Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Microb Biotechnol ; 17(4): e14444, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38564168

ABSTRACT

Assisted reproductive techniques are routinely used in livestock species to increase and enhance productivity. Ovarian hyperstimulation is a process that currently relies on administering pituitary-derived follicle-stimulating hormone (FSH) or equine chorionic gonadotropin in combination with other hormones to promote the maturation of multiple follicles and thereby achieve superovulation. The use of partially purified preparations of FSH extracted from natural sources is associated with suboptimal and variable results. Recombinant FSH (rFSH) has been produced in a variety of heterologous organisms. However, attaining a bioactive rFSH of high quality and at low cost for use in livestock remains challenging. Here we report the production and characterization of a single chain bovine rFSH consisting of the ß- and α-subunit fused by a polypeptide linker (scbFSH) using Leishmania tarentolae as heterologous expression system. This unicellular eukaryote is non-pathogenic to mammals, can be grown in bioreactors using simple and inexpensive semisynthetic media at 26°C and does not require CO2 or bovine serum supplementation. Stable cell lines expressing scbFSH in an inducible fashion were generated and characterized for their productivity. Different culture conditions and purification procedures were evaluated, and the recombinant product was biochemically and biologically characterized, including bioassays in an animal model. The results demonstrate that L. tarentolae is a suitable host for producing a homogeneous, glycosylated and biologically active form of scbFSH with a reasonable yield.


Subject(s)
Leishmania , Female , Animals , Horses , Leishmania/genetics , Biological Assay , Bioreactors , Cell Line , Follicle Stimulating Hormone , Mammals
2.
Drug Dev Res ; 83(2): 253-263, 2022 04.
Article in English | MEDLINE | ID: mdl-31958156

ABSTRACT

African trypanosomiasis is a major problem for human and animal health in endemic countries, where it threatens millions of people and affects economic development. New drugs are needed to overcome the toxicity, administration, low efficacy, and resistance issues of the current chemotherapy. Robust, simple, and economical high-throughput, whole-cell-based assays are required to accelerate the identification of novel chemical entities. With this aim, we generated a bioluminescent cell line of the bloodstream stage of Trypanosoma brucei brucei and established a screening assay. Trypanosomes were stably transfected to constitutively express a thermostable red-shifted luciferase. The growth phenotype and drug sensitivity of the reporter cell line were essentially identical to that of the parental cell line. The endogenous luciferase activity, measured by a simple bioluminescence assay, proved to be proportional to parasite number and metabolic status. The assay, optimized to detect highly potent compounds in a 96-well-plate format, was validated by screening a small compound library (inter-assay values for Z' factor and coefficient variation were 0.77 and 5.8%, respectively). With a hit-confirmation ratio of ~97%, the assay was potent enough to identify several hits with EC50 ≤ 10 µM. Preliminary tests indicated that the assay can be scaled up to a 384-well-plate format without compromising its robustness. In summary, we have generated reporter trypanosomes and a simple, robust, and affordable bioluminescence screening assay with great potential to speed up the early-phase drug discovery against African trypanosomes.


Subject(s)
Trypanocidal Agents , Trypanosoma brucei brucei , Animals , Drug Discovery , Humans , Luciferases/genetics , Luminescent Measurements , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/genetics
3.
PLoS Pathog ; 15(9): e1008065, 2019 09.
Article in English | MEDLINE | ID: mdl-31557263

ABSTRACT

Most known thioredoxin-type proteins (Trx) participate in redox pathways, using two highly conserved cysteine residues to catalyze thiol-disulfide exchange reactions. Here we demonstrate that the so far unexplored Trx2 from African trypanosomes (Trypanosoma brucei) lacks protein disulfide reductase activity but functions as an effective temperature-activated and redox-regulated chaperone. Immunofluorescence microscopy and fractionated cell lysis revealed that Trx2 is located in the mitochondrion of the parasite. RNA-interference and gene knock-out approaches showed that depletion of Trx2 impairs growth of both mammalian bloodstream and insect stage procyclic parasites. Procyclic cells lacking Trx2 stop proliferation under standard culture conditions at 27°C and are unable to survive prolonged exposure to 37°C, indicating that Trx2 plays a vital role that becomes augmented under heat stress. Moreover, we found that Trx2 contributes to the in vivo infectivity of T. brucei. Remarkably, a Trx2 version, in which all five cysteines were replaced by serine residues, complements for the wildtype protein in conditional knock-out cells and confers parasite infectivity in the mouse model. Characterization of the recombinant protein revealed that Trx2 can coordinate an iron sulfur cluster and is highly sensitive towards spontaneous oxidation. Moreover, we discovered that both wildtype and mutant Trx2 protect other proteins against thermal aggregation and preserve their ability to refold upon return to non-stress conditions. Activation of the chaperone function of Trx2 appears to be triggered by temperature-mediated structural changes and inhibited by oxidative disulfide bond formation. Our studies indicate that Trx2 acts as a novel chaperone in the unique single mitochondrion of T. brucei and reveal a new perspective regarding the physiological function of thioredoxin-type proteins in trypanosomes.


Subject(s)
Protozoan Proteins/metabolism , Thioredoxins/metabolism , Trypanosoma brucei brucei/metabolism , Animals , Gene Knockdown Techniques , Genes, Protozoan , Humans , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mutation , Oxidation-Reduction , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thioredoxins/antagonists & inhibitors , Thioredoxins/genetics , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/pathogenicity
4.
Methods Mol Biol ; 1955: 105-118, 2019.
Article in English | MEDLINE | ID: mdl-30868522

ABSTRACT

Trypanosomatids are unicellular organisms that colonize a wide diversity of environments and hosts. For instance, Trypanosoma cruzi is a human pathogen responsible for Chagas diseases, while Leishmania tarentolae infects amphibians and became a biotechnological tool suitable for recombinant protein expression. T. cruzi antigens are needed for the development of improved epitope-based methods for diagnosis and treatment of Chagas disease. Molecular cloning for the production of recombinant proteins offers the possibility to obtain T. cruzi antigens at high yield and purity. L. tarentolae appears as the ideal expression host to obtain recombinant T. cruzi antigens with a structure and posttranslational modifications typical of trypanosomatids. In this chapter, we present a protocol for the analytical to mid-scale production of recombinant T. cruzi antigens, using L. tarentolae as expression host (LEXSY® inducible system).


Subject(s)
Antigens, Protozoan/genetics , Cloning, Molecular/methods , Leishmania/genetics , Trypanosoma cruzi/genetics , Chagas Disease/parasitology , Genetic Vectors/genetics , Humans , Plasmids/genetics , Recombinant Proteins/genetics , Transfection/methods
5.
J Biol Chem ; 294(9): 3235-3248, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30593501

ABSTRACT

Trypanosomes are flagellated protozoan parasites (kinetoplastids) that have a unique redox metabolism based on the small dithiol trypanothione (T(SH)2). Although GSH may still play a biological role in trypanosomatid parasites beyond being a building block of T(SH)2, most of its functions are replaced by T(SH)2 in these organisms. Consequently, trypanosomes have several enzymes adapted to using T(SH)2 instead of GSH, including the glutaredoxins (Grxs). However, the mechanistic basis of Grx specificity for T(SH)2 is unknown. Here, we combined fast-kinetic and biophysical approaches, including NMR, MS, and fluorescent tagging, to study the redox function of Grx1, the only cytosolic redox-active Grx in trypanosomes. We observed that Grx1 reduces GSH-containing disulfides (including oxidized trypanothione) in very fast reactions (k > 5 × 105 m-1 s-1). We also noted that disulfides without a GSH are much slower oxidants, suggesting a strongly selective binding of the GSH molecule. Not surprisingly, oxidized Grx1 was also reduced very fast by T(SH)2 (4.8 × 106 m-1 s-1); however, GSH-mediated reduction was extremely slow (39 m-1 s-1). This kinetic selectivity in the reduction step of the catalytic cycle suggests that Grx1 uses preferentially a dithiol mechanism, forming a disulfide on the active site during the oxidative half of the catalytic cycle and then being rapidly reduced by T(SH)2 in the reductive half. Thus, the reduction of glutathionylated substrates avoids GSSG accumulation in an organism lacking GSH reductase. These findings suggest that Grx1 has played an important adaptive role during the rewiring of the thiol-redox metabolism of kinetoplastids.


Subject(s)
Biological Evolution , Glutaredoxins/metabolism , Sulfhydryl Compounds/metabolism , Trypanosoma/metabolism , Animals , Catalytic Domain , Glutaredoxins/chemistry , Humans , Kinetics , Oxidation-Reduction
6.
Sci Rep ; 8(1): 13716, 2018 09 12.
Article in English | MEDLINE | ID: mdl-30209332

ABSTRACT

Glutaredoxins (Grx) are small proteins conserved throughout all the kingdoms of life that are engaged in a wide variety of biological processes and share a common thioredoxin-fold. Among them, class II Grx are redox-inactive proteins involved in iron-sulfur (FeS) metabolism. They contain a single thiol group in their active site and use low molecular mass thiols such as glutathione as ligand for binding FeS-clusters. In this study, we investigated molecular aspects of 1CGrx1 from the pathogenic parasite Trypanosoma brucei brucei, a mitochondrial class II Grx that fulfills an indispensable role in vivo. Mitochondrial 1CGrx1 from trypanosomes differs from orthologues in several features including the presence of a parasite-specific N-terminal extension (NTE) whose role has yet to be elucidated. Previously we have solved the structure of a truncated form of 1CGrx1 containing only the conserved glutaredoxin domain but lacking the NTE. Our aim here is to investigate the effect of the NTE on the conformation of the protein. We therefore solved the NMR structure of the full-length protein, which reveals subtle but significant differences with the structure of the NTE-less form. By means of different experimental approaches, the NTE proved to be intrinsically disordered and not involved in the non-redox dependent protein dimerization, as previously suggested. Interestingly, the portion comprising residues 65-76 of the NTE modulates the conformational dynamics of the glutathione-binding pocket, which may play a role in iron-sulfur cluster assembly and delivery. Furthermore, we disclosed that the class II-strictly conserved loop that precedes the active site is critical for stabilizing the protein structure. So far, this represents the first communication of a Grx containing an intrinsically disordered region that defines a new protein subgroup within class II Grx.


Subject(s)
Iron-Sulfur Proteins/metabolism , Regulatory Sequences, Nucleic Acid/physiology , Sulfur/metabolism , Trypanosoma brucei brucei/metabolism , Trypanosoma/metabolism , Amino Acid Sequence , Catalytic Domain/physiology , Glutaredoxins/metabolism , Glutathione/metabolism , Oxidation-Reduction , Protein Conformation , Protein Multimerization/physiology
7.
Redox Biol ; 15: 532-547, 2018 05.
Article in English | MEDLINE | ID: mdl-29413965

ABSTRACT

Trypanosoma brucei glutaredoxin 2 (Grx2) is a dithiol glutaredoxin that is specifically located in the mitochondrial intermembrane space. Bloodstream form parasites lacking Grx2 or both, Grx2 and the cytosolic Grx1, are viable in vitro and infectious to mice suggesting that neither oxidoreductase is needed for survival or infectivity to mammals. A 37 °C to 39 °C shift changes the cellular redox milieu of bloodstream cells to more oxidizing conditions and induces a significantly stronger growth arrest in wildtype parasites compared to the mutant cells. Grx2-deficient cells ectopically expressing the wildtype form of Grx2 with its C31QFC34 active site, but not the C34S mutant, regain the sensitivity of the parental strain, indicating that the physiological role of Grx2 requires both active site cysteines. In the procyclic insect stage of the parasite, Grx2 is essential. Both alleles can be replaced if procyclic cells ectopically express authentic or C34S, but not C31S/C34S Grx2, pointing to a redox role that relies on a monothiol mechanism. RNA-interference against Grx2 causes a virtually irreversible proliferation defect. The cells adopt an elongated morphology but do not show any significant alteration in the cell cycle. The growth retardation is attenuated by high glucose concentrations. Under these conditions, procyclic cells obtain ATP by substrate level phosphorylation suggesting that Grx2 might regulate a respiratory chain component.


Subject(s)
Adaptation, Physiological/genetics , Glutaredoxins/genetics , Trypanosoma brucei brucei/metabolism , Trypanosomiasis, African/metabolism , Adenosine Triphosphate/metabolism , Alleles , Animals , Catalytic Domain , Cell Proliferation/genetics , Cytosol/metabolism , Glutaredoxins/chemistry , Glutaredoxins/metabolism , Hot Temperature , Humans , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/parasitology , Mitochondrial Membranes/metabolism , Mutation , Oxidation-Reduction , Trypanosoma brucei brucei/pathogenicity , Trypanosomiasis, African/parasitology , Trypanosomiasis, African/pathology
8.
Antioxid Redox Signal ; 28(6): 463-486, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29048199

ABSTRACT

SIGNIFICANCE: Major pathogenic enterobacteria and protozoan parasites from the phylum Euglenozoa, such as trypanosomatids, are endowed with glutathione (GSH)-spermidine (Sp) derivatives that play important roles in signaling and metal and thiol-redox homeostasis. For some Euglenozoa lineages, the GSH-Sp conjugates represent the main redox cosubstrates around which entire new redox systems have evolved. Several proteins underwent molecular adaptations to synthesize and utilize the new polyamine-based thiols. Recent Advances: The genomes of closely related organisms have recently been sequenced, which allows mining and analysis of gene sequences that belong to these peculiar redox systems. Similarly, the three-dimensional structures of several of these proteins have been solved, which allows for comparison with their counterparts in classical redox systems that rely on GSH/glutaredoxin and thioredoxin. CRITICAL ISSUES: The evolutionary and structural aspects related to the emergence and use of GSH-Sp conjugates in Euglenozoa are reviewed focusing on unique structural specializations that proteins developed to use N1,N8-bisglutathionylspermidine (trypanothione) as redox cosubstrate. An updated overview on the biochemical and biological significance of the major enzymatic activities is also provided. FUTURE DIRECTIONS: A thiol-redox system strictly dependent on trypanothione is a feature unique to trypanosomatids. The physicochemical properties of the polyamine-GSH conjugates were a major driving force for structural adaptation of proteins that use these thiols as ligand and redox cofactor. In fact, the structural differences of indispensable components of this system can be exploited toward selective drug development. Future research should clarify whether additional cellular processes are regulated by the trypanothione system. Antioxid. Redox Signal. 28, 463-486.


Subject(s)
Glutaredoxins/genetics , Sulfhydryl Compounds/chemistry , Thioredoxins/genetics , Trypanosomatina/metabolism , Evolution, Molecular , Glutaredoxins/chemistry , Glutaredoxins/metabolism , Oxidation-Reduction , Polyamines/chemistry , Polyamines/metabolism , Spermidine/chemistry , Spermidine/metabolism , Sulfhydryl Compounds/metabolism , Thioredoxins/chemistry , Thioredoxins/metabolism , Trypanosomatina/chemistry , Trypanosomatina/genetics
9.
Antioxid Redox Signal ; 27(18): 1491-1504, 2017 Dec 20.
Article in English | MEDLINE | ID: mdl-28463568

ABSTRACT

AIMS: New drugs are needed to treat flatworm infections that cause severe human diseases such as schistosomiasis. The unique flatworm enzyme thioredoxin glutathione reductase (TGR), structurally different from the human enzyme, is a key drug target. Structural studies of the flatworm Echinococcus granulosus TGR, free and complexed with AuI-MPO, a novel gold inhibitor, together with inhibition assays were performed. RESULTS: AuI-MPO is a potent TGR inhibitor that achieves 75% inhibition at a 1:1 TGR:Au ratio and efficiently kills E. granulosus in vitro. The structures revealed salient insights: (i) unique monomer-monomer interactions, (ii) distinct binding sites for thioredoxin and the glutaredoxin (Grx) domain, (iii) a single glutathione disulfide reduction site in the Grx domain, (iv) rotation of the Grx domain toward the Sec-containing redox active site, and (v) a single gold atom bound to Cys519 and Cys573 in the AuI-TGR complex. Structural modeling suggests that these residues are involved in the stabilization of the Sec-containing C-terminus. Consistently, Cys→Ser mutations in these residues decreased TGR activities. Mass spectroscopy confirmed these cysteines are the primary binding site. INNOVATION: The identification of a primary site for gold binding and the structural model provide a basis for gold compound optimization through scaffold adjustments. CONCLUSIONS: The structural study revealed that TGR functions are achieved not only through a mobile Sec-containing redox center but also by rotation of the Grx domain and distinct binding sites for Grx domain and thioredoxin. The conserved Cys519 and Cys573 residues targeted by gold assist catalysis through stabilization of the Sec-containing redox center. Antioxid. Redox Signal. 27, 1491-1504.


Subject(s)
Echinococcus granulosus/enzymology , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , NADH, NADPH Oxidoreductases/chemistry , NADH, NADPH Oxidoreductases/metabolism , Organogold Compounds/pharmacology , Animals , Binding Sites/drug effects , Cysteine/metabolism , Echinococcus granulosus/chemistry , Echinococcus granulosus/genetics , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Glutaredoxins/metabolism , Helminth Proteins/chemistry , Helminth Proteins/genetics , Helminth Proteins/metabolism , Models, Molecular , Multienzyme Complexes/genetics , Mutation , NADH, NADPH Oxidoreductases/genetics , Organogold Compounds/chemistry , Protein Binding , Protein Conformation
10.
Mol Biochem Parasitol ; 206(1-2): 13-9, 2016.
Article in English | MEDLINE | ID: mdl-26975431

ABSTRACT

The trace element selenium is found in polypeptides as selenocysteine, the 21(st) amino acid that is co-translationally inserted into proteins at a UGA codon. In proteins, selenocysteine usually plays a role as an efficient redox catalyst. Trypanosomatids previously examined harbor a full set of genes encoding the machinery needed for selenocysteine biosynthesis and incorporation into three selenoproteins: SelK, SelT and, the parasite-specific, Seltryp. We investigated the selenoproteome of kinetoplastid species in recently sequenced genomes and assessed the in vivo relevance of selenoproteins for African trypanosomes. Database mining revealed that SelK, SelT and Seltryp genes are present in most kinetoplastids, including the free-living species Bodo saltans, and Seltryp was lost in the subgenus Viannia from the New World Leishmania. Homology and sinteny with bacterial sulfur dioxygenases and sulfur transferases suggest a putative role for Seltryp in sulfur metabolism. A Trypanosoma brucei selenocysteine synthase (SepSecS) null-mutant, in which selenoprotein synthesis is abolished, displayed similar sensitivity to oxidative stress induced by a short-term exposure to high concentrations of methylglyoxal or H2O2 to that of the parental wild-type cell line. Importantly, the infectivity of the SepSecS knockout cell line was not impaired when tested in a mouse infection model and compensatory effects via up-regulation of proteins involved in thiol-redox metabolism were not observed. Collectively, our data show that selenoproteins are not required for survival of African trypanosomes in a mammalian host and exclude a role for selenoproteins in parasite antioxidant defense and/or virulence. On this basis, selenoproteins can be disregarded as drug target candidates.


Subject(s)
Kinetoplastida/metabolism , Protozoan Proteins/genetics , Selenocysteine/metabolism , Selenoproteins/genetics , Transferases/genetics , Trypanosoma brucei brucei/metabolism , Animals , Data Mining , Databases, Genetic , Gene Deletion , Gene Expression Regulation , Host-Parasite Interactions , Hydrogen Peroxide/pharmacology , Kinetoplastida/classification , Kinetoplastida/drug effects , Kinetoplastida/growth & development , Mice , Phylogeny , Proteome/genetics , Proteome/metabolism , Protozoan Proteins/metabolism , Pyruvaldehyde/pharmacology , Selenoproteins/deficiency , Transferases/deficiency , Trypanosoma brucei brucei/classification , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/growth & development , Trypanosomiasis, African/parasitology , Trypanosomiasis, African/pathology
11.
Antioxid Redox Signal ; 24(4): 205-216, 2016 01 27.
Article in English | MEDLINE | ID: mdl-26381228

ABSTRACT

AIMS: Members of the thioredoxin (Trx) protein family participate mainly in redox pathways and have not been associated with Fe/S binding, in contrast to some closely related glutaredoxins (Grxs). Cestode parasites possess an unusual diversity of Trxs and Trx-related proteins with unexplored functions. In this study, we addressed the biochemical characterization of a new class of Trx-related protein (IsTRP) and a classical monothiol Grx (EgGrx5) from the human pathogen Echinococcus granulosus. RESULTS: The dimeric form of IsTRP coordinates Fe2S2 in a glutathione-independent manner; instead, Fe/S binding relies on the CXXC motif conserved among Trxs. This novel binding mechanism allows holo-IsTRP to be highly resistant to oxidation. IsTRP lacks canonical reductase activities. Mitochondrially targeted IsTRP aids growth of a Grx5 null yeast strain. Similar complementation assays performed with EgGrx5 revealed functional conservation for class II Grxs, despite the presence of nonconserved structural elements. IsTRP is a cestode lineage-specific protein highly expressed in the gravid adult worm, which releases the infective stage critical for dissemination. INNOVATION: IsTRP is the first member from the Trx family to be reported to bind Fe/S. We disclose a novel mechanism of Fe/S coordination within the Trx folding unit, which renders the cluster highly resistant to oxidation-mediated disassembly. CONCLUSION: We demonstrate that IsTRP defines a new protein family within the Trx superfamily, confirm the conservation of function for class II Grx from nonphylogenetically related species, and highlight the versatility of the Trx folding unit to acquire Fe/S binding as a recurrent emergent function. Antioxid. Redox Signal. 00, 000-000.

12.
Molecules ; 20(7): 11793-807, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-26132905

ABSTRACT

Parasitic flatworms cause serious infectious diseases that affect humans and livestock in vast regions of the world, yet there are few effective drugs to treat them. Thioredoxin glutathione reductase (TGR) is an essential enzyme for redox homeostasis in flatworm parasites and a promising pharmacological target. We purified to homogeneity and characterized the TGR from the tapeworm Mesocestoides vogae (syn. M. corti). This purification revealed absence of conventional TR and GR. The glutathione reductase activity of the purified TGR exhibits a hysteretic behavior typical of flatworm TGRs. Consistently, M. vogae genome analysis revealed the presence of a selenocysteine-containing TGR and absence of conventional TR and GR. M. vogae thioredoxin and glutathione reductase activities were inhibited by 3,4-bis(phenylsulfonyl)-1,2,5-oxadiazole N2-oxide (VL16E), an oxadiazole N-oxide previously identified as an inhibitor of fluke and tapeworm TGRs. Finally, we show that mice experimentally infected with M. vogae tetrathyridia and treated with either praziquantel, the reference drug for flatworm infections, or VL16E exhibited a 28% reduction of intraperitoneal larvae numbers compared to vehicle treated mice. Our results show that oxadiazole N-oxide is a promising chemotype in vivo and highlights the convenience of M. vogae as a model for rapid assessment of tapeworm infections in vivo.


Subject(s)
Cestoda/drug effects , Cestode Infections/parasitology , Multienzyme Complexes/metabolism , NADH, NADPH Oxidoreductases/metabolism , Oxadiazoles/pharmacology , Amino Acid Sequence , Animals , Cestoda/metabolism , Mesocestoides , Mice , Molecular Sequence Data , Multienzyme Complexes/chemistry , NADH, NADPH Oxidoreductases/chemistry , Sequence Homology, Amino Acid
13.
Mol Divers ; 18(1): 1-12, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24136658

ABSTRACT

In this study, we report a strategy using dynamic combinatorial chemistry for targeting the thioredoxin (Trx)-reductase catalytic site on Trx glutathione reductase (TGR), a pyridine nucleotide thiol-disulfide oxido-reductase. We chose Echinococcus granulosus TGR since it is a bottleneck enzyme of platyhelminth parasites and a validated pharmacological target. A dynamic combinatorial library (DCL) was constructed based on thiol-disulfide reversible exchange. We demonstrate the use of 5-thio-2-nitrobenzoic acid (TNB) as a non-covalent anchor fragment in a DCL templated by E. granulosus TGR. The heterodimer of TNB and bisthiazolidine (2af) was identified, upon library analysis by HPLC (IC50 = 24 µM). Furthermore, 14 analogs were synthetically prepared and evaluated against TGR. This allowed the study of a structure-activity relationship and the identification of a disulfide TNB-tricyclic bisthiazolidine (2aj) as the best enzyme inhibitor in these series, with an IC50 = 24 µM. Thus, our results validate the use of DCL for targeting thiol-disulfide oxido-reductases.


Subject(s)
Catalytic Domain , Combinatorial Chemistry Techniques , Drug Discovery , Echinococcus granulosus/enzymology , Enzyme Inhibitors/pharmacology , Multienzyme Complexes/antagonists & inhibitors , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Animals , Dimerization , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Inhibitory Concentration 50 , Multienzyme Complexes/chemistry , NADH, NADPH Oxidoreductases/chemistry , Nitrobenzoates/chemistry , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Sulfhydryl Compounds/chemistry , Thiazolidines/chemical synthesis , Thiazolidines/chemistry , Thiazolidines/pharmacology
14.
Antioxid Redox Signal ; 19(7): 735-45, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-22909029

ABSTRACT

SIGNIFICANCE: Platyhelminth parasites cause chronic infections that are a major cause of disability, mortality, and economic losses in developing countries. Maintaining redox homeostasis is a major adaptive problem faced by parasites and its disruption can shift the biochemical balance toward the host. Platyhelminth parasites possess a streamlined thiol-based redox system in which a single enzyme, thioredoxin glutathione reductase (TGR), a fusion of a glutaredoxin (Grx) domain to canonical thioredoxin reductase (TR) domains, supplies electrons to oxidized glutathione (GSSG) and thioredoxin (Trx). TGR has been validated as a drug target for schistosomiasis. RECENT ADVANCES: In addition to glutathione (GSH) and Trx reduction, TGR supports GSH-independent deglutathionylation conferring an additional advantage to the TGR redox array. Biochemical and structural studies have shown that the TR activity does not require the Grx domain, while the glutathione reductase and deglutathionylase activities depend on the Grx domain, which receives electrons from the TR domains. The search for TGR inhibitors has identified promising drug leads, notably oxadiazole N-oxides. CRITICAL ISSUES: A conspicuous feature of platyhelminth TGRs is that their Grx-dependent activities are temporarily inhibited at high GSSG concentrations. The mechanism underlying the phenomenon and its biological relevance are not completely understood. FUTURE DIRECTIONS: The functional diversity of Trxs and Grxs encoded in platyhelminth genomes remains to be further assessed to thoroughly understand the TGR-dependent redox network. Optimization of TGR inhibitors and identification of compounds targeting other parasite redox enzymes are good options to clinically develop relevant drugs for these neglected, but important diseases.


Subject(s)
Cestode Infections/parasitology , Helminth Proteins/metabolism , Multienzyme Complexes/metabolism , NADH, NADPH Oxidoreductases/metabolism , Platyhelminths/enzymology , Animals , Cestode Infections/immunology , Host-Parasite Interactions , Humans , Metabolic Networks and Pathways , Oxidation-Reduction , Oxidative Stress , Platyhelminths/immunology , Reactive Oxygen Species/metabolism
15.
PLoS One ; 7(4): e35033, 2012.
Article in English | MEDLINE | ID: mdl-22536349

ABSTRACT

Parasitic flatworms are responsible for serious infectious diseases that affect humans as well as livestock animals in vast regions of the world. Yet, the drug armamentarium available for treatment of these infections is limited: praziquantel is the single drug currently available for 200 million people infected with Schistosoma spp. and there is justified concern about emergence of drug resistance. Thioredoxin glutathione reductase (TGR) is an essential core enzyme for redox homeostasis in flatworm parasites. In this work, we searched for flatworm TGR inhibitors testing compounds belonging to various families known to inhibit thioredoxin reductase or TGR and also additional electrophilic compounds. Several furoxans and one thiadiazole potently inhibited TGRs from both classes of parasitic flatworms: cestoda (tapeworms) and trematoda (flukes), while several benzofuroxans and a quinoxaline moderately inhibited TGRs. Remarkably, five active compounds from diverse families possessed a phenylsulfonyl group, strongly suggesting that this moiety is a new pharmacophore. The most active inhibitors were further characterized and displayed slow and nearly irreversible binding to TGR. These compounds efficiently killed Echinococcus granulosus larval worms and Fasciola hepatica newly excysted juveniles in vitro at a 20 µM concentration. Our results support the concept that the redox metabolism of flatworm parasites is precarious and particularly susceptible to destabilization, show that furoxans can be used to target both flukes and tapeworms, and identified phenylsulfonyl as a new drug-hit moiety for both classes of flatworm parasites.


Subject(s)
Anticestodal Agents/pharmacology , Antiplatyhelmintic Agents/pharmacology , Echinococcus granulosus/drug effects , Fasciola hepatica/drug effects , Helminth Proteins/antagonists & inhibitors , Multienzyme Complexes/antagonists & inhibitors , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Animals , Anticestodal Agents/chemistry , Anticestodal Agents/toxicity , Antiplatyhelmintic Agents/chemistry , Antiplatyhelmintic Agents/toxicity , Cell Line , Drug Evaluation, Preclinical , Echinococcus granulosus/enzymology , Fasciola hepatica/enzymology , Fibroblasts/drug effects , Helminth Proteins/chemistry , Humans , Larva/drug effects , Larva/enzymology , Lymphocytes/drug effects , Mice , Models, Molecular , Multienzyme Complexes/chemistry , NADH, NADPH Oxidoreductases/chemistry , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Oxadiazoles/toxicity , Quantum Theory , Quinoxalines/chemistry , Quinoxalines/pharmacology , Quinoxalines/toxicity , Structure-Activity Relationship , Thiadiazoles/chemistry , Thiadiazoles/pharmacology , Thiadiazoles/toxicity
16.
Parasitology ; 139(2): 271-83, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22216900

ABSTRACT

Infection by larval Echinococcus granulosus is usually characterized by tight inflammatory control. However, various degrees of chronic granulomatous inflammation are also observed, reaching a high point in infection of cattle by the most prevalent parasite strain worldwide, which is not well adapted to this host species. In this context, epithelioid and multinucleated giant macrophages surround the parasite, and the secreted products of these cells often associate with the larval wall. The phagocyte-specific S100 proteins, S100A8, S100A9 and S100A12, are important non-conventionally secreted amplifiers of inflammatory responses. We have analysed by proteomics and immunohistochemistry the presence of these proteins at the E. granulosus larva-host interface. We found that, in the context of inflammatory control as observed in human infections, the S100 proteins are not abundant, but S100A9 and S100A8 can be expressed by eosinophils distal to the parasite. In the granulomatous inflammation context as observed in cattle infections, we found that S100A12 is one of the most abundant host-derived, parasite-associated proteins, while S100A9 and S100A8 are not present at similarly high levels. As expected, S100A12 derives mostly from the epithelioid and multinucleated giant cells. S100A12, as well as cathepsin K and matrix metalloproteinase-9, also expressed by E. granulosus-elicited epithelioid cells, are connected to the Th17 arm of immunity, which may therefore be involved in this granulomatous response.


Subject(s)
Echinococcosis/veterinary , Echinococcus granulosus/physiology , Gene Expression Regulation/immunology , Phagocytes/metabolism , S100 Proteins/metabolism , Animals , Cattle , Echinococcosis/immunology , Echinococcosis/parasitology , Humans , Larva/physiology , Mice , Proteomics , S100 Proteins/genetics , Species Specificity
17.
J Biol Chem ; 286(7): 4959-67, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21051543

ABSTRACT

In most organisms, thioredoxin (Trx) and/or glutathione (GSH) systems are essential for redox homeostasis and deoxyribonucleotide synthesis. Platyhelminth parasites have a unique and simplified thiol-based redox system, in which the selenoprotein thioredoxin-glutathione reductase (TGR), a fusion of a glutaredoxin (Grx) domain to canonical thioredoxin reductase domains, is the sole enzyme supplying electrons to oxidized glutathione (GSSG) and Trx. This enzyme has recently been validated as a key drug target for flatworm infections. In this study, we show that TGR possesses GSH-independent deglutathionylase activity on a glutathionylated peptide. Furthermore, we demonstrate that deglutathionylation and GSSG reduction are mediated by the Grx domain by a monothiolic mechanism and that the glutathionylated TGR intermediate is resolved by selenocysteine. Deglutathionylation and GSSG reduction via Grx domain, but not Trx reduction, are inhibited at high [GSSG]/[GSH] ratios. We found that Trxs (cytosolic and mitochondrial) provide alternative pathways for deglutathionylation and GSSG reduction. These pathways are operative at high [GSSG]/[GSH] and function in a complementary manner to the Grx domain-dependent one. Despite the existence of alternative pathways, the thioredoxin reductase domains of TGR are an obligate electron route for both the Grx domain- and the Trx-dependent pathways. Overall, our results provide an explanation for the unique array of thiol-dependent redox pathways present in parasitic platyhelminths. Finally, we found that TGR is inhibited by 1-hydroxy-2-oxo-3-(N-3-methyl-aminopropyl)-3-methyl-1-triazene (NOC-7), giving further evidence for NO donation as a mechanism of action for oxadiazole N-oxide TGR inhibitors. Thus, NO donors aimed at TGR could disrupt the entire redox homeostasis of parasitic flatworms.


Subject(s)
Cestoda/metabolism , Glutathione/metabolism , Helminth Proteins/metabolism , Multienzyme Complexes/metabolism , NADH, NADPH Oxidoreductases/metabolism , Thioredoxins/metabolism , Animals , Hydrazines/pharmacology , Multienzyme Complexes/antagonists & inhibitors , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Nitric Oxide Donors/pharmacology , Oxidation-Reduction/drug effects , Peptides/metabolism , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/physiology
18.
BMC Genomics ; 11: 237, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20385027

ABSTRACT

BACKGROUND: The thioredoxin and/or glutathione pathways occur in all organisms. They provide electrons for deoxyribonucleotide synthesis, function as antioxidant defenses, in detoxification, Fe/S biogenesis and participate in a variety of cellular processes. In contrast to their mammalian hosts, platyhelminth (flatworm) parasites studied so far, lack conventional thioredoxin and glutathione systems. Instead, they possess a linked thioredoxin-glutathione system with the selenocysteine-containing enzyme thioredoxin glutathione reductase (TGR) as the single redox hub that controls the overall redox homeostasis. TGR has been recently validated as a drug target for schistosomiasis and new drug leads targeting TGR have recently been identified for these platyhelminth infections that affect more than 200 million people and for which a single drug is currently available. Little is known regarding the genomic structure of flatworm TGRs, the expression of TGR variants and whether the absence of conventional thioredoxin and glutathione systems is a signature of the entire platyhelminth phylum. RESULTS: We examine platyhelminth genomes and transcriptomes and find that all platyhelminth parasites (from classes Cestoda and Trematoda) conform to a biochemical scenario involving, exclusively, a selenium-dependent linked thioredoxin-glutathione system having TGR as a central redox hub. In contrast, the free-living platyhelminth Schmidtea mediterranea (Class Turbellaria) possesses conventional and linked thioredoxin and glutathione systems. We identify TGR variants in Schistosoma spp. derived from a single gene, and demonstrate their expression. We also provide experimental evidence that alternative initiation of transcription and alternative transcript processing contribute to the generation of TGR variants in platyhelminth parasites. CONCLUSIONS: Our results indicate that thioredoxin and glutathione pathways differ in parasitic and free-living flatworms and that canonical enzymes were specifically lost in the parasitic lineage. Platyhelminth parasites possess a unique and simplified redox system for diverse essential processes, and thus TGR is an excellent drug target for platyhelminth infections. Inhibition of the central redox wire hub would lead to overall disruption of redox homeostasis and disable DNA synthesis.


Subject(s)
Glutathione/metabolism , Platyhelminths/genetics , Platyhelminths/physiology , Thioredoxins/metabolism , Animals , Metabolic Networks and Pathways , Multienzyme Complexes , NADH, NADPH Oxidoreductases , Phylogeny , Promoter Regions, Genetic , Transcription, Genetic
19.
J Biol Chem ; 283(26): 17898-907, 2008 Jun 27.
Article in English | MEDLINE | ID: mdl-18408002

ABSTRACT

Platyhelminth parasites are a major health problem in developing countries. In contrast to their mammalian hosts, platyhelminth thiol-disulfide redox homeostasis relies on linked thioredoxin-glutathione systems, which are fully dependent on thioredoxin-glutathione reductase (TGR), a promising drug target. TGR is a homodimeric enzyme comprising a glutaredoxin domain and thioredoxin reductase (TR) domains with a C-terminal redox center containing selenocysteine (Sec). In this study, we demonstrate the existence of functional linked thioredoxin-glutathione systems in the cytosolic and mitochondrial compartments of Echinococcus granulosus, the platyhelminth responsible for hydatid disease. The glutathione reductase (GR) activity of TGR exhibited hysteretic behavior regulated by the [GSSG]/[GSH] ratio. This behavior was associated with glutathionylation by GSSG and abolished by deglutathionylation. The K(m) and k(cat) values for mitochondrial and cytosolic thioredoxins (9.5 microm and 131 s(-1), 34 microm and 197 s(-1), respectively) were higher than those reported for mammalian TRs. Analysis of TGR mutants revealed that the glutaredoxin domain is required for the GR activity but did not affect the TR activity. In contrast, both GR and TR activities were dependent on the Sec-containing redox center. The activity loss caused by the Sec-to-Cys mutation could be partially compensated by a Cys-to-Sec mutation of the neighboring residue, indicating that Sec can support catalysis at this alternative position. Consistent with the essential role of TGR in redox control, 2.5 microm auranofin, a known TGR inhibitor, killed larval worms in vitro. These studies establish the selenium- and glutathione-dependent regulation of cytosolic and mitochondrial redox homeostasis through a single TGR enzyme in platyhelminths.


Subject(s)
Cytosol/metabolism , Glutathione/chemistry , Mitochondria/metabolism , Multienzyme Complexes/chemistry , NADH, NADPH Oxidoreductases/chemistry , Oxidation-Reduction , Selenium/chemistry , Animals , Cytosol/chemistry , Echinococcus granulosus/metabolism , Enzyme Inhibitors/pharmacology , Homeostasis , Kinetics , Models, Biological , Platyhelminths/metabolism , Protein Structure, Tertiary , Selenocysteine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...