Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
ACS Infect Dis ; 5(1): 90-101, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30375858

ABSTRACT

Malaria is one of the most challenging human infectious diseases, and both prevention and control have been hindered by the development of Plasmodium falciparum resistance to existing therapies. Several new compounds with novel mechanisms are in clinical development for the treatment of malaria, including DSM265, an inhibitor of Plasmodium dihydroorotate dehydrogenase. To explore the mechanisms by which resistance might develop to DSM265 in the field, we selected for DSM265-resistant P. falciparum parasites in vitro. Any of five different amino acid changes led to reduced efficacy on the parasite and to decreased DSM265 binding to P. falciparum DHODH. The DSM265-resistant parasites retained full sensitivity to atovaquone. All but one of the observed mutations were in the DSM265 binding site, and the remaining C276F was in the adjacent flavin cofactor site. The C276F mutation was previously identified in a recrudescent parasite during a Phase IIa clinical study. We confirmed that this mutation (and the related C276Y) accounted for the full level of observed DSM265 resistance by regenerating the mutation using CRISPR/Cas9 genome editing. X-ray structure analysis of the C276F mutant enzyme showed that conformational changes of nearby residues were required to accommodate the larger F276 residue, which in turn led to a restriction in the size of the DSM265 binding pocket. These findings underscore the importance of developing DSM265 as part of a combination therapy with other agents for successful use against malaria.


Subject(s)
Antimalarials/pharmacology , Drug Resistance/genetics , Oxidoreductases Acting on CH-CH Group Donors/genetics , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Point Mutation , Pyrimidines/pharmacology , Triazoles/pharmacology , Amino Acid Sequence , Animals , Binding Sites , CRISPR-Cas Systems , Crystallography, X-Ray , Dihydroorotate Dehydrogenase , Gene Editing , Humans , Malaria, Falciparum , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Plasmodium falciparum/enzymology
2.
Structure ; 26(11): 1499-1512.e5, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30197036

ABSTRACT

Trypanosoma brucei is a neglected tropical disease endemic to Africa. The polyamine spermidine is essential for post-translational hypusine modification of eukaryotic initiation factor 5A (eIF5A), which is catalyzed by deoxyhypusine synthase (TbDHS). In trypanosomatids, deoxyhypusine synthase (DHS) activity is dependent on heterotetramer formation between two paralogs, DHSc and DHSp, both with minimal activity on their own due to missing catalytic residues. We determined the X-ray structure of TbDHS showing a single functional shared active site is formed at the DHSc/DHSp heterodimer interface, with deficiencies in one subunit complemented by the other. Each heterodimer contains two NAD+ binding sites, one housed in the functional catalytic site and the second bound in a remnant dead site that lacks key catalytic residues. Functional analysis of these sites by site-directed mutagenesis identified long-range contributions to the catalytic site from the dead site. Differences between trypanosomatid and human DHS that could be exploited for drug discovery were identified.


Subject(s)
NAD/metabolism , Oxidoreductases Acting on CH-NH Group Donors/chemistry , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Trypanosoma brucei brucei/enzymology , Animals , Catalytic Domain , Crystallography, X-Ray , Humans , Models, Molecular , Protein Multimerization , Protozoan Proteins/chemistry , Species Specificity , Substrate Specificity , Trypanosoma brucei brucei/chemistry
3.
Mol Microbiol ; 106(3): 439-451, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28836704

ABSTRACT

Lipoate is an essential cofactor for enzymes that are important for central metabolism and other processes. In malaria parasites, scavenged lipoate from the human host is required for survival. The Plasmodium falciparum mitochondrion contains two enzymes (PfLipL1 and PfLipL2) that are responsible for activating mitochondrial proteins through the covalent attachment of lipoate (lipoylation). Lipoylation occurs via a novel redox-gated mechanism that remains poorly understood. We show that PfLipL1 functions as a redox switch that determines which downstream proteins will be activated. Based on the lipoate redox state, PfLipL1 either functions as a canonical lipoate ligase or as a lipoate activating enzyme which works in conjunction with PfLipL2. We demonstrate that PfLipL2 is a lipoyltransferase and is a member of a novel clade of lipoate attachment enzymes. We show that a LipL2 enzyme from Chlamydia trachomatis has similar activity, demonstrating conservation between intracellular pathogens from different phylogenetic kingdoms and supporting the hypothesis that an early ancestor of malaria parasites once contained a chlamydial endosymbiont. Redox-dependent lipoylation may regulate processes such as central metabolism and oxidative defense pathways.


Subject(s)
Lipoylation/genetics , Lipoylation/physiology , Chlamydia/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Nucleotidyltransferases , Oxidation-Reduction , Peptide Synthases/genetics , Plasmodium/metabolism , Plasmodium falciparum/genetics , Protozoan Proteins/metabolism , Sequence Alignment
4.
Proteins ; 85(9): 1777-1783, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28543853

ABSTRACT

Plasmodium falciparum lipoate protein ligase 1 (PfLipL1) is an ATP-dependent ligase that belongs to the biotin/lipoate A/B protein ligase family (PFAM PF03099). PfLipL1 is the only known canonical lipoate ligase in Pf and functions as a redox switch between two lipoylation routes in the parasite mitochondrion. Here, we report the crystal structure of a deletion construct of PfLipL1 (PfLipL1Δ243-279 ) bound to lipoate, and validate the lipoylation activity of this construct in both an in vitro lipoylation assay and a cell-based lipoylation assay. This characterization represents the first step in understanding the redox dependence of the lipoylation mechanism in malaria parasites. Proteins 2017; 85:1777-1783. © 2017 Wiley Periodicals, Inc.


Subject(s)
Peptide Synthases/chemistry , Plasmodium falciparum/enzymology , Protein Conformation , Protozoan Proteins/chemistry , Adenosine Triphosphate/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Protein Binding
5.
Mol Microbiol ; 94(1): 156-71, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25116855

ABSTRACT

Lipoate scavenging from the human host is essential for malaria parasite survival. Scavenged lipoate is covalently attached to three parasite proteins: the H-protein and the E2 subunits of branched chain amino acid dehydrogenase (BCDH) and α-ketoglutarate dehydrogenase (KDH). We show mitochondrial localization for the E2 subunits of BCDH and KDH, similar to previously localized H-protein, demonstrating that all three lipoylated proteins reside in the parasite mitochondrion. The lipoate ligase 1, LipL1, has been shown to reside in the mitochondrion and it catalyses the lipoylation of the H-protein; however, we show that LipL1 alone cannot lipoylate BCDH or KDH. A second mitochondrial protein with homology to lipoate ligases, LipL2, does not show ligase activity and is not capable of lipoylating any of the mitochondrial substrates. Instead, BCDH and KDH are lipoylated through a novel mechanism requiring both LipL1 and LipL2. This mechanism is sensitive to redox conditions where BCDH and KDH are exclusively lipoylated under strong reducing conditions in contrast to the H-protein which is preferentially lipoylated under less reducing conditions. Thus, malaria parasites contain two different routes of mitochondrial lipoylation, an arrangement that has not been described for any other organism.


Subject(s)
Mitochondrial Proteins/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Humans , Ketoglutarate Dehydrogenase Complex/genetics , Ketoglutarate Dehydrogenase Complex/metabolism , Lipoylation , Malaria, Falciparum/parasitology , Mitochondrial Proteins/genetics , Oxidation-Reduction , Peptide Synthases/genetics , Peptide Synthases/metabolism , Plasmodium falciparum/chemistry , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Protozoan Proteins/genetics
6.
Bioorg Med Chem Lett ; 24(3): 911-6, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24398298

ABSTRACT

The enoyl acyl-carrier protein reductase (ENR) enzyme of the apicomplexan parasite family has been intensely studied for antiparasitic drug design for over a decade, with the most potent inhibitors targeting the NAD(+) bound form of the enzyme. However, the higher affinity for the NADH co-factor over NAD(+) and its availability in the natural environment makes the NADH complex form of ENR an attractive target. Herein, we have examined a benzimidazole family of inhibitors which target the NADH form of Francisella ENR, but despite good efficacy against Toxoplasma gondii, the IC50 for T. gondii ENR is poor, with no inhibitory activity at 1 µM. Moreover similar benzimidazole scaffolds are potent against fungi which lack the ENR enzyme and as such we believe that there may be significant off target effects for this family of inhibitors.


Subject(s)
Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Drug Delivery Systems , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Toxoplasma/drug effects , Antiparasitic Agents/chemistry , Antiparasitic Agents/pharmacology , Crystallography, X-Ray , Enzyme Activation/drug effects , Inhibitory Concentration 50 , Microbial Sensitivity Tests , Molecular Structure
7.
Biochemistry ; 52(51): 9155-66, 2013 Dec 23.
Article in English | MEDLINE | ID: mdl-24295325

ABSTRACT

Many microbial pathogens rely on a type II fatty acid synthesis (FASII) pathway that is distinct from the type I pathway found in humans. Enoyl-acyl carrier protein reductase (ENR) is an essential FASII pathway enzyme and the target of a number of antimicrobial drug discovery efforts. The biocide triclosan is established as a potent inhibitor of ENR and has been the starting point for medicinal chemistry studies. We evaluated a series of triclosan analogues for their ability to inhibit the growth of Toxoplasma gondii, a pervasive human pathogen, and its ENR enzyme (TgENR). Several compounds that inhibited TgENR at low nanomolar concentrations were identified but could not be further differentiated because of the limited dynamic range of the TgENR activity assay. Thus, we adapted a thermal shift assay (TSA) to directly measure the dissociation constant (Kd) of the most potent inhibitors identified in this study as well as inhibitors from previous studies. Furthermore, the TSA allowed us to determine the mode of action of these compounds in the presence of the reduced nicotinamide adenine dinucleotide (NADH) or nicotinamide adenine dinucleotide (NAD⁺) cofactor. We found that all of the inhibitors bind to a TgENR-NAD⁺ complex but that they differed in their dependence on NAD⁺ concentration. Ultimately, we were able to identify compounds that bind to the TgENR-NAD⁺ complex in the low femtomolar range. This shows how TSA data combined with enzyme inhibition, parasite growth inhibition data, and ADMET predictions allow for better discrimination between potent ENR inhibitors for the future development of medicine.


Subject(s)
Antiprotozoal Agents/pharmacology , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Protozoan Proteins/antagonists & inhibitors , Toxoplasma/enzymology , Triclosan/analogs & derivatives , Antiprotozoal Agents/adverse effects , Antiprotozoal Agents/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Drug Design , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/chemistry , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/chemistry , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/parasitology , High-Throughput Screening Assays , Hot Temperature , Humans , Inhibitory Concentration 50 , Kinetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Molecular Conformation , Molecular Docking Simulation , NAD/chemistry , NAD/metabolism , Oxidation-Reduction , Protein Unfolding , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Toxoplasma/drug effects , Toxoplasma/growth & development , Triclosan/adverse effects , Triclosan/chemistry , Triclosan/pharmacology
8.
ChemMedChem ; 8(7): 1138-60, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23776166

ABSTRACT

Through our focused effort to discover new and effective agents against toxoplasmosis, a structure-based drug design approach was used to develop a series of potent inhibitors of the enoyl-acyl carrier protein (ACP) reductase (ENR) enzyme in Toxoplasma gondii (TgENR). Modifications to positions 5 and 4' of the well-known ENR inhibitor triclosan afforded a series of 29 new analogues. Among the resulting compounds, many showed high potency and improved physicochemical properties in comparison with the lead. The most potent compounds 16 a and 16 c have IC50 values of 250 nM against Toxoplasma gondii tachyzoites without apparent toxicity to the host cells. Their IC50 values against recombinant TgENR were found to be 43 and 26 nM, respectively. Additionally, 11 other analogues in this series had IC50 values ranging from 17 to 130 nM in the enzyme-based assay. With respect to their excellent in vitro activity as well as improved drug-like properties, the lead compounds 16 a and 16 c are deemed to be excellent starting points for the development of new medicines to effectively treat Toxoplasma gondii infections.


Subject(s)
Antiprotozoal Agents/pharmacology , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Toxoplasma/enzymology , Toxoplasmosis/drug therapy , Triclosan/pharmacology , Animals , Antiprotozoal Agents/chemical synthesis , Antiprotozoal Agents/chemistry , Caco-2 Cells , Disease Models, Animal , Dose-Response Relationship, Drug , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Mice , Models, Molecular , Molecular Structure , Parasitic Sensitivity Tests , Permeability/drug effects , Plasmodium falciparum/drug effects , Structure-Activity Relationship , Toxoplasma/drug effects , Triclosan/chemical synthesis , Triclosan/chemistry
9.
Bioorg Med Chem Lett ; 23(12): 3551-5, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23664871

ABSTRACT

The enoyl acyl-carrier protein reductase (ENR) enzyme is harbored within the apicoplast of apicomplexan parasites providing a significant challenge for drug delivery, which may be overcome through the addition of transductive peptides, which facilitates crossing the apicoplast membranes. The binding site of triclosan, a potent ENR inhibitor, is occluded from the solvent making the attachment of these linkers challenging. Herein, we have produced 3 new triclosan analogs with bulky A- and B-ring motifs, which protrude into the solvent allowing for the future attachment of molecular transporters for delivery.


Subject(s)
Carrier Proteins/chemistry , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/chemistry , Triclosan/analogs & derivatives , Binding Sites , Carrier Proteins/metabolism , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Models, Molecular , Plasmodium falciparum/metabolism , Toxoplasma/metabolism , Triclosan/chemical synthesis , Triclosan/chemistry , Triclosan/pharmacology
10.
Bioorg Med Chem Lett ; 23(7): 2035-43, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23453069

ABSTRACT

Triclosan is a potent inhibitor of Toxoplasma gondii enoyl reductase (TgENR), which is an essential enzyme for parasite survival. In view of triclosan's poor druggability, which limits its therapeutic use, a new set of B-ring modified analogs were designed to optimize its physico-chemical properties. These derivatives were synthesized and evaluated by in vitro assay and TgENR enzyme assay. Some analogs display improved solubility, permeability and a comparable MIC50 value to that of triclosan. Modeling of these inhibitors revealed the same overall binding mode with the enzyme as triclosan, but the B-ring modifications have additional interactions with the strongly conserved Asn130.


Subject(s)
Drug Design , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Toxoplasma/enzymology , Triclosan/pharmacology , Dose-Response Relationship, Drug , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Triclosan/chemical synthesis , Triclosan/chemistry
11.
ChemMedChem ; 8(3): 442-61, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23341167

ABSTRACT

Malaria is a potentially fatal disease caused by Plasmodium parasites and poses a major medical risk in large parts of the world. The development of new, affordable antimalarial drugs is of vital importance as there are increasing reports of resistance to the currently available therapeutics. In addition, most of the current drugs used for chemoprophylaxis merely act on parasites already replicating in the blood. At this point, a patient might already be suffering from the symptoms associated with the disease and could additionally be infectious to an Anopheles mosquito. These insects act as a vector, subsequently spreading the disease to other humans. In order to cure not only malaria but prevent transmission as well, a drug must target both the blood- and pre-erythrocytic liver stages of the parasite. P. falciparum (Pf) enoyl acyl carrier protein (ACP) reductase (ENR) is a key enzyme of plasmodial type II fatty acid biosynthesis (FAS II). It has been shown to be essential for liver-stage development of Plasmodium berghei and is therefore qualified as a target for true causal chemoprophylaxis. Using virtual screening based on two crystal structures of PfENR, we identified a structurally novel class of FAS inhibitors. Subsequent chemical optimization yielded two compounds that are effective against multiple stages of the malaria parasite. These two most promising derivatives were found to inhibit blood-stage parasite growth with IC(50) values of 1.7 and 3.0 µM and lead to a more prominent developmental attenuation of liver-stage parasites than the gold-standard drug, primaquine.


Subject(s)
Antimalarials/chemistry , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Fatty Acids/biosynthesis , Antimalarials/chemical synthesis , Antimalarials/toxicity , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/toxicity , HeLa Cells , Humans , Molecular Docking Simulation , Plasmodium berghei/drug effects , Plasmodium berghei/enzymology , Protein Structure, Tertiary , Structure-Activity Relationship
12.
Antimicrob Agents Chemother ; 56(5): 2666-82, 2012 May.
Article in English | MEDLINE | ID: mdl-22354304

ABSTRACT

Toxoplasma gondii is a protozoan parasite that can damage the human brain and eyes. There are no curative medicines. Herein, we describe our discovery of N-benzoyl-2-hydroxybenzamides as a class of compounds effective in the low nanomolar range against T. gondii in vitro and in vivo. Our lead compound, QQ-437, displays robust activity against the parasite and could be useful as a new scaffold for development of novel and improved inhibitors of T. gondii. Our genome-wide investigations reveal a specific mechanism of resistance to N-benzoyl-2-hydroxybenzamides mediated by adaptin-3ß, a large protein from the secretory protein complex. N-Benzoyl-2-hydroxybenzamide-resistant clones have alterations of their secretory pathway, which traffics proteins to micronemes, rhoptries, dense granules, and acidocalcisomes/plant-like vacuole (PLVs). N-Benzoyl-2-hydroxybenzamide treatment also alters micronemes, rhoptries, the contents of dense granules, and, most markedly, acidocalcisomes/PLVs. Furthermore, QQ-437 is active against chloroquine-resistant Plasmodium falciparum. Our studies reveal a novel class of compounds that disrupts a unique secretory pathway of T. gondii, with the potential to be used as scaffolds in the search for improved compounds to treat the devastating diseases caused by apicomplexan parasites.


Subject(s)
Adaptor Proteins, Vesicular Transport/antagonists & inhibitors , Antiprotozoal Agents/pharmacology , Benzamides/pharmacology , Protozoan Proteins/antagonists & inhibitors , Toxoplasma/drug effects , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Antimalarials/chemical synthesis , Antimalarials/pharmacology , Antiprotozoal Agents/chemical synthesis , Benzamides/chemical synthesis , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/parasitology , Humans , Inhibitory Concentration 50 , Organelles/drug effects , Organelles/genetics , Organelles/metabolism , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protein Transport/drug effects , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Quantitative Structure-Activity Relationship , Secretory Pathway/drug effects , Secretory Pathway/physiology , Toxoplasma/genetics , Toxoplasma/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...