Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (136)

Search Parameters:
Keywords = trypanosomatids

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 1369 KiB  
Article
Blood Parasites (Haemosporida, Trypanosomatida) in Culex pipiens: A Study and Review of Hibernating and Active Mosquitoes
by Kristina Valavičiūtė-Pocienė, Margarita Kazak, Tatjana Iezhova, Gabrielė Kalinauskaitė and Rasa Bernotienė
Microbiol. Res. 2024, 15(4), 2184-2198; https://doi.org/10.3390/microbiolres15040146 - 25 Oct 2024
Viewed by 522
Abstract
Culex pipiens mosquitoes (Diptera: Culicidae) are widespread during warm periods and actively feed on blood while serving as vectors for various human and animal pathogens. Culex mosquitoes overwinter as adults in temperate zones, raising the question of whether hibernating Cx. pipiens can act [...] Read more.
Culex pipiens mosquitoes (Diptera: Culicidae) are widespread during warm periods and actively feed on blood while serving as vectors for various human and animal pathogens. Culex mosquitoes overwinter as adults in temperate zones, raising the question of whether hibernating Cx. pipiens can act as pathogen reservoirs. In this study, hibernating mosquitoes and mosquitoes collected during the warm season were tested for the presence of trypanosomatids and avian haemosporidian parasites using PCR. Midgut preparations were made from Cx. pipiens females in order to search for trypanosomatids morphologically. In total, 1037 Cx. pipiens mosquitoes (556 collected during the warm season and 481 overwintering mosquitos) were investigated. The parasite prevalence differed for mosquitoes collected during the warm season and hibernating ones for both Haemosporida (2.9% in warm-season and no infections in overwintering mosquitoes) and Trypanosomatida (1.6% and 0.4%, respectively) parasites. A phylogenetic analysis confirmed that the trypanosomatids found in hibernating mosquitoes were monoxenous and were not parasites of vertebrates. The peak prevalence of Haemosporida parasites was detected in July (4.9%) and August (2.8%), and for Trypanosomatida, it was detected in May (3.5%). The results of the present study show that overwintering Cx. pipiens mosquitoes are questionable reservoirs for avian haemosporidian parasites, but some monoxenous trypanosomatids can be found in overwintering females. Full article
(This article belongs to the Special Issue Veterinary Microbiology and Diagnostics)
Show Figures

Figure 1

Figure 1
<p>Average number of collected <span class="html-italic">Culex pipiens</span> females (left axis), with the average number of mosquitoes infected with Haemosporida and Trypanosomatida parasites (right axis) during the warm season in 2021–2023. Average ± standard error.</p>
Full article ">Figure 2
<p>Trypanosomatids found in <span class="html-italic">Culex pipiens</span> midgut preparations: (<b>A</b>)—<span class="html-italic">Trypanosoma culicavium</span> (GenBank No. PP946100); (<b>B</b>)—<span class="html-italic">Trypanosoma trinaperronei</span> (PP946104); (<b>C</b>)—monoxenous trypanosomatid (PP948731). Long arrows—parasite nuclei; arrowheads—kinetoplast; short arrows—free flagellum. Scale bar = 10 µm.</p>
Full article ">Figure 3
<p>Bayesian phylogenetic tree of <span class="html-italic">Trypanosoma</span> (<b>A</b>) and other Trypanosomatidae (<b>B</b>) using fragments of 18S rRNA. The tree was rooted using <span class="html-italic">Cryptobia catostomi</span>. The Genbank accession numbers of samples obtained during this investigation are written in bold text.</p>
Full article ">
12 pages, 1756 KiB  
Article
Untargeted Liquid Chromatography–High-Resolution Mass Spectrometry Metabolomic Investigation Reveals Altered Lipid Content in Leishmania infantum Lacking Lipid Droplet Protein Kinase
by Juliana Martins Ribeiro, Gisele André Baptista Canuto, Alisson Samuel Portes Caldeira, Ezequias Pessoa de Siqueira, Carlos Leomar Zani, Silvane Maria Fonseca Murta and Tânia Maria de Almeida Alves
Trop. Med. Infect. Dis. 2024, 9(9), 208; https://doi.org/10.3390/tropicalmed9090208 - 10 Sep 2024
Viewed by 825
Abstract
Leishmaniasis is a complex disease caused by different species of Leishmania. To date, no vaccine for humans or ideal therapy has been developed owing to the limited efficacy and toxicity of available drugs, as well as the emergence of resistant strains. Therefore, [...] Read more.
Leishmaniasis is a complex disease caused by different species of Leishmania. To date, no vaccine for humans or ideal therapy has been developed owing to the limited efficacy and toxicity of available drugs, as well as the emergence of resistant strains. Therefore, it is necessary to identify novel therapeutic targets and discover therapeutic options for leishmaniasis. In this study, we evaluated the impact of deleting the lipid droplet protein kinase (LDK) enzyme in Leishmania infantum using an untargeted metabolomics approach performed using liquid chromatography and high-resolution mass spectrometry. LDK is involved in lipid droplet biogenesis in trypanosomatids. Thirty-nine lipid metabolites altered in the stationary and logarithmic growth phases were noted and classified into five classes: (1) sterols, (2) fatty and conjugated acids, (3) ceramides, (4) glycerophosphocholine and its derivatives, and (5) glycerophosphoethanolamine and its derivatives. Our data demonstrated that glycerophosphocholine and its derivatives were the most affected after LDK deletion, suggesting that the absence of this enzyme promotes the remodeling of lipid composition in L. infantum, thus contributing to a better understanding of the function of LDK in this parasite. Full article
(This article belongs to the Special Issue Advances in Parasitic Neglected Tropical Diseases)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) PCA model of all samples and Pareto-scaled QCs, (<b>B</b>) PLS-DA model of all samples and Pareto-scaled QCs (quality parameters: R<sup>2</sup> = 0.96 and Q<sup>2</sup> = 0.64), (<b>C</b>) heat map of the entire metabolome dataset, and (<b>D</b>) HCA of the sample groups and QCs. Labels: pink, quality control (QC); red, Cas9 stationary phase; green, Cas9 logarithmic phase; blue, ΔLDK stationary phase; and cyan, ΔLDK logarithmic phase.</p>
Full article ">Figure 2
<p>Full lipid network with node size scaled by fold change for comparison between <span class="html-italic">L. infantum</span> ΔLDK and Cas9. (<b>Up</b>) Stationary phase and (<b>down</b>) logarithm phase. Lipids are colored by class, and edge colors indicate reaction types connecting two nodes. Larger nodes correspond to greater abundance in the ΔLDK group, and small nodes correspond to greater abundance in the Cas9 group.</p>
Full article ">
5 pages, 185 KiB  
Editorial
Spotlight on Leishmaniasis Research: Insights from the Special Issue “Emerging Topics in Leishmaniasis Research”
by Sandra Regina Maruyama
Trop. Med. Infect. Dis. 2024, 9(9), 200; https://doi.org/10.3390/tropicalmed9090200 - 2 Sep 2024
Viewed by 782
Abstract
Leishmaniases, caused by dixenous trypanosomatids from the Leishmaniinae subfamily (over 20 Leishmania species), manifest in three primary clinical forms: visceral (VL), cutaneous (CL), and mucocutaneous (MCL) [...] Full article
(This article belongs to the Special Issue Emerging Topics in Leishmaniasis Research)
14 pages, 2012 KiB  
Article
Free Radical Production Induced by Nitroimidazole Compounds Lead to Cell Death in Leishmania infantum Amastigotes
by Julia Andrés-Rodríguez, María-Cristina González-Montero, Nerea García-Fernández, Estefanía Calvo-Álvarez, María-Yolanda Pérez-Pertejo, Rosa-María Reguera-Torres, Rafael Balaña-Fouce and Carlos García-Estrada
Molecules 2024, 29(17), 4041; https://doi.org/10.3390/molecules29174041 - 26 Aug 2024
Viewed by 797
Abstract
Leishmania infantum is the vector-borne trypanosomatid parasite causing visceral leishmaniasis in the Mediterranean basin. This neglected tropical disease is treated with a limited number of obsolete drugs that are not exempt from adverse effects and whose overuse has promoted the emergence of resistant [...] Read more.
Leishmania infantum is the vector-borne trypanosomatid parasite causing visceral leishmaniasis in the Mediterranean basin. This neglected tropical disease is treated with a limited number of obsolete drugs that are not exempt from adverse effects and whose overuse has promoted the emergence of resistant pathogens. In the search for novel antitrypanosomatid molecules that help overcome these drawbacks, drug repurposing has emerged as a good strategy. Nitroaromatic compounds have been found in drug discovery campaigns as promising antileishmanial molecules. Fexinidazole (recently introduced for the treatment of stages 1 and 2 of African trypanosomiasis), and pretomanid, which share the nitroimidazole nitroaromatic structure, have provided antileishmanial activity in different studies. In this work, we have tested the in vitro efficacy of these two nitroimidazoles to validate our 384-well high-throughput screening (HTS) platform consisting of L. infantum parasites emitting the near-infrared fluorescent protein (iRFP) as a biomarker of cell viability. These molecules showed good efficacy in both axenic and intramacrophage amastigotes and were poorly cytotoxic in RAW 264.7 and HepG2 cultures. Fexinidazole and pretomanid induced the production of ROS in axenic amastigotes but were not able to inhibit trypanothione reductase (TryR), thus suggesting that these compounds may target thiol metabolism through a different mechanism of action. Full article
Show Figures

Figure 1

Figure 1
<p>Chemical structure of the nitroimidazoles fexinidazole and pretomanid.</p>
Full article ">Figure 2
<p>Evolution of fluorescence emission by axenic amastigotes of <span class="html-italic">L. infantum</span>-iRFP throughout the culture time from 0 h to 72 h in the presence of different concentrations (from 4 μM to 0.0312 μM) of either fexinidazole or pretomanid, 0.1% (<span class="html-italic">v</span>/<span class="html-italic">v</span>) DMSO (negative control), and 18 µM Amp B (positive control). The fluorescence emitted at 0 h was subtracted from the rest of the values. Results show the mean values ± SD of three independent experiments with at least three technical replicates each.</p>
Full article ">Figure 3
<p>Dose-response curves adjusted with the Sigma Plot 10.1 statistical package showing the effect of (<b>a</b>) fexinidazole and (<b>b</b>) pretomanid on <span class="html-italic">L. infantum</span>-iRFP axenic and intramacrophage amastigotes (<b>left</b> panels), and HepG2 and RAW 264.7 cells (<b>right</b> panels). Graphs were prepared with viability data obtained from cells after 72 h of incubation in the presence of different concentrations of these compounds: from 4 μM to 0.0078 μM with one-half serial dilutions for parasites, or from 200 μM to 1.56 μM with one-half serial dilutions for mammalian cells. The <span class="html-italic">y</span>-axis represents the percentage of cell viability relative to the negative control, while the <span class="html-italic">x</span>-axis, in logarithmic scale, represents the concentration (μM) of the different nitroimidazole molecules and amphotericin B. Results show the mean values ± SD of three independent experiments with at least three technical replicates each.</p>
Full article ">Figure 4
<p>Representative flow cytometry graphs showing ROS production by the axenic amastigote cultures of <span class="html-italic">L. infantum</span>-iRFP stained with DCFH-DA after the addition of 0.01% (<span class="html-italic">v</span>/<span class="html-italic">v</span>) H<sub>2</sub>O<sub>2</sub> (positive control), 0.03% (<span class="html-italic">v</span>/<span class="html-italic">v</span>) DMSO, 0.04 μM fexinidazole, and 0.28 μM pretomanid. Histograms represent the distribution of fluorescence intensity for FL6-A: B525-A. Results show the mean values of two independent experiments with three technical replicates each.</p>
Full article ">Figure 5
<p>Time-dependent inhibition of TryR activity with a fixed concentration of T[S]<sub>2</sub> (0.075 mM) and NADPH (0.20 mM). Thioridazine (0.1 mM) was used as a positive control. Fexinidazole and pretomanid were tested at concentrations ranging from 6.25 μM to 100 μM. The calculated specific enzymatic activity for the negative control (without inhibitor) was 4.83 × 10<sup>−3</sup> μmol/mg · min. (ΔA/t = ɛ · d · c; ɛ = 14,150 M<sup>−1</sup> cm<sup>−1</sup>; d = 0.34 cm). Results show the mean values ± SD of two independent experiments with three technical replicates each.</p>
Full article ">
19 pages, 11217 KiB  
Article
Neglected Tropical Diseases: A Chemoinformatics Approach for the Use of Biodiversity in Anti-Trypanosomatid Drug Discovery
by Marilia Valli, Thiago H. Döring, Edgard Marx, Leonardo L. G. Ferreira, José L. Medina-Franco and Adriano D. Andricopulo
Biomolecules 2024, 14(8), 1033; https://doi.org/10.3390/biom14081033 - 20 Aug 2024
Cited by 1 | Viewed by 1704
Abstract
The development of new treatments for neglected tropical diseases (NTDs) remains a major challenge in the 21st century. In most cases, the available drugs are obsolete and have limitations in terms of efficacy and safety. The situation becomes even more complex when considering [...] Read more.
The development of new treatments for neglected tropical diseases (NTDs) remains a major challenge in the 21st century. In most cases, the available drugs are obsolete and have limitations in terms of efficacy and safety. The situation becomes even more complex when considering the low number of new chemical entities (NCEs) currently in use in advanced clinical trials for most of these diseases. Natural products (NPs) are valuable sources of hits and lead compounds with privileged scaffolds for the discovery of new bioactive molecules. Considering the relevance of biodiversity for drug discovery, a chemoinformatics analysis was conducted on a compound dataset of NPs with anti-trypanosomatid activity reported in 497 research articles from 2019 to 2024. Structures corresponding to different metabolic classes were identified, including terpenoids, benzoic acids, benzenoids, steroids, alkaloids, phenylpropanoids, peptides, flavonoids, polyketides, lignans, cytochalasins, and naphthoquinones. This unique collection of NPs occupies regions of the chemical space with drug-like properties that are relevant to anti-trypanosomatid drug discovery. The gathered information greatly enhanced our understanding of biologically relevant chemical classes, structural features, and physicochemical properties. These results can be useful in guiding future medicinal chemistry efforts for the development of NP-inspired NCEs to treat NTDs caused by trypanosomatid parasites. Full article
Show Figures

Figure 1

Figure 1
<p>Drugs used for NTDs: (<b>A</b>) drugs for trypanosomatid diseases, (<b>B</b>) drugs for other NTDs.</p>
Full article ">Figure 2
<p>Current clinical pipeline for Chagas disease, leishmaniasis, and HAT: (<b>A</b>) advanced clinical trials, (<b>B</b>) early stages of clinical development. DNDi-2319 uppercase: phosphothionate bases; lowercase: phosphodiester bases. VL: visceral leishmaniasis; PKDL: post-kala-azar dermal leishmaniasis; CL: cutaneous leishmaniasis; LAmB: liposomal amphotericin B.</p>
Full article ">Figure 3
<p>Strategy used to build the dataset used in this study.</p>
Full article ">Figure 4
<p>Profile of the dataset with 678 compounds regarding ring count, structural alerts, and calculated synthetic accessibility: (<b>A</b>) ring count considering any ring size, (<b>B</b>) number of rings in each structure of the dataset, (<b>C</b>) Brenk structural alerts, (<b>D</b>) synthetic accessibility scores using the SwissADME webserver (University of Lausanne and the SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland).</p>
Full article ">Figure 5
<p>Investigation of molecular properties and structural complexity of the dataset compounds: (<b>A</b>) violations of Lipinski’s rule of five, (<b>B</b>) number of stereogenic centers.</p>
Full article ">Figure 6
<p>Chemical diversity analysis of the dataset: (<b>A</b>) structural similarity chart (centroid clustered) indicating the most important regions of similarity (from dark blue to dark red, respectively, 0% to 100% similarity), (<b>B</b>) 3D PCA showing the chemical diversity of the NPs with their corresponding source in distinct colors. The first three components capture 94.8% of the total variance.</p>
Full article ">Figure 7
<p>Scatter plots of associations between lipophilicity (clogP) and molecular weight (MW) and biological activity (IC<sub>50</sub>): (<b>A</b>) clogP versus MW for the entire dataset and (<b>B</b>) clogP versus IC<sub>50</sub> values for a subset of 243 compounds with anti-<span class="html-italic">T. cruzi</span> activity.</p>
Full article ">Figure 8
<p>Box plot of the distribution of MW versus water solubility scores (insoluble &lt; −10 &lt; poorly &lt; −6 &lt; moderately &lt; −4 &lt; soluble &lt; −2 &lt; very &lt; 0 &lt; highly). Red lines indicate the mean, black lines indicate the median, and dots indicate the outliers. Dashed lines indicate the upper and lower quartiles.</p>
Full article ">Figure 9
<p>2D PCA performed using rotatable bonds (nRotB), hydrogen-bond acceptors (HBA), hydrogen-bond donors (HBD), and molecular weight (MW). Solid dot colors represent nRotB and smooth colors represent the fraction of sp<sup>3</sup> hybridized carbon atoms related to the total carbon count (Csp<sup>3</sup>). # = number.</p>
Full article ">Figure 10
<p>(<b>A</b>) Structures of leucinostatin A (<b>89</b>), leucinostatin B (<b>90</b>), and leucinostatin F (<b>91</b>), (<b>B</b>) similarity network for leucinostatin F (<b>91</b>).</p>
Full article ">Figure 11
<p>Structure of drug-like compounds <b>76</b>, <b>154</b>, <b>155</b>, <b>33,</b> and <b>355</b>.</p>
Full article ">Figure 12
<p>(<b>A</b>) Structure of CHT (<b>97</b>), ERGT (<b>98</b>), sesquiterpene (<b>126</b>), β-sitosterol (<b>234</b>) and stigmasterol (<b>235</b>), (<b>B</b>) similarity network for compound <b>97</b>.</p>
Full article ">Figure 13
<p>(<b>A</b>) Structure of compound <b>197</b>, a chalcone derivative, (<b>B</b>) similarity chart for compound <b>197</b>.</p>
Full article ">Figure 14
<p>(<b>A</b>) Structure of compound <b>567</b> (<b>B</b>) similarity chart for compound <b>567</b>.</p>
Full article ">Figure 15
<p>Structure of drug-like terpenoids <b>418</b>, <b>420,</b> and <b>596</b>.</p>
Full article ">
10 pages, 416 KiB  
Article
Molecular Detection and Analysis of Trypanosoma (Megatrypanum) spp. Diversity in Tabanidae (Diptera) Collected in Lithuania
by Jurga Turčinavičienė, Rasa Bernotienė and Andrius Petrašiūnas
Insects 2024, 15(8), 581; https://doi.org/10.3390/insects15080581 - 30 Jul 2024
Viewed by 840
Abstract
Trypanosomatids from the Tabanidae family have not been studied in Lithuania in any detail. In this study, a nested PCR amplifying the DNA fragment coding the SSU rRNA was used to determine the Trypanosoma spp. prevalence and diversity in the Tabanidae family collected [...] Read more.
Trypanosomatids from the Tabanidae family have not been studied in Lithuania in any detail. In this study, a nested PCR amplifying the DNA fragment coding the SSU rRNA was used to determine the Trypanosoma spp. prevalence and diversity in the Tabanidae family collected in Lithuania in 2018–2019. In total, 101 Tabanidae individuals were investigated from six areas in Lithuania, and 14 different species were identified. The overall positivity of Trypanosoma spp. DNA in tabanids was 50.5% (51/101). Tabanus maculicornis was the most abundant species and yielded the highest prevalence of trypanosomatids (84.62%, 22/26), while Hybomitra nitidifrons showed a high prevalence as well, reaching 77.8% (14/18). In flies of some species (Hybomitra lapponica and Hybomitra lurida), Trypanosoma was detected for the first time. Nine different haplotypes were detected as being distributed in different tabanid species. Analysis showed that most sequences obtained during our study were identical or extremely close to two major T. theileri subclades: TthI and TthII. Our data analysis suggests the presence of different Trypanosoma genotypes in the same tabanid species, meaning that different lineages of Trypanosoma could be more related to the vertebrate host and not the fly species. This is the first study of trypanosomatid parasites in tabanids from Lithuania, and our results are valuable in providing data on the diversity of these parasites in different Tabanidae species. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Median-joining network of <span class="html-italic">Trypanosoma</span> spp. found in insects (tabanids, culicids and <span class="html-italic">Glossina fuscipes fuscipes</span>), <span class="html-italic">T</span>. <span class="html-italic">trinaperronei</span> found in white-tailed deer and <span class="html-italic">T. melophagium</span> inferred using SSU rRNA (748 bp) sequences and median-joining network method for intraspecific phylogenies. Network demonstrates evidence of TthI (circle 4) and TthII (circles 1 and 2) phylogenetic lineages, where most sequences belong. Numbers in circles correspond to those in <a href="#insects-15-00581-t001" class="html-table">Table 1</a>.</p>
Full article ">
23 pages, 2711 KiB  
Review
Targeting Trypanothione Metabolism in Trypanosomatids
by María-Cristina González-Montero, Julia Andrés-Rodríguez, Nerea García-Fernández, Yolanda Pérez-Pertejo, Rosa M. Reguera, Rafael Balaña-Fouce and Carlos García-Estrada
Molecules 2024, 29(10), 2214; https://doi.org/10.3390/molecules29102214 - 9 May 2024
Cited by 3 | Viewed by 1637
Abstract
Infectious diseases caused by trypanosomatids, including African trypanosomiasis (sleeping sickness), Chagas disease, and different forms of leishmaniasis, are Neglected Tropical Diseases affecting millions of people worldwide, mainly in vulnerable territories of tropical and subtropical areas. In general, current treatments against these diseases are [...] Read more.
Infectious diseases caused by trypanosomatids, including African trypanosomiasis (sleeping sickness), Chagas disease, and different forms of leishmaniasis, are Neglected Tropical Diseases affecting millions of people worldwide, mainly in vulnerable territories of tropical and subtropical areas. In general, current treatments against these diseases are old-fashioned, showing adverse effects and loss of efficacy due to misuse or overuse, thus leading to the emergence of resistance. For these reasons, searching for new antitrypanosomatid drugs has become an urgent necessity, and different metabolic pathways have been studied as potential drug targets against these parasites. Considering that trypanosomatids possess a unique redox pathway based on the trypanothione molecule absent in the mammalian host, the key enzymes involved in trypanothione metabolism, trypanothione reductase and trypanothione synthetase, have been studied in detail as druggable targets. In this review, we summarize some of the recent findings on the molecules inhibiting these two essential enzymes for Trypanosoma and Leishmania viability. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Schematic representation of a trypanosomatid amastigote. The main metabolic pathways that have been considered as potential drug targets are indicated in different colors: polyamine metabolism (blue color); methyl cycle metabolism (green color); modification of the topological state of DNA (yellow color); redox metabolism (red color); sterol biosyntyhesis (black color); folate metabolism (purple color); purine salvage pathway (orange color); folate metabolism (gray color). Abbreviations: AdoMet: S-adenosylmethionine; C: cytosol; dcAdoMet: decarboxylated AdoMet; ERG: ergosterol; FOL: folate; G: glycosome; GLY: glycolysis; K: kinetoplast; M: mitochondrion; N: nucleus; PUT: putrescine; PSP: purine salvage pathway; Spd: spermidine; TOP: DNA topoisomerase; T(SH)<sub>2</sub>: reduced trypanothione; TS<sub>2</sub>: oxidized trypanothione.</p>
Full article ">Figure 2
<p>Biochemical structure of trypanothione in the reduced [T(SH)<sub>2</sub>] and oxidized [TS<sub>2</sub>] forms. The spermidine molecule joining two molecules of glutathione is highlighted in blue.</p>
Full article ">Figure 3
<p>Biosynthesis of trypanothione (blue color) and antioxidant pathways (purple color) in trypanosomatids. AdoMetDC: S-adenosylmethionine decarboxylase; APx: ascorbate-dependent heme-peroxidase; ARG: arginase (not in <span class="html-italic">T. cruzi</span> and <span class="html-italic">T. brucei</span>); CBS: cystathione beta-synthase; CGL: cystathionine gamma-lyase; CS: cysteine synthase; FeSODs: iron–superoxyde dismutases; GPx-I and GPx-II: non-selenium glutathione peroxidases; GSH: reduced glutathione; GSH1: γ-glutamylcysteine synthetase; GSH2: glutathione synthetase; GSP: mono-glutathionylspermidine; GspS: GSP synthetase; GSSG: oxidized glutathione; MAT: methionine adenosyl transferase; MST: mercaptopyruvate sulfurtransferase; ODC: ornithine decarboxylase (not in <span class="html-italic">T. cruzi</span>); RR: ribonucleotide reductase; SAT: serine acetyltransferase; SpS: spermidine synthase; TryR: trypanothione reductase; TryS: trypanothione synthetase; TXNPx: tryparedoxin peroxidase (figure adapted from [<a href="#B7-molecules-29-02214" class="html-bibr">7</a>,<a href="#B8-molecules-29-02214" class="html-bibr">8</a>,<a href="#B71-molecules-29-02214" class="html-bibr">71</a>]).</p>
Full article ">Figure 4
<p>Chemical structure of different TryR inhibitors binding to the wide TS<sub>2</sub> cavity.</p>
Full article ">Figure 5
<p>Chemical structures of different TryR inhibitors binding to the Cys52 and Cys57 in the catalytic site, or the NADPH-binding cavity, or disassembling the dimer, or acting as mixed inhibitors.</p>
Full article ">Figure 6
<p>Chemical structures of different “subversive” substrates for TryR.</p>
Full article ">Figure 7
<p>Chemical structures of different TryS inhibitors.</p>
Full article ">
13 pages, 3232 KiB  
Article
Emergent and Known Honey Bee Pathogens through Passive Surveillance in the Republic of Kosovo
by Beqë Hulaj, Anna Granato, Fulvio Bordin, Izedin Goga, Xhavit Merovci, Mauro Caldon, Armend Cana, Laura Zulian, Rosa Colamonico and Franco Mutinelli
Appl. Sci. 2024, 14(3), 987; https://doi.org/10.3390/app14030987 - 24 Jan 2024
Cited by 1 | Viewed by 1646
Abstract
In recent years, honey bee colony losses in the Republic of Kosovo remained largely unknown. From 2019 to 2021, 81 apiaries with different disease suspicions were investigated in the framework of honey bee disease passive surveillance. Fifty-nine of the eighty-one apiaries were tested [...] Read more.
In recent years, honey bee colony losses in the Republic of Kosovo remained largely unknown. From 2019 to 2021, 81 apiaries with different disease suspicions were investigated in the framework of honey bee disease passive surveillance. Fifty-nine of the eighty-one apiaries were tested for Vairimorpha ceranae, Vairimorpha apis, trypanosomatids Lotmaria passim, and Crithidia mellificae. All samples were positive for V. ceranae (100%) whereas L. passim was found with a lower frequency (11.9%). V. apis and C. mellificae were not found. Thirteen of the eighty-one apiaries were tested for seven viruses (ABPV, CBPV, DWV, BQCV, SBV, IAPV, KBV) and five of them were found (ABPV, CBPV, DWV, BQCV, SBV). The most frequently detected viruses in honey bees and Varroa mites were DWV (100%) followed by BQCV, ABPV, SBV, and CBPV (92.3%, 69.2%, 30.8%, and 7.7%, respectively). Varroa mite samples had different degrees of co-infection by viruses. Nine of the eighty-one apiaries consisted of brood combs with larvae, eight of them were AFB positive, ERIC I genotype, and one EFB positive. This paper represents the first molecular investigation (PCR) and detection of the honey bee viruses ABPV, CBPV, DWV, BQCV, and SBV as well as V. ceranae, L. passim, and M. plutonius in the Republic of Kosovo. Full article
(This article belongs to the Section Food Science and Technology)
Show Figures

Figure 1

Figure 1
<p>Distribution of detected pathogens by municipality in the Republic of Kosovo. The numbers correspond to the 25 involved municipalities (1—Suharekë; 2—Shtime; 3—Prishtinë; 4—Deҫan; 5—Kamenicë; 6—Podujevë; 7—Vushtrri; 8—Drenas; 9—Ferizaj; 10—Prizren; 11—Mitrovicë; 12—Novobërdë; 13—Hani i Elezit; 14—Malishevë; 15—Gjilan; 16—Lipjan; 17—Graqanicë; 18—Skenderaj; 19—Viti; 20—Fushë Kosovë; 21—Obiliq; 22—Pejë; 23—Gjakovë; 24—Junik; 25—Istog). Each symbol indicates the presence of a different pathogen individually or in combination. Their percentage of detection is also indicated.</p>
Full article ">Figure 2
<p>Distribution of detected honey bee viruses in <span class="html-italic">Varroa</span> mites by municipality in the Republic of Kosovo. The numbers correspond to the 25 involved municipalities. Each symbol indicates the presence of a different virus detected in <span class="html-italic">Varroa</span> specimens. Their percentage of detection is also indicated.</p>
Full article ">Figure 3
<p>PCR products of eight bee pathogens after 7% acrylamide gel electrophoresis and silver nitrate staining. In this figure, PCR products from some of the analysed apiaries are shown: Suharekë, Drenas, Prizren, Podujevë, Novobërdë, Deçan, Malishevë, Viti. Eight bee pathogens were amplified by PCR: (<b>A</b>) <span class="html-italic">V. ceranae</span> (218 bp); (<b>B</b>) <span class="html-italic">V. apis</span> (321 bp); (<b>C</b>) <span class="html-italic">L. passim</span> (254 bp); (<b>D</b>) <span class="html-italic">P. larvae</span> (1096 bp); (<b>E</b>) <span class="html-italic">C. mellificae</span> (177 bp); (<b>F</b>) IAPV (767 bp); (<b>G</b>) KBV (659 bp); (<b>H</b>) <span class="html-italic">M. plutonius</span> (832 bp). M: 100 bp DNA Ladder (Invitrogen™); PTC: pathogen positive control; NTC: no template control.</p>
Full article ">
26 pages, 2847 KiB  
Review
Polyamine Metabolism for Drug Intervention in Trypanosomatids
by Yolanda Pérez-Pertejo, Carlos García-Estrada, María Martínez-Valladares, Sankaranarayanan Murugesan, Rosa M. Reguera and Rafael Balaña-Fouce
Pathogens 2024, 13(1), 79; https://doi.org/10.3390/pathogens13010079 - 16 Jan 2024
Cited by 3 | Viewed by 2235
Abstract
Neglected tropical diseases transmitted by trypanosomatids include three major human scourges that globally affect the world’s poorest people: African trypanosomiasis or sleeping sickness, American trypanosomiasis or Chagas disease and different types of leishmaniasis. Different metabolic pathways have been targeted to find antitrypanosomatid drugs, [...] Read more.
Neglected tropical diseases transmitted by trypanosomatids include three major human scourges that globally affect the world’s poorest people: African trypanosomiasis or sleeping sickness, American trypanosomiasis or Chagas disease and different types of leishmaniasis. Different metabolic pathways have been targeted to find antitrypanosomatid drugs, including polyamine metabolism. Since their discovery, the naturally occurring polyamines, putrescine, spermidine and spermine, have been considered important metabolites involved in cell growth. With a complex metabolism involving biosynthesis, catabolism and interconversion, the synthesis of putrescine and spermidine was targeted by thousands of compounds in an effort to produce cell growth blockade in tumor and infectious processes with limited success. However, the discovery of eflornithine (DFMO) as a curative drug against sleeping sickness encouraged researchers to develop new molecules against these diseases. Polyamine synthesis inhibitors have also provided insight into the peculiarities of this pathway between the host and the parasite, and also among different trypanosomatid species, thus allowing the search for new specific chemical entities aimed to treat these diseases and leading to the investigation of target-based scaffolds. The main molecular targets include the enzymes involved in polyamine biosynthesis (ornithine decarboxylase, S-adenosylmethionine decarboxylase and spermidine synthase), enzymes participating in their uptake from the environment, and the enzymes involved in the redox balance of the parasite. In this review, we summarize the research behind polyamine-based treatments, the current trends, and the main challenges in this field. Full article
(This article belongs to the Special Issue Leishmaniasis: Transmission, Pathogenesis and Treatment)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Chemical structure of DFMO (eflornithine) and mechanism of action as irreversible inhibitor of ornithine decarboxylase (ODC) in <span class="html-italic">T. brucei</span>.</p>
Full article ">Figure 2
<p>Schematic representation of a bloodstream <span class="html-italic">T. brucei</span> trypomastigote (<b>left picture</b>) and an intracellular <span class="html-italic">T. cruzi</span> and <span class="html-italic">Leishmania</span> amastigote (<b>right picture</b>). The major metabolic pathways that have been considered as potential drug targets are indicated. Some organelles are also represented: C: cytosol; G: glycosome; K: kinetoplast; M: mitochondrion; N: nucleus.</p>
Full article ">Figure 3
<p>The canonic polyamine biosynthetic pathway of mammals (hosts of trypanosomatids). The enzymes involved in polyamine biosynthesis are shown in black boxes, and the metabolites are in empty circles. The full polyamine metabolic pathway includes biosynthesis and interconversion routes that starts from the essential amino acid L-methionine and from the semi-essential L-arginine. Putrescine, spermidine and spermine are synthesized from L-ornithine and AdoMetDc after decarboxylation, the activity of both enzymes being closely regulated by the cells. Both enzymes have been addressed as druggable targets for therapeutic intervention. Note that regardless of trypanosomatids, there is not any connection between polyamines and glutathione in mammals. Abbreviations of the enzymes and transporters and their corresponding EC numbers are given in alphabetic order: AdoMetDC, S-adenosylmethione decarboxylase (EC 4.1.1.50); APAO: N 1-acetylpolyamine oxidase (EC 1.5.3.13); Arginase (EC 3.5.3.1); DHS, deoxyhypusine synthase (EC: 2.5.1.46); DOHH deoxyhypusine hydroxylase (EC 1.14.99.29); MAT, methionine adenosyltransferase (EC 2.5.1.6); ODC, ornithine decarboxylase (EC 4.1.1.17); POT1, putrescine transport 1 (EC 7.6.2.16); SpdS, spermidine synthase (EC 2.5.1.16); SpmS, spermine synthase (EC 2.5.1.22). Abbreviations for the metabolites are also given in alphabetic order: AdoMet, S-adenosylmethionine; dcAdoMet, decarboxylated S-adenosylmethionine; eIF5A, eukaryotic translation initiation factor 5A; dh-eIF5A, deoxyhypusine-eIF5A; h-eIF5A, hypusine-eIF5A.</p>
Full article ">Figure 4
<p>The polyamine biosynthetic pathway of trypanosomatid (African, American trypanosomes and <span class="html-italic">Leishmania</span>) is much simpler than the host pathway and lacks spermine and the whole interconversion pathway. However, it is in part devoted to ROS detoxification by means of the conjugation of spermidine to glutathione to form trypanothione (see <a href="#pathogens-13-00079-f003" class="html-fig">Figure 3</a>). Polyamine biosynthesis has some specific peculiarities involving: the lack of a true arginase in <span class="html-italic">T. brucei</span> and <span class="html-italic">T. cruzi</span> and the lack of the <span class="html-italic">odc</span> encoding gene in <span class="html-italic">T. cruzi</span> that make this parasite auxotrophic for putrescine and/or spermidine. Abbreviations of the enzymes and transporters and their corresponding EC numbers are given in alphabetic order: AdoMetDC, S-adenosylmethione decarboxylase (EC 4.1.1.50); Arginase (EC 3.5.3.1); DHS, deoxyhypusine synthase (EC: 2.5.1.46); DOHH deoxyhypusine hydroxylase (EC 1.14.99.29); MAT, methionine adenosyltransferase (EC 2.5.1.6); ODC, ornithine decarboxylase (EC 4.1.1.17); POT1, putrescine transport 1 (EC 7.6.2.16); SpdS, spermidine synthase (EC 2.5.1.16); Abbreviations for the metabolites are also given in alphabetic order: AdoMet, S-adenosylmethionine; dcAdoMet, decarboxylated S-adenosylmethionine; eIF5A, eukaryotic translation initiation factor 5A; dh-eIF5A, deoxyhypusine-eIF5A; h-eIF5A, hypusine-eIF5A.</p>
Full article ">Figure 5
<p>Host–parasite interplay in <span class="html-italic">Leishmania</span> infections. L-arginine plays a key role in macrophage activation and parasite survival during infection. Within the host macrophage, L-arginine is substrate of inducible nitric oxide synthase (iNOS) and arginase. iNOS/arginase balance is transcriptionally controlled by interleukins and is modulated at a biochemical level too. On the one hand, the classic M1 proinflammatory activation of macrophages responds to Th1 cytokines such as TNFa and IFNg and IL1, IL2 and IL10 interleukins. Alternative M2 anti-inflammatory activation of macrophages responds to Th2 cytokines such as TGFb, IL4, IL10 and IL13. M1 activation induces iNOS in macrophages, responsible for L-arginine cleavage to NO and citrulline byproduct. NO will promote the cascade production of nitrogen reactive species (RNOS) such as peroxynitrite radical (ONOO-). On the other hand, L-arginine cleavage by iNOS is a two-step enzymatic process that produces an important intermediate <span class="html-italic">N</span>-hydroxy-L-arginine (NOHA), which, before its complete hydrolysis to NO, can interfere arginase activity, preventing L-ornithine and polyamine production. Th2 response implies an increase in arginase activity, resulting in the formation of L-ornithine and polyamines, which can be used by the parasite. Inside the parasitophorous vacuole, <span class="html-italic">Leishmania</span> amastigotes can obtain L-arginine and polyamine from the host using the corresponding active transporters. <span class="html-italic">Leishmania</span> can synthesize putrescine and spermidine from L-arginine and L-methionine, which are essentials for the parasite, but <span class="html-italic">T. cruzi</span> is auxothroph for putrescine since it lacks genes encoding for both, a true arginase and ODC. * Absent in <span class="html-italic">T. brucei</span> and <span class="html-italic">T. cruzi</span>; ** Absent in <span class="html-italic">T. cruzi</span>.</p>
Full article ">Figure 6
<p>Trypanothione biosynthetic pathway and redox balance in trypanosomatids. Reduced trypanothione is a complex formed by two molecules of reduced glutathione bridged by their glycine residues with spermidine. Trypanothione can be oxidized by ROS to the oxidized form, establishing a sulfur redox balance enzymatically controlled by trypanothione reductase. The singularity of this ROS scavenger in trypanosomatids is an interesting druggable target for drug intervention in trypanosomatids. Abbreviations of the enzymes and transporters and their corresponding EC numbers are given in alphabetic order: <span class="html-italic">γ</span>GCS, <span class="html-italic">γ</span>-glutamylcysteine synthetase (EC 6.3.2.2); GS, glutathione synthase (EC 6.3.2.3); GSS, glutathionyl spermidine synthetase (EC 6.3.1.8); TryR, trypanothione reductase (EC 1.8.1.12); TryS, trypanothione synthase (EC 6.3.1.9). Abbreviations for the metabolites are also given in alphabetic order: <span class="html-italic">γ</span>GC, <span class="html-italic">γ</span>-glutamylcysteine; Gsp, glutathionylspermidine; GSH, reduced glutathione; ROS, reactive oxygen species; T(SH)2, reduced trypanothione; TS2, oxidized trypanothione.</p>
Full article ">Figure 7
<p>ODC inhibitors. DFMO (<b>1</b>) and other different chemical scaffolds inhibiting <span class="html-italic">Trypanosoma</span> ODC.</p>
Full article ">Figure 8
<p>Polyamine uptake inhibitors. MGBG (<b>5</b>), the diamidines CGP40215A (<b>6</b>) and CGP48664A (<b>7</b>), MDL 73811 (<b>8</b>), UTSam568 (<b>9</b>).</p>
Full article ">Figure 9
<p>AdoMetDC inhibitors. Triclabendazole (<b>10</b>), isotretinoin (<b>11</b>)<b>,</b> promazine (<b>12</b>), chlorpromazine (<b>13</b>) and chlomipramine (<b>14</b>).</p>
Full article ">Figure 10
<p>Polyamine transport system inhibitors. Ant4 (<b>15</b>), GW5074 (<b>16</b>).</p>
Full article ">
14 pages, 2631 KiB  
Article
Solid Lipid Nanoparticles Enhancing the Leishmanicidal Activity of Delamanid
by Javier Santamaría-Aguirre, Daniela Jacho, Miguel A. Méndez, Ana Poveda, Javier Carrión and Mónica L. Fanarraga
Pharmaceutics 2024, 16(1), 41; https://doi.org/10.3390/pharmaceutics16010041 - 27 Dec 2023
Cited by 1 | Viewed by 2263
Abstract
Leishmaniasis, a zoonotic parasitic disease transmitted by infected sandflies, impacts nearly 1 million people yearly and is endemic in many countries across Asia, Africa, the Americas, and the Mediterranean; despite this, it remains a neglected disease with limited effective treatments, particularly in impoverished [...] Read more.
Leishmaniasis, a zoonotic parasitic disease transmitted by infected sandflies, impacts nearly 1 million people yearly and is endemic in many countries across Asia, Africa, the Americas, and the Mediterranean; despite this, it remains a neglected disease with limited effective treatments, particularly in impoverished communities with limited access to healthcare. This study aims to repurpose approved drugs for an affordable leishmaniasis treatment. After the screening of potential drug candidates by reviewing databases and utilizing molecular docking analysis, delamanid was chosen to be incorporated into solid lipid nanoparticles (SLNPs). Both in cellulo and in vivo tests confirmed the successful payload release within macrophages and through the epidermis following topical application on murine skin. The evaluation of macrophages infected with L. infantum amastigotes showed that the encapsulated delamanid exhibited greater leishmanicidal activity compared with the free drug. The process of encapsulating delamanid in SLNPs, as demonstrated in this study, places a strong emphasis on employing minimal technology, ensuring energy efficiency, cost-effectiveness, and reproducibility. It enables consistent, low-cost production of nanomedicines, even on a small scale, offering a promising step toward more accessible and effective leishmaniasis treatments. Full article
(This article belongs to the Section Drug Targeting and Design)
Show Figures

Figure 1

Figure 1
<p>Molecular structure of delamanid and impact on promastigotes. (<b>a</b>) Chemical structure of delamanid, a bicyclic nitroimidazole compound approved for the treatment of multi-drug resistant tuberculosis, exhibiting high activity against <span class="html-italic">Leishmania</span> parasites. (<b>b</b>) Untreated <span class="html-italic">L. infantum</span> promastigotes after 7-day culture. (<b>c</b>) Morphological alterations induced by delamanid in <span class="html-italic">L. infantum</span> promastigotes after 2 days of treatment.</p>
Full article ">Figure 2
<p>SLNP synthesis and characterization. (<b>a</b>) Schematic representation of SLNP synthesis using the solvent–antisolvent method. (<b>b</b>) Characterization of hydrodynamic diameter. Data from three distinct batches (in blue, green and orange colors) exhibit high reproducibility in particle size, as determined using Dynamic Light Scattering (DLS), with a polydispersion index below 0.25. (<b>c</b>) In vitro drug release profiles indicate sustained release over one week. (<b>d</b>) The SLNP dispersion was diluted at a ratio of 1 to 10 in PBS, applied to a grid, and left to air-dry at room temperature overnight before observation under TEM.</p>
Full article ">Figure 3
<p>Liberation of payload from SLNPs containing dye and delamanid. (<b>a</b>) After 24 h of exposure to SLNPs, the macrophages display robust cytoplasmic fluorescence, indicating the successful release of the encapsulated dye intracellularly (Inset #1). (<b>b</b>–<b>d</b>) Confocal microscopy images of skin sections taken at sequential time points after the topical administration of the SLNPs show distinct patterns. (<b>b</b>) Immediately after application, the released compound is visible in the corneal stratum (Inset #2, arrow). (<b>c</b>) In 2 h, the dye penetrated the basal layer of the dermis, which was visible intracellularly (Inset #3, arrow). (<b>d</b>) A notable concentration of the dye around the hair follicles is detected at 24 h. The red channel shows the fluorescence of the dye (Inset #4, arrows). Nuclei were stained with Dapi (blue channel). The green channel represents tissue autofluorescence.</p>
Full article ">Figure 4
<p>Effect of SLNP Treatment on Amastigotes. (<b>a</b>,<b>b</b>) Subcellular localization of SLNPs in infected macrophages. TEM images of an ultrathin section of an infected macrophage treated with SLNPs. Black arrows delineate the localization of intracellular dead parasites. Insets #5a and #5b offer magnified views of the areas enclosed in Inset #5. Inset #6 highlights the specified region in (<b>b</b>). White arrows indicate intracellular SLNPs, some observed within a putative phagosome (Inset #5a), while others are freely present in the cytoplasm (Insets #5b, #6). The presence of magnetic nanoparticles inside serves as the identifier for the SLNPs. (<b>c</b>) The use of SLNPs containing delamanid (SLNP Dm) significantly reduces the Infection Index of macrophages infected with <span class="html-italic">L. infantum</span> in comparison with the control groups, which include unloaded SLNPs and the plain drug (Dm). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
7 pages, 254 KiB  
Editorial
Antileishmanial and Antitrypanosomes Drugs for the Current Century
by José María Alunda
Microorganisms 2024, 12(1), 43; https://doi.org/10.3390/microorganisms12010043 - 26 Dec 2023
Viewed by 1455
Abstract
Human infections by trypanosomatids are widely distributed and prevalent in the tropical and subtropical regions. Diseases caused by Trypanosoma and Leishmania have variable clinical outcomes, ranging from self-healing to fatality, and are considered Neglected Tropical Diseases (NTD). In addition, animal trypanosomiases have a [...] Read more.
Human infections by trypanosomatids are widely distributed and prevalent in the tropical and subtropical regions. Diseases caused by Trypanosoma and Leishmania have variable clinical outcomes, ranging from self-healing to fatality, and are considered Neglected Tropical Diseases (NTD). In addition, animal trypanosomiases have a significant impact on animal health and production, apart from their potential role as reservoirs in zoonotic species. Control of these infections is progressing and, in some cases (such as human African trypanomiasis (HAT)), significant reductions have been achieved. In the absence of effective vaccination, chemotherapy is the most used control method. Unfortunately, the therapeutic arsenal is scarce, old, and of variable efficacy, and reports of resistance to most antiparasitic agents have been published. New drugs, formulations, or combinations are needed to successfully limit the spread and severity of these diseases within a One Health framework. In this Special Issue, contributions regarding the identification and validation of drug targets, underlying mechanisms of action and resistance, and potential new molecules are presented. These research contributions are complemented by an update revision of the current chemotherapy against African Trypanosoma species, and a critical review of the shortcomings of the prevailing model of drug discovery and development. Full article
16 pages, 4495 KiB  
Article
Safety and Immunogenicity of an In Vivo Muscle Electroporation Delivery System for DNA-hsp65 Tuberculosis Vaccine in Cynomolgus Monkeys
by Monique Ribeiro de Lima, Ana Cristina C. S. Leandro, Andreia Lamoglia de Souza, Marcio Mantuano Barradas, Eric Henrique Roma, Ana Teresa Gomes Fernandes, Gabrielle Galdino-Silva, Joyce Katiuccia M. Ramos Carvalho, Renato Sergio Marchevsky, Janice M. C. Oliveira Coelho, Eduardo Dantas Casillo Gonçalves, John L. VandeBerg, Celio Lopes Silva and Maria da Gloria Bonecini-Almeida
Vaccines 2023, 11(12), 1863; https://doi.org/10.3390/vaccines11121863 - 18 Dec 2023
Cited by 1 | Viewed by 1980
Abstract
A Bacille Calmette–Guérin (BCG) is still the only licensed vaccine for the prevention of tuberculosis, providing limited protection against Mycobacterium tuberculosis infection in adulthood. New advances in the delivery of DNA vaccines by electroporation have been made in the past decade. We evaluated [...] Read more.
A Bacille Calmette–Guérin (BCG) is still the only licensed vaccine for the prevention of tuberculosis, providing limited protection against Mycobacterium tuberculosis infection in adulthood. New advances in the delivery of DNA vaccines by electroporation have been made in the past decade. We evaluated the safety and immunogenicity of the DNA-hsp65 vaccine administered by intramuscular electroporation (EP) in cynomolgus macaques. Animals received three doses of DNA-hsp65 at 30-day intervals. We demonstrated that intramuscular electroporated DNA-hsp65 vaccine immunization of cynomolgus macaques was safe, and there were no vaccine-related effects on hematological, renal, or hepatic profiles, compared to the pre-vaccination parameters. No tuberculin skin test conversion nor lung X-ray alteration was identified. Further, low and transient peripheral cellular immune response and cytokine expression were observed, primarily after the third dose of the DNA-hsp65 vaccine. Electroporated DNA-hsp65 vaccination is safe but provides limited enhancement of peripheral cellular immune responses. Preclinical vaccine trials with DNA-hsp65 delivered via EP may include a combination of plasmid cytokine adjuvant and/or protein prime–boost regimen, to help the induction of a stronger cellular immune response. Full article
(This article belongs to the Special Issue Research Progress of New Tuberculosis Vaccines and Vaccine Design)
Show Figures

Figure 1

Figure 1
<p>Vaccination scheme. Cynomolgus macaques were immunized with electroporated DNA-<span class="html-italic">hsp65</span> vaccine delivered in three doses at 30-day intervals by <span class="html-italic">in vivo</span> muscular electroporation. Clinical, hematological, biochemical, and immunological profiles were evaluated as represented.</p>
Full article ">Figure 2
<p>Tuberculin skin test. Cynomolgus macaques were immunized with electroporated DNA-<span class="html-italic">hsp65</span> vaccine delivered in three doses at 30-day intervals by <span class="html-italic">in vivo</span> muscular electroporation. Reaction to the tuberculin skin test was performed 30 days after the last dose of the vaccine. (<b>A</b>) No reaction (grade zero), (<b>B</b>) bruises (grade 1), and (<b>C</b>) number and frequency of reactive tuberculin skin tests.</p>
Full article ">Figure 3
<p>Frequency of immune cells in the peripheral blood. Frequency of CD4<sup>+</sup> and CD8<sup>+</sup> T cells, B cells, NK cells, myeloid dendritic and plasmocytoid dendritic cells in unvaccinated (open circles) and electroporated DNA-<span class="html-italic">hsp65</span> vaccinated (black circles) cynomolgus macaques. Arrows represent the three doses of vaccine.</p>
Full article ">Figure 4
<p>Frequency of central and effector memory cells in the peripheral blood. Frequency of CD4<sup>+</sup> and CD8<sup>+</sup> T CM and EF cells in unvaccinated (open circles) and electroporated DNA-<span class="html-italic">hsp65</span> vaccinated (black circles) cynomolgus macaques. Arrows represent the three doses of vaccine.</p>
Full article ">Figure 5
<p>Frequency of activation marker CD69<sup>+</sup> in peripheral blood. Frequency of CD4<sup>+</sup>/CD69<sup>+</sup>, CD8<sup>+</sup>/CD69<sup>+</sup>, and CD19<sup>+</sup>/CD69<sup>+</sup> cells in unvaccinated (open circles) and electroporated DNA-<span class="html-italic">hsp65</span> vaccinated (black circles) cynomolgus macaques. Arrows represent the three doses of vaccine.</p>
Full article ">Figure 6
<p>Frequency of proliferation, lytic and apoptotic markers in peripheral blood. Frequency of CD4<sup>+</sup>, CD8<sup>+</sup>, and CD19<sup>+</sup> cells expressing Ki67<sup>+</sup>, BCL2<sup>+</sup>, and granzyme/perforin markers in unvaccinated (open circles) and electroporated DNA-<span class="html-italic">hsp65</span> vaccinated (black circles) cynomolgus macaques. Arrows represent the three doses of vaccine.</p>
Full article ">Figure 7
<p>De novo cytokine expression in immune cells. Expression of IFN-γ, TNF-α, IL10, and IL-12 in CD4<sup>+</sup> and CD8<sup>+</sup> T cells, NK cells, and CD19<sup>+</sup> and CD14<sup>+</sup> cells was identified after stimulus of the cells with HSP65 peptide pool in unvaccinated (open circles) and electroporated DNA-<span class="html-italic">hsp65</span> vaccinated (black circles) cynomolgus macaques. Arrows represent the three doses of vaccine.</p>
Full article ">Figure 8
<p>De novo cytokines quantification in supernatant. TNF-α, IL-6, IL-5, IFN-γ, IL-2, and IL-4 were measured in the supernatant of stimulated cell culture with HSP65 peptides pool by CBA in unvaccinated (open circles) and electroporated DNA-<span class="html-italic">hsp65</span> vaccinated (black circles) cynomolgus macaques. Arrows represent the three doses of vaccine.</p>
Full article ">
24 pages, 5606 KiB  
Article
Implications of Flagellar Attachment Zone Proteins TcGP72 and TcFLA-1BP in Morphology, Proliferation, and Intracellular Dynamics in Trypanosoma cruzi
by Normanda Souza-Melo, Carolina de Lima Alcantara, Juliana Cunha Vidal, Gustavo Miranda Rocha and Wanderley de Souza
Pathogens 2023, 12(11), 1367; https://doi.org/10.3390/pathogens12111367 - 18 Nov 2023
Cited by 1 | Viewed by 1766
Abstract
The highly adaptable parasite Trypanosoma cruzi undergoes complex developmental stages to exploit host organisms effectively. Each stage involves the expression of specific proteins and precise intracellular structural organization. These morphological changes depend on key structures that control intracellular components’ growth and redistribution. In [...] Read more.
The highly adaptable parasite Trypanosoma cruzi undergoes complex developmental stages to exploit host organisms effectively. Each stage involves the expression of specific proteins and precise intracellular structural organization. These morphological changes depend on key structures that control intracellular components’ growth and redistribution. In trypanosomatids, the flagellar attachment zone (FAZ) connects the flagellum to the cell body and plays a pivotal role in cell expansion and structural rearrangement. While FAZ proteins are well-studied in other trypanosomatids, there is limited knowledge about specific components, organization, and function in T. cruzi. This study employed the CRISPR/Cas9 system to label endogenous genes and conduct deletions to characterize FAZ-specific proteins during epimastigote cell division and metacyclogenesis. In T. cruzi, these proteins exhibited distinct organization compared to their counterparts in T. brucei. TcGP72 is anchored to the flagellar membrane, while TcFLA-1BP is anchored to the membrane lining the cell body. We identified unique features in the organization and function of the FAZ in T. cruzi compared to other trypanosomatids. Deleting these proteins had varying effects on intracellular structures, cytokinesis, and metacyclogenesis. This study reveals specific variations that directly impact the success of cell division and differentiation of this parasite. Full article
Show Figures

Figure 1

Figure 1
<p>Bioinformatics analysis and domain organization of TcGP72 and TcFLA-1BP. (<b>A</b>) Phylogenetic tree from protein sequences of trypanosomatids. Amino acid sequences were aligned using Clustal Omega <a href="https://www.ebi.ac.uk/Tools/msa/clustalo/" target="_blank">https://www.ebi.ac.uk/Tools/msa/clustalo/</a> (accessed on 22 April 2020, and the aligned sequences were used to generate a phylogenetic tree (SeaView 5.4). (<b>B</b>) Diagram of the conserved domains found in TcGP72 and TcFLA-1BP orthologous to <span class="html-italic">Trypanosoma brucei</span>. TcGP72 contains a transmembrane domain (TM) at the N-terminus and an NHL-repeat in the extracellular domain. TcFLA-1BP is structured with a transmembrane domain (TM) at the N-terminus, followed by NHL-repeat-containing extracellular domain, TM, and a 45-amino acid tail (C45) at the C-terminus. Numbers indicate the amino acid residues. The domains in the diagram were labeled based on the information deposited in the TritrypDB database. Domain organization is shown for TcGP72 and FLA-1BP fused to c-Myc and mNeonGreen tags at the C-terminus (3xMyc::mNG::3xMyc), named TcGP72::mNG and TcFLA-1BP::mNG. Tb: <span class="html-italic">Trypanosoma brucei</span>. Tc: <span class="html-italic">Trypanosoma cruzi</span>. Tev: <span class="html-italic">Trypanosoma evansi</span>. TvY486: <span class="html-italic">Trypanosoma vivax</span>. DQO04: <span class="html-italic">Trypanosoma grayi</span>. ADEAN: <span class="html-italic">Angomonas deanei</span>. CFAC1: <span class="html-italic">Crithidia fasciculata</span>. LbrM: <span class="html-italic">Leishmania braziliensis</span>. LAMA: <span class="html-italic">Leishmania amazonensis</span>. LINF: <span class="html-italic">Leishmania infantum</span>. LdCL: <span class="html-italic">Leishmania donovani</span>. LmjF: <span class="html-italic">Leishmania major strain</span>.</p>
Full article ">Figure 2
<p>Analysis of the expression of fluorescent proteins. (<b>A</b>) Representative histograms showing the expression of fluorescent proteins in the mutant parasites. The peak with a black dashed line represents T7Cas9 lineage (control), and peaks with blue and red lines show TcGP72::mNG and TcFLA-1BP::mNG parasites, respectively. (<b>B</b>) Western blot analysis of fluorescent parasite (epimastigote) lysates expressing C-Myc and mNeonGreen fusion proteins (equivalent to 10<sup>7</sup> cells). Blots were probed with anti-Myc (AcMyc) antibodies and re-probed with anti-aldolase (AcAldo). kDa: molecular weight marker. The asterisk (*) shows the tagged protein bands. TcGP72::mNG: 98.2 kDa. TcFLA-1BP::mNG 116 kDa. TcAldolase: 29.3 kDa.</p>
Full article ">Figure 3
<p>Localization of TcGP72::mNG and TcFLA-1BP::mNG proteins. (<b>A</b>) The fluorescent parasites generated with CRISPR/Cas9 were analyzed with fluorescence microscopy. Direct mNeonGreen fluorescence (green) reveals the cellular distribution of TcGP72::mNG (<b>c</b>,<b>d</b>) and TcFLA-1BP::mNG (<b>e</b>,<b>f</b>). TcGP72::mNG localized with distributed reticulated on the cell body and flagellum. In contrast, TcFLA-1BP::mNG was localized in just the FAZ. To confirm the FAZ domain localization of the TcFLA-1BP, we performed TcGP72 knockout in the TcFLA-1BP::mNG parasite (TcFLA-1BP::mNGΔGP72). TcFLA-1BP::mNGΔGP72 (<b>g</b>,<b>h</b>) parasites showed the detached flagellum and tagging exclusively on the side of the cell body along the FAZ. DAPI for DNA (blue). K: kinetoplast, N: nucleus. (<b>B</b>) Scanning electron microscopy shows TcGP72::mNG parasite (<b>c</b>,<b>d</b>) morphology with flagellum detachment in contrast to the T7Cas9 control parasite (<b>a</b>,<b>b</b>). The dotted region shows the magnified area, and the yellow arrows show the FAZ.</p>
Full article ">Figure 4
<p>TcFLA-1BP targets the FAZ in amastigotes and trypomastigote (TCT) forms. (<b>A</b>) Fluorescence microscopy of TcFLA-1BP::mNG and T7Cas9 control in intracellular amastigotes (IAs) with FAZ labeled with anti-L3B2 (FAZ-1 <span class="html-italic">T. brucei</span>; red) and anti-mNG (mNeonGreen tag; green). (<b>B</b>) Localization in tissue culture derived TCTs. Nuclei and kinetoplasts were stained with Hoechst (blue). Scale bars: 2 and 5 µm.</p>
Full article ">Figure 5
<p>Depletion of TcGP72 and TcFLA−1BP. Illustration of gene knockout strategy (not to scale). (<b>A</b>) Diagram showing the TcGP72 and TcFLA-1BP locus and PCR primers (arrows) used to confirm the presence of the TcGP72 and TcFLA-1BP coding sequence (blue box) or the correct integration of the drug-resistant genes (green box). An arrowhead marks Cas9 cleavage sites. Yellow and purple boxes represent the untranslated regions (UTRs) of endogenous genes and donor DNAs. (<b>B</b>,<b>C</b>) PCR products were visualized on agarose gel. T7Cas9, parental cell line; TcGP72<sup>−/−</sup>, double knockout parasite to TcGP72; TcGP72<sup>−/−</sup>, double knockout parasite to TcFLA-1BP. P1 (forward primer) anneals to the 5′ UTR of TcGP72 or TcFLA-1BP. P2 (reverse primer) anneals to the coding sequence of TcGP72 or TcFLA-1BP. P5 (reverse primer) anneals to the end of the resistance gene coding sequence (blasticidin or hygromicin).</p>
Full article ">Figure 6
<p>TcFLA-1BP and TcGP72 knockout in <span class="html-italic">T. cruzi</span> causes partial or total flagellum detachment and impairs assembly of the flagellum, morphogenesis, organelle positioning, and cell growth. (<b>A</b>) Fluorescence microscopy of TcGP72::mNG and TcFLA-1BP::mNG with FAZ labeled with anti-L3B2 (FAZ-1 <span class="html-italic">T. brucei</span>; green) or anti-2F6 (PFR; red). Nuclei and kinetoplasts were stained with Hoechst (blue). Scale bars: 2 µm. T7Cas9 (<b>a</b>–<b>d</b>). TcGP72<sup>−/−</sup> (<b>e</b>–<b>h</b>). TcFLA-1BP<sup>−/−</sup> (<b>i</b>–<b>l</b>). (<b>B</b>) SEM showing the morphology of knockout parasites. TcGP72<sup>−/−</sup> (<b>c</b>,<b>d</b>) with total flagellum detachment and TcFLA-1BP<sup>−/−</sup> (<b>e</b>,<b>f</b>) partial detachment relative to the T7Cas9 control parasite (<b>a</b>,<b>b</b>). (<b>C</b>) TEM (<b>a</b>,<b>c</b>,<b>e</b>) and negative staining TEM (<b>b</b>,<b>d</b>,<b>f</b>) analysis. (<b>a</b>,<b>c</b>,<b>e</b>) Axial section of epimastigote forms showing the cell body (CB), kinetoplast (K), nuclei (N), flagellum (F), and FAZ (yellow arrows). (<b>b</b>,<b>d</b>,<b>f</b>) Negatively stained epimastigote forms showing the subpellicular microtubule of the cell body (CB) with a fully detached flagellum as in TcGP72<sup>−/−</sup> or a partially detached flagellum as in TcFLA-1BP<sup>−/−</sup>. (<b>D</b>) Violin plots show flagellum length, cell size, and the relation between flagellum length and cell size in the epimastigote forms. The mean values are indicated with a black line. Statistically significant differences are indicated with <span class="html-italic">p</span>-values. n = 300 cells. The line chart depicts the growth curves of the knockout parasites. Statistically significant differences are indicated with asterisks for TcGP72<sup>−/−</sup> and pound signs for TcFLA-1BP<sup>−/−</sup> in relation to the T7Cas9 control (* or # <span class="html-italic">p</span> &lt; 0.05 and ** or ## <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 7
<p>TcGP72 knockout parasites display rosette formation, protrusions, and projection along the flagellum. (<b>A</b>) SEM of TcGP72<sup>−/−</sup> epimastigotes. (<b>a</b>) Rosette formation. (<b>b</b>) Flagellar membrane protrusions (yellow arrowhead) and (<b>c</b>,<b>d</b>) flagellar structures bizarrely filled with amorphous material. The dotted region (<b>c</b>) shows the magnified area (<b>d</b>). (<b>B</b>) TEM shows (<b>a</b>,<b>b</b>) the membrane projections (black arrowhead) and (<b>a</b>,<b>c</b>,<b>d</b>) protrusions (yellow arrowhead) of the flagellum and the cell body membrane in TcGP72 knockout parasites. F: Flagellum.</p>
Full article ">Figure 8
<p>TcGP72 and TcFLA-1BP knockout in <span class="html-italic">T. cruzi</span> epimastigotes show cell cycle defects. Cell cycle comparative analysis with flow cytometry of the knockout parasites. (<b>A</b>) Representative histograms showing the DNA content of TcGP72<sup>−/−</sup>, TcFLA-1BP<sup>−/−</sup>, and the T7Cas9 control. The peak in gray represents the T7Cas9 parasites and peaks in red and blue represent depleted TcFLA-1BP and TcGP72, respectively. The bar graph represents the percentage of cells in each cell cycle phase in three independent experiments. Statistical analysis was performed with two-way ANOVA with Bonferroni correction for multiple testing. (<b>B</b>) Quantification of nuclei, the kinetoplast, and the flagellum in cells labeled for immunofluorescence microscopy with anti-2F6 (PFR) and Hoechst stain of knockout parasites (n = 300 cells). (<b>C</b>) SEM showing the abnormal morphology of knockout parasites. TcGP72<sup>−/−</sup> (<b>a</b>,<b>b</b>) and TcFLA-1BP<sup>−/−</sup> (<b>c</b>,<b>d</b>).</p>
Full article ">Figure 9
<p>TcGP72 and TcFLA−1BP knockout <span class="html-italic">T. cruzi</span> show metacyclogenesis defects. (<b>A</b>) Metacyclic TCTs were purified on a DEAE cellulose column stained with Giemsa and observed under a microscope. The white arrowhead represents the kinetoplast, the yellow arrowhead represents the nucleus, and the blue arrowhead represents the flagellum. (<b>B</b>) Scanning microscopy images of TcGP72<sup>−/−</sup> parasites forming an adhesion network at the bottom of the culture flask during the metacyclogenesis assay.</p>
Full article ">Figure 10
<p>A simplified view of the flagellum and cell body adhesion mediated by the interaction of TcGP72 and FLA-1BP in <span class="html-italic">Trypanosoma cruzi</span>. In <span class="html-italic">T. cruzi</span>, the flagellum membrane interacts with the cell body membrane, forming an adhesive network of proteins. This region is called the flagellar attachment zone (FAZ). The FAZ is divided into 3 main domains: (1) FAZ flagellar domain, (2) FAZ intercellular domain, and (3) FAZ filament domain. The components of the FAZ are distributed along the domains. Both TcGP72 and FLA1BP are essential for this adhesion at different intensities. In our model, TcGP72 is present on the flagellar membrane, and TcFLA-1BP is present on the cell membrane. TcGP72 is anchored to the flagellum by the intracellular domain at the N-terminus. TcFLA-1BP is anchored to the cell body by the two intracellular domains (N- and C-terminus). TcGP72 and TcFLA-1BP interact via the extracellular domains directed towards the intercellular domain of FAZ, thereby mediating flagellum/cell body adhesion. The numerals 1 and 2, highlighted in blue, denote the transmembrane regions. This interaction happens via the NHL-repeat domains present in the extracellular regions. The absence of TcGP72 and TcFLA-1BP causes partial or complete detachment of the flagellum. Both proteins have N- and O-glycosylation sites. Extra: extracellular. Intra: intracellular. FM: flagellum membrane. PM: plasma membrane.</p>
Full article ">
11 pages, 2554 KiB  
Article
Assessment of Cross-Reactivity of Chimeric Trypanosoma cruzi Antigens with Crithidia sp. LVH-60A: Implications for Accurate Diagnostics
by Emily F. Santos, Ramona T. Daltro, Carlos G. Regis-Silva, Tycha B. S. Pavan, Fabrícia A. de Oliveira, Ângela M. da Silva, Roque P. Almeida, Noilson L. S. Gonçalves, Daniel D. Sampaio, Faber N. Santos, Fabricio K. Marchini, Paola A. F. Celedon, Nilson I. T. Zanchin and Fred L. N. Santos
Diagnostics 2023, 13(22), 3470; https://doi.org/10.3390/diagnostics13223470 - 17 Nov 2023
Cited by 4 | Viewed by 1576
Abstract
This study focuses on developing accurate immunoassays for diagnosing Chagas disease (CD), a challenging task due to antigenic similarities between Trypanosoma cruzi and other parasites, leading to cross-reactivity. To address this challenge, chimeric recombinant T. cruzi antigens (IBMP-8.1, IBMP-8.2, IBMP-8.3, and IBMP-8.4) were [...] Read more.
This study focuses on developing accurate immunoassays for diagnosing Chagas disease (CD), a challenging task due to antigenic similarities between Trypanosoma cruzi and other parasites, leading to cross-reactivity. To address this challenge, chimeric recombinant T. cruzi antigens (IBMP-8.1, IBMP-8.2, IBMP-8.3, and IBMP-8.4) were synthesized to enhance specificity and reduce cross-reactivity in tests. While these antigens showed minimal cross-reactivity with leishmaniasis, their performance with other trypanosomatid infections was unclear. This study aimed to assess the diagnostic potential of these IBMP antigens for detecting CD in patients with Crithidia sp. LVH-60A, a parasite linked to visceral leishmaniasis-like symptoms in Brazil. This study involved seven Crithidia sp. LVH-60A patients and three Leishmania infantum patients. The results indicated that these IBMP antigens displayed 100% sensitivity, with specificity ranging from 87.5% to 100%, and accuracy values between 90% and 100%. No cross-reactivity was observed with Crithidia sp. LVH-60A, and only one L. infantum-positive sample showed limited cross-reactivity with IBMP-8.1. This study suggests that IBMP antigens offer promising diagnostic performance, with minimal cross-reactivity in regions where T. cruzi and other trypanosomatids are prevalent. However, further research with a larger number of Crithidia sp. LVH-60A-positive samples is needed to comprehensively evaluate antigen cross-reactivity. Full article
(This article belongs to the Special Issue Advances in the Diagnosis of Infectious Diseases and Microorganisms)
Show Figures

Figure 1

Figure 1
<p>Flowchart illustrating study design in conformity with the Standards for Reporting of Diagnostic Accuracy Studies (STARD) guidelines. Public domain digital map was freely obtained from the Brazilian Institute of Geography and Statistics (IBGE) cartographic database in shapefile format (.shp), which was subsequently reformatted and analyzed using QGIS version 3.22.16 (Geographic Information System, Open Source Geospatial Foundation Project. <a href="http://qgis.osgeo.org" target="_blank">http://qgis.osgeo.org</a> accessed on 27 September 2023).</p>
Full article ">Figure 2
<p>Reaction patterns of chimeric antigens in latent class analysis (LCA) used in anti-<span class="html-italic">T. cruzi</span> ELISA tests. LCS, latent class status; NR, nonreactive; PP, a posteriori probability; R, reactive; P1, P2, P3, P4, and P5, reaction response; N, number of samples.</p>
Full article ">Figure 3
<p>Reactivity index values from <span class="html-italic">Crithidia</span> sp. LVH-60A- and <span class="html-italic">Leishmania infantum</span>-positive serum samples assayed with four IBMP chimeric antigens. RI = 1.0, cutoff; RI = 1.0 ± 10% (shaded area), gray zone. Horizontal lines for each group of results: geometric means; GZ, gray zone; CR, cross-reactivity; RI, reactivity index.</p>
Full article ">
17 pages, 1503 KiB  
Article
In Silico and In Vitro Search for Dual Inhibitors of the Trypanosoma brucei and Leishmania major Pteridine Reductase 1 and Dihydrofolate Reductase
by Katharina Possart, Fabian C. Herrmann, Joachim Jose and Thomas J. Schmidt
Molecules 2023, 28(22), 7526; https://doi.org/10.3390/molecules28227526 - 10 Nov 2023
Cited by 3 | Viewed by 1781
Abstract
The parasites Trypanosoma brucei (Tb) and Leishmania major (Lm) cause the tropical diseases sleeping sickness, nagana, and cutaneous leishmaniasis. Every year, millions of humans, as well as animals, living in tropical to subtropical climates fall victim to these illnesses’ [...] Read more.
The parasites Trypanosoma brucei (Tb) and Leishmania major (Lm) cause the tropical diseases sleeping sickness, nagana, and cutaneous leishmaniasis. Every year, millions of humans, as well as animals, living in tropical to subtropical climates fall victim to these illnesses’ health threats. The parasites’ frequent drug resistance and widely spread natural reservoirs heavily impede disease prevention and treatment. Due to pteridine auxotrophy, trypanosomatid parasites have developed a peculiar enzyme system consisting of dihydrofolate reductase-thymidylate synthase (DHFR-TS) and pteridine reductase 1 (PTR1) to support cell survival. Extending our previous studies, we conducted a comparative study of the T. brucei (TbDHFR, TbPTR1) and L. major (LmDHFR, LmPTR1) enzymes to identify lead structures with a dual inhibitory effect. A pharmacophore-based in silico screening of three natural product databases (approximately 4880 compounds) was performed to preselect possible inhibitors. Building on the in silico results, the inhibitory potential of promising compounds was verified in vitro against the recombinant DHFR and PTR1 of both parasites using spectrophotometric enzyme assays. Twelve compounds were identified as dual inhibitors against the Tb enzymes (0.2 μM < IC50 < 85.1 μM) and ten against the respective Lm enzymes (0.6 μM < IC50 < 84.5 μM). These highly promising results may represent the starting point for the future development of new leads and drugs utilizing the trypanosomatid pteridine metabolism as a target. Full article
Show Figures

Figure 1

Figure 1
<p>Chemical structures <b>1</b>–<b>10</b> of the in silico hits identified from the Phytolab database that displayed in vitro inhibitory activity against the target enzymes. Squares from left to right: <span class="html-italic">Tb</span>PTR1, <span class="html-italic">Tb</span>DHFR, <span class="html-italic">Lm</span>PTR1, <span class="html-italic">Lm</span>DHFR, <span class="html-italic">h</span>DHFR; red: active; blue: inactive; empty: not tested.</p>
Full article ">Figure 2
<p>Chemical structures <b>11</b>–<b>17</b> of the in silico hits identified from the AnalytiCon Discovery database that displayed in vitro inhibitory activity against the target enzymes. Squares from left to right: <span class="html-italic">Tb</span>PTR1, <span class="html-italic">Tb</span>DHFR, <span class="html-italic">Lm</span>PTR1, <span class="html-italic">Lm</span>DHFR, <span class="html-italic">h</span>DHFR; red: active; blue: inactive; empty: not tested.</p>
Full article ">Figure 3
<p>Chemical structures <b>18</b>–<b>22</b> of the in silico hits identified from the Specs Natural Products (Specs NP) database that displayed in vitro inhibitory activity against the target enzymes. Squares from left to right: <span class="html-italic">Tb</span>PTR1, <span class="html-italic">Tb</span>DHFR, <span class="html-italic">Lm</span>PTR1, <span class="html-italic">Lm</span>DHFR, <span class="html-italic">h</span>DHFR; red: active; blue: inactive; empty: not tested.</p>
Full article ">Figure 4
<p>Best scoring docking conformation for Compound <b>17</b> in the binding pocket of <span class="html-italic">Tb</span>PTR1 (ID: “2X9G”) with co-crystallized NADP (carbon atoms colored in yellow). The molecular surface is colored according to lipophilicity, with lipophilic areas in yellow and hydrophilic areas in blue. Co-crystallized solvent not shown. Note that the reactive β-carbon of the enone system is not far away from the SH group of Cys168 in this docking pose (red circles; 4.6 Å). From this orientation, it could easily assume a position suitable for a Michael addition without much change.</p>
Full article ">Figure 5
<p>Best scoring docking conformations for Compound <b>11</b> in the binding pocket of (<b>a</b>) <span class="html-italic">Tb</span>PTR1 (ID: “4CMK”) and (<b>b</b>) <span class="html-italic">Tb</span>DHFR (ID: “3QFX”) with co-crystallized NADP/NADPH (carbon atoms colored in yellow). The molecular surface is colored according to lipophilicity, with lipophilic areas in yellow and hydrophilic areas in blue. Co-crystallized solvent not shown.</p>
Full article ">
Back to TopTop