Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Redox Biol ; 34: 101547, 2020 07.
Article in English | MEDLINE | ID: mdl-32388269

ABSTRACT

Hydroperoxide reduction in African trypanosomes relies on 2-Cys-peroxiredoxins (Prxs) and glutathione peroxidase-type enzymes (Pxs) which both obtain their reducing equivalents from the trypanothione/tryparedoxin couple and thus act as tryparedoxin peroxidases. While the cytosolic forms of the peroxidases are essential, the mitochondrial mPrx and Px III appear dispensable in bloodstream Trypanosoma brucei. This led to the suggestion that in this developmental stage which is characterized by a mitochondrion that lacks an active respiratory chain, only one of the two peroxidases might be required. Here we show that bloodstream cells in which the Px III gene is deleted and mPrx is down-regulated by RNA interference, proliferate as the parental cells indicating that both mitochondrial peroxidases are dispensable. However, when we raised the culture temperature to 39 °C, mPrx-depleted cells died indicating that under conditions mimicking a fever situation in the mammalian host, the protein becomes essential. In contrast, depletion of mPrx in insect stage procyclic T. brucei causes a proliferation defect under standard conditions at 27 °C, in the absence of any stress. In the absence of mPrx, a tryparedoxin-coupled roGFP2 biosensor expressed in the mitochondrial matrix is unable to respond to antimycin A treatment. Thus mPrx reduces mitochondrial H2O2 with the generation of trypanothione disulfide and acts as peroxidase. However, mPrx-depleted procyclic cells neither display any alteration in the cytosolic or mitochondrial trypanothione redox state nor increased sensitivity towards exogenous oxidative stressors suggesting that the peroxidase activity is not the crucial physiological function. After prolonged mPrx-depletion, the cells almost stop proliferation and display a highly elongated shape and diminished MitoTracker Red staining. In contrast to the situation in the mammalian bloodstream T. brucei and Leishmania, mPrx appears to play a constitutive role for the morphology, mitochondrial function and proliferation of the insect stage of African trypanosomes.


Subject(s)
Hydrogen Peroxide , Trypanosoma brucei brucei , Animals , Hydrogen Peroxide/metabolism , Mitochondria/metabolism , Oxidation-Reduction , Peroxiredoxins/genetics , Peroxiredoxins/metabolism
2.
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
3.
Angew Chem Int Ed Engl ; 58(11): 3640-3644, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30605929

ABSTRACT

Trypanosomal and leishmanial infections claim tens of thousands of lives each year. The metabolism of these unicellular eukaryotic parasites differs from the human host and their enzymes thus constitute promising drug targets. Tryparedoxin (Tpx) from Trypanosoma brucei is the essential oxidoreductase in the parasite's hydroperoxide-clearance cascade. In vitro and in vivo functional assays show that a small, selective inhibitor efficiently inhibits Tpx. With X-ray crystallography, SAXS, analytical SEC, SEC-MALS, MD simulations, ITC, and NMR spectroscopy, we show how covalent binding of this monofunctional inhibitor leads to Tpx dimerization. Intra- and intermolecular inhibitor-inhibitor, protein-protein, and inhibitor-protein interactions stabilize the dimer. The behavior of this efficient antitrypanosomal molecule thus constitutes an exquisite example of chemically induced dimerization with a small, monovalent ligand that can be exploited for future drug design.


Subject(s)
Antiprotozoal Agents/chemistry , Bacterial Proteins/chemistry , Enzyme Inhibitors/chemistry , Oxidoreductases/chemistry , Thioredoxins/chemistry , Trypanosoma brucei brucei/enzymology , Animals , Antiprotozoal Agents/metabolism , Drug Design , Enzyme Inhibitors/metabolism , Glutathione/analogs & derivatives , Glutathione/chemistry , Humans , Hydrogen Peroxide/metabolism , Molecular Dynamics Simulation , Oxidation-Reduction , Protein Binding , Protein Conformation , Protein Multimerization , Spermidine/analogs & derivatives , Spermidine/chemistry , Trypanosoma/metabolism , Trypanosoma/parasitology
4.
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
5.
ACS Med Chem Lett ; 7(12): 1073-1076, 2016 Dec 08.
Article in English | MEDLINE | ID: mdl-27994740

ABSTRACT

Dipeptidyl nitroalkenes are potent reversible inhibitors of cysteine proteases. Inhibitor 11 resulted to be the most potent one with Ki values of 0.49 and 0.44 nM against rhodesain and cruzain, respectively. According to enzymatic dilution and dialysis experiments, as well as computational and NMR studies, dipeptidyl nitroalkenes are tightly binding covalent reversible inhibitors.

6.
Mol Biochem Parasitol ; 207(2): 84-8, 2016 06.
Article in English | MEDLINE | ID: mdl-27262262

ABSTRACT

Px IV is a distant relative of the known glutathione peroxidase-type enzymes of African trypanosomes. Immunofluorescence microscopy of bloodstream cells expressing C-terminally Myc6-tagged Px IV revealed a mitochondrial localization. Recombinant Px IV possesses very low activity as glutathione peroxidase but catalyzes the trypanothione/tryparedoxin-dependent reduction of hydrogen peroxide and, even more efficiently, of arachidonic acid hydroperoxide. Neither overexpression in bloodstream cells nor the deletion of both alleles in bloodstream or procyclic parasites affected the in vitro proliferation. Trypanosoma brucei Px IV shares 58% of all residues with TcGPXII. The orthologous enzymes have in common their substrate preference for fatty acid hydroperoxides. However, the T. cruzi protein has been reported to be localized in the endoplasmic reticulum and to be specific for glutathione as reducing agent. Taken together, our data show that Px IV is a low abundant tryparedoxin peroxidase of T. brucei that is not essential, at least under culture conditions.


Subject(s)
Lipid Peroxides/metabolism , Peroxidases/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/enzymology , Animals , Catalysis , Glutathione/metabolism , Hydrogen Peroxide/metabolism , Peroxidases/genetics , Protozoan Proteins/genetics , Substrate Specificity , Thioredoxins/metabolism , Trypanosomiasis/parasitology
7.
Mol Microbiol ; 99(1): 172-87, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26374473

ABSTRACT

African trypanosomes express three virtually identical glutathione peroxidase (Px)-type enzymes that occur in the cytosol (Px I and II) and mitochondrion (Px III) and detoxify fatty acid-derived hydroperoxides. Selective deletion of the genes revealed that procyclic Trypanosoma brucei lacking either the cytosolic or mitochondrial enzyme proliferate nearly as wild-type parasites, whereas the knockout of the complete genomic locus is lethal. Flow cytometry and immunofluorescence analyses revealed that the Px I-III-deficient parasites lose their mitochondrial membrane potential, which is followed by a loss of the lysosomal signal but not the glycosomal one. Mitochondrial damage and cell lysis are prevented by Trolox, ubiquinone derivatives and the iron chelator deferoxamine, whereas starch-deferoxamine is inefficient. In glucose-rich medium, cell death is attenuated suggesting that oxidants generated by the respiratory chain contribute to the lethal phenotype. Thus, the Px-type peroxidases protect procyclic cells from an iron-mediated oxidative membrane damage that originates at the mitochondrion. This contrasts with the situation in bloodstream cells, where the lysosome is the primarily affected organelle. Strikingly, either the cytosolic or the mitochondrial form of the peroxidases is required and sufficient to protect the mitochondrion and prevent cell lysis.


Subject(s)
Iron/toxicity , Mitochondria/drug effects , Peroxidases/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/enzymology , Trypanosoma brucei brucei/metabolism , Cell Survival/drug effects , Culture Media/chemistry , Gene Deletion , Membrane Potential, Mitochondrial/drug effects , Mitochondria/physiology , Peroxidases/genetics , Protozoan Proteins/genetics , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/growth & development
8.
Mol Biochem Parasitol ; 204(2): 93-105, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26854591

ABSTRACT

As constituents of their unusual trypanothione-based thiol metabolism, African trypanosomes express two dithiol glutaredoxins (Grxs), a cytosolic Grx1 and a mitochondrial Grx2, with so far unknown biological functions. As revealed by gel shift assays, in the mammalian bloodstream form of Trypanosoma brucei, Grx1 is in the fully reduced state. Upon diamide treatment of the cells, Grx1 forms an active site disulfide bridge that is rapidly re-reduced after stress removal; Cys76, a conserved non-active site Cys remains in the thiol state. Deletion of both grx1 alleles does not result in any proliferation defect of neither the procyclic insect form nor the bloodstream form, even not under various stress conditions. In addition, the Grx1-deficient parasites are fully infectious in the mouse model. A functional compensation by Grx2 is unlikely as identical levels of Grx2 were found in wildtype and Grx1-deficient cells. In the classical hydroxyethyl disulfide assay, Grx1-deficient bloodstream cells display 50-60% of the activity of wildtype cells indicating that the cytosolic oxidoreductase accounts for a major part of the total deglutathionylation capacity of the parasite. Intriguingly, at elevated temperature, proliferation of the Grx1-deficient bloodstream parasites is significantly less affected compared to wildtype cells. When cultured for three days at 39°C, only 51% of the cells in the wildtype population retained normal morphology with single mitochondrial and nuclear DNA (1K1N), whereas 27% of the cells displayed ≥2K2N. In comparison, 64% of the Grx1-deficient cells kept the 1K1N phenotype and only 18% had ≥2K2N. The data suggest that Grx1 plays a role in the regulation of the thermotolerance of the parasites by (in)directly interfering with the progression of the cell cycle, a process that may comprise protein (de)glutathionylation step(s).


Subject(s)
Glutaredoxins/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/enzymology , Trypanosoma brucei brucei/physiology , Trypanosomiasis, African/parasitology , Animals , Female , Glutaredoxins/genetics , Hot Temperature , Humans , Mice , Mice, Inbred BALB C , Protozoan Proteins/genetics , Trypanosoma brucei brucei/genetics , Trypanosomiasis, African/blood
9.
ChemMedChem ; 9(8): 1880-91, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24788386

ABSTRACT

The causative agents of the parasitic disease human African trypanosomiasis belong to the family of trypanosomatids. These parasitic protozoa exhibit a unique thiol redox metabolism that is based on the flavoenzyme trypanothione reductase (TR). TR was identified as a potential drug target and features a large active site that allows a multitude of possible ligand orientations, which renders rational structure-based inhibitor design highly challenging. Herein we describe the synthesis, binding properties, and kinetic analysis of a new series of small-molecule inhibitors of TR. The conjunction of biological activities, mutation studies, and virtual ligand docking simulations led to the prediction of a binding mode that was confirmed by crystal structure analysis. The crystal structures revealed that the ligands bind to the hydrophobic wall of the so-called "mepacrine binding site". The binding conformation and potency of the inhibitors varied for TR from Trypanosoma brucei and T. cruzi.


Subject(s)
Antiprotozoal Agents/chemistry , Enzyme Inhibitors/chemistry , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Protozoan Proteins/antagonists & inhibitors , Antiprotozoal Agents/metabolism , Antiprotozoal Agents/pharmacology , Binding Sites , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Ligands , Molecular Dynamics Simulation , NADH, NADPH Oxidoreductases/metabolism , Protein Binding , Protein Structure, Tertiary , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/enzymology , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/enzymology
10.
Biochem Pharmacol ; 85(1): 38-45, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23085438

ABSTRACT

Malaria causes millions of death cases per year. Since Plasmodium falciparum rapidly develops drug resistance, it is of high importance to investigate potential drug targets which may lead to novel rational therapy approaches. Here we report on the interaction of translationally controlled tumor protein of P. falciparum (PfTCTP) with the anti-malarial drug artemisinin. Furthermore, we investigated the crystal structure of PfTCTP. Using mass spectrometry, bioinformatic approaches and surface plasmon resonance spectroscopy, we identified novel binding sites of artemisinin which are in direct neighborhood to amino acids 19-46, 108-134 and 140-163. The regions covered by these residues are known to be functionally important for TCTP function. We conclude that interaction of artemisinin with TCTP may be at least in part explain the antimalarial activity of artemisinin.


Subject(s)
Antimalarials/chemistry , Artemisinins/chemistry , Biomarkers, Tumor/chemistry , Plasmodium falciparum/metabolism , Protozoan Proteins/chemistry , Binding Sites , Biomarkers, Tumor/metabolism , Computer Simulation , Crystallography, X-Ray , Humans , Molecular Docking Simulation , Molecular Structure , Protein Binding , Protozoan Proteins/metabolism , Recombinant Proteins/chemistry , Surface Plasmon Resonance , Tumor Protein, Translationally-Controlled 1
11.
J Biol Chem ; 285(45): 35224-37, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20826822

ABSTRACT

Trypanosoma brucei, the causative agent of African sleeping sickness, possesses two dithiol glutaredoxins (Grx1 and Grx2). Grx1 occurs in the cytosol and catalyzes protein deglutathionylations with k(cat)/K(m)-values of up to 2 × 10(5) M(-1) S(-1). It accelerates the reduction of ribonucleotide reductase by trypanothione although less efficiently than the parasite tryparedoxin and has low insulin disulfide reductase activity. Despite its classical CPYC active site, Grx1 forms dimeric iron-sulfur complexes with GSH, glutathionylspermidine, or trypanothione as non-protein ligands. Thus, contrary to the generally accepted assumption, replacement of the Pro is not a prerequisite for cluster formation. T. brucei Grx2 shows an unusual CQFC active site, and orthologues occur exclusively in trypanosomatids. Grx2 is enriched in mitoplasts, and fractionated digitonin lysis resulted in a co-elution with cytochrome c, suggesting localization in the mitochondrial intermembrane space. Grx2 catalyzes the reduction of insulin disulfide but not of ribonucleotide reductase and exerts deglutathionylation activity 10-fold lower than that of Grx1. RNA interference against Grx2 caused a growth retardation of procyclic cells consistent with an essential role. Grx1 and Grx2 are constitutively expressed with cellular concentrations of about 2 µM and 200 nM, respectively, in both the mammalian bloodstream and insect procyclic forms. Trypanothione reduces the disulfide form of both proteins with apparent rate constants that are 3 orders of magnitude higher than those with glutathione. Grx1 and, less efficiently, also Grx2 catalyze the reduction of GSSG by trypanothione. Thus, the Grxs play exclusive roles in the trypanothione-based thiol redox metabolism of African trypanosomes.


Subject(s)
Glutaredoxins/metabolism , Glutathione/analogs & derivatives , Protozoan Proteins/metabolism , Spermidine/analogs & derivatives , Trypanosoma brucei brucei/enzymology , Animals , Catalytic Domain , Cytochromes c/genetics , Cytochromes c/metabolism , Glutaredoxins/genetics , Glutathione/genetics , Glutathione/metabolism , Mitochondrial Membranes/enzymology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidation-Reduction , Protozoan Proteins/genetics , Spermidine/metabolism , Thioredoxins/genetics , Thioredoxins/metabolism
12.
Int J Parasitol ; 39(10): 1059-62, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19477177

ABSTRACT

Trypanosomatids, the causative agents of several tropical diseases, have a unique thiol metabolism based on trypanothione [bis(glutathionyl)spermidine]. Enzymes of the pathway are attractive drug target molecules but the availability of trypanothione remains an obstacle. Here, we present a convenient method for the production of trypanothione and trypanothione disulfide in >200mg quantities using a mutant of Crithidia fasciculata trypanothione synthetase in which Cys59 has been replaced by an alanine residue. The reagent costs less than 1% of the commercial price of trypanothione disulfide. The protocol also allows the synthesis of related glutathione conjugates. It will greatly facilitate the thorough analysis of this parasite's metabolism and drug screening approaches against trypanothione-dependent enzymes.


Subject(s)
Amide Synthases/genetics , Crithidia fasciculata/enzymology , Glutathione/analogs & derivatives , Spermidine/analogs & derivatives , Trypanocidal Agents/metabolism , Trypanosoma/enzymology , Disulfides/metabolism , Drug Design , Glutathione/biosynthesis , Molecular Sequence Data , Spermidine/biosynthesis , Substrate Specificity
13.
J Biol Chem ; 283(41): 27785-27798, 2008 Oct 10.
Article in English | MEDLINE | ID: mdl-18669638

ABSTRACT

African trypanosomes encode three monothiol glutaredoxins (1-C-Grx). 1-C-Grx1 occurs exclusively in the mitochondrion, and 1-C-Grx2 and -3 are predicted to be mitochondrial and cytosolic proteins, respectively. All three 1-C-Grx are expressed in both the mammalian bloodstream and the insect procyclic form of Trypanosoma brucei, with the highest levels found in stationary phase and starving parasites. In the rudimentary mitochondrion of bloodstream cells, 1-C-Grx1 reaches concentrations above 200 microm/subunit. Recombinant T. brucei 1-C-Grx1 exists as a noncovalent homodimer, whereas 1-C-Grx2 and 1-C-Grx3 are monomeric proteins. In vitro, dimeric 1-C-Grx1 coordinated an H(2)O(2)-sensitive [2Fe-2S] cluster that required GSH as an additional ligand. Both bloodstream and procyclic trypanosomes were refractory to down-regulation of 1-C-Grx1 expression by RNA interference. In procyclic parasites, the 1-c-grx1 alleles could only be deleted if an ectopic copy of the gene was expressed. A 5-10-fold overexpression of 1-C-Grx1 in both parasite forms did not yield a growth phenotype under optimal culture conditions. However, exposure of these cells to the iron chelator deferoxamine or H(2)O(2), but not to iron or menadione, impaired cell growth. Treatment of wild-type bloodstream parasites with deferoxamine and H(2)O(2) caused a 2-fold down- and up-regulation of 1-C-Grx1, respectively. The results point to an essential role of the mitochondrial 1-C-Grx1 in the iron metabolism of these parasites.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Glutaredoxins/biosynthesis , Iron/metabolism , Metalloproteins/biosynthesis , Mitochondrial Proteins/biosynthesis , Protozoan Proteins/biosynthesis , Trypanosoma brucei brucei/enzymology , Trypanosomiasis, African/enzymology , Animals , Cytosol/enzymology , Deferoxamine/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Glutaredoxins/genetics , Humans , Hydrogen Peroxide/pharmacology , Isoenzymes/biosynthesis , Isoenzymes/genetics , Metalloproteins/genetics , Mitochondrial Proteins/genetics , Oxidants/pharmacology , Protozoan Proteins/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Siderophores/pharmacology , Sulfur/metabolism , Trypanosoma brucei brucei/genetics , Trypanosomiasis, African/genetics , Up-Regulation/drug effects , Up-Regulation/physiology , Vitamin K 3/pharmacology , Vitamins/pharmacology
14.
Biol Chem ; 389(1): 21-32, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18095866

ABSTRACT

African trypanosomes encode three monothiol glutaredoxins (1-C-Grx1 to 3). 1-C-Grx1 has a putative CAYS active site and Cys181 as single additional cysteine. The recombinant protein forms non-covalent homodimers. As observed for other monothiol glutaredoxins, Trypanosoma brucei 1-C-Grx1 was not active in the glutaredoxin assay with hydroxyethyl disulfide and glutathione nor catalyzed the reduction of insulin disulfide. In addition, it lacked peroxidase activity and did not catalyze protein (de)glutathionylation. Upon oxidation, 1-C-Grx1 forms an intramolecular disulfide bridge and, to a minor degree, covalent dimers. Both disulfide forms are reduced by the parasite trypanothione/tryparedoxin system. 1-C-Grx1 shows mitochondrial localization. The total cellular concentration is at least 5 microm. Thus, 1-C-Grx1 is an abundant protein especially in the rudimentary organelle of the mammalian form of the parasite. Expression of 1-C-Grx1 in Grx5-deficient yeast cells with its authentic presequence targeted the protein to the mitochondria and partially restored the growth phenotype and aconitase activity of the mutant, and conferred resistance against hydroperoxides and diamide. The parasite Grx2 and 3 failed to substitute for Grx5. This is surprising because even bacterial and plant 1-Cys-glutaredoxins efficiently revert the defects, and may be due to the lack of two basic residues conserved in all but the trypanosomatid proteins.


Subject(s)
Glutaredoxins/metabolism , Trypanosoma brucei brucei/genetics , Amino Acid Sequence , Animals , Blotting, Western , Chromatography, Gel , Cloning, Molecular , Codon/genetics , DNA, Protozoan/genetics , DNA, Protozoan/metabolism , Disulfides/metabolism , Ethanol/analogs & derivatives , Ethanol/metabolism , Genetic Complementation Test , Glutaredoxins/chemistry , Glutaredoxins/genetics , Insulin/metabolism , Mitochondria/enzymology , Mitochondria/metabolism , Molecular Sequence Data , Oxidation-Reduction , Oxidoreductases/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Reducing Agents/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Subcellular Fractions/metabolism , Sulfhydryl Compounds/metabolism , Trypanosoma brucei brucei/growth & development , Trypanosoma brucei brucei/metabolism
15.
J Biol Chem ; 282(12): 8678-94, 2007 Mar 23.
Article in English | MEDLINE | ID: mdl-17242409

ABSTRACT

Trypanosomatids, the causative agents of several tropical diseases, lack glutathione reductase and thioredoxin reductase but have a trypanothione reductase instead. The main low molecular weight thiols are trypanothione (N(1),N(8)-bis-(glutathionyl)spermidine) and glutathionyl-spermidine, but the parasites also contain free glutathione. To elucidate whether trypanosomes employ S-thiolation for regulatory or protection purposes, six recombinant parasite thiol redox proteins were studied by ESI-MS and MALDI-TOF-MS for their ability to form mixed disulfides with glutathione or glutathionylspermidine. Trypanosoma brucei mono-Cys-glutaredoxin 1 is specifically thiolated at Cys(181). Thiolation of this residue induced formation of an intramolecular disulfide bridge with the putative active site Cys(104). This contrasts with mono-Cys-glutaredoxins from other sources that have been reported to be glutathionylated at the active site cysteine. Both disulfide forms of the T. brucei protein were reduced by tryparedoxin and trypanothione, whereas glutathione cleaved only the protein disulfide. In the glutathione peroxidase-type tryparedoxin peroxidase III of T. brucei, either Cys(47) or Cys(95) became glutathionylated but not both residues in the same protein molecule. T. brucei thioredoxin contains a third cysteine (Cys(68)) in addition to the redox active dithiol/disulfide. Treatment of the reduced protein with GSSG caused glutathionylation of Cys(68), which did not affect its capacity to catalyze reduction of insulin disulfide. Reduced T. brucei tryparedoxin possesses only the redox active Cys(32)-Cys(35) couple, which upon reaction with GSSG formed a disulfide. Also glyoxalase II and Trypanosoma cruzi trypanothione reductase were not sensitive to thiolation at physiological GSSG concentrations.


Subject(s)
Glutathione/analogs & derivatives , Glutathione/chemistry , Spermidine/analogs & derivatives , Sulfhydryl Compounds/chemistry , Amino Acid Sequence , Animals , Cysteine/chemistry , Disulfides/chemistry , Insulin/chemistry , Molecular Sequence Data , Oxidation-Reduction , Sequence Homology, Amino Acid , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spermidine/chemistry , Trypanosoma
16.
J Biol Chem ; 280(15): 14385-94, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15664987

ABSTRACT

Trypanosoma brucei, the causative agent of African sleeping sickness, encodes three nearly identical cysteine homologues of the classical selenocysteine-containing glutathione peroxidases. Although one of the sequences, peroxidase III, carries both putative mitochondrial and glycosomal targeting signals, the proteins are detectable only in the cytosol and mitochondrion of mammalian bloodstream and insect procyclic T. brucei. The enzyme is a trypanothione/tryparedoxin peroxidase as are the 2 Cys-peroxiredoxins of the parasite. Hydrogen peroxide, thymine hydroperoxide, and linoleic acid hydroperoxide are reduced with second order rate constants of 8.7 x 10(4), 7.6 x 10(4), and 4 x 10(4) m(-1) s(-1), respectively, and represent probable physiological substrates. Phosphatidylcholine hydroperoxide is a very weak substrate and, in the absence of Triton X-100, even an inhibitor of the enzyme. The substrate preference clearly contrasts with that of the closely related T. cruzi enzyme, which reduces phosphatidylcholine hydroperoxides but not H(2)O(2). RNA interference causes severe growth defects in bloodstream and procyclic cells in accordance with the peroxidases being essential in both developmental stages. Thus, the cellular functions of the glutathione peroxidase-type enzymes cannot be taken over by the 2 Cys-peroxiredoxins that also occur in the cytosol and mitochondrion of the parasite.


Subject(s)
Glutathione Peroxidase/chemistry , Peroxidases/chemistry , Peroxidases/physiology , Protozoan Proteins/chemistry , Protozoan Proteins/physiology , Trypanosoma brucei brucei/enzymology , Animals , Blotting, Northern , Blotting, Western , Cytosol/metabolism , Dose-Response Relationship, Drug , Hydrogen Peroxide/pharmacology , Kinetics , Linoleic Acids/pharmacology , Lipid Peroxides/pharmacology , Mitochondria/metabolism , Octoxynol/pharmacology , Phosphatidylcholines/chemistry , RNA Interference , Recombinant Proteins/chemistry , Subcellular Fractions/metabolism , Substrate Specificity , Thymine/chemistry , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...