville MJ, Ryan MT, Stroud DA, and Stojanovski D
- Subjects
- Apoptosis, Apoptosis Regulatory Proteins metabolism, Cell Line, Copper Transport Proteins metabolism, Humans, Membrane Transport Proteins deficiency, Mitochondrial Precursor Protein Import Complex Proteins, Oxidative Stress, Protein Interaction Maps, Deaf-Blind Disorders physiopathology, Dystonia physiopathology, Electron Transport Complex IV metabolism, Intellectual Disability physiopathology, Membrane Transport Proteins metabolism, Neurons metabolism, Optic Atrophy physiopathology, Protein Multimerization
- Abstract
Human Tim8a and Tim8b are members of an intermembrane space chaperone network, known as the small TIM family. Mutations in TIMM8A cause a neurodegenerative disease, Mohr-Tranebjærg syndrome (MTS), which is characterised by sensorineural hearing loss, dystonia and blindness. Nothing is known about the function of hTim8a in neuronal cells or how mutation of this protein leads to a neurodegenerative disease. We show that hTim8a is required for the assembly of Complex IV in neurons, which is mediated through a transient interaction with Complex IV assembly factors, in particular the copper chaperone COX17. Complex IV assembly defects resulting from loss of hTim8a leads to oxidative stress and changes to key apoptotic regulators, including cytochrome c, which primes cells for death. Alleviation of oxidative stress with Vitamin E treatment rescues cells from apoptotic vulnerability. We hypothesise that enhanced sensitivity of neuronal cells to apoptosis is the underlying mechanism of MTS., Competing Interests: YK, AA, TJ, CP, DD, KF, TS, AF, NC, DT, DT, MM, MR, DS, DS No competing interests declared, (© 2019, Kang et al.)
- Published
- 2019
- Full Text
- View/download PDF
48. Coxiella burnetii utilizes both glutamate and glucose during infection with glucose uptake mediated by multiple transporters.
- Author
-
Kuba M, Neha N, De Souza DP, Dayalan S, Newson JPM, Tull D, McConville MJ, Sansom FM, and Newton HJ
- Subjects
- Animals, Bacterial Proteins metabolism, Biological Transport, Coxiella burnetii pathogenicity, Gluconeogenesis physiology, Glycolysis physiology, HeLa Cells, Humans, Lepidoptera microbiology, Membrane Transport Proteins metabolism, THP-1 Cells, Coxiella burnetii metabolism, Glucose metabolism, Glutamic Acid metabolism, Host-Pathogen Interactions, Q Fever microbiology, Virulence physiology
- Abstract
Coxiella burnetii is a Gram-negative bacterium which causes Q fever, a complex and life-threatening infection with both acute and chronic presentations. C. burnetii invades a variety of host cell types and replicates within a unique vacuole derived from the host cell lysosome. In order to understand how C. burnetii survives within this intracellular niche, we have investigated the carbon metabolism of both intracellular and axenically cultivated bacteria. Both bacterial populations were shown to assimilate exogenous [13C]glucose or [13C]glutamate, with concomitant labeling of intermediates in glycolysis and gluconeogenesis, and in the TCA cycle. Significantly, the two populations displayed metabolic pathway profiles reflective of the nutrient availabilities within their propagated environments. Disruption of the C. burnetii glucose transporter, CBU0265, by transposon mutagenesis led to a significant decrease in [13C]glucose utilization but did not abolish glucose usage, suggesting that C. burnetii express additional hexose transporters which may be able to compensate for the loss of CBU0265. This was supported by intracellular infection of human cells and in vivo studies in the insect model showing loss of CBU0265 had no impact on intracellular replication or virulence. Using this mutagenesis and [13C]glucose labeling approach, we identified a second glucose transporter, CBU0347, the disruption of which also showed significant decreases in 13C-label incorporation but did not impact intracellular replication or virulence. Together, these analyses indicate that C. burnetii may use multiple carbon sources in vivo and exhibits greater metabolic flexibility than expected., (© 2019 The Author(s).)
- Published
- 2019
- Full Text
- View/download PDF
49. Autocrine IFN-I inhibits isocitrate dehydrogenase in the TCA cycle of LPS-stimulated macrophages.
- Author
-
De Souza DP, Achuthan A, Lee MK, Binger KJ, Lee MC, Davidson S, Tull DL, McConville MJ, Cook AD, Murphy AJ, Hamilton JA, and Fleetwood AJ
- Subjects
- Animals, Autocrine Communication, Citric Acid Cycle, Humans, Interleukin-10 metabolism, Isocitrate Dehydrogenase genetics, Isocitrate Dehydrogenase metabolism, Lipopolysaccharides pharmacology, Macrophage Activation, Macrophages drug effects, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Receptor, Interferon alpha-beta deficiency, Receptor, Interferon alpha-beta genetics, Receptor, Interferon alpha-beta metabolism, Succinates metabolism, Interferon Type I metabolism, Isocitrate Dehydrogenase antagonists & inhibitors, Macrophages immunology, Macrophages metabolism
- Abstract
Macrophage activation in response to LPS is coupled to profound metabolic changes, typified by accumulation of the TCA cycle intermediates citrate, itaconate, and succinate. We have identified that endogenous type I IFN controls the cellular citrate/α-ketoglutarate ratio and inhibits expression and activity of isocitrate dehydrogenase (IDH); and, via 13C-labeling studies, demonstrated that autocrine type I IFN controls carbon flow through IDH in LPS-activated macrophages. We also found that type I IFN-driven IL-10 contributes to inhibition of IDH activity and itaconate synthesis in LPS-stimulated macrophages. Our findings have identified the autocrine type I IFN pathway as being responsible for the inhibition of IDH in LPS-stimulated macrophages.
- Published
- 2019
- Full Text
- View/download PDF
50. A Family of Dual-Activity Glycosyltransferase-Phosphorylases Mediates Mannogen Turnover and Virulence in Leishmania Parasites.
- Author
-
Sernee MF, Ralton JE, Nero TL, Sobala LF, Kloehn J, Vieira-Lara MA, Cobbold SA, Stanton L, Pires DEV, Hanssen E, Males A, Ward T, Bastidas LM, van der Peet PL, Parker MW, Ascher DB, Williams SJ, Davies GJ, and McConville MJ
- Subjects
- Crystallography, X-Ray, Gene Transfer, Horizontal, Glycosyltransferases chemistry, Glycosyltransferases genetics, Mannans, Mannosyltransferases chemistry, Mannosyltransferases genetics, Models, Molecular, Oligosaccharides, Phosphorylases chemistry, Phosphorylases genetics, Protein Conformation, Thermotolerance, Virulence, Glycosyltransferases classification, Glycosyltransferases metabolism, Leishmania enzymology, Mannosyltransferases metabolism, Phosphorylases classification, Phosphorylases metabolism
- Abstract
Parasitic protists belonging to the genus Leishmania synthesize the non-canonical carbohydrate reserve, mannogen, which is composed of β-1,2-mannan oligosaccharides. Here, we identify a class of dual-activity mannosyltransferase/phosphorylases (MTPs) that catalyze both the sugar nucleotide-dependent biosynthesis and phosphorolytic turnover of mannogen. Structural and phylogenic analysis shows that while the MTPs are structurally related to bacterial mannan phosphorylases, they constitute a distinct family of glycosyltransferases (GT108) that have likely been acquired by horizontal gene transfer from gram-positive bacteria. The seven MTPs catalyze the constitutive synthesis and turnover of mannogen. This metabolic rheostat protects obligate intracellular parasite stages from nutrient excess, and is essential for thermotolerance and parasite infectivity in the mammalian host. Our results suggest that the acquisition and expansion of the MTP family in Leishmania increased the metabolic flexibility of these protists and contributed to their capacity to colonize new host niches., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.