Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 133
Filter
Add more filters










Publication year range
1.
Mol Biochem Parasitol ; 259: 111628, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38719028

ABSTRACT

Apicomplexan parasites are the primary causative agents of many human diseases, including malaria, toxoplasmosis, and cryptosporidiosis. These opportunistic pathogens undergo complex life cycles with multiple developmental stages, wherein many key steps are regulated by phosphorylation mechanisms. The genomes of apicomplexan pathogens contain protein kinases from different groups including tyrosine kinase-like (TKL) family proteins. Although information on the role of TKL kinases in apicomplexans is quite limited, recent studies have revealed the important role of this family of proteins in apicomplexan biology. TKL kinases in these protozoan pathogens show unique organization with many novel domains thus making them attractive candidates for drug development. In this mini review, we summarize the current understanding of the role of TKL kinases in human apicomplexan pathogens' (Toxoplasma gondii, Plasmodium falciparum and Cryptosporidium parvum) biology and pathogenesis.


Subject(s)
Apicomplexa , Cryptosporidium parvum , Plasmodium falciparum , Protozoan Proteins , Toxoplasma , Humans , Toxoplasma/enzymology , Toxoplasma/genetics , Cryptosporidium parvum/enzymology , Cryptosporidium parvum/genetics , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Plasmodium falciparum/enzymology , Plasmodium falciparum/genetics , Apicomplexa/enzymology , Apicomplexa/genetics , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/chemistry , Phosphorylation
2.
Cell Rep ; 43(6): 114263, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38814783

ABSTRACT

The protozoan parasite Cryptosporidium is a leading cause of diarrhea in young children. The parasite's life cycle involves a coordinated and timely progression from asexual to sexual stages, leading to the formation of the transmissible oocyst. Underlying molecular signaling mechanisms orchestrating sexual development are not known. Here, we describe the function of a signaling kinase in Cryptosporidium male gametogenesis. We reveal the expression of Cryptosporidium parvum calcium-dependent protein kinase 5 (CDPK5) during male gamete development and its important role in the egress of mature gametes. Genetic ablation of this kinase results in viable parasites, indicating that this gene is dispensable for parasite survival. Interestingly, cdpk5 deletion decreases parasite virulence and impacts oocyst shedding in immunocompromised mice. Using phosphoproteomics, we identify possible CDPK5 substrates and biological processes regulated by this kinase. Collectively, these findings illuminate parasite cell biology by revealing a mechanism controlling male gamete production and a potential target to block disease transmission.


Subject(s)
Gametogenesis , Protozoan Proteins , Animals , Male , Mice , Virulence , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Cryptosporidium parvum/pathogenicity , Cryptosporidium parvum/enzymology , Protein Kinases/metabolism , Protein Kinases/genetics , Cryptosporidiosis/parasitology , Humans , Signal Transduction
3.
Bioorg Med Chem ; 50: 116458, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34687983

ABSTRACT

Parasitic diseases remain a major public health concern for humans, claiming millions of lives annually. Although different treatments are required for these diseases, drug usage is limited due to the development of resistance and toxicity, which necessitate alternative therapies. It has been shown in the literature that parasitic lactate dehydrogenases (LDH) and malate dehydrogenases (MDH) have unique pharmacological selective and specificity properties compared to other isoforms, thus highlighting them as viable therapeutic targets involved in aerobic and anaerobic glycolytic pathways. LDH and MDH are important therapeutic targets for invasive parasites because they play a critical role in the progression and development of parasitic diseases. Any strategy to impede these enzymes would be fatal to the parasites, paving the way to develop and discover novel antiparasitic agents. This review aims to highlight the importance of parasitic LDH and MDH as therapeutic drug targets in selected obligate apicoplast parasites. To the best of our knowledge, this review presents the first comprehensive review of LDH and MDH as potential antiparasitic targets for drug development studies.


Subject(s)
Antiparasitic Agents/pharmacology , Drug Development , L-Lactate Dehydrogenase/antagonists & inhibitors , Malate Dehydrogenase/antagonists & inhibitors , Animals , Antiparasitic Agents/chemical synthesis , Antiparasitic Agents/chemistry , Cryptosporidium parvum/drug effects , Cryptosporidium parvum/enzymology , Humans , L-Lactate Dehydrogenase/metabolism , Malate Dehydrogenase/metabolism , Molecular Structure , Parasitic Sensitivity Tests , Plasmodium/drug effects , Plasmodium/enzymology , Schistosoma/drug effects , Schistosoma/enzymology , Toxoplasma/drug effects , Toxoplasma/enzymology , Trichomonas vaginalis/drug effects , Trichomonas vaginalis/enzymology
4.
mBio ; 12(2)2021 03 09.
Article in English | MEDLINE | ID: mdl-33688009

ABSTRACT

The apicomplexan parasite Cryptosporidium parvum contains an expanded family of 22 insulinase-like proteases (INS), a feature that contrasts with their otherwise streamlined genome. Here, we examined the function of INS1, which is most similar to the human insulinase protease that cleaves a variety of small peptide substrates. INS1 is an M16A clan member and contains a signal peptide, an N-terminal domain with the HXXEH active site, followed by three inactive domains. Unlike previously studied C. parvum INS proteins that are expressed in sporozoites and during merogony, INS1 was expressed exclusively in macrogamonts, where it was localized in small cytoplasmic vesicles. Although INS1 did not colocalize with the oocyst wall protein recognized by the antibody OW50, immune-electron microscopy indicated that INS1 resides in small vesicles in the secretory system. Notably, these small INS1-positive vesicles were often in close proximity to large OW50-positive vacuoles resembling wall-forming bodies, which contain precursors for oocyst wall formation. Genetic deletion of INS1, or replacement with an active-site mutant, resulted in lower formation of macrogamonts in vitro and reduced oocyst shedding in vivo Our findings reveal that INS1 functions in the formation or maturation of macrogamonts and that its loss results in attenuated virulence in immunocompromised mice.IMPORTANCE Cryptosporidiosis is a debilitating diarrheal disease in young children in developing countries. The absence of effective treatments or vaccines makes this infection very difficult to manage in susceptible populations. Although the oral dose of oocysts needed to cause infection is low, infected individuals shed very high numbers of oocysts, readily contaminating the environment. Our studies demonstrate that the protease INS1 is important for formation of female sexual stages and that in its absence, parasites produce fewer oocysts and are attenuated in immunocompromised mice. These findings suggest that mutants lacking INS1, or related proteases, are useful for further characterizing the pathway that leads to macrogamont maturation and oocyst wall formation.


Subject(s)
Cryptosporidium parvum/enzymology , Cryptosporidium parvum/physiology , Insulysin/genetics , Insulysin/metabolism , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Animals , Cryptosporidiosis/parasitology , Cryptosporidium parvum/growth & development , Cryptosporidium parvum/ultrastructure , Feces/parasitology , Female , Gene Deletion , Immunocompromised Host , Life Cycle Stages/genetics , Life Cycle Stages/physiology , Mice , Mice, SCID , Microscopy, Electron , Oocysts/physiology , Oocysts/ultrastructure , Receptors, Interferon/genetics , Vacuoles/parasitology , Vacuoles/ultrastructure , Interferon gamma Receptor
5.
J Mol Model ; 27(2): 35, 2021 Jan 09.
Article in English | MEDLINE | ID: mdl-33423140

ABSTRACT

Compound P131 has been established to inhibit Cryptosporidium parvum's inosine monophosphate dehydrogenase (CpIMPDH). Its inhibitory activity supersedes that of paromomycin, which is extensively used in treating cryptosporidiosis. Through the per-residue energy decomposition approach, crucial moieties of P131 were identified and subsequently adopted to create a pharmacophore model for virtual screening in the ZINC database. This search generated eight ADMET-compliant hits that were examined thoroughly to fit into the active site of CpIMPDH via molecular docking. Three compounds ZINC46542062, ZINC58646829, and ZINC89780094, with favorable docking scores of - 8.3 kcal/mol, - 8.2 kcal/mol, and - 7.5 kcal/mol, were selected. The potential inhibitory mechanism of these compounds was probed using molecular dynamics simulation and Molecular Mechanics Generalized Poisson Boltzmann Surface Area (MM/PBSA) analyses. Results revealed that one of the hits (ZINC46542062) exhibited a lower binding free energy of - 39.52 kcal/mol than P131, which had - 34.6 kcal/mol. Conformational perturbation induced by the binding of the identified hits to CpIMPDH was similar to P131, suggesting a similarity in inhibitory mechanisms. Also, in silico investigation of the properties of the hit compounds implied superior physicochemical properties with regards to their synthetic accessibility, lipophilicity, and number of hydrogen bond donors and acceptors in comparison with P131. ZINC46542062 was identified as a promising hit compound with the highest binding affinity to the target protein and favorable physicochemical and pharmacokinetic properties relative to P131. The identified compounds can serve as a basis for conducting further experimental investigations toward the development of anticryptosporidials, which can overcome the challenges of existing therapeutic options. Graphical abstract P131 and the identified compounds docked in the NAD+ binding site of Cryptosporidium parvum IMPDH.


Subject(s)
Cryptosporidium parvum/enzymology , Enzyme Inhibitors/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , IMP Dehydrogenase/metabolism , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Thermodynamics
6.
mBio ; 11(5)2020 10 20.
Article in English | MEDLINE | ID: mdl-33082260

ABSTRACT

UDP-N-acetylglucosamine (UDP-GlcNAc), the main product of the hexosamine biosynthetic pathway, is an important metabolite in protozoan parasites since its sugar moiety is incorporated into glycosylphosphatidylinositol (GPI) glycolipids and N- and O-linked glycans. Apicomplexan parasites have a hexosamine pathway comparable to other eukaryotic organisms, with the exception of the glucosamine-phosphate N-acetyltransferase (GNA1) enzymatic step that has an independent evolutionary origin and significant differences from nonapicomplexan GNA1s. By using conditional genetic engineering, we demonstrate the requirement of GNA1 for the generation of a pool of UDP-GlcNAc and for the development of intraerythrocytic asexual Plasmodium falciparum parasites. Furthermore, we present the 1.95 Å resolution structure of the GNA1 ortholog from Cryptosporidium parvum, an apicomplexan parasite which is a leading cause of diarrhea in developing countries, as a surrogate for P. falciparum GNA1. The in-depth analysis of the crystal shows the presence of specific residues relevant for GNA1 enzymatic activity that are further investigated by the creation of site-specific mutants. The experiments reveal distinct features in apicomplexan GNA1 enzymes that could be exploitable for the generation of selective inhibitors against these parasites, by targeting the hexosamine pathway. This work underscores the potential of apicomplexan GNA1 as a drug target against malaria.IMPORTANCE Apicomplexan parasites cause a major burden on global health and economy. The absence of treatments, the emergence of resistances against available therapies, and the parasite's ability to manipulate host cells and evade immune systems highlight the urgent need to characterize new drug targets to treat infections caused by these parasites. We demonstrate that glucosamine-6-phosphate N-acetyltransferase (GNA1), required for the biosynthesis of UDP-N-acetylglucosamine (UDP-GlcNAc), is essential for P. falciparum asexual blood stage development and that the disruption of the gene encoding this enzyme quickly causes the death of the parasite within a life cycle. The high-resolution crystal structure of the GNA1 ortholog from the apicomplexan parasite C. parvum, used here as a surrogate, highlights significant differences from human GNA1. These divergences can be exploited for the design of specific inhibitors against the malaria parasite.


Subject(s)
Erythrocytes/parasitology , Glucosamine 6-Phosphate N-Acetyltransferase/metabolism , Plasmodium falciparum/enzymology , Plasmodium falciparum/growth & development , Amino Acid Sequence , Binding Sites , Biosynthetic Pathways , Cryptosporidium parvum/enzymology , Cryptosporidium parvum/genetics , Crystallography, X-Ray , Genetic Engineering , Glucosamine 6-Phosphate N-Acetyltransferase/genetics , Humans , Malaria, Falciparum/parasitology , Plasmodium falciparum/genetics
7.
Bioorg Med Chem Lett ; 30(24): 127543, 2020 12 15.
Article in English | MEDLINE | ID: mdl-32931912

ABSTRACT

Inosine-5'-monophosphate dehydrogenase (IMPDH) is a potential target for microorganisms. However, identifying inhibitor design determinants for IMPDH orthologs continues to evolve. Herein, a series of mycophenolic anilide inhibitors of Cryptosporidium parvum and human IMPDHs are reported. Furthermore, molecular docking of 12 (e.g. SH-19; CpIMPDH Ki,app = 0.042 ± 0.015 µM, HsIMPDH2 Ki,app = 0.13 ± 0.05 µM) supports different binding modes with the two enzymes. For CpIMPDH the inhibitor extends into a pocket in an adjacent subunit. In contrast, docking suggests the inhibitor interacts with Ser276 in the NAD binding site in HsIMPDH2, as well as an adjacent pocket within the same subunit. These results provide further guidance for generating IMPDH inhibitors for enzymes found in an array of pathogenic microorganisms, including Mycobacterium tuberculosis.


Subject(s)
Anilides/pharmacology , Antiparasitic Agents/pharmacology , Cryptosporidium parvum/enzymology , Enzyme Inhibitors/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , Anilides/chemistry , Antiparasitic Agents/chemistry , Binding Sites/drug effects , Cryptosporidiosis/drug therapy , Cryptosporidiosis/parasitology , Cryptosporidium parvum/metabolism , Enzyme Inhibitors/chemistry , Humans , IMP Dehydrogenase/metabolism , Molecular Docking Simulation , Phenols/chemistry , Phenols/pharmacology
8.
Acta Parasitol ; 65(3): 704-715, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32347536

ABSTRACT

PURPOSE: Cryptosporidium parvum is an Apicomplexa parasite that causes watery diarrhea (cryptosporidiosis), especially in children and immunocompromised adults (the latter in a very severe form). No effective treatment exists against infection by this parasite. Phosphatases participate in the regulation of various cellular functions and are thus considered potential therapeutic targets in many diseases. The aim of the present study was to indirectly identify and in silico characterize a protein phosphatase 2C of C. parvum. METHODS: Western blot and indirect immunofluorescence microscopy were performed with a polyclonal antibody against Leishmania major PP2C. Possible cross-reactivity with LmPP2C was assessed by in silico sequence homology to analyze phylogenetic relationships between distinct C. parvum PP2Cs. In addition, another bioinformatics approach was used to predict the possible relationship and function of C. parvum PP2C in the regulation of several cellular processes. RESULTS: Western blotting showed a protein of approximately 72 kDa. With immunofluorescence, PP2C was localized in the nucleus of oocysts (with some additional labeling in the cytoplasm) and at the apical region of sporozoites. By aligning C. parvum PP2C with known ortholog sequences and carrying out PPI analysis, a determination could be made of the degree of conservation of these enzymes, their possible relationship, and their function in the regulation of several cellular processes associated with a likely nuclear location. CONCLUSION: Microscopic localization by immunofluorescence identified CpPP2C at the nucleus in oocysts and at the apical end of the sporozoite body. Hence, this enzyme could be associated with proteins that have an important role in the regulation of transcription and other processes orchestrated by MAPK kinases, according to in silico analysis.


Subject(s)
Cryptosporidium parvum/enzymology , Phylogeny , Protein Phosphatase 2C/chemistry , Protein Phosphatase 2C/genetics , Animals , Animals, Newborn/parasitology , Antibodies, Protozoan/immunology , Blotting, Western , Cattle , Cryptosporidiosis/parasitology , Cryptosporidium parvum/genetics , Fluorescent Antibody Technique , Protozoan Proteins/chemistry , Protozoan Proteins/genetics
9.
Mol Biochem Parasitol ; 237: 111277, 2020 05.
Article in English | MEDLINE | ID: mdl-32348840

ABSTRACT

Cryptosporidiosis is an obligate intracellular pathogen causing diarrhea. Merozoite egress is essential for infection to spread between host cells. However, the mechanisms of egress have yet to be defined. We hypothesized that Cyclic GMP-Dependent Protein Kinase G (PKG) may be involved in Cryptosporidium egress. In this study, Cryptosporidium parvum PKG was silenced by using antisense RNA sequences. PKG-silencing significantly inhibited egress of merozoites from infected HCT-8 cells into the supernatant and led to retention of intracellular forms within the host cells. This data identifies PKG as a key mediator of merozoite egress, a key step in the parasite lifecycle.


Subject(s)
Cryptosporidium parvum/genetics , Cyclic GMP-Dependent Protein Kinases/genetics , Host-Parasite Interactions/genetics , Merozoites/genetics , Protozoan Proteins/genetics , Cell Line , Cryptosporidium parvum/enzymology , Cryptosporidium parvum/growth & development , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/metabolism , Epithelial Cells/parasitology , Gene Expression , Gene Silencing , Humans , Merozoites/enzymology , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
10.
Parasitol Res ; 118(11): 3159-3171, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31486948

ABSTRACT

Cryptosporidiosis is a significant cause of gastroenteritis in both humans and livestock in developing countries. The only FDA-approved drug available against the same is nitazoxanide, with questionable efficacy in malnourished children and immunocompromised patients. Recent in vitro studies have indicated the viability of Triacsin C as a potential drug candidate, which targets the parasite's long-chain fatty acyl coenzyme A synthetase enzyme (LC-FACS), a critical component of the fatty acid metabolism pathway. We have used this molecule as a baseline to propose more potent versions thereof. We have applied a combined approach of substructure replacement, literature search, and database screening to come up with 514 analogs of Triacsin C. A virtual screening protocol was carried out which lead us to identify a potential hit compound. This was further subjected to a 100-ns molecular dynamics simulation in complex to determine its stability and binding characteristics. After which, the ADME/tox properties were predicted to assess its viability as a drug. The molecule R134 was identified as the best hit due to its highest average binding affinity, stability in complex when subjected to MD simulations, and reasonable predicted ADMET (Absorption, Distribution, Metabolism, Excretion and Toxicity) properties comparable to those of the Triacsin C parent molecule. We have proposed R134 as a putative drug candidate against the Cryptosporidium parvum LC-FACS enzyme isoforms, following an in silico protocol. We hope the results will be helpful when planning future in vitro experiments for identifying drugs against Cryptosporidium.


Subject(s)
Antiprotozoal Agents/pharmacology , Coenzyme A Ligases/antagonists & inhibitors , Cryptosporidium parvum/enzymology , Cryptosporidium parvum/metabolism , Drug Discovery/methods , Triazenes/pharmacology , Acyl Coenzyme A/metabolism , Animals , Antiprotozoal Agents/chemistry , Child , Cryptosporidiosis/drug therapy , Cryptosporidiosis/parasitology , Fatty Acids/metabolism , Gastroenteritis/parasitology , Humans , Molecular Dynamics Simulation , Protein Isoforms , Triazenes/chemistry
11.
J Antibiot (Tokyo) ; 72(12): 934-942, 2019 12.
Article in English | MEDLINE | ID: mdl-31296916

ABSTRACT

We have found cyclophane-type adenosine derivatives having p-quinone amide moieties (1 and 2) as weak inhibitors of Cryptosporidium parvum inosine 5'-monophosphate dehydrogenase (CpIMPDH) from the Hokkaido University Chemical Library via the luciferase-based high-throughput screening. To obtain more potent inhibitors, we synthesized four new derivatives free from cyclophane rings (3-6). The N-H derivatives 3 and 5 showed more potent activities (24.4 and 11.1 µM, respectively) in the presence of dithiothreitol (DTT), whereas the N-methyl derivative 4 indicated more potent activity (2.1 µM) without DTT. Conformational analysis of compounds 3 and 4 suggested that N-H amide 3 binds to IMP-binding site in the DTT mediated manner.


Subject(s)
Cryptosporidium parvum/enzymology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , Antiparasitic Agents/chemistry , Antiparasitic Agents/pharmacology , Binding Sites , Dithiothreitol/chemistry , Enzyme Inhibitors/chemical synthesis , High-Throughput Screening Assays , Humans , IMP Dehydrogenase/genetics , IMP Dehydrogenase/metabolism , Structure-Activity Relationship
12.
PLoS Pathog ; 15(7): e1007953, 2019 07.
Article in English | MEDLINE | ID: mdl-31356619

ABSTRACT

Cryptosporidium parvum is a highly prevalent zoonotic and anthroponotic protozoan parasite that causes a diarrheal syndrome in children and neonatal livestock, culminating in growth retardation and mortalities. Despite the high prevalence of C. parvum, there are no fully effective and safe drugs for treating infections, and there is no vaccine. We have previously reported that the bacterial-like C. parvum lactate dehydrogenase (CpLDH) enzyme is essential for survival, virulence and growth of C. parvum in vitro and in vivo. In the present study, we screened compound libraries and identified inhibitors against the enzymatic activity of recombinant CpLDH protein in vitro. We tested the inhibitors for anti-Cryptosporidium effect using in vitro infection assays of HCT-8 cells monolayers and identified compounds NSC158011 and NSC10447 that inhibited the proliferation of intracellular C. parvum in vitro, with IC50 values of 14.88 and 72.65 µM, respectively. At doses tolerable in mice, we found that both NSC158011 and NSC10447 consistently significantly reduced the shedding of C. parvum oocysts in infected immunocompromised mice's feces, and prevented intestinal villous atrophy as well as mucosal erosion due to C. parvum. Together, our findings have unveiled promising anti-Cryptosporidium drug candidates that can be explored further for the development of the much needed novel therapeutic agents against C. parvum infections.


Subject(s)
Antiprotozoal Agents/pharmacology , Cryptosporidium parvum/drug effects , Cryptosporidium parvum/enzymology , Enzyme Inhibitors/pharmacology , L-Lactate Dehydrogenase/antagonists & inhibitors , Protozoan Proteins/antagonists & inhibitors , Animals , Cell Line , Cryptosporidiosis/drug therapy , Cryptosporidiosis/parasitology , Cryptosporidiosis/pathology , Cryptosporidium parvum/pathogenicity , Host-Parasite Interactions/drug effects , Humans , L-Lactate Dehydrogenase/chemistry , L-Lactate Dehydrogenase/genetics , Mice , Mice, Knockout , Molecular Docking Simulation , Parasitic Sensitivity Tests , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
13.
Article in English | MEDLINE | ID: mdl-31231619

ABSTRACT

Cryptosporidium parvum is a globally important zoonotic parasite capable of causing severe to deadly diarrhea in humans and animals. Its small genome (~9.1 Mb) encodes not only a highly streamlined metabolism, but also a 25-kb, 3-module fatty acid synthase (CpFAS1) and a 40-kb, 7-module polyketide synthase (CpPKS1). The two megasynthases contain a C-terminal reductase domain to release the final products with predicted chain lengths of ≥C22 for CpFAS1 or C28 to C38 for CpPKS1.The parasite genome also encodes a discrete thioesterase ortholog, suggesting its role to be an alternative tool in releasing the final products from CpFAS1 and/or CpPKS1, or as an editor to remove non-reactive residues or aberrant intermediates, or to control starter units as seen in other parasites. In this study, we have confirmed that this C. parvum thioesterase is a type II thioesterase (thus named as CpTEII). CpTEII contains motifs and a catalytic triad characteristic to the type II thioesterase family. CpTEII is expressed during the entire parasite life cycle stages with the highest levels of expression in the later developmental stages. CpTEII showed the highest hydrolytic activity toward C10:0 decanoyl-CoA, so we speculated that CpTEII may mainly act as an editor to remove non-reactive residues and/or aberrant medium acyl chain from CpFAS1 and/or CpPKS1. However, we cannot rule out the possibility that CpTEII may also participate in the release of final products from CpFAS1 because of its moderate activity on C20:0, C:22:0 and C24:0 acyl-CoA thioesters (i.e., ~20-30% activity vs. decanoyl-CoA).


Subject(s)
Cryptosporidium parvum/genetics , Cryptosporidium parvum/metabolism , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Thiolester Hydrolases/genetics , Thiolester Hydrolases/metabolism , Acyl Coenzyme A , Animals , Cloning, Molecular , Cryptosporidium parvum/enzymology , Gene Expression Regulation , Genome, Protozoan , Oxidoreductases/metabolism , Polyketide Synthases/metabolism , Recombinant Proteins , Zoonoses/parasitology
14.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 4): 290-298, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30950830

ABSTRACT

S-Adenosyl-L-methionine (AdoMet), the primary methyl donor in most biological methylation reactions, is produced from ATP and methionine in a multistep reaction catalyzed by AdoMet synthetase. The diversity of group-transfer reactions that involve AdoMet places this compound at a key crossroads in amino-acid, nucleic acid and lipid metabolism, and disruption of its synthesis has adverse consequences for all forms of life. The family of AdoMet synthetases is highly conserved, and structures of this enzyme have been determined from organisms ranging from bacteria to humans. Here, the structure of an AdoMet synthetase from the infectious parasite Cryptosporidium parvum has been determined as part of an effort to identify structural differences in this enzyme family that can guide the development of species-selective inhibitors. This enzyme form has a less extensive subunit interface than some previously determined structures, and contains some key structural differences from the human enzyme in an allosteric site, presenting an opportunity for the design of selective inhibitors against the AdoMet synthetase from this organism.


Subject(s)
Cryptosporidium parvum/enzymology , Methionine Adenosyltransferase/chemistry , Allosteric Regulation , Amino Acid Sequence , Crystallization , Humans , Models, Molecular , Protein Multimerization , Sequence Homology, Amino Acid , Structural Homology, Protein
15.
Proteins ; 87(9): 730-737, 2019 09.
Article in English | MEDLINE | ID: mdl-31017332

ABSTRACT

Cladosporin (CLD) is a fungal metabolite that kills the malaria parasite via inhibiting its cytoplasmic lysyl-tRNA synthetase (KRS) and abrogating protein translation. Here we provide structural and drug selectivity analyses on CLD interacting residues in apo and holo KRSs from Plasmodium falciparum, Homo sapiens, Cryptosporidium parvum, and Mycobacterium ulcerans. We show that both gross and subtle alterations in protein backbone and sidechains drive the active site structural plasticity that allows integration of CLD in KRSs. The ligand-induced fit of CLD in PfKRS is marked by closure and stabilization of three disordered loops and one alpha helix. However, these structural rearragements are not evident in KRS-CLD complexes from H. sapiens, C. parvum, or M. ulcerans. Strikingly, CLD fits into the MuKRS active site due to a remarkable rotameric alteration in its clash-prone methionine residue that provides accommodation for the methyl moiety in CLD. Although the high concentrations of drugs used for Hs, Cp, and MuKRS-CLD complexes in co-crystallization studies enable elucidation of a structural framework for understanding drug binding in KRSs, we propose that these data should be concurrently assessed via biochemical studies of potency and drug selectivity given the poor cell-based activity of CLD against human and bacterial cells. Our comprehensive analyses of KRS-CLD interactions, therefore, highlight vital issues in structure-based drug discovery studies.


Subject(s)
Isocoumarins/metabolism , Lysine-tRNA Ligase/metabolism , Plasmodium falciparum/enzymology , Cryptosporidium parvum/enzymology , Isocoumarins/chemistry , Lysine-tRNA Ligase/chemistry , Mycobacterium ulcerans/enzymology , Protein Binding
16.
Proc Natl Acad Sci U S A ; 116(14): 7015-7020, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30894487

ABSTRACT

Malaria and cryptosporidiosis, caused by apicomplexan parasites, remain major drivers of global child mortality. New drugs for the treatment of malaria and cryptosporidiosis, in particular, are of high priority; however, there are few chemically validated targets. The natural product cladosporin is active against blood- and liver-stage Plasmodium falciparum and Cryptosporidium parvum in cell-culture studies. Target deconvolution in P. falciparum has shown that cladosporin inhibits lysyl-tRNA synthetase (PfKRS1). Here, we report the identification of a series of selective inhibitors of apicomplexan KRSs. Following a biochemical screen, a small-molecule hit was identified and then optimized by using a structure-based approach, supported by structures of both PfKRS1 and C. parvum KRS (CpKRS). In vivo proof of concept was established in an SCID mouse model of malaria, after oral administration (ED90 = 1.5 mg/kg, once a day for 4 d). Furthermore, we successfully identified an opportunity for pathogen hopping based on the structural homology between PfKRS1 and CpKRS. This series of compounds inhibit CpKRS and C. parvum and Cryptosporidium hominis in culture, and our lead compound shows oral efficacy in two cryptosporidiosis mouse models. X-ray crystallography and molecular dynamics simulations have provided a model to rationalize the selectivity of our compounds for PfKRS1 and CpKRS vs. (human) HsKRS. Our work validates apicomplexan KRSs as promising targets for the development of drugs for malaria and cryptosporidiosis.


Subject(s)
Cryptosporidiosis , Cryptosporidium parvum/enzymology , Enzyme Inhibitors/pharmacology , Lysine-tRNA Ligase/antagonists & inhibitors , Malaria, Falciparum , Plasmodium falciparum/enzymology , Protozoan Proteins/antagonists & inhibitors , Animals , Cryptosporidiosis/drug therapy , Cryptosporidiosis/enzymology , Disease Models, Animal , Enzyme Inhibitors/chemistry , Humans , Lysine-tRNA Ligase/metabolism , Malaria, Falciparum/drug therapy , Malaria, Falciparum/enzymology , Mice, SCID , Protozoan Proteins/metabolism
17.
J Eukaryot Microbiol ; 66(3): 460-468, 2019 05.
Article in English | MEDLINE | ID: mdl-30222231

ABSTRACT

Cryptosporidium parvum is one of the major species causing mild to severe cryptosporidiosis in humans and animals. We have previously observed that 2-deoxy-d-glucose (2DG) could inhibit both the enzyme activity of C. parvum hexokinase (CpHK) and the parasite growth in vitro. However, the action and fate of 2DG in C. parvum was not fully investigated. In the present study, we showed that, although 2DG could be phosphorylated by CpHK to form 2DG-6-phosphate (2DG6P), the anti-cryptosporidial activity of 2DG was mainly attributed to the action of 2DG on CpHK, rather than the action of 2DG or 2DG6P on the downstream enzyme glucose-6-phosphate isomerase (CpGPI) nor 2DG6P on CpHK. These observations further supported the hypothesis that CpHK could serve as a drug target in the parasite. We also screened 1,200 small molecules consisting of marketed drugs against CpHK, from which four drugs were identified as CpHK inhibitors with micromolar level of anti-cryptospordial activities at concentrations nontoxic to the host cells (i.e. hexachlorphene, thimerosal, alexidine dihydrochloride, and ebselen with EC50  = 0.53, 1.77, 8.1 and 165 µM, respectively). The anti-CpHK activity of the four existing drugs provided us new reagents for studying the enzyme properties of the parasite hexokinase.


Subject(s)
Antiprotozoal Agents/pharmacology , Cryptosporidium parvum/drug effects , Deoxyglucose/pharmacology , Glucose-6-Phosphate/analogs & derivatives , Hexokinase/metabolism , Protozoan Proteins/metabolism , Cryptosporidium parvum/enzymology , Glucose-6-Phosphate/metabolism , Glucose-6-Phosphate Isomerase/metabolism , Phosphorylation
18.
J Enzyme Inhib Med Chem ; 34(1): 171-178, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30451014

ABSTRACT

Inosine 5'-monophosphate dehydrogenase (IMPDH) is an essential enzyme for the production of guanine nucleotides. Disruption of IMPDH activity has been explored as a therapeutic strategy for numerous purposes, such as for anticancer, immunosuppression, antiviral, and antimicrobial therapy. In the present study, we established a luciferase-based high-throughput screening system to identify IMPDH inhibitors from our chemical library of known bioactive small molecules. The screening of 1400 compounds resulted in the discovery of three irreversible inhibitors: disulfiram, bronopol, and ebselen. Each compound has a distinct chemical moiety that differs from other reported IMPDH inhibitors. Further evaluation revealed that these compounds are potent inhibitors of IMPDHs with kon values of 0.7 × 104 to 9.3 × 104 M-1·s-1. Both disulfiram and bronopol exerted similar degree of inhibition to protozoan and mammalian IMPDHs. Ebselen showed an intriguing difference in mode of inhibition for different IMPDHs, with reversible and irreversible inhibition to each Cryptosporidium parvum IMPDH and human IMPDH type II, respectively. In the preliminary efficacy experiment against cryptosporidiosis in severe combined immunodeficiency (SCID) mouse, a decrease in the number of oocyst shed was observed upon the oral administration of disulfiram and bronopol, providing an early clinical proof-of-concept for further utilization of these compounds as IMPDH inhibitors.


Subject(s)
Drug Discovery , Drug Repositioning , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , IMP Dehydrogenase/antagonists & inhibitors , Animals , Azoles/chemistry , Azoles/isolation & purification , Azoles/pharmacology , Cryptosporidium parvum/enzymology , Disulfiram/chemistry , Disulfiram/isolation & purification , Disulfiram/pharmacology , Enzyme Inhibitors/chemistry , Humans , IMP Dehydrogenase/metabolism , Isoindoles , Kinetics , Mice , Mice, SCID , Organoselenium Compounds/chemistry , Organoselenium Compounds/isolation & purification , Organoselenium Compounds/pharmacology , Proof of Concept Study , Propylene Glycols/chemistry , Propylene Glycols/isolation & purification , Propylene Glycols/pharmacology , Small Molecule Libraries
19.
Comput Biol Chem ; 77: 44-51, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30240985

ABSTRACT

Cryptosporidium parvum is a protozoan parasite which causes waterborne diseases known as Cryptosporidiosis. It is an acute enteric diarrheal disease being severe in the case of immunocompromised individuals and children. C. parvum mainly depends on the glycolysis process for energy production and LDH (Lactate Dehydrogenase) is a key controller of this process. In this study from different in-silico approaches such as structure-based, ligand-based and de novo drug design; a total of 40 compounds were selected for docking studies against LDH. The study reported a compound CHEMBL1784973 from Pathogen Box as the best inhibitor in terms of docking score and pharmacophoric features. Furthermore, the binding mode of the best-reported inhibitor was validated through molecular dynamics simulation for a time interval of 70 ns in water environment. The findings resulted in the stable conformation of the inhibitor in the active site of the protein. This study will be helpful for experimental validation.


Subject(s)
Cryptosporidium parvum/enzymology , L-Lactate Dehydrogenase/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Databases, Factual , Drug Design , Drug Evaluation, Preclinical , L-Lactate Dehydrogenase/metabolism , Ligands , Models, Molecular , Molecular Structure , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
20.
Int J Parasitol ; 48(8): 649-656, 2018 07.
Article in English | MEDLINE | ID: mdl-29530646

ABSTRACT

Cryptosporidium is a highly prevalent protozoan parasite that is the second leading cause of childhood morbidity and mortality due to diarrhoea in developing countries, and causes a serious diarrheal syndrome in calves, lambs and goat kids worldwide. Development of fully effective drugs against Cryptosporidium has mainly been hindered by the lack of genetic tools for functional characterization and validation of potential molecular drug targets in the parasite. Herein, we report the development of a morpholino-based in vivo approach for Cryptosporidium parvum gene knockdown to facilitate determination of the physiological roles of the parasite's genes in a murine model. We show that, when administered intraperitoneally at non-toxic doses, morpholinos targeting C. parvum lactate dehydrogenase (CpLDH) and sporozoite 60K protein (Cp15/60) were able to specifically and sustainably down-regulate the expression of CpLDH and Cp15/60 proteins, respectively, in C. parvum-infected interferon-γ knockout mice. Over a period of 6 days of daily administration of target morpholinos, CpLDH and Cp15/60 proteins were down-regulated by 20- to 50-fold, and 10- to 20-fold, respectively. Knockdown of CpLDH resulted in approximately 80% reduction in oocyst load in the feces of mice, and approximately 70% decrease in infectivity of the sporozoites excysted from the shed oocysts. Cp15/60 knockdown did not affect oocyst shedding nor infectivity but, nevertheless, provided a proof-of-principle for the resilience of the morpholino-mediated C. parvum gene knockdown system in vivo. Together, our findings provide a genetic tool for deciphering the physiological roles of C. parvum genes in vivo, and validate CpLDH as an essential gene for the growth and viability of C. parvum in vivo.


Subject(s)
Cryptosporidium parvum/enzymology , L-Lactate Dehydrogenase/metabolism , Morpholinos/metabolism , Oocysts , Animals , Cryptosporidiosis/parasitology , Cryptosporidium parvum/genetics , Cryptosporidium parvum/metabolism , DNA, Protozoan , Down-Regulation , Feces/parasitology , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , Gene Silencing , L-Lactate Dehydrogenase/genetics , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...