Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 265
Filtrar
1.
ACS Infect Dis ; 10(5): 1739-1752, 2024 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-38647213

RESUMEN

Reverse analogs of the phosphonohydroxamic acid antibiotic fosmidomycin are potent inhibitors of the nonmevalonate isoprenoid biosynthesis enzyme 1-deoxy-d-xylulose 5-phosphate reductoisomerase (DXR, IspC) of Plasmodium falciparum. Some novel analogs with large phenylalkyl substituents at the hydroxamic acid nitrogen exhibit nanomolar PfDXR inhibition and potent in vitro growth inhibition of P. falciparum parasites coupled with good parasite selectivity. X-ray crystallographic studies demonstrated that the N-phenylpropyl substituent of the newly developed lead compound 13e is accommodated in a subpocket within the DXR catalytic domain but does not reach the NADPH binding pocket of the N-terminal domain. As shown for reverse carba and thia analogs, PfDXR selectively binds the S-enantiomer of the new lead compound. In addition, some representatives of the novel inhibitor subclass are nanomolar Escherichia coli DXR inhibitors, whereas the inhibition of Mycobacterium tuberculosis DXR is considerably weaker.


Asunto(s)
Isomerasas Aldosa-Cetosa , Antimaláricos , Fosfomicina , Ácidos Hidroxámicos , Complejos Multienzimáticos , Plasmodium falciparum , Fosfomicina/farmacología , Fosfomicina/análogos & derivados , Fosfomicina/química , Isomerasas Aldosa-Cetosa/antagonistas & inhibidores , Isomerasas Aldosa-Cetosa/metabolismo , Isomerasas Aldosa-Cetosa/química , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/enzimología , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/química , Antimaláricos/farmacología , Antimaláricos/química , Complejos Multienzimáticos/antagonistas & inhibidores , Complejos Multienzimáticos/metabolismo , Complejos Multienzimáticos/química , Cristalografía por Rayos X , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/síntesis química , Relación Estructura-Actividad , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/enzimología , Modelos Moleculares , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/enzimología , Dominio Catalítico , Oxidorreductasas/antagonistas & inhibidores , Oxidorreductasas/metabolismo
2.
J Agric Food Chem ; 72(14): 7684-7693, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38532701

RESUMEN

Fosmidomycin (FOS) is a natural product inhibiting the DXR enzyme in the MEP pathway and has stimulated interest for finding more suitable FOS analogues. Herein, two series of FOS analogue hydroxamate-containing bisphosphonates as proherbicides were designed, with bisphosphonate replacing the phosphonic unit in FOS while retaining the hydroxamate (BPF series) or replacing it with retro-hydroxamate (BPRF series). The BPF series were synthesized through a three-step reaction sequence including Michael addition of vinylidenebisphosphonate, N-acylation, and deprotection, and the BPRF series were synthesized with a retro-Claisen condensation incorporated into the reaction sequence. Evaluation on model plants demonstrated several compounds having considerable herbicidal activities, and in particular, compound 8m exhibited multifold activity enhancement as compared to the control FOS. The proherbicide properties were comparatively validated. Furthermore, DXR enzyme assay, dimethylallyl pyrophosphate rescue, and molecular docking verified 8m to be a promising proherbicide candidate targeting the DXR enzyme. In addition, 8m also displayed good antimalarial activities.


Asunto(s)
Isomerasas Aldosa-Cetosa , Antimaláricos , Fosfomicina , Fosfomicina/análogos & derivados , Difosfonatos , Simulación del Acoplamiento Molecular , Fosfomicina/farmacología , Isomerasas Aldosa-Cetosa/metabolismo
3.
PLoS Pathog ; 20(1): e1011557, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38277417

RESUMEN

A proposed treatment for malaria is a combination of fosmidomycin and clindamycin. Both compounds inhibit the methylerythritol 4-phosphate (MEP) pathway, the parasitic source of farnesyl and geranylgeranyl pyrophosphate (FPP and GGPP, respectively). Both FPP and GGPP are crucial for the biosynthesis of several essential metabolites such as ubiquinone and dolichol, as well as for protein prenylation. Dietary prenols, such as farnesol (FOH) and geranylgeraniol (GGOH), can rescue parasites from MEP inhibitors, suggesting the existence of a missing pathway for prenol salvage via phosphorylation. In this study, we identified a gene in the genome of P. falciparum, encoding a transmembrane prenol kinase (PolK) involved in the salvage of FOH and GGOH. The enzyme was expressed in Saccharomyces cerevisiae, and its FOH/GGOH kinase activities were experimentally validated. Furthermore, conditional knockout parasites (Δ-PolK) were created to investigate the biological importance of the FOH/GGOH salvage pathway. Δ-PolK parasites were viable but displayed increased susceptibility to fosmidomycin. Their sensitivity to MEP inhibitors could not be rescued by adding prenols. Additionally, Δ-PolK parasites lost their capability to utilize prenols for protein prenylation. Experiments using culture medium supplemented with whole/delipidated human plasma in transgenic parasites revealed that human plasma has components that can diminish the effectiveness of fosmidomycin. Mass spectrometry tests indicated that both bovine supplements used in culture and human plasma contain GGOH. These findings suggest that the FOH/GGOH salvage pathway might offer an alternate source of isoprenoids for malaria parasites when de novo biosynthesis is inhibited. This study also identifies a novel kind of enzyme related to isoprenoid metabolism.


Asunto(s)
Diterpenos , Fosfomicina/análogos & derivados , Hemiterpenos , Parásitos , Pentanoles , Humanos , Animales , Bovinos , Parásitos/metabolismo , Fosfatos , Terpenos/farmacología , Terpenos/metabolismo
4.
PLoS Pathog ; 18(9): e1010803, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36103572

RESUMEN

Efforts to control the global malaria health crisis are undermined by antimalarial resistance. Identifying mechanisms of resistance will uncover the underlying biology of the Plasmodium falciparum malaria parasites that allow evasion of our most promising therapeutics and may reveal new drug targets. We utilized fosmidomycin (FSM) as a chemical inhibitor of plastidial isoprenoid biosynthesis through the methylerythritol phosphate (MEP) pathway. We have thus identified an unusual metabolic regulation scheme in the malaria parasite through the essential glycolytic enzyme, glyceraldehyde 3-phosphate dehydrogenase (GAPDH). Two parallel genetic screens converged on independent but functionally analogous resistance alleles in GAPDH. Metabolic profiling of FSM-resistant gapdh mutant parasites indicates that neither of these mutations disrupt overall glycolytic output. While FSM-resistant GAPDH variant proteins are catalytically active, they have reduced assembly into the homotetrameric state favored by wild-type GAPDH. Disrupted oligomerization of FSM-resistant GAPDH variant proteins is accompanied by altered enzymatic cooperativity and reduced susceptibility to inhibition by free heme. Together, our data identifies a new genetic biomarker of FSM-resistance and reveals the central role of GAPDH in MEP pathway control and antimalarial sensitivity.


Asunto(s)
Antimaláricos , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Malaria Falciparum , Parásitos , Animales , Antimaláricos/metabolismo , Biomarcadores/metabolismo , Resistencia a Medicamentos/genética , Fosfomicina/análogos & derivados , Hemo/metabolismo , Humanos , Malaria Falciparum/parasitología , Parásitos/metabolismo , Fosfatos/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Terpenos/metabolismo
5.
Microbiology (Reading) ; 168(3)2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35358034

RESUMEN

Burkholderia cenocepacia infections are difficult to treat and there is an urgent need for alternative (combination) treatments. The use of anti-virulence therapies in combination with antibiotics is a possible strategy to increase the antimicrobial susceptibility of the pathogen and to slow down the development of resistance. In the present study we evaluated the ß-lactam and colistin-potentiating activity, and anti-virulence effect of the non-mevalonate pathway inhibitor FR900098 against B. cenocepacia in various in vitro and in vivo models. In addition, we evaluated whether repeated exposure to FR900098 alone or when combined with ceftazidime leads to increased resistance. FR900098 potentiated the activity of colistin and several ß-lactam antibiotics (aztreonam, cefepime, cefotaxime, ceftazidime, mecillinam and piperacillin) but not of imipenem and meropenem. When used alone or in combination with ceftazidime, FR900098 increased the survival of infected Galleria mellonella and Caenorhabditis elegans. Furthermore, combining ceftazidime with FR900098 resulted in a significant inhibition of the biofilm formation of B. cenocepacia. Repeated exposure to FR900098 in the C. elegans infection model did not lead to decreased activity, and the susceptibility of the evolved B. cenocepacia HI2424 lineages to ceftazidime, FR900098 and the combination of both remained unchanged. In conclusion, FR900098 reduces B. cenocepacia virulence and potentiates ceftazidime in an in vivo C. elegans model, and this activity is not lost during the experimental evolution experiment carried out in the present study.


Asunto(s)
Burkholderia cenocepacia , Fosfomicina , Animales , Burkholderia cenocepacia/genética , Burkholderia cenocepacia/metabolismo , Caenorhabditis elegans , Fosfomicina/análogos & derivados , Fosfomicina/metabolismo , Fosfomicina/farmacología , Virulencia
6.
Molecules ; 26(16)2021 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-34443699

RESUMEN

Three α,α-difluorophosphonate derivatives of fosmidomycin were synthesized from diethyl 1,1-difluorobut-3-enylphosphonate and were evaluated on Escherichia coli. Two of them are among the best 1-deoxy-d-xylulose 5-phosphate reductoisomerase inhibitors, with IC50 in the nM range, much better than fosmidomycin, the reference compound. They also showed an enhanced antimicrobial activity against E. coli on Petri dishes in comparison with the corresponding phosphates and the non-fluorinated phosphonate.


Asunto(s)
Antibacterianos/farmacología , Fosfomicina/análogos & derivados , Ácidos Hidroxámicos/farmacología , Isomerasas Aldosa-Cetosa/antagonistas & inhibidores , Isomerasas Aldosa-Cetosa/metabolismo , Farmacorresistencia Bacteriana/efectos de los fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Fosfomicina/síntesis química , Fosfomicina/química , Fosfomicina/farmacología , Pruebas de Sensibilidad Microbiana
7.
Chem Biol Drug Des ; 98(5): 762-778, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34315189

RESUMEN

The failure to treat everyday bacterial infections is a current threat as pathogens are finding new ways to thwart antibiotics through mechanisms of resistance and intracellular refuge, thus rendering current antibiotic strategies ineffective. Cell-penetrating peptides (CPPs) are providing a means to improve antibiotics that are already approved for use. Through coadministration and conjugation of antibiotics with CPPs, improved accumulation and selectivity with alternative and/or additional modes of action against infections have been observed. Herein, we review the recent progress of this antibiotic-cell-penetrating peptide strategy in combatting sensitive and drug-resistant pathogens. We take a closer look into the specific antibiotics that have been enhanced, and in some cases repurposed as broad-spectrum drugs. Through the addition and conjugation of cell-penetrating peptides to antibiotics, increased permeation across mammalian and/or bacterial membranes and a broader range in bacterial selectivity have been achieved.


Asunto(s)
Antibacterianos/química , Infecciones Bacterianas/tratamiento farmacológico , Péptidos de Penetración Celular/química , Aminoglicósidos/química , Animales , Antibacterianos/farmacología , Péptidos de Penetración Celular/farmacología , Descubrimiento de Drogas , Resistencia a Antineoplásicos , Fosfomicina/análogos & derivados , Fosfomicina/farmacología , Humanos , Pruebas de Sensibilidad Microbiana , Preparaciones Farmacéuticas , Relación Estructura-Actividad
8.
Mol Microbiol ; 116(1): 97-108, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33561903

RESUMEN

The Gram-negative bacterium Pseudomonas aeruginosa is an opportunistic pathogen, responsible for many hospital-acquired infections. The bacterium is quite resistant toward many antibiotics, in particular because of the fine-tuned permeability of its outer membrane (OM). General diffusion outer membrane pores are quite rare in this organism. Instead, its OM contains many substrate-specific porins. Their expression is varying according to growth conditions and virulence. Phosphate limitations, as well as pathogenicity factors, result in the induction of the two mono- and polyphosphate-specific porins, OprP and OprO, respectively, together with an inner membrane uptake mechanism and a periplasmic binding protein. These outer membrane channels could serve as outer membrane pathways for the uptake of phosphonates. Among them are not only herbicides, but also potent antibiotics, such as fosfomycin and fosmidomycin. In this study, we investigated the interaction between OprP and OprO and fosmidomycin in detail. We could demonstrate that fosmidomycin is able to bind to the phosphate-specific binding site inside the two porins. The inhibition of chloride conductance of OprP and OprO by fosmidomycin is considerably less than that of phosphate or diphosphate, but it can be measured in titration experiments of chloride conductance and also in single-channel experiments. The results suggest that fosmidomycin transport across the OM of P. aeruginosa occurs through OprP and OprO. Our data with the ones already known in the literature show that phosphonic acid-containing antibiotics are in general good candidates to treat the infections of P. aeruginosa at the very beginning through a favorable OM transport system.


Asunto(s)
Antibacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Fosfomicina/análogos & derivados , Transporte Iónico/fisiología , Porinas/metabolismo , Pseudomonas aeruginosa/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/genética , Sitios de Unión/fisiología , Cloruros/metabolismo , Farmacorresistencia Bacteriana Múltiple/genética , Fosfomicina/metabolismo , Ácidos Fosforosos/metabolismo , Porinas/genética , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/genética
9.
Artículo en Inglés | MEDLINE | ID: mdl-33495219

RESUMEN

The malaria parasite Plasmodium falciparum contains the apicoplast organelle that synthesizes isoprenoids, which are metabolites necessary for posttranslational modification of Plasmodium proteins. We used fosmidomycin, an antibiotic that inhibits isoprenoid biosynthesis, to identify mechanisms that underlie the development of the parasite's adaptation to the drug at sublethal concentrations. We first determined a concentration of fosmidomycin that reduced parasite growth by ∼50% over one intraerythrocytic developmental cycle (IDC). At this dose, we maintained synchronous parasite cultures for one full IDC and collected metabolomic and transcriptomic data at multiple time points to capture global and stage-specific alterations. We integrated the data with a genome-scale metabolic model of P. falciparum to characterize the metabolic adaptations of the parasite in response to fosmidomycin treatment. Our simulations showed that, in treated parasites, the synthesis of purine-based nucleotides increased, whereas the synthesis of phosphatidylcholine during the trophozoite and schizont stages decreased. Specifically, the increased polyamine synthesis led to increased nucleotide synthesis, while the reduced methyl-group cycling led to reduced phospholipid synthesis and methyltransferase activities. These results indicate that fosmidomycin-treated parasites compensate for the loss of prenylation modifications by directly altering processes that affect nucleotide synthesis and ribosomal biogenesis to control the rate of RNA translation during the IDC. This also suggests that combination therapies with antibiotics that target the compensatory response of the parasite, such as nucleotide synthesis or ribosomal biogenesis, may be more effective than treating the parasite with fosmidomycin alone.


Asunto(s)
Antimaláricos , Apicoplastos , Fosfomicina , Malaria Falciparum , Antimaláricos/uso terapéutico , Fosfomicina/análogos & derivados , Fosfomicina/farmacología , Fosfomicina/uso terapéutico , Humanos , Malaria Falciparum/tratamiento farmacológico , Plasmodium falciparum/genética
10.
Curr Comput Aided Drug Des ; 17(2): 214-224, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32053077

RESUMEN

BACKGROUND: Considering the interesting role in the peptidoglycan biosynthesis pathway, the enzyme UDP-N-acetylglucosamine enolpyruvyl transferase is an attractive target to develop new antibacterial agents. It catalyzes the first key step of this pathway and its inhibition leads to bacterial cell death. Fosfomycin is known as the natural inhibitor of MurA. OBJECTIVE: The study aimed to introduce new inhibitors of MurA by virtual screening of different chemical compounds libraries, and test the best scored "virtual hits" against three pathogenic bacteria: Escherichia coli, Bacillus subtilis and Staphylococcus aureus. METHODS: A virtual screening of the structural analogues of fosfomycin downloaded from the Pub- Chem database was performed. Moreover, French National Chemical Library and ZINC database were also utilized to identify new structures different from fosfomycin. FlexX was the software used for this study. The antibacterial testing was divided into two methods: disk diffusion and broth dilution. RESULTS: A set of virtual hits was found to have better energy score than that of fosfomycin, seven of them were tested in vitro. In addition, the disk diffusion method explored four compounds that exhibited antibacterial activity: CID-21680357 (fosfomycin analogue), AB-00005001, ZINC04658565, and ZINC901335. The testing was continued by broth dilution method for both compounds CID-21680357 and ZINC901335 to determine their minimum inhibitory concentrations, and ZINC901335 had the best value with 457µg/ml against Staphylococcus aureus. CONCLUSION: Four compounds were found and proven in silico and in vitro to have antibacterial activity, namely CID-21680357, AB-00005001, ZINC04658565, and ZINC901335.


Asunto(s)
Transferasas Alquil y Aril/antagonistas & inhibidores , Antibacterianos/química , Inhibidores Enzimáticos/química , Fosfomicina/análogos & derivados , Simulación del Acoplamiento Molecular/métodos , Transferasas Alquil y Aril/metabolismo , Antibacterianos/farmacología , Pruebas Antimicrobianas de Difusión por Disco/métodos , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/farmacología , Escherichia coli/efectos de los fármacos , Escherichia coli/fisiología , Fosfomicina/farmacología , Pruebas de Sensibilidad Microbiana/métodos
11.
Bioorg Chem ; 105: 104280, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33152647

RESUMEN

A series of N-benzylated phosphoramidate esters, containing a 3,4-dihydroxyphenyl Mg2+-chelating group, has been synthesised in five steps as analogues of fosmidomycin, a Plasmodium falciparum 1-deoxy-1-d-xylulose-5-phosphate reductoisomerase (PfDXR) inhibitor. The 3,4-dihydroxyphenyl group effectively replaces the Mg2+-chelating hydroxamic acid group in fosmidomycin. The compounds showed very encouraging anti-parasitic activity with IC50 values of 5.6-16.4 µM against Plasmodium falciparum parasites and IC50 values of 5.2 - 10.2 µM against Trypanosoma brucei brucei (T.b.brucei). Data obtained from in silico docking of the ligands in the PfDXR receptor cavity (3AU9)5 support their potential as PfDXR inhibitors.


Asunto(s)
Amidas/síntesis química , Antimaláricos/síntesis química , Complejos de Coordinación/síntesis química , Magnesio/química , Ácidos Fosfóricos/síntesis química , Plasmodium falciparum/efectos de los fármacos , Antimaláricos/farmacología , Complejos de Coordinación/farmacología , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Fosfomicina/análogos & derivados , Fosfomicina/farmacología , Células HeLa , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Trypanosoma brucei brucei/efectos de los fármacos
12.
Molecules ; 25(20)2020 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-33096817

RESUMEN

Malaria, despite many efforts, remains among the most problematic infectious diseases worldwide, mainly due to the development of drug resistance by Plasmodium falciparum. The antibiotic fosmidomycin (FSM) is also known for its antimalarial activity by targeting the non-mevalonate isoprenoid synthesis pathway, which is essential for the malaria parasites but is absent in mammalians. In this study, we synthesized and evaluated against the chloroquine-resistant P. falciparum FcB1/Colombia strain, a series of FSM analogs, derivatives, and conjugates with other antimalarial agents, such as artemisinin (ART) and aminochloroquinoline (ACQ). The biological evaluation revealed four new compounds with higher antimalarial activity than FSM: two FSM-ACQ derivatives and two FSM-ART conjugates, with 3.5-5.4 and 41.5-23.1 times more potent activities than FSM, respectively.


Asunto(s)
Antimaláricos/farmacología , Artemisininas/farmacología , Fosfomicina/análogos & derivados , Plasmodium falciparum/efectos de los fármacos , Quinolinas/farmacología , Antimaláricos/síntesis química , Antimaláricos/química , Artemisininas/química , Fosfomicina/síntesis química , Fosfomicina/química , Fosfomicina/farmacología , Estructura Molecular , Pruebas de Sensibilidad Parasitaria , Quinolinas/química
13.
Molecules ; 25(9)2020 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-32403408

RESUMEN

Drug resistance is a major healthcare challenge, resulting in a continuous need to develop new inhibitors. The development of these inhibitors requires an understanding of the mechanisms of resistance for a critical mass of occurrences. Recent genome editing technologies based on high-throughput DNA synthesis and sequencing may help to predict mutations resulting in resistance by testing large mutagenesis libraries. Here we describe the rationale of this approach, with examples and relevance to drug development and resistance in malaria.


Asunto(s)
Isomerasas Aldosa-Cetosa/química , Evolución Molecular Dirigida/métodos , Resistencia a Medicamentos/genética , Malaria/tratamiento farmacológico , Mutagénesis , Isomerasas Aldosa-Cetosa/antagonistas & inhibidores , Isomerasas Aldosa-Cetosa/metabolismo , Antibacterianos/farmacología , Escherichia coli/genética , Escherichia coli/metabolismo , Fosfomicina/análogos & derivados , Fosfomicina/farmacología , Biblioteca de Genes , Mutación , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
14.
J Chem Theory Comput ; 16(4): 2751-2765, 2020 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-32167296

RESUMEN

To reach their site of action, it is essential for antibiotic molecules to cross the bacterial outer membrane. The progress of enhanced sampling techniques in molecular dynamics simulations enables us to understand these translocations at an atomic level. To this end, calculations of free energy surfaces for these permeation processes are of key importance. Herein, we investigate the translocation of a variety of anionic solutes through the outer membrane pore OprO of the Gram-negative bacterium Pseudomonas aeruginosa using the metadynamics and umbrella sampling techniques at the all-atom level. Free energy calculations have been performed employing these two distinct methods in order to illustrate the difference in computed free energies, if any. The investigated solutes range from a single atomic chloride ion over a multiatomic monophosphate ion to a more bulky fosmidomycin antibiotic. The role of complexity of the permeating solutes in estimating accurate free energy profiles is demonstrated by performing extensive convergence analysis. For simple monatomic ions, good agreement between the well-tempered metadynamics and the umbrella sampling approaches is achieved, while for the permeation of the monophosphate ion differences start to appear. In the case of larger molecules such as fosmidomycin it is a tough challenge to achieve converged free energy profiles. This issue is mainly due to neglecting orthogonal degrees of freedom during the free energy calculations. Nevertheless, the freely driven metadynamics approach leads to clearly advantageous results. Additionally, atomistic insights of the translocation mechanisms of all three solutes are discussed.


Asunto(s)
Proteínas Bacterianas/química , Simulación de Dinámica Molecular , Porinas/química , Cloruros/química , Entropía , Fosfomicina/análogos & derivados , Fosfomicina/química , Fosfatos/química
15.
PLoS Pathog ; 16(2): e1008316, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32059044

RESUMEN

Malaria parasites rely on a plastid organelle for survival during the blood stages of infection. However, the entire organelle is dispensable as long as the isoprenoid precursor, isopentenyl pyrophosphate (IPP), is supplemented in the culture medium. We engineered parasites to produce isoprenoid precursors from a mevalonate-dependent pathway, creating a parasite line that replicates normally after the loss of the apicoplast organelle. We show that carbon-labeled mevalonate is specifically incorporated into isoprenoid products, opening new avenues for researching this essential class of metabolites in malaria parasites. We also show that essential apicoplast proteins, such as the enzyme target of the drug fosmidomycin, can be deleted in this mevalonate bypass parasite line, providing a new method to determine the roles of other important apicoplast-resident proteins. Several antibacterial drugs kill malaria parasites by targeting basic processes, such as transcription, in the organelle. We used metabolomic and transcriptomic methods to characterize parasite metabolism after azithromycin treatment triggered loss of the apicoplast and found that parasite metabolism and the production of apicoplast proteins is largely unaltered. These results provide insight into the effects of apicoplast-disrupting drugs, several of which have been used to treat malaria infections in humans. Overall, the mevalonate bypass system provides a way to probe essential aspects of apicoplast biology and study the effects of drugs that target apicoplast processes.


Asunto(s)
Hemiterpenos/metabolismo , Ácido Mevalónico/metabolismo , Compuestos Organofosforados/metabolismo , Plasmodium falciparum/metabolismo , Animales , Antibacterianos/farmacología , Apicoplastos/genética , Apicoplastos/fisiología , Azitromicina/metabolismo , Fosfomicina/análogos & derivados , Fosfomicina/farmacología , Humanos , Malaria/metabolismo , Malaria/parasitología , Parásitos/metabolismo , Plastidios/parasitología , Proteínas Protozoarias/metabolismo
16.
mBio ; 10(6)2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31822583

RESUMEN

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. falciparumpgp 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.


Asunto(s)
Antimaláricos/farmacología , Carbono/metabolismo , Resistencia a Medicamentos/efectos de los fármacos , Fosfomicina/análogos & derivados , Monoéster Fosfórico Hidrolasas/metabolismo , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/metabolismo , Fosfomicina/farmacología , Glucólisis/efectos de los fármacos , Humanos , Lactatos/farmacología , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/metabolismo , Azúcares Ácidos/farmacología
17.
J Comput Aided Mol Des ; 33(10): 927-940, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31654265

RESUMEN

Proteins of the independent mevalonate pathway for isoprenoid biosynthesis are important targets for the development of new antibacterial compounds as this pathway is present in most pathogenic organisms such as Mycobacterium tuberculosis, DPlasmodium falciparum and Escherichia coli, but is not present in mammalian species, including humans. Deoxy-D-xylulose 5-phosphate reductoisomerase (DXR) is an important target in this pathway and the most effective DXR inhibitor to date is fosmidomycin, which is used to treat malaria and, more recently, tuberculosis. Recently, Armstrong C. M. et al. showed that a mutant of DXR, S222T, induces a loss of the fosmidomycin inhibition efficiency, even though the bacteria culture is still viable and able to produce isoprenoids. As this represents a potential fosmidomycin-resistant mutation, it is important to understand the mechanism of this apparent mutation-induced resistance to fosmidomycin. Here, we used molecular dynamics simulations and Molecular Mechanics/Poisson Boltzmann Surface Area analysis to understand the structural and energetic basis of the resistance. Our results suggest that the point mutation results in changes to the structural dynamics of an active site loop that probably protects the active site and facilitates enzymatic reaction. From the simulation analysis, we also showed that the mutation results in changes in the interaction energy profiles in a way that can explain the observed activity of the mutant protein toward the natural inhibitor deoxy-D-xylulose 5-phosphate. These results should be taken into consideration in future efforts to develop new therapeutic antibiotic compounds that target DXR.


Asunto(s)
Isomerasas Aldosa-Cetosa/antagonistas & inhibidores , Isomerasas Aldosa-Cetosa/metabolismo , Farmacorresistencia Microbiana , Escherichia coli/enzimología , Fosfomicina/análogos & derivados , Simulación de Dinámica Molecular , Mutación , Isomerasas Aldosa-Cetosa/genética , Antibacterianos/administración & dosificación , Antibacterianos/metabolismo , Sitios de Unión , Escherichia coli/efectos de los fármacos , Fosfomicina/administración & dosificación , Fosfomicina/metabolismo , Ligandos , Modelos Teóricos , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/enzimología , Pentosafosfatos/metabolismo , Conformación Proteica
18.
PLoS Pathog ; 15(10): e1008078, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31622442

RESUMEN

The antibiotic, fosmidomycin (FSM) targets the methylerythritol phosphate (MEP) pathway of isoprenoid synthesis by inhibiting the essential enzyme, 1-deoxy-D-xylulose 5-phosphate reductoisomerase (Dxr) and is lethal to intracellular parasites and bacteria. The obligate intracellular bacterial pathogen, Chlamydia trachomatis, alternates between two developmental forms: the extracellular, infectious elementary body (EB), and the intracellular, replicative form called the reticulate body (RB). Several stressful growth conditions including iron deprivation halt chlamydial cell division and cause development of a morphologically enlarged, but viable form termed an aberrant body (AB). This phenotype constitutes the chlamydial developmental state known as persistence. This state is reversible as removal of the stressor allows the chlamydiae to re-enter and complete the normal developmental cycle. Bioinformatic analysis indicates that C. trachomatis encodes a homolog of Dxr, but its function and the requirement for isoprenoid synthesis in chlamydial development is not fully understood. We hypothesized that chlamydial Dxr (DxrCT) is functional and that the methylerythritol phosphate (MEP) pathway is required for normal chlamydial development. Thus, FSM exposure should be lethal to C. trachomatis. Overexpression of chlamydial Dxr (DxrCT) in Escherichia coli under FSM exposure and in a conditionally lethal dxr mutant demonstrated that DxrCT functions similarly to E. coli Dxr. When Chlamydia-infected cultures were exposed to FSM, EB production was significantly reduced. However, titer recovery assays, electron microscopy, and peptidoglycan labeling revealed that FSM inhibition of isoprenoid synthesis is not lethal to C. trachomatis, but instead induces persistence. Bactoprenol is a critical isoprenoid required for peptidoglycan precursor assembly. We therefore conclude that FSM induces persistence in Chlamydia by preventing bactoprenol production necessary for peptidoglycan precursor assembly and subsequent cell division.


Asunto(s)
Antibacterianos/farmacología , Chlamydia trachomatis/efectos de los fármacos , Fosfomicina/análogos & derivados , Peptidoglicano/biosíntesis , Terpenos/metabolismo , Isomerasas Aldosa-Cetosa/antagonistas & inhibidores , Isomerasas Aldosa-Cetosa/genética , Isomerasas Aldosa-Cetosa/metabolismo , Línea Celular Tumoral , Infecciones por Chlamydia/patología , Chlamydia trachomatis/enzimología , Chlamydia trachomatis/fisiología , Escherichia coli/enzimología , Escherichia coli/genética , Escherichia coli/metabolismo , Fosfomicina/farmacología , Células HeLa , Humanos
19.
Nat Chem Biol ; 15(11): 1049-1056, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31451762

RESUMEN

Fosmidomycin and related molecules comprise a family of phosphonate natural products with potent antibacterial, antimalarial and herbicidal activities. To understand the biosynthesis of these compounds, we characterized the fosmidomycin producer, Streptomyces lavendulae, using biochemical and genetic approaches. We were unable to elicit production of fosmidomycin, instead observing the unsaturated derivative dehydrofosmidomycin, which we showed potently inhibits 1-deoxy-D-xylulose-5-phosphate reductoisomerase and has bioactivity against a number of bacteria. The genes required for dehydrofosmidomycin biosynthesis were established by heterologous expression experiments. Bioinformatics analyses, characterization of intermediates and in vitro biochemistry show that the biosynthetic pathway involves conversion of a two-carbon phosphonate precursor into the unsaturated three-carbon product via a highly unusual rearrangement reaction, catalyzed by the 2-oxoglutarate dependent dioxygenase DfmD. The required genes and biosynthetic pathway for dehydrofosmidomycin differ substantially from that of the related natural product FR-900098, suggesting that the ability to produce these bioactive molecules arose via convergent evolution.


Asunto(s)
Productos Biológicos/metabolismo , Fosfomicina/análogos & derivados , Organofosfonatos/metabolismo , Fosfomicina/biosíntesis , Genes Bacterianos , Familia de Multigenes , Streptomyces/genética
20.
Eur J Med Chem ; 181: 111555, 2019 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-31382119

RESUMEN

Thia analogs of fosmidomycin are potent inhibitors of the non-mevalonate isoprenoid biosynthesis enzyme 1-deoxy-d-xylulose 5-phosphate reductoisomerase (IspC, Dxr) of Plasmodium falciparum. Several new thioethers displayed antiplasmodial in vitro activity in the low nanomolar range, without apparent cytotoxic effects in HeLa cells. The (S)-(+)-enantiomer of a typical representative selectively inhibited IspC and the growth of P. falciparum in continuous culture. The inhibitor was stable at pH 7.6 and room temperature, and no racemization was observed under these conditions during a period of up to two days. Oxidation of selected thioethers to sulfones reduced antiplasmodial activity and the inhibitory activity against Escherichia coli, Mycobacterium tuberculosis and P. falciparum IspC orthologs.


Asunto(s)
Antibacterianos/farmacología , Antiprotozoarios/farmacología , Escherichia coli/efectos de los fármacos , Fosfomicina/análogos & derivados , Mycobacterium tuberculosis/efectos de los fármacos , Plasmodium falciparum/efectos de los fármacos , Antibacterianos/síntesis química , Antibacterianos/química , Antiprotozoarios/síntesis química , Antiprotozoarios/química , Células CACO-2 , Relación Dosis-Respuesta a Droga , Escherichia coli/crecimiento & desarrollo , Fosfomicina/síntesis química , Fosfomicina/química , Fosfomicina/farmacología , Células HeLa , Humanos , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Mycobacterium tuberculosis/crecimiento & desarrollo , Pruebas de Sensibilidad Parasitaria , Plasmodium falciparum/crecimiento & desarrollo , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA