Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
BMC Res Notes ; 10(1): 364, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28764751

ABSTRACT

OBJECTIVE: Recent studies into the antifungal activity of NK-cells against the Aspergillus fumigatus have presented differing accounts on their mode of antifungal activity. One of these mechanisms proposed that NK-cells may kill the fungus via the direct effects of exposure to Interferon gamma (IFN-γ). RESULTS: In this study we investigated the direct antifungal effects of recombinant human IFN-γ against a range of pathogenic fungi by measuring cellular damage using an XTT-based assay and cell viability through plate counts. It was found that 32 pg/ml of IFN-γ exhibited a significant but small antifungal effect on A. fumigatus (p = 0.02), Aspergillus flavus (p = 0.04) and Saccharomyces cerevisiae (p = 0.03), inhibiting growth by 6, 11 and 17% respectively. No significant inhibitory effects were observed in Candida species (p > 0.05 for all species tested) or Cryptococus neoformans (p = 0.98). Short term exposure (3 h) to a combination of amphotericin B (1 µg/ml) and IFN-γ (32 pg/ml) increased the effectiveness of amphotericin B against A. fumigatus and S. cerevisiae but not Candida albicans. These data suggest that IFN-γ does not possess strong antifungal activity but can enhance the effect of amphotericin B under some testing conditions against Aspergillus species.


Subject(s)
Amphotericin B/pharmacology , Antifungal Agents/pharmacology , Aspergillus fumigatus/drug effects , Candida albicans/drug effects , Interferon-gamma/pharmacology , Aspergillus flavus/drug effects , Aspergillus flavus/growth & development , Aspergillus fumigatus/growth & development , Candida albicans/growth & development , Cryptococcus neoformans , Drug Combinations , Drug Synergism , Humans , Microbial Sensitivity Tests , Recombinant Proteins/pharmacology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/growth & development
2.
Int J Parasitol ; 47(4): 203-213, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28104352

ABSTRACT

Certain clinical isolates of Tritrichomonas foetus infect the urogenital tract of cattle while others infect the gastrointestinal tract of cats. Previous studies have identified subtle genetic differences between these isolates with the term "genotype" adopted to reflect host origin. The aim of this work was to seek evidence of host-specific adaptation and to clarify the relationship between T. foetus genotypes. To do this we characterised the proteomes of both genotypes using two-dimensional gel electrophoresis (2DE) coupled with LC-MS/MS. Our comparative analysis of the data revealed that both genotypes exhibited largely similar proteoform profiles; however differentiation was possible with 24 spots identified as having a four-fold or greater change. Deeper analysis using 2DE zymography and protease-specific fluorogenic substrates revealed marked differences in cysteine protease (CP) expression profiles between the two genotypes. These variances in CP activities could also account for the pathogenic and histopathological differences previously observed between T. foetus genotypes in cross-infection studies. Our findings highlight the importance of CPs as major determinants of parasite virulence and provide a foundation for future host-parasite interaction studies, with direct implications for the development of vaccines or drugs targeting T. foetus.


Subject(s)
Genotype , Proteome/analysis , Protozoan Proteins/analysis , Tritrichomonas foetus/chemistry , Animals , Cat Diseases/parasitology , Cats , Cattle , Cattle Diseases/parasitology , Chromatography, Liquid , Cysteine Proteases/analysis , Electrophoresis, Gel, Two-Dimensional , Proteomics , Protozoan Infections/parasitology , Tandem Mass Spectrometry , Tritrichomonas foetus/classification , Tritrichomonas foetus/genetics , Tritrichomonas foetus/isolation & purification
3.
Exp Parasitol ; 169: 13-21, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27373432

ABSTRACT

Infection with the apicomplexan parasite Plasmodium falciparum is a major cause of morbidity and mortality worldwide. One of the striking features of this parasite is its ability to remodel and decrease the deformability of host red blood cells, a process that contributes to disease. To further understand the virulence of Pf we investigated the biochemistry and function of a putative Pf S33 proline aminopeptidase (PfPAP). Unlike other P. falciparum aminopeptidases, PfPAP contains a predicted protein export element that is non-syntenic with other human infecting Plasmodium species. Characterization of PfPAP demonstrated that it is exported into the host red blood cell and that it is a prolyl aminopeptidase with a preference for N-terminal proline substrates. In addition genetic deletion of this exopeptidase was shown to lead to an increase in the deformability of parasite-infected red cells and in reduced adherence to the endothelial cell receptor CD36 under flow conditions. Our studies suggest that PfPAP plays a role in the rigidification and adhesion of infected red blood cells to endothelial surface receptors, a role that may make this protein a novel target for anti-disease interventions strategies.


Subject(s)
Aminopeptidases/metabolism , Erythrocyte Deformability/physiology , Plasmodium falciparum/enzymology , Amino Acid Sequence , Aminopeptidases/chemistry , Aminopeptidases/genetics , Aminopeptidases/immunology , Antibodies, Protozoan/immunology , Blotting, Northern , Blotting, Western , Cell Adhesion/physiology , Elasticity , Erythrocyte Membrane/genetics , Erythrocyte Membrane/physiology , Erythrocytes/parasitology , Gene Knockout Techniques , Humans , Microscopy, Atomic Force , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Plasmodium falciparum/genetics , RNA, Protozoan/chemistry , Real-Time Polymerase Chain Reaction , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sequence Alignment , Transfection
4.
Ann Clin Microbiol Antimicrob ; 13: 18, 2014 May 12.
Article in English | MEDLINE | ID: mdl-24884795

ABSTRACT

BACKGROUND: There is a pressing need to identify novel antifungal drug targets to aid in the therapy of life-threatening mycoses and overcome increasing drug resistance. Identifying specific mechanisms of action of membrane-interacting antimicrobial drugs on the model fungus Saccharomyces cerevisiae is one avenue towards addressing this issue. The S. cerevisiae deletion mutants Δizh2, Δizh3, Δaif1 and Δstm1 were demonstrated to be resistant to amphibian-derived antimicrobial peptides (AMPs). The purpose of this study was to examine whether AMPs and polyene antifungals have a similar mode of action; this was done by comparing the relative tolerance of the mutants listed above to both classes of antifungal. FINDINGS: In support of previous findings on solid media it was shown that Δizh2 and Δizh3 mutants had increased resistance to both amphotericin B (1-2 µg ml-1) and nystatin (2.5 - 5 µg ml-1) in liquid culture, after acute exposure. However, Δaif1 and Δstm1 had wild-type levels of susceptibility to these polyenes. The generation of reactive oxygen species (ROS) after exposure to amphotericin B was also reduced in Δizh2 and Δizh3. These data indicated that polyene antifungal and AMPs may act via distinct mechanisms of inducing cell death in S. cerevisiae. CONCLUSIONS: Further understanding of the mechanism(s) involved in causing cell death and the roles of IZH2 and IZH3 in drug susceptibility may help to inform improved drug design and treatment of fungal pathogens.


Subject(s)
Amphotericin B/pharmacology , Antifungal Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Microbial Viability/drug effects , Nystatin/pharmacology , Polyenes/pharmacology , Saccharomyces cerevisiae/drug effects , Gene Deletion , Microbial Sensitivity Tests , Saccharomyces cerevisiae/genetics
5.
Lasers Med Sci ; 29(1): 157-63, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23525830

ABSTRACT

Fungal infection of nails, onychomycosis, is predominantly caused by Trichophyton rubrum. This infection is an important public health concern due to its persistent nature and high recurrence rates. Alternative treatments are urgently required. One such alternative is phototherapy involving the action of photothermal or photochemical processes. The aim of this novel study was to assess which wavelengths within the ultraviolet (UV) spectrum were inhibitory and equally important nail transmissible. Initial irradiations of T. rubrum spore suspensions were carried out using a tunable wavelength lamp system (fluence ≤3.1 J/cm(2)) at wavelengths between 280 and 400 nm (UVC to UVA) to evaluate which wavelengths prevented fungal growth. Light-emitting diodes (LEDs) of defined wavelengths were subsequently chosen with a view to evaluate and potentially implement this technology as a low-cost "in-home" treatment. Our experiments demonstrated that exposure at 280 nm using an LED with a fluence as low as 0.5 J/cm(2) was inhibitory, i.e., no growth following a 2-week incubation (p < 0.05; one-way ANOVA), while exposure to longer wavelengths was not. A key requirement for the use of phototherapy in the treatment of onychomycosis is that it must be nail transmissible. Our results indicate that the treatment with UVC is not feasible given that there is no overlap between the antifungal activity observed at 280 nm and transmission through the nail plate. However, a potential indirect application of this technology could be the decontamination of reservoirs of infection such as the shoes of infected individuals, thus preventing reinfection.


Subject(s)
Onychomycosis/radiotherapy , Trichophyton/radiation effects , Ultraviolet Therapy/methods , Foot Dermatoses/microbiology , Foot Dermatoses/radiotherapy , Humans , Nails/microbiology , Nails/radiation effects , Onychomycosis/microbiology , Optical Phenomena , Phototherapy/methods , Spores, Fungal/radiation effects , Trichophyton/pathogenicity , Ultraviolet Rays
6.
Int J Parasitol ; 42(13-14): 1143-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23123273

ABSTRACT

The parasitic protists in the genus Tritrichomonas cause significant disease in domestic cattle and cats. To assess the genetic diversity of feline and bovine isolates of Tritrichomonas foetus (Riedmüller, 1928) Wenrich and Emmerson, 1933, we used 10 different genetic regions, namely the protein coding genes of cysteine proteases 1, 2 and 4-9 (CP1, 2, 4-9) involved in the pathogenesis of the disease caused by the parasite. The cytosolic malate dehydrogenase 1 (MDH1) and internal transcribed spacer region 2 of the rDNA unit (ITS2) were included as additional markers. The gene sequences were compared with those of Tritrichomonas suis (Davaine, 1875) Morgan and Hawkins, 1948 and Tritrichomonas mobilensisCulberson et al., 1986. The study revealed 100% identity for all 10 genes among all feline isolates (=T. foetus cat genotype), 100% identity among all bovine isolates (=T. foetus cattle genotype) and a genetic distinctness of 1% between the cat and cattle genotypes of T. foetus. The cattle genotype of T. foetus was 100% identical to T. suis at nine loci (CP1, 2, 4-8, ITS2, MDH1). At CP9, three out of four T. suis isolates were identical to the T. foetus cattle genotype, while the T. suis isolate SUI-H3B sequence contained a single unique nucleotide substitution. Tritrichomonas mobilensis was 0.4% and 0.7% distinct from the cat and cattle genotypes of T. foetus, respectively. The genetic differences resulted in amino acid changes in the CP genes, most pronouncedly in CP2, potentially providing a platform for elucidation of genotype-specific host-pathogen interactions of T. foetus. On the basis of this data we judge T. suis and T. foetus to be subjective synonyms. For the first time, on objective nomenclatural grounds, the authority of T. suis is given to Davaine, 1875, rather than the commonly cited Gruby and Delafond, 1843. To maintain prevailing usage of T. foetus, we are suppressing the senior synomym T. suisDavaine, 1875 according to Article 23.9, because it has never been used as a valid name after 1899 and T. foetus is widely discussed as the cause of bovine trichomonosis. Thus bovine, feline and porcine isolates should all be given the name T. foetus. This promotes the stability of T. foetus for the veterinary and economically significant venereal parasite causing bovine trichomonosis.


Subject(s)
Genetic Variation , Tritrichomonas/classification , Tritrichomonas/genetics , Amino Acid Sequence , Animals , Base Sequence , Cats , Cattle , Gene Expression Regulation/physiology , Genotype , Helminth Proteins/genetics , Helminth Proteins/metabolism , Molecular Sequence Data , Phylogeny
7.
Proc Natl Acad Sci U S A ; 107(6): 2449-54, 2010 Feb 09.
Article in English | MEDLINE | ID: mdl-20133789

ABSTRACT

Current therapeutics and prophylactics for malaria are under severe challenge as a result of the rapid emergence of drug-resistant parasites. The human malaria parasite Plasmodium falciparum expresses two neutral aminopeptidases, PfA-M1 and PfA-M17, which function in regulating the intracellular pool of amino acids required for growth and development inside the red blood cell. These enzymes are essential for parasite viability and are validated therapeutic targets. We previously reported the X-ray crystal structure of the monomeric PfA-M1 and proposed a mechanism for substrate entry and free amino acid release from the active site. Here, we present the X-ray crystal structure of the hexameric leucine aminopeptidase, PfA-M17, alone and in complex with two inhibitors with antimalarial activity. The six active sites of the PfA-M17 hexamer are arranged in a disc-like fashion so that they are orientated inwards to form a central catalytic cavity; flexible loops that sit at each of the six entrances to the catalytic cavern function to regulate substrate access. In stark contrast to PfA-M1, PfA-M17 has a narrow and hydrophobic primary specificity pocket which accounts for its highly restricted substrate specificity. We also explicate the essential roles for the metal-binding centers in these enzymes (two in PfA-M17 and one in PfA-M1) in both substrate and drug binding. Our detailed understanding of the PfA-M1 and PfA-M17 active sites now permits a rational approach in the development of a unique class of two-target and/or combination antimalarial therapy.


Subject(s)
Aminopeptidases/chemistry , Drug Design , Plasmodium falciparum/enzymology , Protozoan Proteins/chemistry , Aminopeptidases/antagonists & inhibitors , Aminopeptidases/metabolism , Antimalarials/chemistry , Antimalarials/metabolism , Antimalarials/pharmacology , Catalysis , Catalytic Domain , Crystallography, X-Ray , Hydrophobic and Hydrophilic Interactions , Metals/chemistry , Metals/metabolism , Models, Molecular , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/metabolism , Substrate Specificity
8.
J Biol Chem ; 285(5): 3383-92, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-19923225

ABSTRACT

Helminth pathogens prepare a Th2 type immunological environment in their hosts to ensure their longevity. They achieve this by secreting molecules that not only actively drive type 2 responses but also suppress type 1 responses. Here, we show that the major cysteine proteases secreted from the helminth pathogens Fasciola hepatica (FheCL1) and Schistosoma mansoni (SmCB1) protect mice from the lethal effects of lipopolysaccharide by preventing the release of inflammatory mediators, nitric oxide, interleukin-6, tumor necrosis factor alpha, and interleukin-12, from macrophages. The proteases specifically block the MyD88-independent TRIF-dependent signaling pathway of Toll-like receptor (TLR)4 and TLR3. Microscopical and flow cytometric studies, however, show that alteration of macrophage function by cysteine protease is not mediated by cleavage of components of the TLR4 complex on the cell surface but occurs by degradation of TLR3 within the endosome. This is the first study to describe a parasite molecule that degrades this receptor and pinpoints a novel mechanism by which helminth parasites modulate the innate immune responses of their hosts to suppress the development of Th1 responses.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Cysteine Proteases/chemistry , Macrophages/enzymology , Toll-Like Receptor 3/chemistry , Animals , Cytokines/metabolism , Endotoxins/chemistry , Female , Helminths , Mice , Mice, Inbred BALB C , Myeloid Differentiation Factor 88/metabolism , Nitrites/metabolism , Recombinant Proteins/chemistry , Th1 Cells/metabolism
9.
Trends Biochem Sci ; 35(1): 53-61, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19796954

ABSTRACT

The neutral aminopeptidases M1 alanyl aminopeptidase (PfM1AAP) and M17 leucine aminopeptidase (PfM17LAP) of the human malaria parasite Plasmodium falciparum are targets for the development of novel anti-malarial drugs. Although the functions of these enzymes remain unknown, they are believed to act in the terminal stages of haemoglobin degradation, generating amino acids essential for parasite growth and development. Inhibitors of both enzymes are lethal to P. falciparum in culture and kill the murine malaria P. chabaudi in vivo. Recent biochemical, structural and functional studies provide the substrate specificity and mechanistic binding data needed to guide the development of more potent anti-malarial drugs. Together with biological studies, these data form the rationale for choosing PfM1AAP and PfM17LAP as targets for anti-malarial development.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Antimalarials/pharmacology , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Humans , Malaria, Falciparum/physiopathology , Plasmodium falciparum/enzymology
10.
Expert Rev Anti Infect Ther ; 7(9): 1087-98, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19883329

ABSTRACT

Malaria remains one of the world's most devastating infectious diseases. Drug resistance to all classes of antimalarial agents has now been observed, highlighting the need for new agents that act against novel parasite targets. The complete sequencing of the Plasmodium falciparum genome has allowed the identification of new molecular targets within the parasite that may be amenable to chemotherapeutic intervention. In this review, we investigate four possible targets for the future development of new classes of antimalarial agents. These targets include histone deacetylase, the aspartic proteases or plasmepsins, aminopeptidases and the purine salvage enzyme hypoxanthine-xanthine-guanine phosphoribosyltransferase.


Subject(s)
Antimalarials/chemistry , Drug Design , Enzyme Inhibitors/chemistry , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Aminopeptidases/antagonists & inhibitors , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use , Aspartic Acid Endopeptidases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Hypoxanthine Phosphoribosyltransferase/antagonists & inhibitors , Malaria, Falciparum/parasitology , Pentosyltransferases/antagonists & inhibitors , Plasmodium falciparum/enzymology , Plasmodium falciparum/genetics , Protozoan Proteins/antagonists & inhibitors
11.
Biochemistry ; 48(23): 5435-9, 2009 Jun 16.
Article in English | MEDLINE | ID: mdl-19408962

ABSTRACT

The M17 leucine aminopeptidase of the intraerythrocytic stages of the malaria parasite Plasmodium falciparum (PfLAP) plays a role in releasing amino acids from host hemoglobin that are used for parasite protein synthesis, growth, and development. This enzyme represents a target at which new antimalarials could be designed since metalloaminopeptidase inhibitors prevent the growth of the parasites in vitro and in vivo. A study on the metal ion binding characteristics of recombinant P. falciparum M17 leucine aminopeptidase (rPfLAP) shows that the active site of this exopeptidase contains two metal-binding sites, a readily exchangeable site (site 1) and a tight binding site (site 2). The enzyme retains activity when the metal ion is removed from site 1, while removal of metal ions from both sites results in an inactive apoenzyme that cannot be reactivated by the addition of divalent metal cations. The metal ion at site 1 is readily exchangeable with several divalent metal ions and displays a preference in the order of preference Zn(2+) > Mn(2+) > Co(2+) > Mg(2+). While it is likely that native PfLAP contains a Zn(2+) in site 2, the metal ion located in site 1 may be dependent on the type and concentration of metal ions in the cytosolic compartment of the parasite. Importantly, the type of metal ion present at site 1 influences not only the catalytic efficiency of the enzyme for peptide substrates but also the mode of binding by bestatin, a metal-chelating inhibitor of M17 aminopeptidases with antimalarial activity.


Subject(s)
Enzyme Inhibitors/chemistry , Leucyl Aminopeptidase/chemistry , Plasmodium falciparum/enzymology , Animals , Binding Sites , Catalytic Domain , Enzyme Inhibitors/metabolism , Kinetics , Leucyl Aminopeptidase/metabolism , Metals/chemistry , Metals/metabolism , Plasmodium falciparum/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Structure-Activity Relationship , Substrate Specificity
12.
Proc Natl Acad Sci U S A ; 106(8): 2537-42, 2009 Feb 24.
Article in English | MEDLINE | ID: mdl-19196988

ABSTRACT

Plasmodium falciparum parasites are responsible for the major global disease malaria, which results in >2 million deaths each year. With the rise of drug-resistant malarial parasites, novel drug targets and lead compounds are urgently required for the development of new therapeutic strategies. Here, we address this important problem by targeting the malarial neutral aminopeptidases that are involved in the terminal stages of hemoglobin digestion and essential for the provision of amino acids used for parasite growth and development within the erythrocyte. We characterize the structure and substrate specificity of one such aminopeptidase, PfA-M1, a validated drug target. The X-ray crystal structure of PfA-M1 alone and in complex with the generic inhibitor, bestatin, and a phosphinate dipeptide analogue with potent in vitro and in vivo antimalarial activity, hPheP[CH(2)]Phe, reveals features within the protease active site that are critical to its function as an aminopeptidase and can be exploited for drug development. These results set the groundwork for the development of antimalarial therapeutics that target the neutral aminopeptidases of the parasite.


Subject(s)
CD13 Antigens/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Plasmodium falciparum/enzymology , Animals , CD13 Antigens/chemistry , CD13 Antigens/metabolism , Chromatography, High Pressure Liquid , Crystallography, X-Ray , Drug Design , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/chemistry , Models, Molecular , Protein Conformation , Substrate Specificity
13.
PLoS Negl Trop Dis ; 3(1): e369, 2009.
Article in English | MEDLINE | ID: mdl-19172172

ABSTRACT

The helminth parasite Fasciola hepatica secretes cathepsin L cysteine proteases to invade its host, migrate through tissues and digest haemoglobin, its main source of amino acids. Here we investigated the importance of pH in regulating the activity and functions of the major cathepsin L protease FheCL1. The slightly acidic pH of the parasite gut facilitates the auto-catalytic activation of FheCL1 from its inactive proFheCL1 zymogen; this process was approximately 40-fold faster at pH 4.5 than at pH 7.0. Active mature FheCL1 is very stable at acidic and neutral conditions (the enzyme retained approximately 45% activity when incubated at 37 degrees C and pH 4.5 for 10 days) and displayed a broad pH range for activity peptide substrates and the protein ovalbumin, peaking between pH 5.5 and pH 7.0. This pH profile likely reflects the need for FheCL1 to function both in the parasite gut and in the host tissues. FheCL1, however, could not cleave its natural substrate Hb in the pH range pH 5.5 and pH 7.0; digestion occurred only at pH

Subject(s)
Cathepsins/metabolism , Fasciola hepatica/enzymology , Hemoglobins/metabolism , Animals , Hydrogen-Ion Concentration
14.
FASEB J ; 22(11): 4022-32, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18708590

ABSTRACT

During helminth infections, alternatively activated macrophages (AAMacs) are key to promoting Th2 responses and suppressing Th1-driven inflammatory pathology. Th2 cytokines IL-4 and/or IL-13 are believed to be important in the induction and activation of AAMacs. Using murine models for the helminth infections caused by Fasciola hepatica (Fh) and Schistosoma mansoni (Sm), we show that a secreted antioxidant, peroxiredoxin (Prx), induces alternative activation of macrophages. These activated, Ym1-expressing macrophages enhanced the secretion of IL-4, IL-5, and IL-13 from naive CD4(+) T cells. Administration of recombinant FhPrx and SmPrx to wild-type and IL-4(-/-) and IL-13(-/-) mice induced the production of AAMacs. In addition, Prx stimulated the expression of markers of AAMacs (particularly, Ym1) in vitro, and therefore can act independently of IL-4/IL-13 signaling. The immunomodulatory property of Prx is not due to its antioxidant activity, as an inactive recombinant variant with active site Cys residues replaced by Gly could also induce AAMacs and Th2 responses. Immunization of mice with recombinant Prx or passive transfer of anti-Prx antibodies prior to infection with Fh not only blocked the induction of AAMacs but also the development of parasite-specific Th2 responses. We propose that Prx activates macrophages as an initial step in the induction of Th2 responses by helminth parasites and is thereby a novel pathogen-associated molecular pattern.


Subject(s)
Fasciola hepatica/immunology , Fascioliasis/immunology , Helminth Proteins/immunology , Macrophage Activation/immunology , Macrophages/immunology , Peroxiredoxins/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Th2 Cells/immunology , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/immunology , Cells, Cultured , Disease Models, Animal , Fasciola hepatica/enzymology , Fasciola hepatica/genetics , Fascioliasis/enzymology , Fascioliasis/genetics , Female , Helminth Proteins/genetics , Helminth Proteins/metabolism , Immunization , Immunization, Passive , Interleukin-13/biosynthesis , Interleukin-13/immunology , Interleukin-4/biosynthesis , Interleukin-4/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Schistosoma mansoni/enzymology , Schistosoma mansoni/genetics , Schistosomiasis mansoni/enzymology , Schistosomiasis mansoni/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Th2 Cells/metabolism
15.
Mol Cell Proteomics ; 7(6): 1111-23, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18296439

ABSTRACT

Cathepsin L proteases secreted by the helminth pathogen Fasciola hepatica have functions in parasite virulence including tissue invasion and suppression of host immune responses. Using proteomics methods alongside phylogenetic studies we characterized the profile of cathepsin L proteases secreted by adult F. hepatica and hence identified those involved in host-pathogen interaction. Phylogenetic analyses showed that the Fasciola cathepsin L gene family expanded by a series of gene duplications followed by divergence that gave rise to three clades associated with mature adult worms (Clades 1, 2, and 5) and two clades specific to infective juvenile stages (Clades 3 and 4). Consistent with these observations our proteomics studies identified representatives from Clades 1, 2, and 5 but not from Clades 3 and 4 in adult F. hepatica secretory products. Clades 1 and 2 account for 67.39 and 27.63% of total secreted cathepsin Ls, respectively, suggesting that their expansion was positively driven and that these proteases are most critical for parasite survival and adaptation. Sequence comparison studies revealed that the expansion of cathepsin Ls by gene duplication was followed by residue changes in the S2 pocket of the active site. Our biochemical studies showed that these changes result in alterations in substrate binding and suggested that the divergence of the cathepsin L family produced a repertoire of enzymes with overlapping and complementary substrate specificities that could cleave host macromolecules more efficiently. Although the cathepsin Ls are produced as zymogens containing a prosegment and mature domain, all secreted enzymes identified by MS were processed to mature active enzymes. The prosegment region was highly conserved between the clades except at the boundary of prosegment and mature enzyme. Despite the lack of conservation at this section, sites for exogenous cleavage by asparaginyl endopeptidases and a Leu-Ser[downward arrow]His motif for autocatalytic cleavage by cathepsin Ls were preserved.


Subject(s)
Cathepsins/metabolism , Cysteine Endopeptidases/metabolism , Fasciola hepatica/enzymology , Gene Expression Regulation , Proteomics/methods , Amino Acid Sequence , Animals , Binding Sites , Cathepsin L , Databases, Factual , Endopeptidases/chemistry , Mass Spectrometry/methods , Molecular Sequence Data , Phylogeny , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Virulence Factors/metabolism
16.
J Biol Chem ; 283(15): 9896-908, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18160404

ABSTRACT

The helminth parasite Fasciola hepatica secretes cysteine proteases to facilitate tissue invasion, migration, and development within the mammalian host. The major proteases cathepsin L1 (FheCL1) and cathepsin L2 (FheCL2) were recombinantly produced and biochemically characterized. By using site-directed mutagenesis, we show that residues at position 67 and 205, which lie within the S2 pocket of the active site, are critical in determining the substrate and inhibitor specificity. FheCL1 exhibits a broader specificity and a higher substrate turnover rate compared with FheCL2. However, FheCL2 can efficiently cleave substrates with a Pro in the P2 position and degrade collagen within the triple helices at physiological pH, an activity that among cysteine proteases has only been reported for human cathepsin K. The 1.4-A three-dimensional structure of the FheCL1 was determined by x-ray crystallography, and the three-dimensional structure of FheCL2 was constructed via homology-based modeling. Analysis and comparison of these structures and our biochemical data with those of human cathepsins L and K provided an interpretation of the substrate-recognition mechanisms of these major parasite proteases. Furthermore, our studies suggest that a configuration involving residue 67 and the "gatekeeper" residues 157 and 158 situated at the entrance of the active site pocket create a topology that endows FheCL2 with its unusual collagenolytic activity. The emergence of a specialized collagenolytic function in Fasciola likely contributes to the success of this tissue-invasive parasite.


Subject(s)
Fasciola hepatica/enzymology , Helminth Proteins/chemistry , Models, Molecular , Virulence Factors/chemistry , Animals , Binding Sites/physiology , Cathepsins , Crystallography, X-Ray , Fasciola hepatica/genetics , Fasciola hepatica/pathogenicity , Helminth Proteins/genetics , Humans , Hydrogen-Ion Concentration , Protein Structure, Secondary/physiology , Protein Structure, Tertiary/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sequence Homology, Amino Acid , Structural Homology, Protein , Structure-Activity Relationship , Substrate Specificity/physiology , Virulence Factors/genetics
17.
J Med Chem ; 50(24): 6024-31, 2007 Nov 29.
Article in English | MEDLINE | ID: mdl-17960925

ABSTRACT

Previous studies have pinpointed the M17 leucyl aminopeptidase of Plasmodium falciparum (PfLAP) as a target for the development of new antimalarials. This metallo-exopeptidase functions in the terminal stages of hemoglobin digestion and is inhibited by bestatin, a natural analog of Phe-Leu. By screening novel phosphinate dipeptide analogues for inhibitory activity against recombinant PfLAP, we have discovered two compounds, 4 (hPheP[CH2]Phe) and 5 (hPheP[CH2]Tyr), with inhibitory constants better than bestatin. These compounds are fast, tight-binding inhibitors that make improved contacts within the active site of PfLAP. Both compounds inhibit the growth of P. falciparum in vitro, exhibiting IC50 values against the chloroquine-resistant clone Dd2 of 20-40 and 12-23 muM, respectively. While bestatin exhibited some in vivo activity against Plasmodium chabaudi chabaudi, compound 4 reduced parasite burden by 92%. These studies establish the PfLAP as a prime target for the development of antimalarial drugs and provide important new lead compounds.


Subject(s)
Antimalarials/chemical synthesis , Dipeptides/chemical synthesis , Leucyl Aminopeptidase/antagonists & inhibitors , Phosphinic Acids/chemical synthesis , Plasmodium falciparum/drug effects , Amino Acid Sequence , Animals , Antimalarials/chemistry , Antimalarials/pharmacology , Dipeptides/chemistry , Dipeptides/pharmacology , Kinetics , Leucine/analogs & derivatives , Leucine/chemistry , Leucyl Aminopeptidase/chemistry , Models, Molecular , Molecular Sequence Data , Phosphinic Acids/chemistry , Phosphinic Acids/pharmacology , Plasmodium chabaudi/drug effects , Plasmodium falciparum/enzymology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Structure-Activity Relationship
18.
J Biol Chem ; 282(42): 30817-26, 2007 Oct 19.
Article in English | MEDLINE | ID: mdl-17720817

ABSTRACT

A member of the M18 family of aspartyl aminopeptidases is expressed by all intra-erythrocytic stages of the human malaria parasite Plasmodium falciparum (PfM18AAP), with highest expression levels in rings. Functionally active recombinant enzyme, rPfM18AAP, and native enzyme in cytosolic extracts of malaria parasites are 560-kDa octomers that exhibit optimal activity at neutral pH and require the presence of metal ions to maintain enzymatic activity and stability. Like the human aspartyl aminopeptidase, the exopeptidase activity of PfM18AAP is exclusive to N-terminal acidic amino acids, glutamate and aspartate, making this enzyme of particular interest and suggesting that it may function alongside the malaria cytosolic neutral aminopeptidases in the release of amino acids from host hemoglobin-derived peptides. Whereas immunocytochemical studies using transgenic P. falciparum parasites show that PfM18AAP is expressed in the cytosol, immunoblotting experiments revealed that the enzyme is also trafficked out of the parasite into the surrounding parasitophorous vacuole. Antisense-mediated knockdown of PfM18AAP results in a lethal phenotype as a result of significant intracellular damage and validates this enzyme as a target at which novel antimalarial drugs could be directed. Novel phosphinic derivatives of aspartate and glutamate showed modest inhibition of rPfM18AAP but did not inhibit malaria growth in culture. However, we were able to draw valuable observations concerning the structure-activity relationship of these inhibitors that can be employed in future inhibitor optimization studies.


Subject(s)
Antimalarials/chemistry , Enzyme Inhibitors/chemistry , Glutamyl Aminopeptidase/chemistry , Phosphinic Acids/chemistry , Plasmodium falciparum/enzymology , Protozoan Proteins/chemistry , Amino Acids/metabolism , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use , Cytosol/enzymology , DNA, Antisense/genetics , DNA, Antisense/pharmacology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Erythrocytes/enzymology , Erythrocytes/parasitology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Glutamyl Aminopeptidase/antagonists & inhibitors , Glutamyl Aminopeptidase/genetics , Hemoglobins/metabolism , Humans , Hydrogen-Ion Concentration , Metals/chemistry , Metals/metabolism , Peptides/metabolism , Phenotype , Phosphinic Acids/pharmacology , Phosphinic Acids/therapeutic use , Plasmodium falciparum/genetics , Protein Transport/drug effects , Protein Transport/genetics , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Structure-Activity Relationship , Substrate Specificity/drug effects , Substrate Specificity/genetics , Vacuoles/enzymology , Vacuoles/parasitology
19.
J Biol Chem ; 282(22): 16532-43, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17403677

ABSTRACT

A protease secreted by the parasitic helminth Fasciola hepatica, a 37-kDa procathepsin L1 (FheproCL1), autocatalytically processes and activates to its mature enzyme (FheCL1) over a wide pH range of 7.3 to 4.0, although activation is more rapid at low pH. Maturation initiates with cleavages of a small proportion of molecules within the central region of the prosegment, possibly by intramolecular events. However, activation to fully mature enzymes is achieved by a precise intermolecular cleavage at a Leu-12-Ser-11 downward arrowHis-10 sequence within the nonconserved C-terminal region of the prosegment. The importance of this cleavage site in enzyme activation was demonstrated using an active site variant FheproCL1Gly26 (Cys26 to Gly26) and a double variant FheproCL1Pro-12/Gly26 (Leu-12 to Pro-12), and although both of these variants cannot autocatalytically process, the former is susceptible to trans-processing at a Leu-12-Ser-11 downward arrowHis-10 sequence by pre-activated FheCL1, but the latter is not. Another F. hepatica secreted protease FheCL2, which, unlike FheCL1, can readily accept proline in the S2 subsite of its active site, can trans-process the double variant FheproCL1Pro-12/Gly26 by cleavage at the Pro-12-Ser-11 downward arrowHis-10 sequence. Furthermore, the autoactivation of a variant enzyme with a single replacement, FheproCL1Pro-12, was very slow but was increased 40-fold in the presence of FheCL2. These studies provide a molecular insight into the regulation of FheproCL1 autocatalysis.


Subject(s)
Cathepsins/chemistry , Dicrocoelium/enzymology , Enzyme Precursors/chemistry , Helminth Proteins/chemistry , Protein Processing, Post-Translational , Amino Acid Substitution , Animals , Catalysis , Cathepsins/genetics , Enzyme Activation/genetics , Enzyme Precursors/genetics , Helminth Proteins/genetics , Hydrogen-Ion Concentration , Leucine/chemistry , Leucine/genetics , Mutation, Missense , Proline/chemistry , Proline/genetics , Protein Processing, Post-Translational/genetics
20.
J Biol Chem ; 282(3): 2069-80, 2007 Jan 19.
Article in English | MEDLINE | ID: mdl-17107951

ABSTRACT

Amino acids generated from the catabolism of hemoglobin by intra-erythrocytic malaria parasites are not only essential for protein synthesis but also function in maintaining an osmotically stable environment, and creating a gradient by which amino acids that are rare or not present in hemoglobin are drawn into the parasite from host serum. We have proposed that a Plasmodium falciparum M17 leucyl aminopeptidase (PfLAP) generates and regulates the internal pool of free amino acids and therefore represents a target for novel antimalarial drugs. This enzyme has been expressed in insect cells as a functional 320-kDa homo-hexamer that is optimally active at neutral or alkaline pH, is dependent on metal ions for activity, and exhibits a substrate preference for N-terminally exposed hydrophobic amino acids, particularly leucine. PfLAP is produced by all stages in the intra-erythrocytic developmental cycle of malaria but was most highly expressed by trophozoites, a stage at which hemoglobin degradation and parasite protein synthesis are elevated. The enzyme was located by immunohistochemical methods and by transfecting malaria cells with a PfLAP-green fluorescent protein construct, to the cytosolic compartment of the cell at all developmental stages, including segregated merozoites. Amino acid dipeptide analogs, such as bestatin and its derivatives, are potent inhibitors of the protease and also block the growth of P. falciparum malaria parasites in culture. This study provides a biochemical basis for the antimalarial activity of aminopeptidase inhibitors. Availability of functionally active recombinant PfLAP, coupled with a simple enzymatic readout, will aid medicinal chemistry and/or high throughput approaches for the future design/discovery of new antimalarial drugs.


Subject(s)
Amino Acids/chemistry , Antimalarials/pharmacology , Leucyl Aminopeptidase/physiology , Plasmodium falciparum/enzymology , Animals , Cytosol/metabolism , Hydrogen-Ion Concentration , Immunohistochemistry , Ions , Kinetics , Leucine/chemistry , Leucyl Aminopeptidase/chemistry , Merozoites/chemistry , Phylogeny , Plasmids/metabolism , Recombinant Proteins/chemistry , Trophozoites/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...