[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
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

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (226)

Search Parameters:
Keywords = nucleoside analogues

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 1862 KiB  
Article
Thymidine Analogue Mutations with M184V Significantly Decrease Phenotypic Susceptibility of HIV-1 Subtype C Reverse Transcriptase to Islatravir
by Hyeonah Byun, Maria Antonia Papathanasopoulos, Kim Steegen and Adriaan Erasmus Basson
Viruses 2024, 16(12), 1888; https://doi.org/10.3390/v16121888 - 6 Dec 2024
Viewed by 886
Abstract
Islatravir (ISL) is the first-in-class nucleoside reverse transcriptase translocation inhibitor (NRTtI) with novel modes of action. Data on ISL resistance are currently limited, particularly to HIV-1 non-B subtypes. This study aimed to assess prevalent nucleos(t)ide reverse transcriptase inhibitor (NRTI)-resistant mutations in HIV-1 subtype [...] Read more.
Islatravir (ISL) is the first-in-class nucleoside reverse transcriptase translocation inhibitor (NRTtI) with novel modes of action. Data on ISL resistance are currently limited, particularly to HIV-1 non-B subtypes. This study aimed to assess prevalent nucleos(t)ide reverse transcriptase inhibitor (NRTI)-resistant mutations in HIV-1 subtype C for their phenotypic resistance to ISL. Prevalent single and combinations of NRTI-resistant mutations were selected from a routine HIV-1 genotypic drug resistance testing database and introduced into HIV-1 subtype C-like pseudoviruses, which were then tested for ISL susceptibility. Single NRTI-resistant mutations were susceptible or showed only a low level of resistance to ISL. This included thymidine analogue mutations (TAMs, i.e., M41L, D67N, K70R, T215FY, and K219EQ) and non-TAMs (i.e., A62V, K65R, K70ET, L74IV, A114S, Y115F, and M184V). Combinations of M184V with one or more additional NRTI-resistant mutations generally displayed reduced ISL susceptibilities. This was more prominent for combinations that included M184V+TAMs, and particularly M184V+TAM-2 mutations. Combinations that included M184V+K65R did not impact significantly on ISL susceptibility. Our study suggests that ISL would be effective in treating people living with HIV (PLWH) failing tenofovir disoproxil fumarate (TDF)/lamivudine (3TC) or TDF/emtricitabine (FTC)-containing regimens, but would be less effective in PLH failing zidovudine (AZT) with 3TC or FTC-containing regimens. Full article
Show Figures

Figure 1

Figure 1
<p>Variation in IC<sub>50</sub> and fold-change of ISL against wild-type PSV. (<b>A</b>) Multiple independent in vitro assays (<span class="html-italic">n</span> = 41) were conducted to determine the IC<sub>50</sub> of ISL against the wild-type HIV-1 subtype C virus (i.e., p8.9MJ4). The mean IC<sub>50</sub> = 8.32 nM ± 4.99 nM (median IC<sub>50</sub> = 7.27 nM, IQR 4.16–12.88). (<b>B</b>) Each IC<sub>50</sub> value was divided by the mean IC<sub>50</sub> value to determine the fold-change (FC). The mean FC was, therefore, 1.0 ± 0.6 FC (median FC = 0.87, IQR 0.54–1.55). Technical cut-off (TCO): obtained from the 99th percentile of the IC<sub>50</sub> values. (<b>C</b>) Multiple independent in vitro assays (<span class="html-italic">n</span> = 13) were conducted to determine the IC<sub>50</sub> value of ISL against the wild-type HIV-1 subtype B virus (i.e., p8.9NSX). The mean IC<sub>50</sub> concentration was shown to be 7.91 nM ± 5.51 nM (median IC<sub>50</sub> = 5.90 nM, IQR 2.58–13.32). (<b>D</b>) Each IC<sub>50</sub> value was divided by the mean IC<sub>50</sub> value to obtain a mean FC value of 1 (median FC = 0.74, IQR 0.33–1.68).</p>
Full article ">Figure 2
<p>Fold-change in IC<sub>50</sub> of single mutants in subtype C PSVs compared to the wild-type PSV. Following the in vitro phenotypic activity assays, the IC<sub>50</sub> values for each mutant was compared against the mean IC<sub>50</sub> of the MJ4 wild-type PSV, allowing the determination of fold changes. The 99th percentile of variation in the wild-type IC<sub>50</sub> value, calculated to be 2.2 (TCO), served as the threshold for categorizing mutants as either susceptible or having a decreased susceptibility to ISL. Among the single mutants, M41L, K65R, D67N, K70E/R/T, L74I, A114S, Y115F, T215F, and K219E/Q demonstrated susceptibility to ISL. In contrast, A62V, L74V, and T215Y exhibited potential low-level resistance, and M184V exhibited potential-low- to low-level resistance. Susceptible (<span class="html-italic">n</span>), potential-low-level resistance (<span class="html-italic">n</span>), low-level resistance (<span class="html-italic">n</span>), intermediate resistance (<span class="html-italic">n</span>), and high-level resistance (<span class="html-italic">n</span>). Data are shown as median bar graphs with the IQR as error bars.</p>
Full article ">Figure 3
<p>Single mutant L74V in wild-type HIV-1 subtype B and C laboratory-adapted strains. A phenotypic activity assay was conducted to determine whether ISL had similar potencies against the single mutant L74V in different wild-type strains and subtypes of HIV-1. IC<sub>50</sub> values are expressed as median fold-change differences to the IC<sub>50</sub> of the relevant control subtype, with the IQR as error bars. The TCO for each subtype is indicated on the graph by a dotted line.</p>
Full article ">Figure 4
<p>IC<sub>50</sub> values of L74V in laboratory-adapted PSVs and inter-subtype Kruskal–Wallis test. Site-directed mutagenesis was performed to introduce the L74V mutant into the wild-type p8.9NSX and p8.9MJ4, and laboratory-adapted strains. (<b>A</b>) Median IC<sub>50</sub> values of the L74V mutation in laboratory-adapted strains show that the LTNP5 PSV has the highest median IC<sub>50</sub> of 9.76 nM (IQR 6.27–9.98). DS9 had the lowest median IC<sub>50</sub> of 2.31 nM (IQR 1.78–4.20). Data are shown as median bar graphs with the IQR. (<b>B</b>) Inter-subtype non-parametric statistical analysis was performed using a Kruskal–Wallis multiple comparisons test. The grid shows the <span class="html-italic">p</span>-values of the comparisons in IC<sub>50</sub> values. No significant differences (<span class="html-italic">p</span> &gt; 0.05) were observed.</p>
Full article ">Figure 5
<p>Fold-change in IC<sub>50</sub> of mutation combinations in subtype C PSVs compared to the wild-type MJ4 PSV. Following the in vitro phenotypic activity assays, the IC<sub>50</sub> value for each mutant was compared against the mean IC<sub>50</sub> of the MJ4 wild-type PSV, allowing for the determination of fold changes. The 99th percentile of variation in the wild-type IC<sub>50</sub> value, calculated to be 2.2 (TCO), served as the threshold for categorizing mutants as either susceptible or resistant to ISL. It was observed that the combination of NRTI mutations generally increased resistance to ISL. The A114S/M184V mutation combination showed a very high level of resistance to ISL. Its IC<sub>50</sub> value was greater than the highest ISL concentration tested, and consequently, its FC value was &gt; 60. Susceptible (<span class="html-italic">n</span>), potential-low-level resistance (<span class="html-italic">n</span>), low-level resistance (<span class="html-italic">n</span>), intermediate resistance (<span class="html-italic">n</span>), and high-level resistance (<span class="html-italic">n</span>). Data are shown as median bar graphs with the IQR as error bars.</p>
Full article ">
14 pages, 3007 KiB  
Article
Production of Value-Added Arabinofuranosyl Nucleotide Analogues from Nucleoside by an In Vitro Enzymatic Synthetic Biosystem
by Yuxue Liu, Xiaojing Zhang, Erchu Yang, Xiaobei Liu, Weiwei Su, Zhenyu Wang and Hailei Wang
Biomolecules 2024, 14(11), 1440; https://doi.org/10.3390/biom14111440 - 13 Nov 2024
Viewed by 882
Abstract
Arabinofuranosyl nucleotide analogue (arabinoside) and the derived compounds, a family of nucleoside analogues, exhibit diverse, typically biological activities and are widely used as antibacterial, antiviral, anti-inflammatory, and antitumor drugs in both clinical and preclinical trials. Despite their long and rich history in medicinal [...] Read more.
Arabinofuranosyl nucleotide analogue (arabinoside) and the derived compounds, a family of nucleoside analogues, exhibit diverse, typically biological activities and are widely used as antibacterial, antiviral, anti-inflammatory, and antitumor drugs in both clinical and preclinical trials. Despite their long and rich history in medicinal chemistry, the biosynthesis of arabinoside has only been sporadically designed and studied and has remained a challenging task. In this study, an in vitro synthetic enzymatic biosystem was designed and constructed for the production of arabinoside from low-cost nucleoside, based on a phosphorolysis -isomerization-dephosphorylation enzymatic cascade conversion routes. The enzymatic system achieves the biosynthesis of arabinoside by isomerizing the ribose part of nucleoside to arabinose. The reaction conditions affecting the yield of arabinoside were investigated and optimized, including meticulous enzyme selection, key enzyme dosage, the concentration of orthophosphate, and reaction time. Under the optimized conditions, we achieved the production of 0.12 mM of arabinofuranosylguanine from 0.5 mM of guanosine, representing 24% of the theoretical yield. Furthermore, this biosystem also demonstrated the capability to produce other arabinosides, such as vidarabine, spongouridine, and hypoxanthine arabinofuranoside from corresponding nucleosides. Overall, our biosynthesis approach provides a pathway for the biosynthesis of arabinoside. Full article
(This article belongs to the Section Synthetic Biology and Bioengineering)
Show Figures

Figure 1

Figure 1
<p>A multistep biocatalytic cascade for the biosynthesis of arabinoside from nucleoside: (<b>A</b>) Diagram of the multistep biocatalytic cascade. (<b>B</b>,<b>C</b>) The changes in Gibbs free energy for each individual reaction in this multistep biocatalytic cascade for Ara-A and Ara-U. The changes in Gibbs free energy for each individual reaction of converting R1P to A1P in this multistep biocatalytic cascade under conditions characterized by a pH of 7.5 and an ionic strength of 0.25 M (<a href="https://equilibrator.weizmann.ac.il/" target="_blank">https://equilibrator.weizmann.ac.il/</a> (accessed on 12 September 2024)).</p>
Full article ">Figure 2
<p>Activity analysis of NPs: (<b>A</b>) Phosphorolysis ability of NPs towards arabinosides. (<b>B</b>) Phosphorolysis and dephosphorylation activity of PNPs toward guanosine and Ara-G.</p>
Full article ">Figure 3
<p>Proof-of-concept production of Ara-G from guanosine: (<b>A</b>) HPLC chromatograms of the peaks of Ara-G, guanosine, and guanine in different reactions. (<b>B</b>) Quantification of Ara-G production in different reactions. Values shown are the means of triplicate determinations.</p>
Full article ">Figure 4
<p>Optimization of reaction conditions for Ara-G production: (<b>A</b>) Time course of Ara-G production. (<b>B</b>) Effect of single-enzyme concentrates on Ara-G production. Gray, control. Blue, the concentration of <span class="html-italic">Kl</span>PNP was changed. Orange, the concentration of <span class="html-italic">Bc</span>PPM was changed. Yellow, the concentration of <span class="html-italic">Ec</span>RpiA was changed. Green, the concentration of <span class="html-italic">Ec</span>KdsD was changed. <span class="html-italic">n</span> = 3 independent experiments. Data are presented as mean values ± SD.</p>
Full article ">Figure 5
<p>Validation of the feasibility of arabinoside production from nucleosides: (<b>A</b>) HPLC chromatograms of the production of Ara-A. Blue line, the authentic standard of nucleobases and arabinosides; orange line, reaction sample. (<b>B</b>) The quantification of arabinoside production in the reaction system.</p>
Full article ">Scheme 1
<p>Examples of nucleoside and arabinoside investigated in the present study.</p>
Full article ">Scheme 2
<p>Chemical synthesis of nelarabine and Ara-U (<b>A</b>,<b>B</b>) [<a href="#B11-biomolecules-14-01440" class="html-bibr">11</a>,<a href="#B13-biomolecules-14-01440" class="html-bibr">13</a>].</p>
Full article ">Scheme 3
<p>General scheme of the biosynthesis of arabinosides (<b>A</b>–<b>C</b>): NP, nucleoside phosphorylase; NDT, 2′-deoxyribosyltransferase; RK, ribokinase; PPM, phosphopentomutase; 6PGDH, 6-phosphogluconate dehydrogenase; and API, D-arabinose 5-phosphate isomerase.</p>
Full article ">
15 pages, 3623 KiB  
Review
Nucleoside Analogues for Chagas Disease and Leishmaniasis Therapy: Current Status and Future Perspectives
by Emmanuel Awucha Nwoke, Silvester Lowe, Fawaz Aldabbagh, Karunakaran Kalesh and Hachemi Kadri
Molecules 2024, 29(22), 5234; https://doi.org/10.3390/molecules29225234 - 5 Nov 2024
Viewed by 1043
Abstract
Chagas disease and leishmaniasis are two neglected tropical diseases that affect millions of people in low- and middle-income tropical countries. These diseases caused by protozoan parasites pose significant global health challenges, which have been exacerbated by the recent COVID-19 pandemic. There is an [...] Read more.
Chagas disease and leishmaniasis are two neglected tropical diseases that affect millions of people in low- and middle-income tropical countries. These diseases caused by protozoan parasites pose significant global health challenges, which have been exacerbated by the recent COVID-19 pandemic. There is an urgent need for novel therapeutics as current treatments are limited by toxicity and drug resistance. Nucleoside analogues, which have been extensively studied and successfully applied in antiviral and antitumor therapies, hold potential that has yet to be fully explored for treating these neglected diseases. In this review, we discuss the use of nucleoside analogues as promising therapeutic agents for Chagas disease and leishmaniasis. After briefly examining the pathology, progression, and current treatment options for these diseases, we provide a comprehensive analysis of the status of nucleoside analogues and explore their prospects. By outlining the current landscape and future directions, this review aims to guide research and development efforts towards more effective nucleoside-based treatments for Chagas disease and leishmaniasis. Full article
(This article belongs to the Special Issue Heterocyclic Chemistry with Applications (Second Edition))
Show Figures

Figure 1

Figure 1
<p>Current treatments for Chagas disease and leishmaniasis.</p>
Full article ">Figure 2
<p>Nucleoside analogues with anti-Chagas disease activity.</p>
Full article ">Figure 3
<p>Nucleoside analogues with anti-leishmanial activity.</p>
Full article ">Figure 4
<p>Mechanism of action of allopurinol in <span class="html-italic">Leishmania</span> and <span class="html-italic">T. cruzi</span>.</p>
Full article ">
16 pages, 3210 KiB  
Article
Widely Targeted Metabolomics Method Reveals Differences in Volatile and Nonvolatile Metabolites in Three Different Varieties of Raw Peanut by GC–MS and HPLC–MS
by Jiantao Fu, Yuxing An, Dao Yao, Lijun Chen, Liwen Zhou, Dachun Shen, Sixing Dai, Yinglin Lu and Donglei Sun
Molecules 2024, 29(22), 5230; https://doi.org/10.3390/molecules29225230 - 5 Nov 2024
Viewed by 815
Abstract
The aim of the present study was to comprehensively analyze and identify the metabolites of different varieties of raw peanut, as well as provide a reference for the utilization of different varieties of peanuts. In this study, three varieties of peanuts, namely ZKH1H, [...] Read more.
The aim of the present study was to comprehensively analyze and identify the metabolites of different varieties of raw peanut, as well as provide a reference for the utilization of different varieties of peanuts. In this study, three varieties of peanuts, namely ZKH1H, ZKH13H, and CFD, were investigated via ultrahigh-performance liquid chromatography (UPLC) and widely targeted metabolomics methods based on tandem mass spectrometry (MS) and solid-phase microextraction-gas chromatography–mass spectrometry (SPME-GC–MS). In total, 417 nonvolatile and 55 volatile substances were detected. The nonvolatile substances were classified into the following 10 categories: organic acids and derivatives (28.9%); organic oxygen compounds (21.9%); lipids and lipid-like molecules (12.6%); organoheterocyclic compounds (9.9%); nucleosides, nucleotides, and analogues (9.4%); benzenoids (7.8%); phenylpropanoids and polyketides (6.1%); organic nitrogen compounds (2.7%); lignans, neolignans, and related compounds (0.5%); and alkaloids and their derivatives (0.3%). The volatile compounds (VOCs) were classified into the following eight categories: organic oxygen compounds (24.1%); organic cyclic compounds (20.4%); organic nitrogen compounds (13%); organic acids and their derivatives (13%); lipids and lipid-like molecules (11.2%); benzenoids (11.1%); hydrocarbons (3.7%); and homogeneous non-metallic compounds (3.7%). Differentially abundant metabolites among the different peanut varieties (ZKH13H vs. CFD, ZKH1H vs. CFD, and ZKH1H vs. ZKH13H) were investigated via multivariate statistical analyses, which identified 213, 204, and 157 nonvolatile differentially abundant metabolites, respectively, and 12, 11, and 10 volatile differentially abundant metabolites, respectively. KEGG metabolic pathway analyses of the differential non-VOCs revealed that the most significant metabolic pathways among ZKH13H vs. CFD, ZKH1H vs. CFD, and ZKH1H vs. ZKH13H were galactose metabolism, purine metabolism, and aminoacyl-tRNA, while the nitrogen metabolism pathway was identified as a significant metabolic pathway for the VOCs. The present findings provide a theoretical foundation for the development and utilization of these three peanut species, as well as for the breeding of new peanut varieties. Full article
Show Figures

Figure 1

Figure 1
<p>The appearance (<b>A</b>), length (<b>B</b>), width (<b>C</b>), and length/width ratio (<b>D</b>) of the three varieties of peanuts. The data are presented as the mean ± standard deviation (SD).</p>
Full article ">Figure 2
<p>Different main nonvolatile metabolites in the three varieties of peanut samples were identified through a widely targeted metabolic method and method reliability evaluation. (<b>A</b>) Classification of the 417 nonvolatile metabolites detected in the three varieties of peanut samples. (<b>B</b>) PCA score plot. (<b>C</b>) Heatmap analysis.</p>
Full article ">Figure 3
<p>K-means clustering analysis of all nonvolatile metabolites of the three varieties of peanut samples.</p>
Full article ">Figure 4
<p>Differential nonvolatile metabolites obtained by comparison of each pair of varieties. Score plots of OPLS-DA pairwise comparisons of differentially abundant metabolites in (<b>A</b>) ZKH1H vs. CFD, (<b>D</b>) ZKH1H vs. ZKH13H, and (<b>G</b>) ZKH13H vs. CFD. Volcano plots showing the differential nonvolatile metabolite expression levels in (<b>B</b>) ZKH1H vs. CFD, (<b>E</b>) ZKH1H vs. ZKH13H, and (<b>H</b>) ZKH13H vs. CFD. Classification of the differential nonvolatile metabolites obtained by comparison of each pair of varieties in (<b>C</b>) ZKH1H vs. CFD, (<b>F</b>) ZKH1H vs. ZKH13H, and (<b>I</b>) ZKH13H vs. CFD.</p>
Full article ">Figure 5
<p>KEGG pathway annotation of the differential nonvolatile metabolites obtained by comparing each of the two groups in (<b>A</b>) ZKH1H vs. CFD, (<b>B</b>) ZKH1H vs. ZKH13H, and (<b>C</b>) ZKH13H vs. CFD. (<b>D</b>) Intersection of the 5 pathways with the most significant differences in the 3 comparison groups.</p>
Full article ">Figure 6
<p>Multivariate statistical analysis of VOCs in the three varieties of peanut samples. (<b>A</b>) Classification of the 55 nonvolatile metabolites detected in the three varieties of peanut samples. (<b>B</b>) PCA score plot. (<b>C</b>) Heatmap analysis.</p>
Full article ">Figure 7
<p>Differential VOCs obtained by comparison of each pair of varieties. Score plots of OPLS-DA pairwise comparisons of differentially abundant metabolites in (<b>A</b>) ZKH1H vs. CFD, (<b>D</b>) ZKH1H vs. ZKH13H, and (<b>G</b>) ZKH13H vs. CFD. Volcano plots showing the differential nonvolatile metabolite expression levels in (<b>B</b>) ZKH1H vs. CFD, (<b>E</b>) ZKH1H vs. ZKH13H, and (<b>H</b>) ZKH13H vs. CFD. Correlation analysis of differential volatile metabolites in (<b>C</b>) ZKH1H vs. CFD, (<b>F</b>) ZKH1H vs. ZKH13H, and (<b>I</b>) ZKH13H vs. CFD.</p>
Full article ">Figure 8
<p>Differential VOCs from GC-MS were enriched in distinct KEGG pathways by comparing each of the two groups. (<b>A</b>) ZKH1H vs. CFD, (<b>B</b>) ZKH1H vs. ZKH13H.</p>
Full article ">
9 pages, 6587 KiB  
Communication
Discovery of Substituted 5-(2-Hydroxybenzoyl)-2-Pyridone Analogues as Inhibitors of the Human Caf1/CNOT7 Ribonuclease
by Ishwinder Kaur, Gopal P. Jadhav, Peter M. Fischer and Gerlof Sebastiaan Winkler
Molecules 2024, 29(18), 4351; https://doi.org/10.3390/molecules29184351 - 13 Sep 2024
Viewed by 936
Abstract
The Caf1/CNOT7 nuclease is a catalytic component of the Ccr4-Not deadenylase complex, which is a key regulator of post-transcriptional gene regulation. In addition to providing catalytic activity, Caf1/CNOT7 and its paralogue Caf1/CNOT8 also contribute a structural function by mediating interactions between the large, [...] Read more.
The Caf1/CNOT7 nuclease is a catalytic component of the Ccr4-Not deadenylase complex, which is a key regulator of post-transcriptional gene regulation. In addition to providing catalytic activity, Caf1/CNOT7 and its paralogue Caf1/CNOT8 also contribute a structural function by mediating interactions between the large, non-catalytic subunit CNOT1, which forms the backbone of the Ccr4-Not complex and the second nuclease subunit Ccr4 (CNOT6/CNOT6L). To facilitate investigations into the role of Caf1/CNOT7 in gene regulation, we aimed to discover and develop non-nucleoside inhibitors of the enzyme. Here, we disclose that the tri-substituted 2-pyridone compound 5-(5-bromo-2-hydroxy-benzoyl)-1-(4-chloro-2-methoxy-5-methyl-phenyl)-2-oxo-pyridine-3-carbonitrile is an inhibitor of the Caf1/CNOT7 nuclease. Using a fluorescence-based nuclease assay, the activity of 16 structural analogues was determined, which predominantly explored substituents on the 1-phenyl group. While no compound with higher potency was identified among this set of structural analogues, the lowest potency was observed with the analogue lacking substituents on the 1-phenyl group. This indicates that substituents on the 1-phenyl group contribute significantly to binding. To identify possible binding modes of the inhibitors, molecular docking was carried out. This analysis suggested that the binding modes of the five most potent inhibitors may display similar conformations upon binding active site residues. Possible interactions include π-π interactions with His225, hydrogen bonding with the backbone of Phe43 and Van der Waals interactions with His225, Leu209, Leu112 and Leu115. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Structure of 5-(5-bromo-2-hydroxybenzoyl)-1-(4-chloro-2-methoxy-5-methylphenyl)-2-oxo-1,2-dihydropyridine-3-carbonitrile, an inhibitor of the human Caf1/CNOT7 nuclease. The reported IC<sub>50</sub> value is 14.6 ± 3.1 μM [<a href="#B37-molecules-29-04351" class="html-bibr">37</a>].</p>
Full article ">Figure 2
<p>Molecular docking of inhibitors into the active site of human Caf1/CNOT7. (<b>A</b>) Catalytic site of the Caf1/CNOT7 enzyme. Shown is the position of the residues coordinating two Mg<sup>2+</sup> ions (bright green) in the active site of <span class="html-italic">Schizosaccharomyces pombe</span> Pop2 protein (PDB 2P51, slate blue) [<a href="#B9-molecules-29-04351" class="html-bibr">9</a>] and the corresponding coordinating residues of human Caf1/CNOT7 (PDB 7VOI, salmon red) [<a href="#B12-molecules-29-04351" class="html-bibr">12</a>]. (<b>B</b>) Model of human Caf1/CNOT7 bound to poly(A) RNA. The RNA was obtained by superposition of the structure of human Caf1/CNOT7 (PDB 7VOI) [<a href="#B12-molecules-29-04351" class="html-bibr">12</a>] and <span class="html-italic">Schizosaccharomyces pombe</span> Pan2 in complex with poly(A) RNA (PDB 6R9J) [<a href="#B45-molecules-29-04351" class="html-bibr">45</a>]. Shown are the surface views of the residues developing; polar interactions (red) and nonpolar interactions (white) with the analogues (<b>1</b>, <b>8</b>, <b>9</b>, <b>11</b>, <b>15</b> and <b>17</b>) in the active site. (<b>C</b>) Molecular docking of <b>1</b> (cyan) into the active site of Caf1/CNOT7. (<b>D</b>) Overlay of <b>1</b> (cyan) with the plausible conformation of the five most potent analogues <b>8</b> (light green), <b>9</b> (light yellow), <b>11</b> (slate blue), <b>15</b> (magenta) and <b>17</b> (white). (<b>E</b>) Molecular docking of <b>17</b> (white) into the active site of Caf1/CNOT7 (<b>F</b>) Overlay of <b>1</b> (cyan) with the plausible conformation of the most potent analogue <b>17</b>, into the active site of Caf1/CNOT7.</p>
Full article ">
13 pages, 782 KiB  
Article
Bacterial Purine Nucleoside Phosphorylases from Mesophilic and Thermophilic Sources: Characterization of Their Interaction with Natural Nucleosides and Modified Arabinofuranoside Analogues
by Irina A. Bychek, Anastasia A. Zenchenko, Maria A. Kostromina, Marat M. Khisamov, Pavel N. Solyev, Roman S. Esipov, Sergey N. Mikhailov and Irina V. Varizhuk
Biomolecules 2024, 14(9), 1069; https://doi.org/10.3390/biom14091069 - 27 Aug 2024
Viewed by 1232
Abstract
The enzymatic synthesis of nucleoside derivatives is an important alternative to multi-step chemical methods traditionally used for this purpose. Despite several undeniable advantages of the enzymatic approach, there are a number of factors limiting its application, such as the limited substrate specificity of [...] Read more.
The enzymatic synthesis of nucleoside derivatives is an important alternative to multi-step chemical methods traditionally used for this purpose. Despite several undeniable advantages of the enzymatic approach, there are a number of factors limiting its application, such as the limited substrate specificity of enzymes, the need to work at fairly low concentrations, and the physicochemical properties of substrates—for example, low solubility. This research conducted by our group is dedicated to the advantages and limitations of using purine nucleoside phosphorylases (PNPs), the main enzymes for the metabolic reutilization of purines, in the synthesis of modified nucleoside analogues. In our work, the substrate specificity of PNP from various bacterial sources (mesophilic and thermophilic) was studied, and the effect of substrate, increased temperature, and the presence of organic solvents on the conversion rate was investigated. Full article
Show Figures

Figure 1

Figure 1
<p>The influence of organic solvents on activity of mesophilic and thermophilic PNPs in the reaction of inosine phosphorolysis (50 mM KH<sub>2</sub>PO<sub>4</sub>, pH 7.5, 25 °C for <span class="html-italic">Ec</span>PNP and <span class="html-italic">Ent</span>PNP, 80 °C for <span class="html-italic">Tth</span>PNP I).</p>
Full article ">Figure 2
<p>Effect of temperature on <span class="html-italic">Ec</span>PNP and <span class="html-italic">Ent</span>PNP activity in the reaction of inosine phosphorolysis. Relative enzyme activity is a percentage of the value obtained at 25 °C (50 mM KH<sub>2</sub>PO<sub>4</sub>, pH 7.5).</p>
Full article ">Figure 3
<p>Long-term stability of <span class="html-italic">Ec</span>PNP at elevated temperatures. Enzyme activity in the reaction of inosine phosphorolysis (50 mM KH<sub>2</sub>PO<sub>4</sub>, pH 7.5) obtained at 25 °C for freshly prepared enzyme solution taken as 100%.</p>
Full article ">
12 pages, 4005 KiB  
Article
Discovery of Novel Amino Acids (Analogues)-Substituted Thiophene[3,2-d]pyrimidine Derivatives as Potent HIV-1 Non-Nucleoside Reverse Transcriptase Inhibitors: Design, Synthesis, and Biological Evaluation
by Zongji Zhuo, Zhao Wang, Lanlan Jing, Tao Zhang, Anchao Ge, Zhenzhen Zhou, Ying Liu, Xin Li, Erik De Clercq, Christophe Pannecouque, Peng Zhan, Xinyong Liu and Dongwei Kang
Int. J. Mol. Sci. 2024, 25(16), 9028; https://doi.org/10.3390/ijms25169028 - 20 Aug 2024
Viewed by 992
Abstract
Inspired by our previous work on the modification of diarylpyrimidine-typed non-nucleoside reverse transcriptase inhibitors (NNRTIs) and the reported crystallographic studies, a series of novel amino acids (analogues)-substituted thiophene[3,2-d]pyrimidine derivatives were designed and synthesized by targeting the solvent-exposed region of the NNRTI-binding [...] Read more.
Inspired by our previous work on the modification of diarylpyrimidine-typed non-nucleoside reverse transcriptase inhibitors (NNRTIs) and the reported crystallographic studies, a series of novel amino acids (analogues)-substituted thiophene[3,2-d]pyrimidine derivatives were designed and synthesized by targeting the solvent-exposed region of the NNRTI-binding pocket. The biological evaluation results showed that compound 5k was the most active inhibitor, exhibiting moderate-to-excellent potency against HIV-1 wild-type (WT) and a panel of NNRTI-resistant strains, with EC50 values ranging from 0.042 μM to 7.530 μM. Of special note, 5k exhibited the most potent activity against single-mutant strains (K103N and E138K), with EC50 values of 0.031 μM and 0.094 μM, being about 4.3-fold superior to EFV (EC50 = 0.132 μM) and 1.9-fold superior to NVP (EC50 = 0.181 μM), respectively. In addition, 5k demonstrated lower cytotoxicity (CC50 = 27.9 μM) and higher selectivity index values. The HIV-1 reverse transcriptase (RT) inhibition assay was further performed to confirm their binding target. Moreover, preliminary structure–activity relationships (SARs) and molecular docking studies were also discussed in order to provide valuable insights for further structural optimizations. In summary, 5k turned out to be a promising NNRTI lead compound for further investigations of treatments for HIV-1 infections. Full article
(This article belongs to the Special Issue Antiviral Drug Discovery)
Show Figures

Figure 1

Figure 1
<p>Chemical structures of six NNRTIs approved by the U.S. FDA.</p>
Full article ">Figure 2
<p>The detailed interactions with WT RT of <b>K-5a2</b> and the four-point pharmacophore mode (PDB code: 6C0J).</p>
Full article ">Figure 3
<p>Design of the novel thiophene[3,2-<span class="html-italic">d</span>]pyrimidine derivatives.</p>
Full article ">Figure 4
<p>(<b>a</b>–<b>c</b>) Predicted binding modes of <b>5k</b> (sliver) with the HIV-1 K103N RT (PDB code: 6C0K), E138K RT (PDB code: 6C0L) and HIV-1 WT RT (PDB code: 6C0J), respectively.</p>
Full article ">Scheme 1
<p>Synthesis of <b>5a</b>–<b>p</b>. Reagents and conditions: (i) DMF, K<sub>2</sub>CO<sub>3</sub>, r.t., 5 h; (ii) BINAP, Pd<sub>2</sub>(dba)<sub>3</sub>, Cs<sub>2</sub>CO<sub>3</sub>, 1,4-dioxane, 120 °C, N<sub>2</sub>, 14 h; (iii) TFA, DCM, r.t., 4 h; (iv) HATU, DIEA, CH<sub>2</sub>Cl<sub>2</sub>, 0 °C to r.t., 8–10 h.</p>
Full article ">
18 pages, 3269 KiB  
Article
Exploring the Mutated Kinases for Chemoenzymatic Synthesis of N4-Modified Cytidine Monophosphates
by Martyna Koplūnaitė, Kamilė Butkutė, Jonita Stankevičiūtė and Rolandas Meškys
Molecules 2024, 29(16), 3767; https://doi.org/10.3390/molecules29163767 - 9 Aug 2024
Viewed by 932
Abstract
Nucleosides, nucleotides, and their analogues are an important class of molecules that are used as substrates in research of enzymes and nucleic acid, or as antiviral and antineoplastic agents. Nucleoside phosphorylation is usually achieved with chemical methods; however, enzymatic phosphorylation is a viable [...] Read more.
Nucleosides, nucleotides, and their analogues are an important class of molecules that are used as substrates in research of enzymes and nucleic acid, or as antiviral and antineoplastic agents. Nucleoside phosphorylation is usually achieved with chemical methods; however, enzymatic phosphorylation is a viable alternative. Here, we present a chemoenzymatic synthesis of modified cytidine monophosphates, where a chemical synthesis of novel N4-modified cytidines is followed by an enzymatic phosphorylation of the nucleosides by nucleoside kinases. To enlarge the substrate scope, multiple mutant variants of Drosophila melanogaster deoxynucleoside kinase (DmdNK) (EC:2.7.1.145) and Bacillus subtilis deoxycytidine kinase (BsdCK) (EC:2.7.1.74) have been created and tested. It has been determined that certain point mutations in the active sites of the kinases alter their substrate specificities noticeably and allow phosphorylation of compounds that had been otherwise not phosphorylated by the wild-type DmdNK or BsdCK. Full article
(This article belongs to the Special Issue Exploring Bioactive Organic Compounds for Drug Discovery, 2nd Edition)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>(<b>a</b>) A dimer of <span class="html-italic">Dm</span>dNK with 2′-deoxycytidine molecules (green) bound in the active centres. Each monomer is coloured in different shades of blue. (<b>b</b>) Interactions between 2′-deoxycytidine (green) and amino acid residues (cyan) in the active site of deoxynucleoside kinase. F114 is omitted for a clearer view. Yellow dashed lines represent hydrogen bonds between the nucleoside and the residues. PDB ID: 1j90.</p>
Full article ">Figure 2
<p>Tertiary structures of non-TK1-like family nucleoside kinases. (<b>a</b>) HSV-1 TK monomer. PDB ID: 4IVP. (<b>b</b>) <span class="html-italic">Hs</span>dCK monomer. PDB ID: 1p61. (<b>c</b>) <span class="html-italic">Dm</span>dNK monomer. PDB ID: 1j90. (<b>d</b>) <span class="html-italic">Bs</span>dCK monomer. Model generated by ColabFold v1.5.5.</p>
Full article ">Figure 3
<p>Structures of the canonical and <span class="html-italic">N</span><sup>4</sup>-modified nucleosides used in the study.</p>
Full article ">Figure 4
<p>The active site of <span class="html-italic">Dm</span>dNK with bound deoxycytidine. (<b>a</b>) Distances between <span class="html-italic">N</span><sup>4</sup> atom of deoxycytidine and V84, M88, and A110 residues of <span class="html-italic">Dm</span>dNK-WT. PDB ID: 1j90. (<b>b</b>) Predicted distances between <span class="html-italic">N</span><sup>4</sup> atom of deoxycytidine and residues at positions 84 and 110, where V84 and A110 are replaced by glycine. Model generated by ColabFold v1.5.5d. (<b>c</b>) Hydrogen bonds between <span class="html-italic">N</span><sup>3</sup> and <span class="html-italic">N</span><sup>4</sup> atoms of deoxycytidine and Q81 residue in <span class="html-italic">Dm</span>dNK-WT. PDB ID: 1j90. (<b>d</b>) Predicted distances between deoxycytidine and the residues at positions 81 and 110, where Q81 is replaced by alanine, and A110 is replaced by glycine. Model generated by ColabFold v1.5.5.</p>
Full article ">Figure 5
<p>The active site of <span class="html-italic">Dm</span>dNK with bound deoxycytidine. (<b>a</b>) Stacking interaction between W57 and cytosine atoms. PDB ID: 1j90. (<b>b</b>) Predicted distances between C5 and C6 atoms of deoxycytidine and residue at position 57, where W57 is replaced by valine. Model generated by ColabFold v1.5.5. (<b>c</b>) Possible interactions between C5 and C6 atoms of deoxycytidine and residue at position 57, where W57 is replaced by phenylalanine. Model generated by ColabFold v1.5.5.</p>
Full article ">Figure 6
<p>Superimposition of <span class="html-italic">Bs</span>dCK (cyan) on <span class="html-italic">Dm</span>dNK (grey) with 2′-deoxycytidine (green) in the active centres. (<b>a</b>) M88 and A110 residues of <span class="html-italic">Dm</span>dNK-WT, and corresponding R70 and D93 residues of <span class="html-italic">Bs</span>dCK-WT near the <span class="html-italic">N</span><sup>4</sup> and C5 positions of 2′-deoxycytidine. (<b>b</b>) M88 and A110 residues of <span class="html-italic">Dm</span>dNK-WT, and M70 and A93 residues of <span class="html-italic">Bs</span>dCK-R70M+D93A mutant variant near the <span class="html-italic">N</span><sup>4</sup> and C5 positions of 2′-deoxycytidine. Grey residues belong to <span class="html-italic">Dm</span>dNK. Cyan residues belong to <span class="html-italic">Bs</span>dCK. Yellow dashed lines show predicted distances between the residues of <span class="html-italic">Bs</span>dCK and 2′-deoxycytidine. <span class="html-italic">Dm</span>dNK PDB ID: 1j90. <span class="html-italic">Bs</span>dCK models generated using ColabFold v.1.5.5 software.</p>
Full article ">Figure 7
<p>(<b>a</b>) Molecular docking of <span class="html-italic">N</span><sup>4</sup>-glycinoyl-2′-deoxycytidine in the active centre of <span class="html-italic">Dm</span>dNK-WT. (<b>b</b>) Molecular docking of <span class="html-italic">N</span>-[1-(β-D-ribofuranosyl)-2-oxo-4-pyrimidinyl]-glycine in the active centre of <span class="html-italic">Dm</span>dNK-WT. PDB ID: 1j90. Molecular docking performed by DiffDock software (<a href="https://huggingface.co/spaces/reginabarzilaygroup/DiffDock-Web" target="_blank">https://huggingface.co/spaces/reginabarzilaygroup/DiffDock-Web</a>; accessed on 6 August 2024).</p>
Full article ">Scheme 1
<p>Preparation of <span class="html-italic">N</span>-[1-(β-D-ribofuranosyl)-2-oxo-4-pyrimidinyl]-amino acids.</p>
Full article ">Scheme 2
<p>Preparation of <span class="html-italic">N</span><sup>4</sup>-hydroxy- and <span class="html-italic">N</span><sup>4</sup>-alkoxycytidines.</p>
Full article ">
13 pages, 1632 KiB  
Article
Phylogenetic Network Analyses Reveal the Influence of Transmission Clustering on the Spread of HIV Drug Resistance in Quebec from 2002 to 2022
by Bluma G. Brenner, Ruxandra-Ilinca Ibanescu, Maureen Oliveira, Guillaume Margaillan, Bertrand Lebouché, Réjean Thomas, Jean Guy Baril, René-Pierre Lorgeoux, Michel Roger, Jean-Pierre Routy and the Montreal Primary HIV Infection (PHI) Cohort Study Group
Viruses 2024, 16(8), 1230; https://doi.org/10.3390/v16081230 - 31 Jul 2024
Viewed by 1027
Abstract
Background: HIV drug resistance (HIV-DR) may jeopardize the benefit of antiretroviral therapy (ART) in treatment and prevention. This study utilized viral phylogenetics to resolve the influence of transmission networks on sustaining the spread of HIV-DR in Quebec spanning 2002 to 2022. Methods: Time [...] Read more.
Background: HIV drug resistance (HIV-DR) may jeopardize the benefit of antiretroviral therapy (ART) in treatment and prevention. This study utilized viral phylogenetics to resolve the influence of transmission networks on sustaining the spread of HIV-DR in Quebec spanning 2002 to 2022. Methods: Time trends in acquired (ADR) and transmitted drug resistance (TDR) were delineated in treatment-experienced (n = 3500) and ART-naïve persons (n = 6011) with subtype B infections. Similarly, non-B-subtype HIV-DR networks were assessed pre- (n = 1577) and post-ART experience (n = 488). Risks of acquisition of resistance-associated mutations (RAMs) were related to clustering using 1, 2–5, vs. 6+ members per cluster as categorical variables. Results: Despite steady declines in treatment failure and ADR since 2007, rates of TDR among newly infected, ART-naive persons remained at 14% spanning the 2007–2011, 2012–2016, and 2017–2022 periods. Notably, half of new infections among men having sex with men and heterosexual groups were linked in large, clustered networks having a median of 35 (14–73 IQR) and 16 (9–26 IQR) members per cluster, respectively. Cluster membership and size were implicated in forward transmission of non-nucleoside reverse transcriptase inhibitor NNRTI RAMs (9%) and thymidine analogue mutations (TAMs) (5%). In contrast, transmission of M184V, K65R, and integrase inhibitors (1–2%) remained rare. Levels of TDR reflected viral replicative fitness. The median baseline viremia in ART-naïve groups having no RAMs, NNRTI RAMs, TAMs, and M184VI were 46.088, 38,447, 20,330, and 6811 copies/mL, respectively (p < 0.0001). Conclusion: Phylogenetics emphasize the need to prioritize ART and pre-exposure prophylaxis strategies to avert the expansion of transmission cascades of HIV-DR. Full article
Show Figures

Figure 1

Figure 1
<p>Frequencies of acquired and transmitted resistance-associated mutations (RAMs) in ART-experienced and ART-naïve groups with subtype B infections. (<b>A</b>) Rates of acquired resistance to PIs (green), NRTIs/TAMs (blue), and NNRTIs (red) among patients failing first ART treatment. (<b>B</b>) Rates of transmitted drug resistance mutations in newly genotyped, ART-naïve persons. Mutations are based on the updated World Health Organization 2009 list. Several mutations, including E138A, E138K, L234I, V318F, and N348I, associated with resistance to newer drugs rilpivirine and doravirine are also depicted.</p>
Full article ">Figure 2
<p>Time trends in the prevalence of acquired and transmitted drug resistance in first genotypes of persons bearing subtype B infections. (<b>A</b>). Frequencies of ADR in treated persons failing treatment (viral load &gt; 100 copies/mL) in the 2007–2011 (blue), 2012–2016 (red) and 2017–2022 (green) periods. (<b>B</b>). Frequencies of TDR in treatment-naïve individuals during these three periods.</p>
Full article ">Figure 3
<p>Influence of clustering on onward spread of TDR in treatment-naïve persons with subtype B infections. Representative large clusters bearing NNRTI RAMs (red nodes) are circled. Viral variants with resistance to NRTI mutations (blue nodes) include smaller clusters. Variants bearing PIs (green), and dual-class or multidrug resistance (MDR) are largely singleton transmissions.</p>
Full article ">Figure 4
<p>The impact of clustering on rates of transmission of resistance to nucleoside reverse transcriptase (RT) inhibitors (NRTIs), non-nucleoside RT inhibitors (NNRTIs), protease inhibitors (PIs), and dual- and triple-class multidrug resistance (MDR).</p>
Full article ">Figure 5
<p>Influence of TDR mutations on viral replicative fitness. Pretreatment viremia (median ± IQR) in ART-naive persons with no RAMs (<span class="html-italic">n</span> = 5132), M184V/I (<span class="html-italic">n</span> = 258), thymidine analogue mutations (TAMs) (<span class="html-italic">n</span> = 359), and NNRTI RAMs present as unique (U) transmissions (<span class="html-italic">n</span> = 104) or within large 6+ clusters (<span class="html-italic">n</span> = 271, nonparametric ANOVA analysis and Dunn’s post hoc comparison tests of statistical differences in viremia between groups). <span class="html-italic">p</span>-values: * <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; 0.0001.</p>
Full article ">Figure 6
<p>The prevalence of transmitted (TDR) and acquired (ADR) drug resistance among genotyped INSTI-naïve (<span class="html-italic">n</span> = 1802) and INSTI-experienced (<span class="html-italic">n</span> = 1461) persons in Quebec.</p>
Full article ">
23 pages, 7117 KiB  
Article
Synthesis of Chiral Acyclic Pyrimidine Nucleoside Analogues from DHAP-Dependent Aldolases
by Mariano Nigro, Israél Sánchez-Moreno, Raúl Benito-Arenas, Ana L. Valino, Adolfo M. Iribarren, Nicolás Veiga, Eduardo García-Junceda and Elizabeth S. Lewkowicz
Biomolecules 2024, 14(7), 750; https://doi.org/10.3390/biom14070750 - 25 Jun 2024
Viewed by 1050
Abstract
Dihydroxyacetone phosphate (DHAP)-dependent aldolases catalyze the aldol addition of DHAP to a variety of aldehydes and generate compounds with two stereocenters. This reaction is useful to synthesize chiral acyclic nucleosides, which constitute a well-known class of antiviral drugs currently used. In such compounds, [...] Read more.
Dihydroxyacetone phosphate (DHAP)-dependent aldolases catalyze the aldol addition of DHAP to a variety of aldehydes and generate compounds with two stereocenters. This reaction is useful to synthesize chiral acyclic nucleosides, which constitute a well-known class of antiviral drugs currently used. In such compounds, the chirality of the aliphatic chain, which mimics the open pentose residue, is crucial for activity. In this work, three DHAP-dependent aldolases: fructose-1,6-biphosphate aldolase from rabbit muscle, rhanmulose-1-phosphate aldolase from Thermotoga maritima, and fuculose-1-phosphate aldolase from Escherichia coli, were used as biocatalysts. Aldehyde derivatives of thymine and cytosine were used as acceptor substrates, generating new acyclic nucleoside analogues containing two new stereocenters with conversion yields between 70% and 90%. Moreover, structural analyses by molecular docking were carried out to gain insights into the diasteromeric excess observed. Full article
Show Figures

Figure 1

Figure 1
<p>Representative acyclic nucleosides with medicinal activities.</p>
Full article ">Figure 2
<p>Stereoselective products generated by the four DHAP-dependent aldolases.</p>
Full article ">Figure 3
<p>Expected acyclic nucleoside analogues produced by three different aldolases.</p>
Full article ">Figure 4
<p>Partial view of the <sup>1</sup>H NMR spectrum of the aldol product (<b>2a+3a</b>) obtained by RAMA bio-catalyzed aldol addition with thyminyl acetaldehyde and DHAP as substrates. The inset shows the different coupling constants of C3 hydrogens (<span class="html-italic">J</span><sub>3–4</sub> = 1.9 Hz) and C3′ hydrogens (<span class="html-italic">J</span><sub>3′–4′</sub> = 6.9 Hz), indicating <b>2a</b> and <b>3a</b> presence, respectively.</p>
Full article ">Figure 5
<p>Coupling constants between neighboring protons [<a href="#B67-biomolecules-14-00750" class="html-bibr">67</a>].</p>
Full article ">Figure 6
<p>Partial view of the <sup>1</sup>H NMR spectrum of the aldol product (<b>4b</b>+<b>5b</b>) obtained by <span class="html-italic">Tm</span>Rhua-1PA bio-catalyzed aldol addition, with <b>1b</b> and DHAP as substrates. The inset shows C3 hydrogen (<span class="html-italic">J</span><sub>3–4</sub> = 1.9 Hz) and C3′ hydrogen (<span class="html-italic">J</span><sub>3′–4′</sub> = 4.7 Hz) signals, indicating <b>4b</b> and <b>5b</b> presence, respectively.</p>
Full article ">Figure 7
<p>Partial view of <sup>13</sup>C NMR spectrum of the aldol products (<b>4b</b>+<b>5b</b>) obtained by<span class="html-italic">Tm</span>Rhua-1PA bio-catalyzed aldol addition and <b>1b</b> as a substrate. The insets show the double signals corresponding to the chiral carbons C3 and C4, confirming the presence of both <b>4b</b> and <b>5b</b> isomers.</p>
Full article ">Figure 8
<p>Partial view of <sup>1</sup>H NMR spectrum of the aldol product (<b>4b</b>+<b>5b</b>) obtained by <span class="html-italic">Ec</span>Fuc-1PA bio-catalyzed aldol addition of <b>1b</b> and DHAP as substrates. The inset shows the C3 (<span class="html-italic">J</span><sub>3–4</sub> = 1.6 Hz) and C3′ (<span class="html-italic">J</span><sub>3′–4′</sub> = 5.5 Hz) hydrogen signals.</p>
Full article ">Figure 9
<p>Comparison between the active sites of two DHAP-dependent aldolases: (<b>a</b>) DHAP anchored in the <span class="html-italic">Ec</span>Fuc-1PA active site and (<b>b</b>) DHAP anchored in the <span class="html-italic">Tm</span>Rhu-1PA active site. Hydrophobicity is mapped onto a Connolly solvent-accessible surface of the receptor. Non-polar hydrogen atoms are omitted for clarity. Atom color code: C (grey), N (blue), O (red), Zn/Co (violet), P (orange), and H (white). The two protein chains are depicted with different colors (green and turquoise).</p>
Full article ">Figure 10
<p>Docking for the cytosine derivative <b>1b</b> with <span class="html-italic">Tm</span>Rhu-1PA, generating Solution 1: (<b>a</b>) DHAP coordinated by the carbonyl group of <b>1b</b> ring, and (<b>b</b>) the -NH<sub>2</sub> group of <b>1b</b> interacting with the lateral hydrophilic pocket and the aldehyde group of the derivate with the upper hydrophilic pocket of the active site. The interactions are represented as dashed lines: green (H-bonds) and grey (coordination bonds). In (<b>b</b>), hydrophobicity is mapped onto a Connolly solvent-accessible surface of the receptor. Non-polar hydrogen atoms are omitted for clarity. Atom color code: C (grey), N (blue), O (red), Zn/Co (violet), P (orange), and H (white). The two protein chains are depicted with different colors (green and turquoise).</p>
Full article ">Figure 11
<p>Docking for the cytosine derivative <b>1b</b> with <span class="html-italic">Tm</span>Rhu-1PA and DHAP generating two possible solutions: (<b>a</b>) Solution 2: the R group is directed toward the upper hydrophilic pocket and (<b>b</b>) Solution 3: the R group is directed toward the lower hydrophilic pocket. The interactions are represented as dashed lines: green (H-bonds), grey (coordination bonds), and pink (pi-mediated interactions). Hydrophobicity is mapped onto a Connolly solvent-accessible surface of the receptor. Non-polar hydrogen atoms are omitted for clarity. Atom color code: C (grey), N (blue), O (red), Co (violet), P (orange), and H (white). The two protein chains are depicted with different colors (green and turquoise).</p>
Full article ">Figure 11 Cont.
<p>Docking for the cytosine derivative <b>1b</b> with <span class="html-italic">Tm</span>Rhu-1PA and DHAP generating two possible solutions: (<b>a</b>) Solution 2: the R group is directed toward the upper hydrophilic pocket and (<b>b</b>) Solution 3: the R group is directed toward the lower hydrophilic pocket. The interactions are represented as dashed lines: green (H-bonds), grey (coordination bonds), and pink (pi-mediated interactions). Hydrophobicity is mapped onto a Connolly solvent-accessible surface of the receptor. Non-polar hydrogen atoms are omitted for clarity. Atom color code: C (grey), N (blue), O (red), Co (violet), P (orange), and H (white). The two protein chains are depicted with different colors (green and turquoise).</p>
Full article ">Figure 12
<p>Docking for the thymine derivative <b>1a</b> with <span class="html-italic">Tm</span>Rhu-1PA: (<b>a</b>) Solution 4: the aldehydic hydrogen shifts its orientation, exposing the <span class="html-italic">re</span> face to the DHAP. (<b>b</b>) Solution 5: the aldehydic hydrogen exposes the <span class="html-italic">si</span> face to the DHAP. The interactions are represented as dashed lines: green (H-bonds), light green (non-conventional H-bonds), grey (coordination bonds), and pink (pi-mediated interactions). Hydrophobicity is mapped onto a Connolly solvent-accessible surface of the receptor. Non-polar hydrogen atoms are omitted for clarity. Atom color code: C (grey), N (blue), O (red), Zn/Co (violet), P (orange), and H (white). The two protein chains are depicted with different colors (green and turquoise).</p>
Full article ">Figure 13
<p>Docking for the cytosine derivative <b>1b</b> attached by the aldehyde group to the metal ion of the active site of <span class="html-italic">Ec</span>Fuc-1PA: (<b>a</b>) Solution 6: the aldehydic hydrogen shifts its orientation, exposing the <span class="html-italic">re</span> face to the DHAP. (<b>b</b>) Solution 7: the aldehydic hydrogen exposes the <span class="html-italic">si</span> face to the DHAP. The interactions are represented as dashed lines: green (H-bonds), light green (non-conventional H-bonds), grey (coordination bonds), and pink (pi-mediated interactions). Hydrophobicity is mapped onto a Connolly solvent-accessible surface of the receptor. Non-polar hydrogen atoms are omitted for clarity. Atom color code: C (grey), N (blue), O (red), Zn/Co (violet), P (orange), and H (white). The two protein chains are depicted with different colors (green and turquoise).</p>
Full article ">Figure 14
<p>Docking for the thymine derivative <b>1a</b> attached by the aldehyde group to the metal ion of the active site of <span class="html-italic">Ec</span>Fuc-1PA: (<b>a</b>) Solution 8: Van der Waals interactions optimized in the hydrophobic pocket of the active site. (<b>b</b>) Solution 9: increase in the intensity of the substrate <b>1a</b>–protein hydrogen bonds while minimizing the steric clash. The interactions are represented as dashed lines: green (H-bonds), light green (non-conventional H-bonds), grey (coordination bonds), and pink (pi-mediated interactions). Hydrophobicity is mapped onto a Connolly solvent-accessible surface of the receptor. Non-polar hydrogen atoms are omitted for clarity. Atom color code: C (grey), N (blue), O (red), Zn/Co (violet), P (orange), and H (white). The two protein chains are depicted with different colors (green and turquoise).</p>
Full article ">
15 pages, 2312 KiB  
Article
Synthesis of Substituted 1,2,4-Triazole-3-Thione Nucleosides Using E. coli Purine Nucleoside Phosphorylase
by Ilya V. Fateev, Sobirdjan A. Sasmakov, Jaloliddin M. Abdurakhmanov, Abdukhakim A. Ziyaev, Shukhrat Sh. Khasanov, Farkhod B. Eshboev, Oybek N. Ashirov, Valeriya D. Frolova, Barbara Z. Eletskaya, Olga S. Smirnova, Maria Ya. Berzina, Alexandra O. Arnautova, Yulia A. Abramchik, Maria A. Kostromina, Alexey L. Kayushin, Konstantin V. Antonov, Alexander S. Paramonov, Valeria L. Andronova, Georgiy A. Galegov, Roman S. Esipov, Shakhnoz S. Azimova, Anatoly I. Miroshnikov and Irina D. Konstantinovaadd Show full author list remove Hide full author list
Biomolecules 2024, 14(7), 745; https://doi.org/10.3390/biom14070745 - 24 Jun 2024
Viewed by 1415
Abstract
1,2,4-Triazole derivatives have a wide range of biological activities. The most well-known drug that contains 1,2,4-triazole as part of its structure is the nucleoside analogue ribavirin, an antiviral drug. Finding new nucleosides based on 1,2,4-triazole is a topical task. The aim of this [...] Read more.
1,2,4-Triazole derivatives have a wide range of biological activities. The most well-known drug that contains 1,2,4-triazole as part of its structure is the nucleoside analogue ribavirin, an antiviral drug. Finding new nucleosides based on 1,2,4-triazole is a topical task. The aim of this study was to synthesize ribosides and deoxyribosides of 1,2,4-triazole-3-thione derivatives and test their antiviral activity against herpes simplex viruses. Three compounds from a series of synthesized mono- and disubstituted 1,2,4-triazole-3-thione derivatives were found to be substrates for E. coli purine nucleoside phosphorylase. Of six prepared nucleosides, the riboside and deoxyriboside of 3-phenacylthio-1,2,4-triazole were obtained at good yields. The yields of the disubstituted 1,2,4-triazol-3-thiones were low due to the effect of bulky substituents at the C3 and C5 positions on the selectivity of enzymatic glycosylation for one particular nitrogen atom in the triazole ring. The results of cytotoxic and antiviral studies on acyclovir-sensitive wild-type strain HSV-1/L2(TK+) and acyclovir-resistant strain (HSV-1/L2/RACV) in Vero E6 cell culture showed that the incorporation of a thiobutyl substituent into the C5 position of 3-phenyl-1,2,4-triazole results in a significant increase in the cytotoxicity of the base and antiviral activity. The highest antiviral activity was observed in the 3-phenacylthio-1-(β-D-ribofuranosyl)-1,2,4-triazole and 5-butylthio-1-(2-deoxy-β-D-ribofuranosyl)-3-phenyl-1,2,4-triazole nucleosides, with their selectivity indexes being significantly higher than that of ribavirin. It was also found that with the increasing lipophilicity of the nucleosides, the activity and toxicity of the tested compounds increased. Full article
Show Figures

Figure 1

Figure 1
<p>Synthesis of 3- and 5-substituted 1,2,4-triazoles.</p>
Full article ">Figure 2
<p>Synthesis of nucleosides <b>8</b>–<b>13</b> using nucleoside phosphorylases. PNP—<span class="html-italic">E. coli</span> purine nucleoside phosphorylase, UP—<span class="html-italic">E. coli</span> uridine nucleoside phosphorylase, Urd—uridine, dUrd—2′-deoxyuridine, Ura—uracil, Pi—inorganic phosphate.</p>
Full article ">Figure 3
<p>The interaction of carbon and hydrogen atoms observed in the <sup>1</sup>H,<sup>13</sup>C-HMBC spectra for two glycosylation variants. Red arrows depict the interactions between the carbon atom bonded to the thio substituent and protons of the molecule, and blue arrows are the interactions of the other carbon atom in triazole ring.</p>
Full article ">Figure 4
<p>Cytotoxic properties CC<sub>50</sub> (<b>A</b>), antiviral activity IC<sub>50</sub> (<b>B</b>), and selectivity index SI (<b>C</b>) of compounds <b>3</b>–<b>13</b> and ribavirin.</p>
Full article ">
18 pages, 765 KiB  
Review
mRNA Technology and Mucosal Immunization
by Antonio Toniolo, Giuseppe Maccari and Giovanni Camussi
Vaccines 2024, 12(6), 670; https://doi.org/10.3390/vaccines12060670 - 17 Jun 2024
Viewed by 1859
Abstract
Current mRNA vaccines are mainly administered via intramuscular injection, which induces good systemic immunity but limited mucosal immunity. Achieving mucosal immunity through mRNA vaccination could diminish pathogen replication at the entry site and reduce interhuman transmission. However, delivering mRNA vaccines to mucosae faces [...] Read more.
Current mRNA vaccines are mainly administered via intramuscular injection, which induces good systemic immunity but limited mucosal immunity. Achieving mucosal immunity through mRNA vaccination could diminish pathogen replication at the entry site and reduce interhuman transmission. However, delivering mRNA vaccines to mucosae faces challenges like mRNA degradation, poor entry into cells, and reactogenicity. Encapsulating mRNA in extracellular vesicles may protect the mRNA and reduce reactogenicity, making mucosal mRNA vaccines possible. Plant-derived extracellular vesicles from edible fruits have been investigated as mRNA carriers. Studies in animals show that mRNA vehiculated in orange-derived extracellular vesicles can elicit both systemic and mucosal immune responses when administered by the oral, nasal, or intramuscular routes. Once lyophilized, these products show remarkable stability. The optimization of mRNA to improve translation efficiency, immunogenicity, reactogenicity, and stability can be obtained through adjustments of the 5′cap region, poly-A tail, codons selection, and the use of nucleoside analogues. Recent studies have also proposed self-amplifying RNA vaccines containing an RNA polymerase as well as circular mRNA constructs. Data from parenterally primed animals demonstrate the efficacy of nasal immunization with non-adjuvanted protein, and studies in humans indicate that the combination of a parenteral vaccine with the natural exposure of mucosae to the same antigen provides protection and reduces transmission. Hence, mucosal mRNA vaccination would be beneficial at least in organisms pre-treated with parenteral vaccines. This practice could have wide applications for the treatment of infectious diseases. Full article
Show Figures

Figure 1

Figure 1
<p>The activation of adaptive immunity by an mRNA construct encapsulated into extracellular vesicles (EVs). EVs protect the mRNA from degradation and allow for its penetration into mucosal epithelial cells and antigen-presenting cells (APCs). Here, it is translated into the encoded protein which, after processing, is displayed on the cell surface complexed with MHC molecules. The interaction of APCs with lymphocytes (Ly) allow them to recognize the foreign protein and become activated.</p>
Full article ">Figure 2
<p>Enhancing mRNA properties through predictive design. (<b>A</b>) 5’ capping: improves molecule stability by shielding it from exonucleases. (<b>B</b>) Codon usage: influences tissue- or organ-specific expression by optimizing translation speed, based on availability of tRNAs. (<b>C</b>) Nucleoside modifications (e.g., replacing uridine with pseudouridine): enhances mRNA stability, reduces activation of innate immunity, improves translation efficiency. (<b>D</b>) 3’ poly-A tail: enhances mRNA stability and translation efficiency.</p>
Full article ">
17 pages, 336 KiB  
Article
In Vitro Evaluation of Synergistic Essential Oils Combination for Enhanced Antifungal Activity against Candida spp.
by Lukáš Hleba, Miroslava Hlebová and Ivana Charousová
Life 2024, 14(6), 693; https://doi.org/10.3390/life14060693 - 28 May 2024
Cited by 3 | Viewed by 1858
Abstract
In recent years, a significant number of infections have been attributed to non-albicidal Candida species (NAC), mainly due to the increasing resistance of NAC to antifungal agents. As only a few antifungal agents are available (azoles, echinocandins, polyenes, allylamines and nucleoside analogues), it [...] Read more.
In recent years, a significant number of infections have been attributed to non-albicidal Candida species (NAC), mainly due to the increasing resistance of NAC to antifungal agents. As only a few antifungal agents are available (azoles, echinocandins, polyenes, allylamines and nucleoside analogues), it is very important to look for possible alternatives to inhibit resistant fungi. One possibility could be essential oils (EOs), which have been shown to have significant antifungal and antibacterial activity. Therefore, in this study, the efficacy of 12 EOs and their combinations was evaluated against four yeasts of the genus Candida (C. albicas, C. glabrata, C. tropicalis and C. parapsilosis). GC-MS and GC-MS FID techniques were used for the chemical analysis of all EOs. VITEK-2XL was used to determine the antifungal susceptibility of the tested Candida spp. strains. The agar disc diffusion method was used for primary screening of the efficacy of the tested EOs. The broth dilution method was used to determine the minimum inhibitory concentrations (MICs) of the most potent EOs. After MIC cultivation, the minimum fungicidal concentration (MFC) was determined on Petri dishes (60 mm). The synergistic effect of combined EOs was evaluated using the checkerboard method and expressed as a fractional inhibitory concentration index (FICI). The results showed that ginger > ho-sho > absinth > dill > fennel > star anise > and cardamom were the most effective EOs. For all Candida species tested, the synergy was mainly observed in these combinations: ginger/fennel for C. albicans FICI 0.25 and C. glabrata, C. tropicalis and C. parapsilosis FICI 0.5 and absinth/fennel for C. albicans FICI 0.3125, C. tropicalis FICI 0.3125 and C. parapsilosis FICI 0.375. Our results suggest that the resistance of fungal pathogens to available antifungals could be reduced by combining appropriate EOs. Full article
20 pages, 2459 KiB  
Article
Co-Delivery of an Innovative Organoselenium Compound and Paclitaxel by pH-Responsive PCL Nanoparticles to Synergistically Overcome Multidrug Resistance in Cancer
by Daniela Mathes, Letícia Bueno Macedo, Taís Baldissera Pieta, Bianca Costa Maia, Oscar Endrigo Dorneles Rodrigues, Julliano Guerin Leal, Marcelo Wendt, Clarice Madalena Bueno Rolim, Montserrat Mitjans and Daniele Rubert Nogueira-Librelotto
Pharmaceutics 2024, 16(5), 590; https://doi.org/10.3390/pharmaceutics16050590 - 26 Apr 2024
Viewed by 1606
Abstract
In this study, we designed the association of the organoselenium compound 5′-Seleno-(phenyl)-3′-(ferulic-amido)-thymidine (AFAT-Se), a promising innovative nucleoside analogue, with the antitumor drug paclitaxel, in poly(ε-caprolactone) (PCL)-based nanoparticles (NPs). The nanoprecipitation method was used, adding the lysine-based surfactant, 77KS, as a pH-responsive adjuvant. The [...] Read more.
In this study, we designed the association of the organoselenium compound 5′-Seleno-(phenyl)-3′-(ferulic-amido)-thymidine (AFAT-Se), a promising innovative nucleoside analogue, with the antitumor drug paclitaxel, in poly(ε-caprolactone) (PCL)-based nanoparticles (NPs). The nanoprecipitation method was used, adding the lysine-based surfactant, 77KS, as a pH-responsive adjuvant. The physicochemical properties presented by the proposed NPs were consistent with expectations. The co-nanoencapsulation of the bioactive compounds maintained the antioxidant activity of the association and evidenced greater antiproliferative activity in the resistant/MDR tumor cell line NCI/ADR-RES, both in the monolayer/two-dimensional (2D) and in the spheroid/three-dimensional (3D) assays. Hemocompatibility studies indicated the safety of the nanoformulation, corroborating the ability to spare non-tumor 3T3 cells and human mononuclear cells of peripheral blood (PBMCs) from cytotoxic effects, indicating its selectivity for the cancerous cells. Furthermore, the synergistic antiproliferative effect was found for both the association of free compounds and the co-encapsulated formulation. These findings highlight the antitumor potential of combining these bioactives, and the proposed nanoformulation as a potentially safe and effective strategy to overcome multidrug resistance in cancer therapy. Full article
(This article belongs to the Section Nanomedicine and Nanotechnology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>AFAT-Se-PTX-PCL-77KS-NP morphology by SEM.</p>
Full article ">Figure 2
<p>Chromatograms obtained after method validation for the active compounds and nanoformulations under analysis. 1—AFAT-Se-PTX-Free, 2—AFAT-Se-PTX-PCL-77 KS-NP, and 3—PCL-77KS-NP. Peaks A and B correspond to AFAT-Se; peaks C and D correspond to PTX.</p>
Full article ">Figure 3
<p>Scavenging activity of AFAT-Se-PTX-Free, AFAT-Se-PTX-PCL-77KS-NP, and PCL-77KS-NP using DPPH and ABTS assays. Results are expressed as mean ± SE of three independent experiments. Statistical analyses were performed using ANOVA followed by the Tukey post hoc test. <sup>a</sup> Significantly different from AFAT-Se-PTX-PCL-77KS-NP (<span class="html-italic">p</span> &lt; 0.05), <sup>b</sup> from PCL-77KS-NP (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Safety profile using non-tumor cell line 3T3 and human mononuclear cells of peripheral blood (PBMC). Results are expressed as mean ± SE of three independent experiments. Statistical analyses were performed using ANOVA followed by the Tukey post hoc test. No significant differences were found between concentrations used in the assays.</p>
Full article ">Figure 5
<p>Hemocompatibility study of AFAT-Se-PTX-Free, AFAT-Se-PTX-PCL-77KS-NP, and PCL-77KS-NP after incubation with human erythrocytes. Results are expressed as mean ± SE of three independent experiments. Statistical analyses were performed using ANOVA followed by the Tukey post hoc test. <sup>a</sup> Significantly different from AFAT-Se-PTX-PCL-77KS-NP (<span class="html-italic">p</span> &lt; 0.05), <sup>b</sup> from PCL-77KS-NP (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>pH-dependent membrane-lytic activity of AFAT-Se-PTX-PCL-NP and PCL-NP (NPs without 77KS), and AFAT-Se-PTX-PCL-77KS-NP and PCL-77KS-NP (NPs with 77KS), after 5 h of incubation with human erythrocytes at pH 5.4, 6.6, and 7.4. Each value represents mean ± SE of three experiments. Statistical analyses were performed using ANOVA followed by the Tukey post hoc test. <sup>a</sup> Significantly different from pH 7.4 (<span class="html-italic">p</span> &lt; 0.05) and <sup>b</sup> from pH 6.6 (<span class="html-italic">p</span> &lt; 0.05). * Indicates significant difference between formulations with and without 77KS (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 7
<p>(<b>A</b>–<b>C</b>) In vitro cell viability in NCI/ADR-RES cell line using 2D cell model by MTT assay after 72 h of treatment. Data are expressed as mean of three independent experiments ± SE. Statistical analyses were performed using ANOVA followed by the Duncan post hoc test. <sup>a</sup> significant difference from AFAT-Se-Free (<span class="html-italic">p</span> &lt; 0.05), <sup>b</sup> significant difference from PTX-Free, * significant difference from AFAT-Se-PTX-Free and <sup>#</sup> significant difference from PCL-77KS-NP. (<b>D</b>) Combination index values for the association of active compounds, AFAT-Se-PTX-Free and AFAT-Se-PTX-PCL-77KS-NP, in NCI/ADR-RES cell line.</p>
Full article ">Figure 8
<p>Cytotoxicity of AFAT-Se-PTX-Free and AFAT-Se-PTX-PCL-77KS-NP against NCI/ADR-RES spheroids. Spheroid area percentage (%) determined in comparison to day 0, which was set as 100%. Statistical analyses were performed using ANOVA followed by the Tukey post hoc test. * significant difference from control, <sup>@</sup> significant difference from AFAT-Se-PTX-Free.</p>
Full article ">Figure 9
<p>Representative images of NCI/ADR-RES spheroids treated with AFAT-Se-PTX-Free and AFAT-Se-PTX-PCL-77KS-NP at concentration of 60 μg/mL AFAT-Se + 12 μg/mL PTX. Images were obtained using an inverted microscope at day 0 (before treatment), and after exposure to the treatments for 2, 5, 7, 9, and 12 days.</p>
Full article ">
20 pages, 23188 KiB  
Article
Boosting Clear Cell Renal Carcinoma-Specific Drug Discovery Using a Deep Learning Algorithm and Single-Cell Analysis
by Yishu Wang, Xiaomin Chen, Ningjun Tang, Mengyao Guo and Dongmei Ai
Int. J. Mol. Sci. 2024, 25(7), 4134; https://doi.org/10.3390/ijms25074134 - 8 Apr 2024
Cited by 1 | Viewed by 2883
Abstract
Clear cell renal carcinoma (ccRCC), the most common subtype of renal cell carcinoma, has the high heterogeneity of a highly complex tumor microenvironment. Existing clinical intervention strategies, such as target therapy and immunotherapy, have failed to achieve good therapeutic effects. In this article, [...] Read more.
Clear cell renal carcinoma (ccRCC), the most common subtype of renal cell carcinoma, has the high heterogeneity of a highly complex tumor microenvironment. Existing clinical intervention strategies, such as target therapy and immunotherapy, have failed to achieve good therapeutic effects. In this article, single-cell transcriptome sequencing (scRNA-seq) data from six patients downloaded from the GEO database were adopted to describe the tumor microenvironment (TME) of ccRCC, including its T cells, tumor-associated macrophages (TAMs), endothelial cells (ECs), and cancer-associated fibroblasts (CAFs). Based on the differential typing of the TME, we identified tumor cell-specific regulatory programs that are mediated by three key transcription factors (TFs), whilst the TF EPAS1/HIF-2α was identified via drug virtual screening through our analysis of ccRCC’s protein structure. Then, a combined deep graph neural network and machine learning algorithm were used to select anti-ccRCC compounds from bioactive compound libraries, including the FDA-approved drug library, natural product library, and human endogenous metabolite compound library. Finally, five compounds were obtained, including two FDA-approved drugs (flufenamic acid and fludarabine), one endogenous metabolite, one immunology/inflammation-related compound, and one inhibitor of DNA methyltransferase (N4-methylcytidine, a cytosine nucleoside analogue that, like zebularine, has the mechanism of inhibiting DNA methyltransferase). Based on the tumor microenvironment characteristics of ccRCC, five ccRCC-specific compounds were identified, which would give direction of the clinical treatment for ccRCC patients. Full article
Show Figures

Figure 1

Figure 1
<p>The workflow of methods and process used in this study.</p>
Full article ">Figure 2
<p>(<b>A</b>) The landscape of cell clusters determined via scRNA−seq. (<b>B</b>) Heatmap of the marker genes in different cell subclusters.</p>
Full article ">Figure 3
<p>(<b>A</b>) UMAP of subtypes of macrophage cells. (<b>B</b>) Evolutionary trajectory of sub-clusters reconstructed the cell differentiation order. (<b>C</b>) Variate expression levels of hub genes in the development of the pseudotime trajectory. (<b>D</b>) Heatmap of top 50 genes which varied as a function of pseudotime.</p>
Full article ">Figure 4
<p>(<b>A</b>) UMAP clustering results of the subclusters of endothelial cells. (<b>B</b>) Evolutionary trajectory of the subclusters, reconstructing the cell differentiation order; numbers 1 to 3 denote the turning points of trajectory development. (<b>C</b>) The expression distribution of significantly expressed genes in different EC subtypes.</p>
Full article ">Figure 5
<p>(<b>A</b>) UMAP of the subclusters of fibroblasts. (<b>B</b>) Evolutionary trajectory of subclusters according to their reconstructed cell differentiation order. (<b>C</b>) GO functional analyses of the subtypes CAF−2 and CAF−3, respectively.</p>
Full article ">Figure 5 Cont.
<p>(<b>A</b>) UMAP of the subclusters of fibroblasts. (<b>B</b>) Evolutionary trajectory of subclusters according to their reconstructed cell differentiation order. (<b>C</b>) GO functional analyses of the subtypes CAF−2 and CAF−3, respectively.</p>
Full article ">Figure 6
<p>(<b>A</b>) UMAP of the subclusters in T cells. (<b>B</b>) The proportion of T cells in six ccRCC samples. (<b>C</b>) Survival analysis of patients stratified according to their expression of the marker genes of T cells in the TCGA dataset.</p>
Full article ">Figure 6 Cont.
<p>(<b>A</b>) UMAP of the subclusters in T cells. (<b>B</b>) The proportion of T cells in six ccRCC samples. (<b>C</b>) Survival analysis of patients stratified according to their expression of the marker genes of T cells in the TCGA dataset.</p>
Full article ">Figure 7
<p>(<b>A</b>) The pathway that EPAS1 takes part in. (<b>B</b>) Structural domain of the protein EPAS1.</p>
Full article ">Figure 8
<p>(<b>A</b>–<b>E</b>): docking models and chemical structures of the five screened compounds.</p>
Full article ">Figure 9
<p>Molecular dynamics simulation results of FDA drugs flufenamic acid and fludarabine. (<b>A</b>–<b>D</b>) Flufenamic acid: root mean square deviation curve (RSMD), root mean square wave curve (RMSF), small molecule root mean square wave curve (RMSF), and statistics of interaction proportion of different residues. (<b>E</b>–<b>H</b>): The same metrics as above, but for fludarabine.</p>
Full article ">Figure 9 Cont.
<p>Molecular dynamics simulation results of FDA drugs flufenamic acid and fludarabine. (<b>A</b>–<b>D</b>) Flufenamic acid: root mean square deviation curve (RSMD), root mean square wave curve (RMSF), small molecule root mean square wave curve (RMSF), and statistics of interaction proportion of different residues. (<b>E</b>–<b>H</b>): The same metrics as above, but for fludarabine.</p>
Full article ">
Back to TopTop