Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.611
Filter
1.
Open Biol ; 14(9): 240128, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39255848

ABSTRACT

Cilia are antenna-like organelles protruding from the surface of many cell types in the human body. Defects in ciliary structure or function often lead to diseases that are collectively called ciliopathies. Cilia and flagella-associated protein 410 (CFAP410) localizes at the basal body of cilia/flagella and plays essential roles in ciliogenesis, neuronal development and DNA damage repair. It remains unknown how its specific basal body location is achieved. Multiple single amino acid mutations in CFAP410 have been identified in patients with various ciliopathies. One of the mutations, L224P, is located in the C-terminal domain (CTD) of human CFAP410 and causes severe spondylometaphyseal dysplasia, axial (SMDAX). However, the molecular mechanism for how the mutation causes the disorder remains unclear. Here, we report our structural studies on the CTD of CFAP410 from three distantly related organisms, Homo sapiens, Trypanosoma brucei and Chlamydomonas reinhardtii. The crystal structures reveal that the three proteins all adopt the same conformation as a tetrameric helical bundle. Our work further demonstrates that the tetrameric assembly of the CTD is essential for the correct localization of CFAP410 in T. brucei, as the L224P mutation that disassembles the tetramer disrupts its basal body localization. Taken together, our studies reveal that the basal body localization of CFAP410 is controlled by the CTD and provide a mechanistic explanation for how the mutation L224P in CFAP410 causes ciliopathies in humans.


Subject(s)
Basal Bodies , Trypanosoma brucei brucei , Basal Bodies/metabolism , Humans , Trypanosoma brucei brucei/metabolism , Trypanosoma brucei brucei/genetics , Models, Molecular , Chlamydomonas reinhardtii/genetics , Chlamydomonas reinhardtii/metabolism , Cilia/metabolism , Crystallography, X-Ray , Mutation , Amino Acid Sequence , Protein Multimerization , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Protozoan Proteins/chemistry
2.
Molecules ; 29(17)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39275072

ABSTRACT

Cruzipain (CZP), the major cysteine protease present in T. cruzi, the ethiological agent of Chagas disease, has attracted particular attention as a therapeutic target for the development of targeted covalent inhibitors (TCI). The vast chemical space associated with the enormous molecular diversity feasible to explore by means of modern synthetic approaches allows the design of CZP inhibitors capable of exhibiting not only an efficient enzyme inhibition but also an adequate translation to anti-T. cruzi activity. In this work, a computer-aided design strategy was developed to combinatorially construct and screen large libraries of 1,4-disubstituted 1,2,3-triazole analogues, further identifying a selected set of candidates for advancement towards synthetic and biological activity evaluation stages. In this way, a virtual molecular library comprising more than 75 thousand diverse and synthetically feasible analogues was studied by means of molecular docking and molecular dynamic simulations in the search of potential TCI of CZP, guiding the synthetic efforts towards a subset of 48 candidates. These were synthesized by applying a Cu(I)-catalyzed azide-alkyne cycloaddition (CuAAC) centered synthetic scheme, resulting in moderate to good yields and leading to the identification of 12 hits selectively inhibiting CZP activity with IC50 in the low micromolar range. Furthermore, four triazole derivatives showed good anti-T. cruzi inhibition when studied at 50 µM; and Ald-6 excelled for its high antitrypanocidal activity and low cytotoxicity, exhibiting complete in vitro biological activity translation from CZP to T. cruzi. Overall, not only Ald-6 merits further advancement to preclinical in vivo studies, but these findings also shed light on a valuable chemical space where molecular diversity might be explored in the search for efficient triazole-based antichagasic agents.


Subject(s)
Cysteine Endopeptidases , Molecular Docking Simulation , Protozoan Proteins , Triazoles , Trypanosoma cruzi , Triazoles/chemistry , Triazoles/pharmacology , Triazoles/chemical synthesis , Cysteine Endopeptidases/chemistry , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/enzymology , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Cysteine Proteinase Inhibitors/chemical synthesis , Molecular Dynamics Simulation , Structure-Activity Relationship , Computer-Aided Design , Drug Design , Humans , Molecular Structure , Trypanocidal Agents/pharmacology , Trypanocidal Agents/chemistry , Trypanocidal Agents/chemical synthesis , Chagas Disease/drug therapy
3.
Parasites Hosts Dis ; 62(3): 302-312, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39218629

ABSTRACT

Plasmodium falciparum apical membrane antigen-1 (PfAMA-1) is a major candidate for the blood-stage malaria vaccine. Genetic polymorphisms of global pfama-1suggest that the genetic diversity of the gene can disturb effective vaccine development targeting this antigen. This study was conducted to explore the genetic diversity and gene structure of pfama-1 among P. falciparum isolates collected in the Khyber Pakhtunkhwa (KP) province of Pakistan. A total of 19 full-length pfama-1 sequences were obtained from KP-Pakistan P. falciparum isolates, and genetic polymorphism and natural selection were investigated. KP-Pakistan pfama-1 exhibited genetic diversity, wherein 58 amino acid changes were identified, most of which were located in ectodomains, and domains I, II, and III. The amino acid changes commonly found in the ectodomain of global pfama-1 were also detected in KP-Pakistan pfama-1. Interestingly, 13 novel amino acid changes not reported in the global population were identified in KP-Pakistan pfama-1. KP-Pakistan pfama-1 shared similar levels of genetic diversity with global pfama-1. Evidence of natural selection and recombination events were also detected in KP-Pakistan pfama-1.


Subject(s)
Antigens, Protozoan , Malaria, Falciparum , Membrane Proteins , Plasmodium falciparum , Polymorphism, Genetic , Protozoan Proteins , Pakistan , Plasmodium falciparum/genetics , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/chemistry , Membrane Proteins/genetics , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/epidemiology , Genetic Variation/genetics , Selection, Genetic , Phylogeny , Recombination, Genetic/genetics
4.
PLoS One ; 19(8): e0306975, 2024.
Article in English | MEDLINE | ID: mdl-39146276

ABSTRACT

Malaria, an ancient mosquito-borne illness caused by Plasmodium parasites, is mostly treated with Artemisinin Combination Therapy (ACT). However, Single Nucleotide Polymorphisms (SNPs) mutations in the P. falciparum Kelch 13 (PfK13) protein have been associated with artemisinin resistance (ART-R). Therefore, this study aims to generate PfK13 recombinant proteins incorporating of two specific SNPs mutations, PfK13-V494I and PfK13-N537I, and subsequently analyze their binding interactions with artemisinin (ART). The recombinant proteins of PfK13 mutations and the Wild Type (WT) variant were expressed utilizing a standard protein expression protocol with modifications and subsequently purified via IMAC and confirmed with SDS-PAGE analysis and Orbitrap tandem mass spectrometry. The binding interactions between PfK13-V494I and PfK13-N537I propeller domain proteins ART were assessed through Isothermal Titration Calorimetry (ITC) and subsequently validated using fluorescence spectrometry. The protein concentrations obtained were 0.3 mg/ml for PfK13-WT, 0.18 mg/ml for PfK13-V494I, and 0.28 mg/ml for PfK13-N537I. Results obtained for binding interaction revealed an increased fluorescence intensity in the mutants PfK13-N537I (83 a.u.) and PfK13-V494I (143 a.u.) compared to PfK13-WT (33 a.u.), indicating increased exposure of surface proteins because of the looser binding between PfK13 protein mutants with ART. This shows that the PfK13 mutations may induce alterations in the binding interaction with ART, potentially leading to reduced effectiveness of ART and ultimately contributing to ART-R. However, this study only elucidated one facet of the contributing factors that could serve as potential indicators for ART-R and further investigation should be pursued in the future to comprehensively explore this complex mechanism of ART-R.


Subject(s)
Artemisinins , Plasmodium falciparum , Protein Binding , Protozoan Proteins , Recombinant Proteins , Artemisinins/pharmacology , Plasmodium falciparum/genetics , Plasmodium falciparum/drug effects , Plasmodium falciparum/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Protozoan Proteins/chemistry , Mutation , Polymorphism, Single Nucleotide , Antimalarials/pharmacology , Drug Resistance/genetics
5.
J Vis Exp ; (210)2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39185871

ABSTRACT

Visual Dynamics (VD) is a web tool that aims to facilitate the use and application of Molecular Dynamics (MD) executed in Gromacs, allowing users without computational familiarity to run short-time simulations for validation, demonstration, and teaching purposes. It is true that quantum methods are the most accurate. However, there is currently no computational feasibility to carry out the experiments that MD performs. The tool described here has continuously received improvements over the course of the last couple of years. This protocol will describe what is needed to run a simulation in VD with a protein-ligand complex previously prepared in ACPYPE and some general directions on the other simulation models available. For the detailed simulation, the FK506-binding protein from Plasmodium vivax complexed with the inhibitor D5 (PDB ID: 4mgv) will be used, and all files used will be provided. Note that this protocol will tell every option to be used to achieve the same results presented, but these options are not necessarily the only ones available.


Subject(s)
Molecular Dynamics Simulation , Plasmodium vivax/physiology , Software , Tacrolimus Binding Proteins/chemistry , Protozoan Proteins/chemistry
6.
BMC Genomics ; 25(1): 804, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39187768

ABSTRACT

BACKGROUND: SET domain-containing histone lysine methyltransferases (HKMTs) and JmjC domain-containing histone demethylases (JHDMs) are essential for maintaining dynamic changes in histone methylation across parasite development and infection. However, information on the HKMTs and JHDMs in human pathogenic piroplasms, such as Babesia duncani and Babesia microti, and in veterinary important pathogens, including Babesia bigemina, Babesia bovis, Theileria annulata and Theileria parva, is limited. RESULTS: A total of 38 putative KMTs and eight JHDMs were identified using a comparative genomics approach. Phylogenetic analysis revealed that the putative KMTs can be divided into eight subgroups, while the JHDMs belong to the JARID subfamily, except for BdJmjC1 (BdWA1_000016) and TpJmjC1 (Tp Muguga_02g00471) which cluster with JmjC domain only subfamily members. The motifs of SET and JmjC domains are highly conserved among piroplasm species. Interspecies collinearity analysis provided insight into the evolutionary duplication events of some SET domain and JmjC domain gene families. Moreover, relative gene expression analysis by RT‒qPCR demonstrated that the putative KMT and JHDM gene families were differentially expressed in different intraerythrocytic developmental stages of B. duncani, suggesting their role in Apicomplexa parasite development. CONCLUSIONS: Our study provides a theoretical foundation and guidance for understanding the basic characteristics of several important piroplasm KMT and JHDM families and their biological roles in parasite differentiation.


Subject(s)
Babesia , Phylogeny , Babesia/genetics , Babesia/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Protozoan Proteins/chemistry , Genomics , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/chemistry , Animals , Humans , Genome, Protozoan , PR-SET Domains/genetics
7.
Biochem J ; 481(16): 1075-1096, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39105673

ABSTRACT

Toxoplasma gondii is a widely distributed apicomplexan parasite causing toxoplasmosis, a critical health issue for immunocompromised individuals and for congenitally infected foetuses. Current treatment options are limited in number and associated with severe side effects. Thus, novel anti-toxoplasma agents need to be identified and developed. 1-Deoxy-d-xylulose 5-phosphate reductoisomerase (DXR) is considered the rate-limiting enzyme in the non-mevalonate pathway for the biosynthesis of the isoprenoid precursors isopentenyl pyrophosphate and dimethylallyl pyrophosphate in the parasite, and has been previously investigated for its key role as a novel drug target in some species, encompassing Plasmodia, Mycobacteria and Escherichia coli. In this study, we present the first crystal structure of T. gondii DXR (TgDXR) in a tertiary complex with the inhibitor fosmidomycin and the cofactor NADPH in dimeric conformation at 2.5 Šresolution revealing the inhibitor binding mode. In addition, we biologically characterize reverse α-phenyl-ß-thia and ß-oxa fosmidomycin analogues and show that some derivatives are strong inhibitors of TgDXR which also, in contrast with fosmidomycin, inhibit the growth of T. gondii in vitro. Here, ((3,4-dichlorophenyl)((2-(hydroxy(methyl)amino)-2-oxoethyl)thio)methyl)phosphonic acid was identified as the most potent anti T. gondii compound. These findings will enable the future design and development of more potent anti-toxoplasma DXR inhibitors.


Subject(s)
Aldose-Ketose Isomerases , Fosfomycin , Multienzyme Complexes , Toxoplasma , Toxoplasma/enzymology , Toxoplasma/drug effects , Aldose-Ketose Isomerases/antagonists & inhibitors , Aldose-Ketose Isomerases/chemistry , Aldose-Ketose Isomerases/metabolism , Aldose-Ketose Isomerases/genetics , Fosfomycin/pharmacology , Fosfomycin/analogs & derivatives , Fosfomycin/chemistry , Crystallography, X-Ray , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , NADP/metabolism , NADP/chemistry , Humans , Models, Molecular , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/chemistry , Oxidoreductases/metabolism
8.
BMC Res Notes ; 17(1): 216, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095914

ABSTRACT

OBJECTIVE: Proteasomes are conserved proteases crucial for proteostasis in eukaryotes and are promising drug targets for protozoan parasites. Yet, the proteasomes of Entamoeba histolytica remain understudied. The study's objective was to analyse the differences in the substrate binding pockets of amoeba proteasomes from those of host, and computational modelling of ß5 catalytic subunit, with the goal of finding selective inhibitors. RESULTS: Comparative sequence analysis revealed differences in substrate binding sites of E. histolytica proteasomes, especially in the S1 and S3 pockets of the catalytic beta subunits, implying differences in substrate preference and susceptibility to inhibitors from host proteasomes. This was strongly supported by significantly lower sensitivity to MG132 mediated inhibition of amoebic proteasome ß5 subunit's chymotryptic activity compared to human proteasomes, also reflected in lower sensitivity of E. histolytica to MG132 for inhibition of proliferation. Computational models of ß4 and ß5 subunits, and a docked ß4-ß5 model revealed a binding pocket between ß4-ß5, similar to that of Leishmania tarentolae. Selective inhibitors for visceral leishmaniasis, LXE408 and compound 8, docked well to this pocket. This functional and sequence-based analysis predicts differences between amoebic and host proteasomes that can be utilized to develop rationally designed, selective inhibitors against E. histolytica.


Subject(s)
Entamoeba histolytica , Proteasome Endopeptidase Complex , Entamoeba histolytica/enzymology , Entamoeba histolytica/metabolism , Proteasome Endopeptidase Complex/metabolism , Humans , Binding Sites , Leupeptins/pharmacology , Substrate Specificity , Protozoan Proteins/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Proteasome Inhibitors/pharmacology , Molecular Docking Simulation , Amino Acid Sequence , Catalytic Domain , Protein Binding , Models, Molecular
9.
PLoS One ; 19(8): e0304842, 2024.
Article in English | MEDLINE | ID: mdl-39116045

ABSTRACT

The retromer is a cellular structure that recruits and recycles proteins inside the cell. In mammalian and yeast, the retromer components have been widely studied, but very little in parasites. In yeast, it is formed by a SNX-BAR membrane remodeling heterodimer and the cargo selecting complex (CSC), composed by three proteins. One of them, the Vps26 protein, possesses a flexible and intrinsically disordered region (IDR), that facilitates interactions with other proteins and contributes to the retromer binding to the endosomal membrane. In Entamoeba histolytica, the protozoan parasite responsible for human amoebiasis, the retromer actively participates during the high mobility and phagocytosis of trophozoites, but the molecular details in these events, are almost unknown. Here, we studied the EhVps26 role in phagocytosis. Bioinformatic analyses of EhVps26 revealed a typical arrestin folding structure of the protein, and a long and charged IDR, as described in other systems. EhVps26 molecular dynamics simulations (MDS) allowed us to predict binding pockets for EhVps35, EhSNX3, and a PX domain-containing protein; these pockets were disorganized in a EhVps26 truncated version lacking the IDR. The AlphaFold2 software predicted the interaction of EhVps26 with EhVps35, EhVps29 and EhSNX3, in a model similar to the reported mammalian crystals. By confocal and transmission electron microscopy, EhVps26 was found in the trophozoites plasma membrane, cytosol, endosomes, and Golgi-like apparatus. During phagocytosis, it followed the erythrocytes pathway, probably participating in cargoes selection and recycling. Ehvps26 gene knocking down evidenced that the EhVps26 protein is necessary for efficient phagocytosis.


Subject(s)
Computational Biology , Entamoeba histolytica , Phagocytosis , Protozoan Proteins , Entamoeba histolytica/metabolism , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/chemistry , Computational Biology/methods , Humans , Molecular Dynamics Simulation , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/chemistry , Protein Binding , Amino Acid Sequence , Erythrocytes/parasitology , Erythrocytes/metabolism
10.
Arch Biochem Biophys ; 760: 110124, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39154815

ABSTRACT

Cryptosporidium parvum (C. parvum), a protozoan parasite, is known to induce significant gastrointestinal disease in humans. Lactate dehydrogenase (LDH), a protein of C. parvum, has been identified as a potential therapeutic target for developing effective drugs against infection. This study utilized a computational drug discovery approach to identify potential drug molecules against the LDH protein of C. parvum. In the present investigation, we conducted a structure-based virtual screening of 55 phytochemicals from the Syzygium aromaticum (S. aromaticum). This process identified four phytochemicals, including Gallotannin 23, Eugeniin, Strictinin, and Ellagitannin, that demonstrated significant binding affinity and dynamic stability with LDH protein. Interestingly, these four compounds have been documented to possess antibacterial, antiviral, anti-inflammatory, and antioxidant properties. The docked complexes were simulated for 100 ns using Desmond to check the dynamic stability. Finally, the free binding energy was computed from the last 10ns MD trajectories. Gallotannin 23 and Ellagitannin exhibited considerable binding affinity and stability with the target protein among all four phytochemicals. These findings suggest that these predicted phytochemicals from S. aromaticum could be further explored as potential hit candidates for developing effective drugs against C. parvum infection. The in vitro and in vivo experimental validation is still required to confirm their efficacy and safety as LDH inhibitors.


Subject(s)
Cryptosporidium parvum , L-Lactate Dehydrogenase , Molecular Dynamics Simulation , Phytochemicals , Syzygium , Cryptosporidium parvum/enzymology , Cryptosporidium parvum/drug effects , Syzygium/chemistry , Phytochemicals/chemistry , Phytochemicals/pharmacology , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/chemistry , L-Lactate Dehydrogenase/metabolism , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/chemistry , Molecular Docking Simulation , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism
11.
J Phys Chem B ; 128(35): 8400-8408, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39172501

ABSTRACT

Plasmodium falciparum apical membrane antigen 1 (PfAMA1) is a surface protein found in two stages of the malaria life cycle. This is a protein involved in a reorientation movement of the parasite so that cell invasion occurs in the so-called "moving junction", relevant when the membranes of the parasite and the host are in contact. The structure of a conformational epitope of domain III of PfAMA1 in complex with the monoclonal antibody Fab F8.12.19 is experimentally known. Here, we used molecular dynamics with enhanced sampling by Hamiltonian replica exchange molecular dynamics (HREMD) to understand the effect of intermolecular interactions, conformational variability, and intrinsically disordered regions on the mechanism of antigen-antibody interaction. Clustering methods and the analysis of conformational variability were used in order to understand the influence of the presence of the partner protein in the complex. The free-state epitope accesses a broader conformational pool, including disordered conformations not seen in the bound state. The simulations suggest an extended conformational selection mechanism in which the antibody stabilizes a conformational set of the epitope existing in the free state. The stabilization of the active conformation occurs mainly through hydrogen bonds: Tyr(H33)-Asp493, His(L94)-Val510, Ser(L93)-Glu511, Tyr(H56)-Asp485, and Tyr(H35)-Asp493. The antibody has a structure with few flexible regions, and only the complementarity determining region (CDR) H3 shows greater plasticity in the presence of the epitope.


Subject(s)
Antigens, Protozoan , Membrane Proteins , Molecular Dynamics Simulation , Plasmodium falciparum , Protozoan Proteins , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Antigens, Protozoan/chemistry , Antigens, Protozoan/immunology , Membrane Proteins/chemistry , Membrane Proteins/immunology , Plasmodium falciparum/immunology , Plasmodium falciparum/chemistry , Protein Conformation , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology
12.
J Chem Inf Model ; 64(17): 6912-6925, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39193724

ABSTRACT

The convergence of biotechnology and artificial intelligence has the potential to transform drug development, especially in the field of therapeutic peptide design. Peptides are short chains of amino acids with diverse therapeutic applications that offer several advantages over small molecular drugs, such as targeted therapy and minimal side effects. However, limited oral bioavailability and enzymatic degradation have limited their effectiveness. With advances in deep learning techniques, innovative approaches to peptide design have become possible. In this work, we demonstrate HYDRA, a hybrid deep learning approach that leverages the distribution modeling capabilities of a diffusion model and combines it with a binding affinity maximization algorithm that can be used for de novo design of peptide binders for various target receptors. As an application, we have used our approach to design therapeutic peptides targeting proteins expressed by Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) genes. The ability of HYDRA to generate peptides conditioned on the target receptor's binding sites makes it a promising approach for developing effective therapies for malaria and other diseases.


Subject(s)
Peptides , Peptides/chemistry , Peptides/metabolism , Plasmodium falciparum/metabolism , Diffusion , Protozoan Proteins/metabolism , Protozoan Proteins/chemistry , Models, Molecular , Deep Learning
13.
Nat Commun ; 15(1): 7487, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39209843

ABSTRACT

CelTOS is a malaria vaccine antigen that is conserved in Plasmodium and other apicomplexan parasites and plays a role in cell-traversal. The structural basis and mechanisms of CelTOS-induced protective immunity to parasites are unknown. Here, CelTOS-specific monoclonal antibodies (mAbs) 7g7 and 4h12 demonstrated multistage activity, protecting against liver infection and preventing parasite transmission to mosquitoes. Both mAbs demonstrated cross-species activity with sterile protection against in vivo challenge with transgenic parasites containing either P. falciparum or P. vivax CelTOS, and with transmission reducing activity against P. falciparum. The mAbs prevented CelTOS-mediated pore formation providing insight into the protective mechanisms. X-ray crystallography and mutant-library epitope mapping revealed two distinct broadly conserved neutralizing epitopes. 7g7 bound to a parallel dimer of CelTOS, while 4h12 bound to a novel antiparallel dimer architecture. These findings inform the design of antibody therapies and vaccines and raise the prospect of a single intervention to simultaneously combat P. falciparum and P. vivax malaria.


Subject(s)
Antibodies, Monoclonal , Antibodies, Protozoan , Malaria Vaccines , Plasmodium falciparum , Plasmodium vivax , Antibodies, Monoclonal/immunology , Animals , Plasmodium falciparum/immunology , Plasmodium vivax/immunology , Malaria Vaccines/immunology , Antibodies, Protozoan/immunology , Mice , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/parasitology , Crystallography, X-Ray , Epitopes/immunology , Malaria, Vivax/prevention & control , Malaria, Vivax/immunology , Malaria, Vivax/parasitology , Antigens, Protozoan/immunology , Humans , Female , Epitope Mapping , Malaria/immunology , Malaria/prevention & control , Malaria/parasitology , Mice, Inbred BALB C , Protozoan Proteins/immunology , Protozoan Proteins/chemistry
14.
Nat Commun ; 15(1): 6642, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103329

ABSTRACT

Plasmodium falciparum is the main causative agent of malaria, a deadly disease that mainly affects children under five years old. Artemisinin-based combination therapies have been pivotal in controlling the disease, but resistance has arisen in various regions, increasing the risk of treatment failure. The non-mevalonate pathway is essential for the isoprenoid synthesis in Plasmodium and provides several under-explored targets to be used in the discovery of new antimalarials. 1-deoxy-D-xylulose-5-phosphate synthase (DXPS) is the first and rate-limiting enzyme of the pathway. Despite its importance, there are no structures available for any Plasmodium spp., due to the complex sequence which contains large regions of high disorder, making crystallisation a difficult task. In this manuscript, we use cryo-electron microscopy to solve the P. falciparum DXPS structure at a final resolution of 2.42 Å. Overall, the structure resembles other DXPS enzymes but includes a distinct N-terminal domain exclusive to the Plasmodium genus. Mutational studies show that destabilization of the cap domain interface negatively impacts protein stability and activity. Additionally, a density for the co-factor thiamine diphosphate is found in the active site. Our work highlights the potential of cryo-EM to obtain structures of P. falciparum proteins that are unfeasible by means of crystallography.


Subject(s)
Cryoelectron Microscopy , Plasmodium falciparum , Plasmodium falciparum/enzymology , Plasmodium falciparum/genetics , Pentosyltransferases/metabolism , Pentosyltransferases/chemistry , Pentosyltransferases/genetics , Pentosyltransferases/ultrastructure , Protein Domains , Models, Molecular , Protozoan Proteins/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/ultrastructure , Transferases
15.
Int J Biol Macromol ; 277(Pt 3): 134428, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39097052

ABSTRACT

Mitogen-activated protein kinase (MAPK) interacting protein kinases (Mnk1 and Mnk2) mediated phosphorylation of the eukaryotic initiation factor eIF4E is an important translation initiation control, in Mnk-mediated oncogenic activity and other disease conditions. Thus, Mnk kinases are an important target for therapy. Trypanosomatids are a class of kinetoplastids, some of which are protozoan parasites and cause diseases in humans. While protein translation initiation is well understood in eukaryotes and prokaryotes, there is a lack of sufficient structural information of this process in trypanosomatids. Here, we report that trypanosomatids have one orthologue of Mnk kinase with low overall sequence homology but high homology in the kinase domain and an additional C-terminal domain containing putative calmodulin binding site(s). We show that while many of the domains and motifs are conserved, homology modeling/structure prediction, docking analysis and molecular dynamics simulation studies suggest that trypanosomatid kMnk kinases, kinase domains are present in DFG-in conformation as opposed to the auto-inhibited DFD-out conformation of un-phosphorylated human Mnk1. Furthermore, we observed that several regulatory features are different in trypanosomatid kMnk kinases. Our study indicates that mechanism and regulation in the kinase domain of trypanosomatid kMnks are likely to be altered, and that they can be important drug targets.


Subject(s)
Protein Serine-Threonine Kinases , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Humans , Amino Acid Sequence , Molecular Dynamics Simulation , Phosphorylation , Molecular Docking Simulation , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Protein Domains , Binding Sites , Protein Binding
16.
BMC Infect Dis ; 24(1): 886, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39210269

ABSTRACT

BACKGROUND: The highly expressed surface antigen 1 (SAG1)-related sequence (SRS) proteins of T. gondii tachyzoites, as a widespread zoonotic parasite, are critical for host cell invasion and represent promising vaccine targets. In this study, we employed a computer-aided multi-method approach for in silico design and evaluation of TgVax452, an epitope-based candidate vaccine against T. gondii tachyzoite-specific SRS proteins. METHODS: Using immunoinformatics web-based tools, structural modeling, and static/dynamic molecular simulations, we identified and screened B- and T-cell immunodominant epitopes and predicted TgVax452's antigenicity, stability, safety, adjuvanticity, and physico-chemical properties. RESULTS: The designed protein possessed 452 residues, a MW of 44.07 kDa, an alkaline pI (6.7), good stability (33.20), solubility (0.498), and antigenicity (0.9639) with no allergenicity. Comprehensive molecular dynamic (MD) simulation analyses confirmed the stable interaction (average potential energy: 3.3799 × 106 KJ/mol) between the TLR4 agonist residues (RS09 peptide) of the TgVax452 in interaction with human TLR4, potentially activating innate immune responses. Also, a dramatic increase was observed in specific antibodies (IgM and IgG), cytokines (IFN-γ), and lymphocyte responses, based on C-ImmSim outputs. Finally, we optimized TgVax452's codon adaptation and mRNA secondary structure for efficient expression in E. coli BL21 expression machinery. CONCLUSION: Our findings suggest that TgVax452 is a promising candidate vaccine against T. gondii tachyzoite-specific SRS proteins and requires further experimental studies for its potential use in preclinical trials.


Subject(s)
Antigens, Protozoan , Computational Biology , Epitopes, T-Lymphocyte , Protozoan Proteins , Protozoan Vaccines , Toxoplasma , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Protozoan Proteins/chemistry , Toxoplasma/immunology , Toxoplasma/genetics , Toxoplasma/chemistry , Protozoan Vaccines/immunology , Protozoan Vaccines/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/genetics , Antigens, Protozoan/chemistry , Animals , Mice , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Female , Antibodies, Protozoan/immunology , Mice, Inbred BALB C , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/chemistry , Humans , Molecular Dynamics Simulation , Immunodominant Epitopes/immunology , Immunodominant Epitopes/genetics , Immunodominant Epitopes/chemistry , Toxoplasmosis/prevention & control , Toxoplasmosis/immunology , Immunoinformatics
17.
Commun Biol ; 7(1): 1070, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39217277

ABSTRACT

In the absence of an efficacious vaccine, chemotherapy remains crucial to prevent and treat malaria. Given its key role in haemoglobin degradation, falcilysin constitutes an attractive target. Here, we reveal the mechanism of enzymatic inhibition of falcilysin by MK-4815, an investigational new drug with potent antimalarial activity. Using X-ray crystallography, we determine two binary complexes of falcilysin in a closed state, bound with peptide substrates from the haemoglobin α and ß chains respectively. An antiparallel ß-sheet is formed between the substrate and enzyme, accounting for sequence-independent recognition at positions P2 and P1. In contrast, numerous contacts favor tyrosine and phenylalanine at the P1' position of the substrate. Cryo-EM studies reveal a majority of unbound falcilysin molecules adopting an open conformation. Addition of MK-4815 shifts about two-thirds of falcilysin molecules to a closed state. These structures give atomic level pictures of the proteolytic cycle, in which falcilysin interconverts between a closed state conducive to proteolysis, and an open conformation amenable to substrate diffusion and products release. MK-4815 and quinolines bind to an allosteric pocket next to a hinge region of falcilysin and hinders this dynamic transition. These data should inform the design of potent inhibitors of falcilysin to combat malaria.


Subject(s)
Antimalarials , Plasmodium falciparum , Plasmodium falciparum/enzymology , Plasmodium falciparum/drug effects , Antimalarials/pharmacology , Antimalarials/chemistry , Protozoan Proteins/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/antagonists & inhibitors , Crystallography, X-Ray , Models, Molecular , Cryoelectron Microscopy , Humans
18.
Parasitol Res ; 123(8): 310, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39207503

ABSTRACT

Babesia orientalis, a protozoan parasite transmitted by the tick Rhipicephalus haemaphysaloides, holds significant economic importance along the Yangtze River. Key factors in the host invasion process include rhoptry neck proteins (RON2, RON4, and RON5) and apical membrane antigen 1 (AMA1). However, the intricacies of the interaction between AMA1 and RONs remain incompletely elucidated in B. orientalis. To better understand these crucial invasion components, the RON4 gene of B. orientalis (BoRON4) was cloned and sequenced. RON4 is 3468 base pairs long, encodes 1155 amino acids, and has a predicted molecular weight of 130 kDa. Bioinformatics analysis revealed a unique region (amino acid residues 109-452) in BoRON4, which demonstrates higher sensitivity to epitope activity. The BoRON4 gene was strategically truncated, amplified, and cloned into the pGEX-6p-1 vector for fusion expression. We successfully used the mouse polyclonal antibody to identify native BoRON4 in B. orientalis lysates. Furthermore, the corresponding BoRON4 protein band was detected in the water buffalo serum infected with B. orientalis, while no such band was observed in the control. Additionally, I-TASSER and Discovery Studio software were used to predict the tertiary structures of BoRON4 and its ligands, CH-PKA and CH-complex. These ligands can serve as lead compounds for the development of anti-babesiosis drugs. In conclusion, BoRON4 emerges as a promising candidate antigen for distinguishing water buffalo infected with B. orientalis from their normal counterparts. This study positions BoRON4 as a potential diagnostic antigen for babesiosis in water buffalo, contributing valuable insights to the field of parasitology.


Subject(s)
Babesia , Protozoan Proteins , Babesia/genetics , Animals , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Babesiosis/parasitology , Babesiosis/diagnosis , Buffaloes/parasitology , Cloning, Molecular , Amino Acid Sequence
19.
Acta Crystallogr D Struct Biol ; 80(Pt 8): 629-638, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39052317

ABSTRACT

Chagas disease is a neglected tropical disease caused by the protozoan parasite Trypanosoma cruzi. It bears a significant global health burden with limited treatment options, thus calling for the development of new and effective drugs. Certain trypanosomal metabolic enzymes have been suggested to be druggable and valid for subsequent inhibition. In this study, the crystal structure of glycerol kinase from T. cruzi, a key enzyme in glycerol metabolism in this parasite, is presented. Structural analysis allowed a detailed description of the glycerol binding pocket, while comparative assessment pinpointed a potential regulatory site which may serve as a target for selective inhibition. These findings advance the understanding of glycerol metabolism in eukaryotes and provide a solid basis for the future treatment of Chagas disease.


Subject(s)
Chagas Disease , Glycerol Kinase , Trypanosoma cruzi , Trypanosoma cruzi/enzymology , Glycerol Kinase/chemistry , Glycerol Kinase/metabolism , Chagas Disease/drug therapy , Chagas Disease/parasitology , Crystallography, X-Ray , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Models, Molecular , Humans , Binding Sites , Glycerol/chemistry , Protein Conformation
20.
Protein J ; 43(4): 819-833, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39009910

ABSTRACT

Plasmodium falciparum (P. falciparum), which causes the most severe form of malaria, if left untreated, has 24 h window in which it can cause severe illness and even death. The aim of this study was to create the most comprehensive and informative secretory-proteome possible by combining high-accuracy and high-sensitivity protein identification technology. In this study, we used Plasmodium falciparum 3D7 (Pf3D7) as the model parasite to develop a label-free quantification proteomic strategy with the main goal of identifying Pf3D7 proteins that are supposed to be secreted outside the infected erythrocytes in the spent media culture during the in-vitro study. The spent culture media supernatant was subjected to differential and ultra-centrifugation steps followed by total protein extraction, estimation, and in-solution digestion using trypsin, digested peptides were analyzed using Nano-LC coupled with ESI for MS/MS. MS/MS spectra were processed using Maxquant software (v2.1.4.0.). Non-infected erythrocytes incubated spent cultured media supernatant were considered as control. Out of discovered 38 proteins, proteins belonging to P. falciparum spp. were EGF-like protein (C0H544), Endoplasmic reticulum chaperone GRP170 (C0H5H0), Small GTP-binding protein sar1 (Q8I1S0), Erythrocyte membrane protein 1, PfEMP1 (Q8I639), aldehyde reductase (Q8ID61), Conserved Plasmodium proteins (Q8IEH3, Q8ILD1), Antigen 332, DBL-like protein (Q8IHN4), Fe-S cluster assembly protein (Q8II78), identified and chosen for further in-depth investigation. This study highlights the value of secretory Plasmodium proteins play crucial roles in various aspects of the disease progression and host-pathogen interactions which can serve as diagnostic markers for malaria infection.


Subject(s)
Biomarkers , Erythrocytes , Plasmodium falciparum , Proteomics , Protozoan Proteins , Plasmodium falciparum/chemistry , Erythrocytes/parasitology , Erythrocytes/chemistry , Erythrocytes/metabolism , Protozoan Proteins/metabolism , Protozoan Proteins/chemistry , Humans , Proteomics/methods , Malaria, Falciparum/parasitology , Malaria, Falciparum/diagnosis , Malaria, Falciparum/blood , Proteome/analysis
SELECTION OF CITATIONS
SEARCH DETAIL