Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 212
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Anal Chem ; 95(2): 1115-1122, 2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36544272

RESUMEN

Venous thromboembolism (VTE) refers to a blood clot that starts in a vein. The risk of developing VTE is highest after major surgery or a major injury, or when someone has heart failure, cancer, or infectious disease (e.g., COVID-19). Without prompt treatment to break up clots and prevent more from forming, VTE can restrict or block blood flow and oxygen, which can damage the body tissue or organs. VTE can occur without any obvious signs, and imaging technologies are used. Alternatively rapid measurement of thrombin generation (TG) and D-dimer could be used to make a fast, portable, and easy-to-use diagnostic platform for VTE. Here, we have demonstrated a diagnostic sensing platform with the ability of simultaneous detection of TG and D-dimer in human plasma. Modifications were made to both the assay protocols to eliminate the need for sample dilution and incubation steps. Using a substantially reduced sample volume, the measurement results show comparable performance to the gold standard method. Our platform is able to deliver accurate and cost-effective results for both TG and D-dimer assays when using undiluted plasma in under 15 min. The assays presented are therefore a good candidate technology for use in a point-of-care platform to diagnose VTE.


Asunto(s)
Productos de Degradación de Fibrina-Fibrinógeno , Trombina , Tromboembolia Venosa , Trombosis de la Vena , Humanos , Biomarcadores , Productos de Degradación de Fibrina-Fibrinógeno/química , Sistemas de Atención de Punto , Trombina/química , Tromboembolia Venosa/diagnóstico , Tromboembolia Venosa/prevención & control , Trombosis de la Vena/diagnóstico
2.
PLoS Pathog ; 17(7): e1009734, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34310651

RESUMEN

Animal African Trypanosomiasis (AAT) is a debilitating livestock disease prevalent across sub-Saharan Africa, a main cause of which is the protozoan parasite Trypanosoma congolense. In comparison to the well-studied T. brucei, there is a major paucity of knowledge regarding the biology of T. congolense. Here, we use a combination of omics technologies and novel genetic tools to characterise core metabolism in T. congolense mammalian-infective bloodstream-form parasites, and test whether metabolic differences compared to T. brucei impact upon sensitivity to metabolic inhibition. Like the bloodstream stage of T. brucei, glycolysis plays a major part in T. congolense energy metabolism. However, the rate of glucose uptake is significantly lower in bloodstream stage T. congolense, with cells remaining viable when cultured in concentrations as low as 2 mM. Instead of pyruvate, the primary glycolytic endpoints are succinate, malate and acetate. Transcriptomics analysis showed higher levels of transcripts associated with the mitochondrial pyruvate dehydrogenase complex, acetate generation, and the glycosomal succinate shunt in T. congolense, compared to T. brucei. Stable-isotope labelling of glucose enabled the comparison of carbon usage between T. brucei and T. congolense, highlighting differences in nucleotide and saturated fatty acid metabolism. To validate the metabolic similarities and differences, both species were treated with metabolic inhibitors, confirming that electron transport chain activity is not essential in T. congolense. However, the parasite exhibits increased sensitivity to inhibition of mitochondrial pyruvate import, compared to T. brucei. Strikingly, T. congolense exhibited significant resistance to inhibitors of fatty acid synthesis, including a 780-fold higher EC50 for the lipase and fatty acid synthase inhibitor Orlistat, compared to T. brucei. These data highlight that bloodstream form T. congolense diverges from T. brucei in key areas of metabolism, with several features that are intermediate between bloodstream- and insect-stage T. brucei. These results have implications for drug development, mechanisms of drug resistance and host-pathogen interactions.


Asunto(s)
Trypanosoma brucei brucei/metabolismo , Trypanosoma congolense/metabolismo , Animales , Reguladores del Metabolismo de Lípidos/farmacología , Ratones , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma congolense/efectos de los fármacos , Tripanosomiasis Africana
3.
PLoS Pathog ; 17(3): e1009204, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33647053

RESUMEN

Trypanosoma brucei, a protist responsible for human African trypanosomiasis (sleeping sickness), is transmitted by the tsetse fly where the procyclic forms of the parasite develop in the proline-rich (1-2 mM) and glucose-depleted digestive tract. Proline is essential for the midgut colonization of the parasite in the insect vector, however other carbon sources could be available and used to feed its central metabolism. Here we show that procyclic trypanosomes can consume and metabolize metabolic intermediates, including those excreted from glucose catabolism (succinate, alanine and pyruvate), with the exception of acetate, which is the ultimate end-product excreted by the parasite. Among the tested metabolites, tricarboxylic acid (TCA) cycle intermediates (succinate, malate and α-ketoglutarate) stimulated growth of the parasite in the presence of 2 mM proline. The pathways used for their metabolism were mapped by proton-NMR metabolic profiling and phenotypic analyses of thirteen RNAi and/or null mutants affecting central carbon metabolism. We showed that (i) malate is converted to succinate by both the reducing and oxidative branches of the TCA cycle, which demonstrates that procyclic trypanosomes can use the full TCA cycle, (ii) the enormous rate of α-ketoglutarate consumption (15-times higher than glucose) is possible thanks to the balanced production and consumption of NADH at the substrate level and (iii) α-ketoglutarate is toxic for trypanosomes if not appropriately metabolized as observed for an α-ketoglutarate dehydrogenase null mutant. In addition, epimastigotes produced from procyclics upon overexpression of RBP6 showed a growth defect in the presence of 2 mM proline, which is rescued by α-ketoglutarate, suggesting that physiological amounts of proline are not sufficient per se for the development of trypanosomes in the fly. In conclusion, these data show that trypanosomes can metabolize multiple metabolites, in addition to proline, which allows them to confront challenging environments in the fly.


Asunto(s)
Glucosa/metabolismo , Prolina/farmacología , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma/efectos de los fármacos , Moscas Tse-Tse/efectos de los fármacos , Animales , Ciclo del Ácido Cítrico/efectos de los fármacos , Insectos Vectores/parasitología , Oxidación-Reducción/efectos de los fármacos , Prolina/metabolismo , Interferencia de ARN/fisiología , Trypanosoma/metabolismo , Trypanosoma brucei brucei/metabolismo , Tripanosomiasis Africana/tratamiento farmacológico , Moscas Tse-Tse/parasitología
4.
PLoS Biol ; 18(6): e3000741, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32520929

RESUMEN

Mitochondrial metabolic remodeling is a hallmark of the Trypanosoma brucei digenetic life cycle because the insect stage utilizes a cost-effective oxidative phosphorylation (OxPhos) to generate ATP, while bloodstream cells switch to aerobic glycolysis. Due to difficulties in acquiring enough parasites from the tsetse fly vector, the dynamics of the parasite's metabolic rewiring in the vector have remained obscure. Here, we took advantage of in vitro-induced differentiation to follow changes at the RNA, protein, and metabolite levels. This multi-omics and cell-based profiling showed an immediate redirection of electron flow from the cytochrome-mediated pathway to an alternative oxidase (AOX), an increase in proline consumption, elevated activity of complex II, and certain tricarboxylic acid (TCA) cycle enzymes, which led to mitochondrial membrane hyperpolarization and increased reactive oxygen species (ROS) levels. Interestingly, these ROS molecules appear to act as signaling molecules driving developmental progression because ectopic expression of catalase, a ROS scavenger, halted the in vitro-induced differentiation. Our results provide insights into the mechanisms of the parasite's mitochondrial rewiring and reinforce the emerging concept that mitochondria act as signaling organelles through release of ROS to drive cellular differentiation.


Asunto(s)
Metabolómica , Mitocondrias/metabolismo , Trypanosoma brucei brucei/crecimiento & desarrollo , Trypanosoma brucei brucei/metabolismo , Adenosina Trifosfato/biosíntesis , Diferenciación Celular/efectos de los fármacos , Línea Celular , Respiración de la Célula/efectos de los fármacos , Transporte de Electrón/efectos de los fármacos , Electrones , Glucosa/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Redes y Vías Metabólicas/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Oxidación-Reducción , Oxidorreductasas/metabolismo , Proteínas de Plantas/metabolismo , Prolina/metabolismo , Proteoma/metabolismo , Proteínas Protozoarias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Transcriptoma/genética , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma brucei brucei/genética
5.
Mol Microbiol ; 116(2): 564-588, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33932053

RESUMEN

Trypanosoma congolense is a principal agent causing livestock trypanosomiasis in Africa, costing developing economies billions of dollars and undermining food security. Only the diamidine diminazene and the phenanthridine isometamidium are regularly used, and resistance is widespread but poorly understood. We induced stable diminazene resistance in T. congolense strain IL3000 in vitro. There was no cross-resistance with the phenanthridine drugs, melaminophenyl arsenicals, oxaborole trypanocides, or with diamidine trypanocides, except the close analogs DB829 and DB75. Fluorescence microscopy showed that accumulation of DB75 was inhibited by folate. Uptake of [3 H]-diminazene was slow with low affinity and partly but reciprocally inhibited by folate and by competing diamidines. Expression of T. congolense folate transporters in diminazene-resistant Trypanosoma brucei brucei significantly sensitized the cells to diminazene and DB829, but not to oxaborole AN7973. However, [3 H]-diminazene transport studies, whole-genome sequencing, and RNA-seq found no major changes in diminazene uptake, folate transporter sequence, or expression. Instead, all resistant clones displayed a moderate reduction in the mitochondrial membrane potential Ψm. We conclude that diminazene uptake in T. congolense proceed via multiple low affinity mechanisms including folate transporters; while resistance is associated with a reduction in Ψm it is unclear whether this is the primary cause of the resistance.


Asunto(s)
Diminazeno/farmacología , Potencial de la Membrana Mitocondrial/fisiología , Tripanocidas/farmacología , Trypanosoma congolense/efectos de los fármacos , Tripanosomiasis Africana/veterinaria , Tripanosomiasis Bovina/tratamiento farmacológico , Animales , Bovinos , Resistencia a Medicamentos/fisiología , Transportadores de Ácido Fólico/metabolismo , Fenantridinas/farmacología , Tripanosomiasis Africana/tratamiento farmacológico , Tripanosomiasis Africana/parasitología , Tripanosomiasis Bovina/parasitología
6.
PLoS Pathog ; 16(11): e1008932, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33141865

RESUMEN

Livestock diseases caused by Trypanosoma congolense, T. vivax and T. brucei, collectively known as nagana, are responsible for billions of dollars in lost food production annually. There is an urgent need for novel therapeutics. Encouragingly, promising antitrypanosomal benzoxaboroles are under veterinary development. Here, we show that the most efficacious subclass of these compounds are prodrugs activated by trypanosome serine carboxypeptidases (CBPs). Drug-resistance to a development candidate, AN11736, emerged readily in T. brucei, due to partial deletion within the locus containing three tandem copies of the CBP genes. T. congolense parasites, which possess a larger array of related CBPs, also developed resistance to AN11736 through deletion within the locus. A genome-scale screen in T. brucei confirmed CBP loss-of-function as the primary mechanism of resistance and CRISPR-Cas9 editing proved that partial deletion within the locus was sufficient to confer resistance. CBP re-expression in either T. brucei or T. congolense AN11736-resistant lines restored drug-susceptibility. CBPs act by cleaving the benzoxaborole AN11736 to a carboxylic acid derivative, revealing a prodrug activation mechanism. Loss of CBP activity results in massive reduction in net uptake of AN11736, indicating that entry is facilitated by the concentration gradient created by prodrug metabolism.


Asunto(s)
Compuestos de Boro/metabolismo , Carboxipeptidasas/metabolismo , Tripanocidas/metabolismo , Trypanosoma brucei brucei/enzimología , Trypanosoma congolense/enzimología , Trypanosoma vivax/enzimología , Tripanosomiasis Africana/veterinaria , Valina/análogos & derivados , Animales , Ácidos Carboxílicos/metabolismo , Resistencia a Medicamentos , Femenino , Ganado , Ratones , Parasitemia/veterinaria , Profármacos/metabolismo , Proteínas Protozoarias/metabolismo , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma congolense/efectos de los fármacos , Trypanosoma vivax/efectos de los fármacos , Tripanosomiasis Africana/tratamiento farmacológico , Tripanosomiasis Africana/parasitología , Valina/metabolismo
7.
J Biol Chem ; 295(24): 8331-8347, 2020 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-32354742

RESUMEN

Introduced about a century ago, suramin remains a frontline drug for the management of early-stage East African trypanosomiasis (sleeping sickness). Cellular entry into the causative agent, the protozoan parasite Trypanosoma brucei, occurs through receptor-mediated endocytosis involving the parasite's invariant surface glycoprotein 75 (ISG75), followed by transport into the cytosol via a lysosomal transporter. The molecular basis of the trypanocidal activity of suramin remains unclear, but some evidence suggests broad, but specific, impacts on trypanosome metabolism (i.e. polypharmacology). Here we observed that suramin is rapidly accumulated in trypanosome cells proportionally to ISG75 abundance. Although we found little evidence that suramin disrupts glycolytic or glycosomal pathways, we noted increased mitochondrial ATP production, but a net decrease in cellular ATP levels. Metabolomics highlighted additional impacts on mitochondrial metabolism, including partial Krebs' cycle activation and significant accumulation of pyruvate, corroborated by increased expression of mitochondrial enzymes and transporters. Significantly, the vast majority of suramin-induced proteins were normally more abundant in the insect forms compared with the blood stage of the parasite, including several proteins associated with differentiation. We conclude that suramin has multiple and complex effects on trypanosomes, but unexpectedly partially activates mitochondrial ATP-generating activity. We propose that despite apparent compensatory mechanisms in drug-challenged cells, the suramin-induced collapse of cellular ATP ultimately leads to trypanosome cell death.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Mitocondrias/metabolismo , Suramina/farmacología , Trypanosoma brucei brucei/metabolismo , Adenosina Trifosfato/metabolismo , Flagelos/efectos de los fármacos , Flagelos/metabolismo , Flagelos/ultraestructura , Glucólisis/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Metaboloma/efectos de los fármacos , Microcuerpos/efectos de los fármacos , Microcuerpos/metabolismo , Microcuerpos/ultraestructura , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Modelos Moleculares , Prolina/metabolismo , Proteoma/metabolismo , ATPasas de Translocación de Protón/metabolismo , Proteínas Protozoarias/metabolismo , Ácido Pirúvico/metabolismo
8.
Artículo en Inglés | MEDLINE | ID: mdl-32340987

RESUMEN

As resistance to artemisinins (current frontline drugs in malaria treatment) emerges in Southeast Asia, there is an urgent need to identify the genetic determinants and understand the molecular mechanisms underpinning such resistance. Such insights could lead to prospective interventions to contain resistance and prevent the eventual spread to other regions where malaria is endemic. Reduced susceptibility to artemisinin in Southeast Asia has been primarily linked to mutations in the Plasmodium falciparum Kelch-13 gene, which is currently widely recognized as a molecular marker of artemisinin resistance. However, two mutations in a ubiquitin hydrolase, UBP-1, have been previously associated with reduced artemisinin susceptibility in a rodent model of malaria, and some cases of UBP-1 mutation variants associated with artemisinin treatment failure have been reported in Africa and SEA. In this study, we employed CRISPR-Cas9 genome editing and preemptive drug pressures to test these artemisinin susceptibility-associated mutations in UBP-1 in Plasmodium berghei sensitive lines in vivo Using these approaches, we show that the V2721F UBP-1 mutation results in reduced artemisinin susceptibility, while the V2752F mutation results in resistance to chloroquine (CQ) and moderately impacts tolerance to artemisinins. Genetic reversal of the V2752F mutation restored chloroquine sensitivity in these mutant lines, whereas simultaneous introduction of both mutations could not be achieved and appears to be lethal. Interestingly, these mutations carry a detrimental growth defect, which would possibly explain their lack of expansion in natural infection settings. Our work provides independent experimental evidence on the role of UBP-1 in modulating parasite responses to artemisinin and chloroquine under in vivo conditions.


Asunto(s)
Antimaláricos , Artemisininas , Malaria Falciparum , África , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Artemisininas/farmacología , Artemisininas/uso terapéutico , Cloroquina/farmacología , Cloroquina/uso terapéutico , Resistencia a Medicamentos/genética , Humanos , Hidrolasas , Malaria Falciparum/tratamiento farmacológico , Mutación/genética , Plasmodium berghei/genética , Plasmodium falciparum , Estudios Prospectivos , Proteínas Protozoarias/genética , Proteínas Protozoarias/uso terapéutico , Ubiquitina/uso terapéutico
9.
PLoS Pathog ; 14(12): e1007475, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30589893

RESUMEN

Bloodstream form African trypanosomes are thought to rely exclusively upon glycolysis, using glucose as a substrate, for ATP production. Indeed, the pathway has long been considered a potential therapeutic target to tackle the devastating and neglected tropical diseases caused by these parasites. However, plasma membrane glucose and glycerol transporters are both expressed by trypanosomes and these parasites can infiltrate tissues that contain glycerol. Here, we show that bloodstream form trypanosomes can use glycerol for gluconeogenesis and for ATP production, particularly when deprived of glucose following hexose transporter depletion. We demonstrate that Trypanosoma brucei hexose transporters 1 and 2 (THT1 and THT2) are localized to the plasma membrane and that knockdown of THT1 expression leads to a growth defect that is more severe when THT2 is also knocked down. These data are consistent with THT1 and THT2 being the primary routes of glucose supply for the production of ATP by glycolysis. However, supplementation of the growth medium with glycerol substantially rescued the growth defect caused by THT1 and THT2 knockdown. Metabolomic analyses with heavy-isotope labelled glycerol demonstrated that trypanosomes take up glycerol and use it to synthesize intermediates of gluconeogenesis, including fructose 1,6-bisphosphate and hexose 6-phosphates, which feed the pentose phosphate pathway and variant surface glycoprotein biosynthesis. We used Cas9-mediated gene knockout to demonstrate a gluconeogenesis-specific, but fructose-1,6-bisphosphatase (Tb927.9.8720)-independent activity, converting fructose 1,6-bisphosphate into fructose 6-phosphate. In addition, we observed increased flux through the tricarboxylic acid cycle and the succinate shunt. Thus, contrary to prior thinking, gluconeogenesis can operate in bloodstream form T. brucei. This pathway, using glycerol as a physiological substrate, may be required in mammalian host tissues.


Asunto(s)
Gluconeogénesis/fisiología , Glicerol/metabolismo , Trypanosoma brucei brucei/metabolismo , Especificidad por Sustrato
10.
PLoS Pathog ; 14(3): e1006953, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29554142

RESUMEN

Transketolase (TKT) is part of the non-oxidative branch of the pentose phosphate pathway (PPP). Here we describe the impact of removing this enzyme from the pathogenic protozoan Leishmania mexicana. Whereas the deletion had no obvious effect on cultured promastigote forms of the parasite, the Δtkt cells were not virulent in mice. Δtkt promastigotes were more susceptible to oxidative stress and various leishmanicidal drugs than wild-type, and metabolomics analysis revealed profound changes to metabolism in these cells. In addition to changes consistent with those directly related to the role of TKT in the PPP, central carbon metabolism was substantially decreased, the cells consumed significantly less glucose, flux through glycolysis diminished, and production of the main end products of metabolism was decreased. Only minor changes in RNA abundance from genes encoding enzymes in central carbon metabolism, however, were detected although fructose-1,6-bisphosphate aldolase activity was decreased two-fold in the knock-out cell line. We also showed that the dual localisation of TKT between cytosol and glycosomes is determined by the C-terminus of the enzyme and by engineering different variants of the enzyme we could alter its sub-cellular localisation. However, no effect on the overall flux of glucose was noted irrespective of whether the enzyme was found uniquely in either compartment, or in both.


Asunto(s)
Leishmania mexicana/patogenicidad , Leishmaniasis Cutánea/metabolismo , Leishmaniasis Cutánea/parasitología , Metaboloma , Transcetolasa/metabolismo , Virulencia , Animales , Glucólisis , Estadios del Ciclo de Vida , Metabolómica , Ratones , Ratones Endogámicos BALB C , Monocitos/metabolismo , Monocitos/parasitología , Estrés Oxidativo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Eliminación de Secuencia , Transcetolasa/genética
11.
PLoS Pathog ; 14(9): e1007315, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30252911

RESUMEN

Kinetoplastid parasites-trypanosomes and leishmanias-infect millions of humans and cause economically devastating diseases of livestock, and the few existing drugs have serious deficiencies. Benzoxaborole-based compounds are very promising potential novel anti-trypanosomal therapies, with candidates already in human and animal clinical trials. We investigated the mechanism of action of several benzoxaboroles, including AN7973, an early candidate for veterinary trypanosomosis. In all kinetoplastids, transcription is polycistronic. Individual mRNA 5'-ends are created by trans splicing of a short leader sequence, with coupled polyadenylation of the preceding mRNA. Treatment of Trypanosoma brucei with AN7973 inhibited trans splicing within 1h, as judged by loss of the Y-structure splicing intermediate, reduced levels of mRNA, and accumulation of peri-nuclear granules. Methylation of the spliced leader precursor RNA was not affected, but more prolonged AN7973 treatment caused an increase in S-adenosyl methionine and methylated lysine. Together, the results indicate that mRNA processing is a primary target of AN7973. Polyadenylation is required for kinetoplastid trans splicing, and the EC50 for AN7973 in T. brucei was increased three-fold by over-expression of the T. brucei cleavage and polyadenylation factor CPSF3, identifying CPSF3 as a potential molecular target. Molecular modeling results suggested that inhibition of CPSF3 by AN7973 is feasible. Our results thus chemically validate mRNA processing as a viable drug target in trypanosomes. Several other benzoxaboroles showed metabolomic and splicing effects that were similar to those of AN7973, identifying splicing inhibition as a common mode of action and suggesting that it might be linked to subsequent changes in methylated metabolites. Granule formation, splicing inhibition and resistance after CPSF3 expression did not, however, always correlate and prolonged selection of trypanosomes in AN7973 resulted in only 1.5-fold resistance. It is therefore possible that the modes of action of oxaboroles that target trypanosome mRNA processing might extend beyond CPSF3 inhibition.


Asunto(s)
Benzoxazoles/farmacología , ARN Protozoario/metabolismo , Tripanocidas/farmacología , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma brucei brucei/metabolismo , Animales , Benzoxazoles/química , Bovinos , Resistencia a Medicamentos/genética , Cabras , Humanos , Ratones , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Procesamiento Postranscripcional del ARN/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Protozoario/genética , Trans-Empalme/efectos de los fármacos , Tripanocidas/química , Trypanosoma brucei brucei/genética , Trypanosoma congolense/efectos de los fármacos , Trypanosoma congolense/genética , Trypanosoma congolense/metabolismo , Trypanosoma vivax/efectos de los fármacos , Trypanosoma vivax/genética , Trypanosoma vivax/metabolismo , Tripanosomiasis/tratamiento farmacológico , Tripanosomiasis/parasitología
12.
PLoS Pathog ; 14(2): e1006850, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29425238

RESUMEN

Recent development of benzoxaborole-based chemistry gave rise to a collection of compounds with great potential in targeting diverse infectious diseases, including human African Trypanosomiasis (HAT), a devastating neglected tropical disease. However, further medicinal development is largely restricted by a lack of insight into mechanism of action (MoA) in pathogenic kinetoplastids. We adopted a multidisciplinary approach, combining a high-throughput forward genetic screen with functional group focused chemical biological, structural biology and biochemical analyses, to tackle the complex MoAs of benzoxaboroles in Trypanosoma brucei. We describe an oxidative enzymatic pathway composed of host semicarbazide-sensitive amine oxidase and a trypanosomal aldehyde dehydrogenase TbALDH3. Two sequential reactions through this pathway serve as the key underlying mechanism for activating a series of 4-aminomethylphenoxy-benzoxaboroles as potent trypanocides; the methylamine parental compounds as pro-drugs are transformed first into intermediate aldehyde metabolites, and further into the carboxylate metabolites as effective forms. Moreover, comparative biochemical and crystallographic analyses elucidated the catalytic specificity of TbALDH3 towards the benzaldehyde benzoxaborole metabolites as xenogeneic substrates. Overall, this work proposes a novel drug activation mechanism dependent on both host and parasite metabolism of primary amine containing molecules, which contributes a new perspective to our understanding of the benzoxaborole MoA, and could be further exploited to improve the therapeutic index of antimicrobial compounds.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Amina Oxidasa (conteniendo Cobre)/metabolismo , Compuestos de Boro/metabolismo , Modelos Biológicos , Profármacos/metabolismo , Tripanocidas/metabolismo , Trypanosoma brucei brucei/enzimología , Activación Metabólica , Aldehído Deshidrogenasa/antagonistas & inhibidores , Aldehído Deshidrogenasa/química , Aldehído Deshidrogenasa/genética , Aldehído Oxidorreductasas/antagonistas & inhibidores , Aldehído Oxidorreductasas/química , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Amina Oxidasa (conteniendo Cobre)/antagonistas & inhibidores , Amina Oxidasa (conteniendo Cobre)/química , Amina Oxidasa (conteniendo Cobre)/genética , Sustitución de Aminoácidos , Animales , Compuestos de Boro/química , Compuestos de Boro/farmacología , Resistencia a Medicamentos , Ensayos Analíticos de Alto Rendimiento , Humanos , Estructura Molecular , Mutación , Filogenia , Profármacos/química , Profármacos/farmacología , Dominios y Motivos de Interacción de Proteínas , Interferencia de ARN , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Tripanocidas/química , Tripanocidas/farmacología , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma brucei brucei/fisiología
13.
Rheumatology (Oxford) ; 59(4): 869-878, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31497857

RESUMEN

OBJECTIVES: Fatty acid oxidation (FAO) and glycolysis have been implicated in immune regulation and activation of macrophages. However, investigation of human monocyte intracellular metabolism in the context of the hypoxic and inflammatory rheumatoid arthritis (RA) synovium is lacking. We hypothesized that exposure of monocytes to the hypoxic and inflammatory RA environment would have a profound impact on their metabolic state, and potential to contribute to disease pathology. METHODS: Human monocytes were isolated from buffy coats and exposed to hypoxia. Metabolic profiling of monocytes was carried out by LC-MS metabolomics. Inflammatory mediator release after LPS or RA-synovial fluid (RA-SF) stimulation was analysed by ELISA. FAO was inhibited by etomoxir or enhanced with exogenous carnitine supplementation. Transcriptomics of RA blood monocytes and RA-SF macrophages was carried out by microarray. RESULTS: Hypoxia exacerbated monocyte-derived CCL20 and IL-1ß release in response to LPS, and increased glycolytic intermediates at the expense of carnitines. Modulation of carnitine identified a novel role for FAO in the production of CCL20 in response to LPS. Transcriptional analysis of RA blood monocytes and RA-SF macrophages revealed that fatty acid metabolism was altered and CCL20 increased when monocytes enter the synovial environment. In vitro analysis of monocytes showed that RA-SF increases carnitine abundance and CCL20 production in hypoxia, which was exacerbated by exogenous carnitine. CONCLUSION: This work has revealed a novel inflammatory mechanism in RA that links FAO to CCL20 production in human monocytes, which could subsequently contribute to RA disease pathogenesis by promoting the recruitment of Th17 cells and osteoclastogenesis.


Asunto(s)
Artritis Reumatoide/metabolismo , Microambiente Celular , Quimiocina CCL20/metabolismo , Ácidos Grasos/metabolismo , Hipoxia/metabolismo , Monocitos/metabolismo , Líquido Sinovial , Carnitina/farmacología , Quimiocina CCL20/efectos de los fármacos , Cromatografía Liquida , Inhibidores Enzimáticos/farmacología , Compuestos Epoxi/farmacología , Perfilación de la Expresión Génica , Humanos , Técnicas In Vitro , Lipopolisacáridos/farmacología , Espectrometría de Masas , Metabolómica , Análisis por Micromatrices , Monocitos/efectos de los fármacos , Membrana Sinovial/metabolismo
14.
Bioinformatics ; 34(2): 317-318, 2018 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-28968802

RESUMEN

MOTIVATION: We recently published MS2LDA, a method for the decomposition of sets of molecular fragment data derived from large metabolomics experiments. To make the method more widely available to the community, here we present ms2lda.org, a web application that allows users to upload their data, run MS2LDA analyses and explore the results through interactive visualizations. RESULTS: Ms2lda.org takes tandem mass spectrometry data in many standard formats and allows the user to infer the sets of fragment and neutral loss features that co-occur together (Mass2Motifs). As an alternative workflow, the user can also decompose a data set onto predefined Mass2Motifs. This is accomplished through the web interface or programmatically from our web service. AVAILABILITY AND IMPLEMENTATION: The website can be found at http://ms2lda.org, while the source code is available at https://github.com/sdrogers/ms2ldaviz under the MIT license. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

15.
Proc Natl Acad Sci U S A ; 113(48): 13738-13743, 2016 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-27856765

RESUMEN

The potential of untargeted metabolomics to answer important questions across the life sciences is hindered because of a paucity of computational tools that enable extraction of key biochemically relevant information. Available tools focus on using mass spectrometry fragmentation spectra to identify molecules whose behavior suggests they are relevant to the system under study. Unfortunately, fragmentation spectra cannot identify molecules in isolation but require authentic standards or databases of known fragmented molecules. Fragmentation spectra are, however, replete with information pertaining to the biochemical processes present, much of which is currently neglected. Here, we present an analytical workflow that exploits all fragmentation data from a given experiment to extract biochemically relevant features in an unsupervised manner. We demonstrate that an algorithm originally used for text mining, latent Dirichlet allocation, can be adapted to handle metabolomics datasets. Our approach extracts biochemically relevant molecular substructures ("Mass2Motifs") from spectra as sets of co-occurring molecular fragments and neutral losses. The analysis allows us to isolate molecular substructures, whose presence allows molecules to be grouped based on shared substructures regardless of classical spectral similarity. These substructures, in turn, support putative de novo structural annotation of molecules. Combining this spectral connectivity to orthogonal correlations (e.g., common abundance changes under system perturbation) significantly enhances our ability to provide mechanistic explanations for biological behavior.


Asunto(s)
Metabolómica/métodos , Espectrometría de Masas en Tándem/métodos , Flujo de Trabajo , Algoritmos , Bases de Datos Factuales , Metabolómica/normas , Espectrometría de Masas en Tándem/normas
16.
Artículo en Inglés | MEDLINE | ID: mdl-29463533

RESUMEN

With the World Health Organization reporting over 30,000 deaths and 200,000 to 400,000 new cases annually, visceral leishmaniasis is a serious disease affecting some of the world's poorest people. As drug resistance continues to rise, there is a huge unmet need to improve treatment. Miltefosine remains one of the main treatments for leishmaniasis, yet its mode of action (MoA) is still unknown. Understanding the MoA of this drug and parasite response to treatment could help pave the way for new and more successful treatments for leishmaniasis. A novel method has been devised to study the metabolome and lipidome of Leishmania donovani axenic amastigotes treated with miltefosine. Miltefosine caused a dramatic decrease in many membrane phospholipids (PLs), in addition to amino acid pools, while sphingolipids (SLs) and sterols increased. Leishmania major promastigotes devoid of SL biosynthesis through loss of the serine palmitoyl transferase gene (ΔLCB2) were 3-fold less sensitive to miltefosine than wild-type (WT) parasites. Changes in the metabolome and lipidome of miltefosine-treated L. major mirrored those of L. donovani A lack of SLs in the ΔLCB2 mutant was matched by substantial alterations in sterol content. Together, these data indicate that SLs and ergosterol are important for miltefosine sensitivity and, perhaps, MoA.


Asunto(s)
Antiprotozoarios/farmacología , Leishmania donovani/metabolismo , Leishmania major/metabolismo , Fosforilcolina/análogos & derivados , Serina C-Palmitoiltransferasa/genética , Esfingolípidos/metabolismo , Esteroles/metabolismo , Ergosterol/metabolismo , Humanos , Leishmaniasis Visceral/tratamiento farmacológico , Leishmaniasis Visceral/parasitología , Lípidos de la Membrana/metabolismo , Metaboloma/efectos de los fármacos , Metaboloma/genética , Fosfolípidos/metabolismo , Fosforilcolina/farmacología
17.
PLoS Pathog ; 12(12): e1006094, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28027318

RESUMEN

Malaria parasites (Plasmodium spp.) encounter markedly different (nutritional) environments during their complex life cycles in the mosquito and human hosts. Adaptation to these different host niches is associated with a dramatic rewiring of metabolism, from a highly glycolytic metabolism in the asexual blood stages to increased dependence on tricarboxylic acid (TCA) metabolism in mosquito stages. Here we have used stable isotope labelling, targeted metabolomics and reverse genetics to map stage-specific changes in Plasmodium berghei carbon metabolism and determine the functional significance of these changes on parasite survival in the blood and mosquito stages. We show that glutamine serves as the predominant input into TCA metabolism in both asexual and sexual blood stages and is important for complete male gametogenesis. Glutamine catabolism, as well as key reactions in intermediary metabolism and CoA synthesis are also essential for ookinete to oocyst transition in the mosquito. These data extend our knowledge of Plasmodium metabolism and point towards possible targets for transmission-blocking intervention strategies. Furthermore, they highlight significant metabolic differences between Plasmodium species which are not easily anticipated based on genomics or transcriptomics studies and underline the importance of integration of metabolomics data with other platforms in order to better inform drug discovery and design.


Asunto(s)
Adaptación Fisiológica/fisiología , Interacciones Huésped-Parásitos/fisiología , Malaria/parasitología , Plasmodium/crecimiento & desarrollo , Plasmodium/metabolismo , Animales , Culicidae , Modelos Animales de Enfermedad , Citometría de Flujo , Cromatografía de Gases y Espectrometría de Masas , Estadios del Ciclo de Vida , Ratones
19.
Proc Natl Acad Sci U S A ; 112(29): 9112-7, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26150481

RESUMEN

Kinetoplastid parasites cause lethal diseases in humans and animals. The kinetoplast itself contains the mitochondrial genome, comprising a huge, complex DNA network that is also an important drug target. Isometamidium, for example, is a key veterinary drug that accumulates in the kinetoplast in African trypanosomes. Kinetoplast independence and isometamidium resistance are observed where certain mutations in the F1-γ-subunit of the two-sector F1Fo-ATP synthase allow for Fo-independent generation of a mitochondrial membrane potential. To further explore kinetoplast biology and drug resistance, we screened a genome-scale RNA interference library in African trypanosomes for isometamidium resistance mechanisms. Our screen identified 14 V-ATPase subunits and all 4 adaptin-3 subunits, implicating acidic compartment defects in resistance; V-ATPase acidifies lysosomes and related organelles, whereas adaptin-3 is responsible for trafficking among these organelles. Independent strains with depleted V-ATPase or adaptin-3 subunits were isometamidium resistant, and chemical inhibition of the V-ATPase phenocopied this effect. While drug accumulation in the kinetoplast continued after V-ATPase subunit depletion, acriflavine-induced kinetoplast loss was specifically tolerated in these cells and in cells depleted for adaptin-3 or endoplasmic reticulum membrane complex subunits, also identified in our screen. Consistent with kinetoplast dispensability, V-ATPase defective cells were oligomycin resistant, suggesting ATP synthase uncoupling and bypass of the normal Fo-A6-subunit requirement; this subunit is the only kinetoplast-encoded product ultimately required for viability in bloodstream-form trypanosomes. Thus, we describe 30 genes and 3 protein complexes associated with kinetoplast-dependent growth. Mutations affecting these genes could explain natural cases of dyskinetoplasty and multidrug resistance. Our results also reveal potentially conserved communication between the compartmentalized two-sector rotary ATPases.


Asunto(s)
ADN de Cinetoplasto/metabolismo , Resistencia a Medicamentos , Mitocondrias/enzimología , ATPasas de Translocación de Protón/metabolismo , Trypanosoma brucei brucei/enzimología , ATPasas de Translocación de Protón Vacuolares/metabolismo , Ácidos/metabolismo , Animales , Compartimento Celular/efectos de los fármacos , Resistencia a Medicamentos/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Membranas Intracelulares/efectos de los fármacos , Membranas Intracelulares/metabolismo , Mitocondrias/efectos de los fármacos , Fenantridinas/química , Fenantridinas/farmacología , Reacción en Cadena de la Polimerasa , Subunidades de Proteína/metabolismo , Interferencia de ARN/efectos de los fármacos , Reproducibilidad de los Resultados , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma brucei brucei/genética
20.
Anal Chem ; 89(14): 7569-7577, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28621528

RESUMEN

In untargeted metabolomics approaches, the inability to structurally annotate relevant features and map them to biochemical pathways is hampering the full exploitation of many metabolomics experiments. Furthermore, variable metabolic content across samples result in sparse feature matrices that are statistically hard to handle. Here, we introduce MS2LDA+ that tackles both above-mentioned problems. Previously, we presented MS2LDA, which extracts biochemically relevant molecular substructures ("Mass2Motifs") from a collection of fragmentation spectra as sets of co-occurring molecular fragments and neutral losses, thereby recognizing building blocks of metabolomics. Here, we extend MS2LDA to handle multiple metabolomics experiments in one analysis, resulting in MS2LDA+. By linking Mass2Motifs across samples, we expose the variability in prevalence of structurally related metabolite families. We validate the differential prevalence of substructures between two distinct samples groups and apply it to fecal samples. Subsequently, within one sample group of urines, we rank the Mass2Motifs based on their variance to assess whether xenobiotic-derived substructures are among the most-variant Mass2Motifs. Indeed, we could ascribe 22 out of the 30 most-variant Mass2Motifs to xenobiotic-derived substructures including paracetamol/acetaminophen mercapturate and dimethylpyrogallol. In total, we structurally characterized 101 Mass2Motifs with biochemically or chemically relevant substructures. Finally, we combined the discovered metabolite families with full scan feature intensity information to obtain insight into core metabolites present in most samples and rare metabolites present in small subsets now linked through their common substructures. We conclude that by biochemical grouping of metabolites across samples MS2LDA+ aids in structural annotation of metabolites and guides prioritization of analysis by using Mass2Motif prevalence.


Asunto(s)
Antihipertensivos/metabolismo , Descubrimiento de Drogas , Metabolómica , Modelos Estadísticos , Adolescente , Anciano , Anciano de 80 o más Años , Antihipertensivos/análisis , Cerveza/análisis , Niño , Cromatografía Liquida , Heces/química , Femenino , Humanos , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Estructura Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA