Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Bioorg Med Chem ; 24(7): 1573-81, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26935942

ABSTRACT

Trypanosomal phosphodiesterases B1 and B2 (TbrPDEB1 and TbrPDEB2) play an important role in the life cycle of Trypanosoma brucei, the causative parasite of human African trypanosomiasis (HAT), also known as African sleeping sickness. Knock down of both enzymes leads to cell cycle arrest and is lethal to the parasite. Recently, we reported the phenylpyridazinone, NPD-001, with low nanomolar IC50 values on both TbrPDEB1 (IC50: 4nM) and TbrPDEB2 (IC50: 3nM) (J. Infect. Dis.2012, 206, 229). In this study, we now report on the first structure activity relationships of a series of phenylpyridazinone analogs as TbrPDEB1 inhibitors. A selection of compounds was also shown to be anti-parasitic. Importantly, a good correlation between TbrPDEB1 IC50 and EC50 against the whole parasite was observed. Preliminary analysis of the SAR of selected compounds on TbrPDEB1 and human PDEs shows large differences which shows the potential for obtaining parasite selective PDE inhibitors. The results of these studies support the pharmacological validation of the Trypanosome PDEB family as novel therapeutic approach for HAT and provide as well valuable information for the design of potent TbrPDEB1 inhibitors that could be used for the treatment of this disease.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Phosphodiesterase Inhibitors/chemical synthesis , Phosphodiesterase Inhibitors/pharmacology , Protozoan Proteins/antagonists & inhibitors , Pyridazines/pharmacology , Tetrazoles/pharmacology , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Dose-Response Relationship, Drug , Humans , Molecular Structure , Parasitic Sensitivity Tests , Phosphodiesterase Inhibitors/chemistry , Protozoan Proteins/metabolism , Pyridazines/chemical synthesis , Pyridazines/chemistry , Structure-Activity Relationship , Tetrazoles/chemical synthesis , Tetrazoles/chemistry , Trypanocidal Agents/chemical synthesis , Trypanosoma brucei brucei/enzymology , Trypanosomiasis, African/drug therapy , Trypanosomiasis, African/parasitology
2.
J Med Chem ; 56(5): 2087-96, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23409953

ABSTRACT

Trypanosoma brucei cyclic nucleotide phosphodiesterase B1 (TbrPDEB1) and TbrPDEB2 have recently been validated as new therapeutic targets for human African trypanosomiasis by both genetic and pharmacological means. In this study we report the crystal structure of the catalytic domain of the unliganded TbrPDEB1 and its use for the in silico screening for new TbrPDEB1 inhibitors with novel scaffolds. The TbrPDEB1 crystal structure shows the characteristic folds of human PDE enzymes but also contains the parasite-specific P-pocket found in the structures of Leishmania major PDEB1 and Trypanosoma cruzi PDEC. The unliganded TbrPDEB1 X-ray structure was subjected to a structure-based in silico screening approach that combines molecular docking simulations with a protein-ligand interaction fingerprint (IFP) scoring method. This approach identified six novel TbrPDEB1 inhibitors with IC50 values of 10-80 µM, which may be further optimized as potential selective TbrPDEB inhibitors.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Phosphodiesterase Inhibitors/isolation & purification , 3',5'-Cyclic-AMP Phosphodiesterases/chemistry , Amino Acid Sequence , Animals , Catalytic Domain , Crystallization , Drug Discovery , High-Throughput Screening Assays , Humans , Ligands , Models, Molecular , Molecular Docking Simulation , Molecular Sequence Data , Phosphodiesterase Inhibitors/therapeutic use , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Sequence Alignment , Trypanosoma brucei brucei/enzymology , Trypanosomiasis, African/drug therapy , X-Ray Diffraction
3.
J Med Chem ; 55(20): 8745-56, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-22963052

ABSTRACT

Trypanosomal phosphodiesterases B1 and B2 (TbrPDEB1 and TbrPDEB2) play an important role in the life cycle of Trypanosoma brucei, the causative parasite of human African trypanosomiasis (HAT), also known as African sleeping sickness. We used homology modeling and docking studies to guide fragment growing into the parasite-specific P-pocket in the enzyme binding site. The resulting catechol pyrazolinones act as potent TbrPDEB1 inhibitors with IC50 values down to 49 nM. The compounds also block parasite proliferation (e.g., VUF13525 (20b): T. brucei rhodesiense IC50 = 60 nM, T. brucei brucei IC50 = 520 nM, T. cruzi = 7.6 µM), inducing a typical multiple nuclei and kinetoplast phenotype without being generally cytotoxic. The mode of action of 20b was investigated with recombinantly engineered trypanosomes expressing a cAMP-sensitive FRET sensor, confirming a dose-response related increase of intracellular cAMP levels in trypanosomes. Our findings further validate the TbrPDEB family as antitrypanosomal target.


Subject(s)
Catechols/chemical synthesis , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Pyrazoles/chemical synthesis , Pyrazolones/chemical synthesis , Tetrazoles/chemical synthesis , Trypanocidal Agents/chemical synthesis , Trypanosoma brucei brucei/drug effects , Binding Sites , Catechols/chemistry , Catechols/pharmacology , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/chemistry , Drug Design , Molecular Docking Simulation , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrazolones/chemistry , Pyrazolones/pharmacology , Structure-Activity Relationship , Tetrazoles/chemistry , Tetrazoles/pharmacology , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/enzymology , Trypanosoma brucei rhodesiense/drug effects , Trypanosoma brucei rhodesiense/enzymology
4.
J Biol Chem ; 287(30): 25640-9, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22613721

ABSTRACT

CsTx-1, the main neurotoxic acting peptide in the venom of the spider Cupiennius salei, is composed of 74 amino acid residues, exhibits an inhibitory cysteine knot motif, and is further characterized by its highly cationic charged C terminus. Venom gland cDNA library analysis predicted a prepropeptide structure for CsTx-1 precursor. In the presence of trifluoroethanol, CsTx-1 and the long C-terminal part alone (CT1-long; Gly-45-Lys-74) exhibit an α-helical structure, as determined by CD measurements. CsTx-1 and CT1-long are insecticidal toward Drosophila flies and destroys Escherichia coli SBS 363 cells. CsTx-1 causes a stable and irreversible depolarization of insect larvae muscle cells and frog neuromuscular preparations, which seem to be receptor-independent. Furthermore, this membranolytic activity could be measured for Xenopus oocytes, in which CsTx-1 and CT1-long increase ion permeability non-specifically. These results support our assumption that the membranolytic activities of CsTx-1 are caused by its C-terminal tail, CT1-long. Together, CsTx-1 exhibits two different functions; as a neurotoxin it inhibits L-type Ca(2+) channels, and as a membranolytic peptide it destroys a variety of prokaryotic and eukaryotic cell membranes. Such a dualism is discussed as an important new mechanism for the evolution of spider venomous peptides.


Subject(s)
Evolution, Molecular , Neurotoxins/chemistry , Spider Venoms/chemistry , Spiders/chemistry , Animals , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cell Membrane/chemistry , Cell Membrane/genetics , Cell Membrane/metabolism , DNA, Complementary/genetics , Drosophila melanogaster , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Female , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Neurotoxins/genetics , Protein Structure, Tertiary , Rana temporaria , Spider Venoms/genetics , Spiders/genetics , Xenopus laevis
5.
J Infect Dis ; 206(2): 229-37, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22291195

ABSTRACT

The development of drugs for neglected infectious diseases often uses parasite-specific enzymes as targets. We here demonstrate that parasite enzymes with highly conserved human homologs may represent a promising reservoir of new potential drug targets. The cyclic nucleotide-specific phosphodiesterases (PDEs) of Trypanosoma brucei, causative agent of the fatal human sleeping sickness, are essential for the parasite. The highly conserved human homologs are well-established drug targets. We here describe what is to our knowledge the first pharmacological validation of trypanosomal PDEs as drug targets. High-throughput screening of a proprietary compound library identified a number of potent hits. One compound, the tetrahydrophthalazinone compound A (Cpd A), was further characterized. It causes a dramatic increase of intracellular cyclic adenosine monophosphate (cAMP). Short-term cell viability is not affected, but cell proliferation is inhibited immediately, and cell death occurs within 3 days. Cpd A prevents cytokinesis, resulting in multinucleated, multiflagellated cells that eventually lyse. These observations pharmacologically validate the highly conserved trypanosomal PDEs as potential drug targets.


Subject(s)
Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Pyridazines/pharmacology , Tetrazoles/pharmacology , Trypanosoma brucei brucei/enzymology , Trypanosomiasis, African/drug therapy , Animals , Cell Proliferation/drug effects , Drug Delivery Systems , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation, Enzymologic , Mice , Molecular Structure , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/therapeutic use , Phosphoric Diester Hydrolases/drug effects , Phosphoric Diester Hydrolases/genetics , Pyridazines/chemistry , Pyridazines/therapeutic use , Tetrazoles/chemistry , Tetrazoles/therapeutic use , Trypanosoma brucei brucei/drug effects
6.
J Biol Chem ; 287(15): 11788-97, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22356915

ABSTRACT

Trypanosoma cruzi phosphodiesterase C (TcrPDEC) is a potential new drug target for the treatment of Chagas disease but has not been well studied. This study reports the enzymatic properties of various kinetoplastid PDECs and the crystal structures of the unliganded TcrPDEC1 catalytic domain and its complex with an inhibitor. Mutations of PDEC during the course of evolution led to inactivation of PDEC in Trypanosoma brucei/Trypanosoma evansi/Trypanosoma congolense, whereas the enzyme is active in all other kinetoplastids. The TcrPDEC1 catalytic domain hydrolyzes both cAMP and cGMP with a K(m) of 23.8 µm and a k(cat) of 31 s(-1) for cAMP and a K(m) of 99.1 µm and a k(cat) of 17 s(-1) for cGMP, thus confirming its dual specificity. The crystal structures show that the N-terminal fragment wraps around the TcrPDEC catalytic domain and may thus regulate its enzymatic activity via direct interactions with the active site residues. A PDE5 selective inhibitor that has an IC(50) of 230 nm for TcrPDEC1 binds to TcrPDEC1 in an orientation opposite to that of sildenafil. This observation, together with the screen of the inhibitory potency of human PDE inhibitors against TcrPDEC, implies that the scaffold of some human PDE inhibitors might be used as the starting model for design of parasite PDE inhibitors. The structural study also identified a unique parasite pocket that neighbors the active site and may thus be valuable for the design of parasite-specific inhibitors.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/chemistry , Phosphodiesterase Inhibitors/chemistry , Phosphoric Diester Hydrolases/chemistry , Protozoan Proteins/chemistry , Sulfonamides/chemistry , Trypanosoma cruzi/enzymology , Amino Acid Sequence , Catalytic Domain , Conserved Sequence , Crystallography, X-Ray , Drug Design , Kinetics , Molecular Sequence Data , Phosphoric Diester Hydrolases/biosynthesis , Phosphoric Diester Hydrolases/metabolism , Protein Binding , Protein Structure, Secondary , Protozoan Proteins/biosynthesis , Protozoan Proteins/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Saccharomyces cerevisiae , Surface Properties
7.
Future Med Chem ; 3(10): 1289-306, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21859303

ABSTRACT

Protozoan infections remain a major unsolved medical problem in many parts of our world. A major obstacle to their treatment is the blatant lack of medication that is affordable, effective, safe and easy to administer. For some of these diseases, including human sleeping sickness, very few compounds are available, many of them old and all of them fraught with toxic side effects. We explore a new concept for developing new-generation antiprotozoan drugs that are based on phosphodiesterase (PDE) inhibitors. Such inhibitors are already used extensively in human pharmacology. Given the high degree of structural similarity between the human and the protozoan PDEs, the vast expertise available in the human field can now be applied to developing disease-specific PDE inhibitors as new antiprotozoan drugs.


Subject(s)
Antiprotozoal Agents/chemistry , Phosphodiesterase Inhibitors/chemistry , Phosphoric Diester Hydrolases/chemistry , 1-Methyl-3-isobutylxanthine/chemistry , 1-Methyl-3-isobutylxanthine/pharmacology , 1-Methyl-3-isobutylxanthine/therapeutic use , Amino Acid Sequence , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Binding Sites , Catalytic Domain , Catechols/chemistry , Catechols/pharmacology , Catechols/therapeutic use , Humans , Leishmania major/enzymology , Molecular Sequence Data , Neglected Diseases/drug therapy , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , Phosphoric Diester Hydrolases/classification , Phosphoric Diester Hydrolases/metabolism , Sequence Alignment , Trypanosoma brucei brucei/enzymology
8.
Handb Exp Pharmacol ; (204): 487-510, 2011.
Article in English | MEDLINE | ID: mdl-21695653

ABSTRACT

Protozoan parasites of the order kinetoplastida are the causative agents of three of the world's most important neglected human diseases: African trypanosomiasis, American trypanosomiasis, and leishmaniasis. Current therapies are limited, with some treatments having serious and sometimes lethal side effects. The growing number of cases that are refractory to treatment is also of concern. With few new drugs in development, there is an unmet medical need for new, more effective, and safer medications. Recent studies employing genetic and pharmacological techniques have begun to shed light on the role of the cyclic nucleotide phosphodiesterases in the life cycle of these pathogens and suggest that these important regulators of cyclic nucleotide signaling may be promising new targets for the treatment of parasitic diseases.


Subject(s)
Leishmaniasis/drug therapy , Phosphodiesterase Inhibitors/therapeutic use , Trypanosomiasis/drug therapy , Animals , Crystallization , Humans , Kinetoplastida/enzymology , Leishmaniasis/enzymology , Nucleotides, Cyclic/physiology , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/physiology , Signal Transduction/physiology , Trypanosomiasis/enzymology
9.
BMC Microbiol ; 11: 4, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-21208463

ABSTRACT

BACKGROUND: Exopolyphosphatases and pyrophosphatases play important but still incompletely understood roles in energy metabolism, and also in other aspects of cell biology such as osmoregulation or signal transduction. Earlier work has suggested that a human exopolyphosphatase, Prune, might exhibit cyclic nucleotide phosphodiesterase activity. RESULTS: The kinetoplastida, a large order of unicellular eukaryotes that contains many important pathogens such as Trypanosoma brucei (human sleeping sickness), Trypanosoma cruzi (Chagas disease) or Leishmania ssp (several clinically dinstinct leishmaniases) all contain several exo- and pyrophosphatases. The current study provides a systematic classification of these enzymes, which now allows to situate the information that is already available on some of these enzymes. It then analyses the exopolyphosphatase TbrPPX1 of T. brucei in detail, using RNA interference and genetic knockouts in an attempt to define its function, and immunofluorescence microscopy to study its subcellular localization.TbrPPX1 is an exopolyphosphatase that does hydrolyze pentasodium triphosphate, but not organic triphosphates such as ATP, pyrophosphate or long-chain polyphosphates. Finally, the study investigates the potential cyclic nucleotide phosphodiesterase activity of TbrPPX1. CONCLUSIONS: All kinetoplastid genomes that are currently available contain genes for an exopolyphosphatase and two classes of pyrophosphatases, one associated with the acidocalcisomes and one cytoplasmic. TbrPPX1 represents the T. brucei exopolyphosphatase. It is located throughout the cytoplasm, and its genetic ablation does not produce a dramatic phenotype. Importantly, TbrPPX1 does not exhibit any cyclic nucleotide specific phosphodiesterase activity, which definitively eliminates it as an additional player in cAMP signalling of the kinetoplastida.


Subject(s)
Acid Anhydride Hydrolases/genetics , Acid Anhydride Hydrolases/metabolism , Genome, Protozoan , Trypanosoma brucei brucei/enzymology , Trypanosoma brucei brucei/genetics , Amino Acid Sequence , Animals , Cell Line , Cloning, Molecular , Female , Gene Expression Regulation , Gene Knockout Techniques , Mice , Molecular Sequence Data , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Pyrophosphatases/genetics , Pyrophosphatases/metabolism , RNA Interference
10.
Amino Acids ; 40(1): 69-76, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20140690

ABSTRACT

Cupiennin 1a, a cytolytic peptide isolated from the venom of the spider Cupiennius salei, exhibits broad membranolytic activity towards bacteria, trypanosomes, and plasmodia, as well as human blood and cancer cells. In analysing the cytolytic activity of synthesised all-D: - and all-L: -cupiennin 1a towards pro- and eukaryotic cells, a stereospecific mode of membrane destruction could be excluded. The importance of negatively charged sialic acids on the outer leaflet of erythrocytes for the binding and haemolytic activity of L: -cupiennin 1a was demonstrated. Reducing the overall negative charges of erythrocytes by partially removing their sialic acids or by protecting them with tri- or pentalysine results in reduced haemolytic activity of the peptide.


Subject(s)
Anti-Infective Agents/pharmacology , Antineoplastic Agents/pharmacology , Cytotoxins/pharmacology , Insecticides/pharmacology , Peptides/pharmacology , Spider Venoms/pharmacology , Spiders/chemistry , Animals , Anti-Infective Agents/chemistry , Antimicrobial Cationic Peptides , Antineoplastic Agents/chemistry , Bacteria/drug effects , Cell Line, Tumor , Cytotoxins/chemistry , Drosophila melanogaster/drug effects , Humans , Insecticides/chemistry , Molecular Structure , Parasites/drug effects , Peptides/chemistry , Spider Venoms/chemistry
11.
Mol Microbiol ; 78(3): 757-69, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20969649

ABSTRACT

The parasitic protozoa Trypanosoma brucei has a complex life cycle. Oxidative phosphorylation is highly active in the procyclic form but absent from bloodstream cells. The mitochondrial genome encodes several gene products that are required for oxidative phosphorylation, but it completely lacks tRNA genes. For mitochondrial translation to occur, the import of cytosolic tRNAs is therefore essential for procyclic T. brucei. Whether the same is true for the bloodstream form has not been studied so far. Here we show that the steady-state levels of mitochondrial tRNAs are essentially the same in both life stages. Editing of the imported tRNA(Trp) also occurs in both forms as well as in mitochondria of Trypanosoma evansi, which lacks a genome and a translation system. These results show that mitochondrial tRNA import is a constitutive process that must be mediated by proteins that are expressed in both forms of the life cycle and that are not encoded in the mitochondrial genome. Moreover, bloodstream cells lacking either mitochondria-specific translation elongation factor Tu or mitochondrial tryptophanyl-tRNA synthetase are not viable indicating that mitochondrial translation is also essential in this stage. Both of these proteins show trypanosomatid-specific features and may therefore be excellent novel drug targets.


Subject(s)
Blood/parasitology , Mitochondria/genetics , Protein Biosynthesis , Protozoan Proteins/genetics , Trypanosoma brucei brucei/growth & development , Trypanosomiasis, African/parasitology , Animals , Animals, Outbred Strains , Biological Transport , Female , Humans , Mice , Mitochondria/metabolism , Molecular Sequence Data , Protozoan Proteins/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/metabolism
12.
Eukaryot Cell ; 9(10): 1466-75, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20693305

ABSTRACT

The precise subcellular localization of the components of the cyclic AMP (cAMP) signaling pathways is a crucial aspect of eukaryotic intracellular signaling. In the human pathogen Trypanosoma brucei, the strict control of cAMP levels by cAMP-specific phosphodiesterases is essential for parasite survival, both in cell culture and in the infected host. Among the five cyclic nucleotide phosphodiesterases identified in this organism, two closely related isoenzymes, T. brucei PDEB1 (TbrPDEB1) (PDEB1) and TbrPDEB2 (PDEB2) are predominantly responsible for the maintenance of cAMP levels. Despite their close sequence similarity, they are distinctly localized in the cell. PDEB1 is mostly located in the flagellum, where it forms an integral part of the flagellar skeleton. PDEB2 is mainly located in the cell body, and only a minor part of the protein localizes to the flagellum. The current study, using transfection of procyclic trypanosomes with green fluorescent protein (GFP) reporters, demonstrates that the N termini of the two enzymes are essential for determining their final subcellular localization. The first 70 amino acids of PDEB1 are sufficient to specifically direct a GFP reporter to the flagellum and to lead to its detergent-resistant integration into the flagellar skeleton. In contrast, the analogous region of PDEB2 causes the GFP reporter to reside predominantly in the cell body. Mutagenesis of selected residues in the N-terminal region of PDEB2 demonstrated that single amino acid changes are sufficient to redirect the reporter from a cell body location to stable integration into the flagellar skeleton.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/chemistry , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Cytoskeleton/enzymology , Flagella/enzymology , Signal Transduction , Trypanosoma brucei brucei/enzymology , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Amino Acid Sequence , Animals , Cyclic AMP/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Molecular Sequence Data , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/metabolism
13.
PLoS Negl Trop Dis ; 3(6): e455, 2009 Jun 09.
Article in English | MEDLINE | ID: mdl-19513125

ABSTRACT

BACKGROUND: Chromosome 9 of Trypanosoma brucei contains two closely spaced, very similar open reading frames for cyclic nucleotide specific phosphodiesterases TbrPDEB1 and TbrPDEB2. They are separated by 2379 bp, and both code for phosphodiesterases with two GAF domains in their N-terminal moieties and a catalytic domain at the C-terminus. METHODS AND FINDINGS: The current study reveals that in the Lister427 strain of T. brucei, these two genes have undergone gene conversion, replacing the coding region for the GAF-A domain of TbrPDEB2 by the corresponding region of the upstream gene TbrPDEB1. As a consequence, these strains express two slightly different versions of TbrPDEB2. TbrPDEB2a represents the wild-type phosphodiesterase, while TbrPDEB2b represents the product of the converted gene. Earlier work on the subcellular localization of TbrPDEB1 and TbrPDEB2 had demonstrated that TbrPDEB1 is predominantly located in the flagellum, whereas TbrPDEB2 partially locates to the flagellum but largely remains in the cell body. The current findings raised the question of whether this dual localization of TbrPDEB2 may reflect the two alleles. To resolve this, TbrPDEB2 of strain STIB247 that is homozygous for TbrPDEB2a was tagged in situ, and its intracellular localization was analyzed. CONCLUSIONS: The results obtained were very similar to those found earlier with Lister427, indicating that the dual localization of TbrPDEB2 reflects its true function and is not simply due to the presence of the two different alleles. Notably, the gene conversion event is unique for the Lister427 strain and all its derivatives. Based on this finding, a convenient PCR test has been developed that allows the stringent discrimination between Lister-derived strains that are common in many laboratories and other isolates. The technique is likely very useful to resolve questions about potential mix-ups of precious field isolates with the ubiquitous Lister strain.


Subject(s)
Gene Conversion , Phosphoric Diester Hydrolases/genetics , Trypanosoma brucei brucei/enzymology , Trypanosoma brucei brucei/genetics , Alleles , Animals , Cytoplasm/enzymology , DNA, Protozoan/genetics , Flagella/enzymology , Polymerase Chain Reaction/methods , Protein Structure, Tertiary/genetics
14.
Anal Biochem ; 382(2): 87-93, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18722997

ABSTRACT

The urgent need for new drug development for African trypanosomiasis is widely recognized. This requires reliable and informative high-throughput assays. Currently, drug action is determined with a fluorimetric/colorimetric assay based on the metabolism of the dye Alamar Blue (resazurin) by live cells. However, this assay does not easily distinguish between cell death and growth arrest, or supply information about the rate at which test compounds affect these parameters. We report here an alternative fluorimetric assay, based on the interaction of propidium iodide with DNA, that allows either real-time monitoring of cell viability or the generation of EC(50) values at a predetermined time-point. The assay is highly sensitive and fluorescence readings easily correlate to numbers of parasites or DNA content. The EC(50) values were highly similar to those obtained with the standard Alamar Blue assay. The procedure lends itself readily to applications in drug development or resistance monitoring.


Subject(s)
Drug Evaluation, Preclinical/methods , Propidium/analysis , Trypanocidal Agents/pharmacology , Animals , Biological Assay , Cell Survival , DNA, Protozoan/metabolism , Indicators and Reagents , Oxazines/analysis , Oxazines/metabolism , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/metabolism , Trypanosomiasis, African/drug therapy , Xanthenes/analysis , Xanthenes/metabolism
15.
Int J Parasitol ; 38(14): 1625-37, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18590734

ABSTRACT

Cyclic nucleotide-specific phosphodiesterases (PDEs) have come into focus as interesting potential targets for PDE inhibitor-based anti-parasitic drugs. Genomes of the various agents of human malaria, most notably Plasmodium falciparum, all contain four genes for class 1 PDEs. The catalytic domains of these enzymes are closely related to those of the 11 human PDE families. This presents the possibility that the available vast expertise in developing drugs against human PDEs might now also be applied to developing compounds that are active against malarial PDEs. The current study identifies four Plasmodium genes that code for PfPDEalpha, PfPDEbeta, PfPDEgamma and PfPDEdelta, respectively. It further demonstrates that the PfPDEalpha polypeptide exists in two versions (PfPDEalphaA and PfPDEalphaB) that are generated by alternative splicing of the primary transcript. All malarial PDEs contain several transmembrane helices in their N-terminal regions, indicating that they are integral membrane proteins. In agreement with this prediction, essentially all PDE activity is associated with the cell membranes. PfPDEalpha was characterized as a cGMP-specific PDE that is not sensitive to a number of standard PDE inhibitors. Genetic ablation of the PfPDE1 gene produced no major phenotype in erythrocyte cultures.


Subject(s)
Membrane Proteins/genetics , Phosphoric Diester Hydrolases/genetics , Plasmodium falciparum/enzymology , Animals , Blotting, Southern , Cyclic GMP/genetics , Enzyme Inhibitors , Gene Expression Regulation, Enzymologic , Malaria/enzymology , Malaria/genetics , Membrane Proteins/metabolism , Molecular Sequence Data , Phosphoric Diester Hydrolases/metabolism , Plasmodium falciparum/genetics , Polymerase Chain Reaction , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
16.
Mol Microbiol ; 66(4): 1029-38, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17944832

ABSTRACT

Human leishmaniasis is a major public health problem in many countries, but chemotherapy is in an unsatisfactory state. Leishmania major phosphodiesterases (LmjPDEs) have been shown to play important roles in cell proliferation and apoptosis of the parasite. Thus LmjPDE inhibitors may potentially represent a novel class of drugs for the treatment of leishmaniasis. Reported here are the kinetic characterization of the LmjPDEB1 catalytic domain and its crystal structure as a complex with 3-isobutyl-1-methylxanthine (IBMX) at 1.55 A resolution. The structure of LmjPDEB1 is similar to that of human PDEs. IBMX stacks against the conserved phenylalanine and forms a hydrogen bond with the invariant glutamine, in a pattern common to most inhibitors bound to human PDEs. However, an extensive structural comparison reveals subtle, but significant differences between the active sites of LmjPDEB1 and human PDEs. In addition, a pocket next to the inhibitor binding site is found to be unique to LmjPDEB1. This pocket is isolated by two gating residues in human PDE families, but constitutes a natural expansion of the inhibitor binding pocket in LmjPDEB1. The structure particularity might be useful for the development of parasite-selective inhibitors for the treatment of leishmaniasis.


Subject(s)
Drug Design , Leishmania major/drug effects , Leishmania major/enzymology , Phosphodiesterase Inhibitors/chemistry , Phosphoric Diester Hydrolases/chemistry , 1-Methyl-3-isobutylxanthine/metabolism , Amino Acid Sequence , Animals , Binding Sites , Catalytic Domain , Crystallization , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/drug effects , Phosphoric Diester Hydrolases/metabolism , Protein Folding
17.
Trends Parasitol ; 23(2): 71-7, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17174157

ABSTRACT

Flagellar-mediated motility is an indispensable function for cell types as evolutionarily distant as mammalian sperm and kinetoplastid parasites, a large group of flagellated protozoa that includes several important human pathogens. Despite the obvious importance of flagellar motility, little is known about the signalling processes that direct the frequency and wave shape of the flagellar beat, or those that provide the motile cell with the necessary environmental cues that enable it to aim its movement. Similarly, the energetics of the flagellar beat and the problem of a sufficient ATP supply along the entire length of the beating flagellum remain to be explored. Recent proteome projects studying the flagella of mammalian sperm and kinetoplastid parasites have provided important information and have indicated a surprising degree of similarities between the flagella of these two cell types.


Subject(s)
Cell Movement/physiology , Flagella/physiology , Spermatozoa/physiology , Trypanosoma/physiology , Animals , Flagella/metabolism , Glycolysis , Humans , Male , Mice , Nucleotides, Cyclic/metabolism , Signal Transduction , Spermatozoa/metabolism , Trypanosoma/metabolism
18.
FASEB J ; 21(3): 720-31, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17167070

ABSTRACT

Cyclic nucleotide specific phosphodiesterases (PDEs) are pivotal regulators of cellular signaling. They are also important drug targets. Besides catalytic activity and substrate specificity, their subcellular localization and interaction with other cell components are also functionally important. In contrast to the mammalian PDEs, the significance of PDEs in protozoal pathogens remains mostly unknown. The genome of Trypanosoma brucei, the causative agent of human sleeping sickness, codes for five different PDEs. Two of these, TbrPDEB1 and TbrPDEB2, are closely similar, cAMP-specific PDEs containing two GAF-domains in their N-terminal regions. Despite their similarity, these two PDEs exhibit different subcellular localizations. TbrPDEB1 is located in the flagellum, whereas TbrPDEB2 is distributed between flagellum and cytoplasm. RNAi against the two mRNAs revealed that the two enzymes can complement each other but that a simultaneous ablation of both leads to cell death in bloodstream form trypanosomes. RNAi against TbrPDEB1 and TbrPDEB2 also functions in vivo where it completely prevents infection and eliminates ongoing infections. Our data demonstrate that TbrPDEB1 and TbrPDEB2 are essential for virulence, making them valuable potential targets for new PDE-inhibitor based trypanocidal drugs. Furthermore, they are compatible with the notion that the flagellum of T. brucei is an important site of cAMP signaling.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/physiology , Trypanosoma brucei brucei/pathogenicity , Virulence/physiology , 3',5'-Cyclic-AMP Phosphodiesterases/classification , Animals , Humans , Trypanosoma brucei brucei/enzymology , Trypanosomiasis, African/parasitology
19.
BMC Microbiol ; 6: 25, 2006 Mar 08.
Article in English | MEDLINE | ID: mdl-16522215

ABSTRACT

BACKGROUND: Leishmania represent a complex of important human pathogens that belong to the systematic order of the kinetoplastida. They are transmitted between their human and mammalian hosts by different bloodsucking sandfly vectors. In their hosts, the Leishmania undergo several differentiation steps, and their coordination and optimization crucially depend on numerous interactions between the parasites and the physiological environment presented by the fly and human hosts. Little is still known about the signalling networks involved in these functions. In an attempt to better understand the role of cyclic nucleotide signalling in Leishmania differentiation and host-parasite interaction, we here present an initial study on the cyclic nucleotide-specific phosphodiesterases of Leishmania major. RESULTS: This paper presents the identification of three class I cyclic-nucleotide-specific phosphodiesterases (PDEs) from L. major, PDEs whose catalytic domains exhibit considerable sequence conservation with, among other, all eleven human PDE families. In contrast to other protozoa such as Dictyostelium, or fungi such as Saccharomyces cerevisiae, Candida ssp or Neurospora, no genes for class II PDEs were found in the Leishmania genomes. LmjPDEA contains a class I catalytic domain at the C-terminus of the polypeptide, with no other discernible functional domains elsewhere. LmjPDEB1 and LmjPDEB2 are coded for by closely related, tandemly linked genes on chromosome 15. Both PDEs contain two GAF domains in their N-terminal region, and their almost identical catalytic domains are located at the C-terminus of the polypeptide. LmjPDEA, LmjPDEB1 and LmjPDEB2 were further characterized by functional complementation in a PDE-deficient S. cerevisiae strain. All three enzymes conferred complementation, demonstrating that all three can hydrolyze cAMP. Recombinant LmjPDEB1 and LmjPDEB2 were shown to be cAMP-specific, with Km values in the low micromolar range. Several PDE inhibitors were found to be active against these PDEs in vitro, and to inhibit cell proliferation. CONCLUSION: The genome of L. major contains only PDE genes that are predicted to code for class I PDEs, and none for class II PDEs. This is more similar to what is found in higher eukaryotes than it is to the situation in Dictyostelium or the fungi that concomitantly express class I and class II PDEs. Functional complementation demonstrated that LmjPDEA, LmjPDEB1 and LmjPDEB2 are capable of hydrolyzing cAMP. In vitro studies with recombinant LmjPDEB1 and LmjPDEB2 confirmed this, and they demonstrated that both are completely cAMP-specific. Both enzymes are inhibited by several commercially available PDE inhibitors. The observation that these inhibitors also interfere with cell growth in culture indicates that inhibition of the PDEs is fatal for the cell, suggesting an important role of cAMP signalling for the maintenance of cellular integrity and proliferation.


Subject(s)
Leishmania major/enzymology , Phosphoric Diester Hydrolases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Proliferation , Enzyme Inhibitors , Gene Expression Regulation, Enzymologic , Phosphoric Diester Hydrolases/genetics , RNA, Protozoan/metabolism , Substrate Specificity
20.
Mol Biochem Parasitol ; 146(1): 38-44, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16343658

ABSTRACT

We previously showed that over-expression of Trypanosoma brucei MRPA, a member of the multidrug resistance protein family in T. brucei, reproducibly resulted in resistance to the anti-trypanosomal drug melarsoprol in vitro. MRPA is predicted to mediate efflux of melarsoprol as a conjugate with trypanothione, a glutathione-spermidine conjugate which is the major small thiol in trypanosomes. Here, we show that depletion of MRPA by RNA interference resulted in moderate hypersensitivity to both melarsoprol and melarsen oxide. Over-expression of MRPA alone is not sufficient to cause melarsoprol resistance in vivo, although it is sufficient in vitro. This discrepancy is not an effect of drug metabolism since over-expression of MRPA alone conferred resistance to melarsoprol and its principle metabolite, melarsen oxide, in vitro. Over-expression of MRPA was not detected in four melarsoprol-resistant trypanosome isolates from sleeping sickness patients.


Subject(s)
Melarsoprol/pharmacology , Membrane Transport Proteins/physiology , Multidrug Resistance-Associated Proteins/physiology , Protozoan Proteins/physiology , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , Trypanosomiasis, African/parasitology , Animals , Arsenicals/pharmacology , Blotting, Western/methods , Cell Line , Electrophoresis, Polyacrylamide Gel , Female , Gene Expression , Humans , Melarsoprol/chemistry , Melarsoprol/therapeutic use , Membrane Transport Proteins/analysis , Membrane Transport Proteins/biosynthesis , Mice , Multidrug Resistance-Associated Proteins/analysis , Multidrug Resistance-Associated Proteins/biosynthesis , Parasitic Sensitivity Tests/methods , Protozoan Proteins/analysis , Protozoan Proteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Treatment Failure , Trypanocidal Agents/chemistry , Trypanocidal Agents/therapeutic use , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/metabolism , Trypanosomiasis, African/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...