[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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (175)

Search Parameters:
Keywords = HCoV-229E

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 740 KiB  
Article
Synthesis and Antiviral Evaluation of 5-(4-Aryl-1,3-butadiyn-1-yl)-uridines and Their Phosphoramidate Pronucleotides
by Evan Saillard, Otmane Bourzikat, Koffi Assa, Vincent Roy and Luigi A. Agrofoglio
Molecules 2025, 30(1), 96; https://doi.org/10.3390/molecules30010096 - 29 Dec 2024
Viewed by 544
Abstract
The emergence of RNA viruses driven by global population growth and international trade highlights the urgent need for effective antiviral agents that can inhibit viral replication. Nucleoside analogs, which mimic natural nucleotides, have shown promise in targeting RNA-dependent RNA polymerases (RdRps). Starting from [...] Read more.
The emergence of RNA viruses driven by global population growth and international trade highlights the urgent need for effective antiviral agents that can inhibit viral replication. Nucleoside analogs, which mimic natural nucleotides, have shown promise in targeting RNA-dependent RNA polymerases (RdRps). Starting from protected 5-iodouridine, we report the synthesis of hitherto unknown C5-substituted-(1,3-diyne)-uridines nucleosides and their phosphoramidate prodrugs. The modifications at C5 include 4-(trifluoromethyl)benzene (a), 4-pentyl-benzene (b), 3,5-dimethoxy-benzene (c), 4-(trifluoromethoxy)benzene (d), 3-aniline (e), 4-pyridine (f), 3-thiophene (g), C6H13 (h), 2-pyrimidine (i), cyclopropyl (j), and phenyl (k) groups. These compounds were synthesized using Sonogashira palladium-catalyzed reactions and nickel–copper-catalyzed C-H activation between various alkynes, yielding between 25% and 67%. The antiviral activities of obtained compounds were measured through HTS against RNA viruses including influenza H1N1 and H3N2, human respiratory syncytial virus (RSV), SARS-CoV-2, Zika, hepatitis C virus (HCV), Hepatitis E virus (HEV), as well as against coronavirus (HCoV-229E). Unfortunately, none of them showed promising antiviral activity, with less than 85% inhibition observed in the cell viability screening of infected cells. Full article
(This article belongs to the Special Issue Bioorganic Chemistry in Europe)
Show Figures

Figure 1

Figure 1
<p>Some modified pyrimidine nucleoside antivirals and target compounds (<b>16a</b>–<b>k</b> and <b>19a</b>–<b>k</b>) [<a href="#B21-molecules-30-00096" class="html-bibr">21</a>].</p>
Full article ">Scheme 1
<p>Reagents and conditions: (i) dry acetone, H<sub>2</sub>SO<sub>4</sub>, 4 Å molecular sieves, r.t., 48 h, 99%; (ii) Ac<sub>2</sub>O, pyridine, r.t., 15 h, 90%; (iii) ethynyltrimethylsilane, CuI/PdCl<sub>2</sub>(PPh<sub>3</sub>)<sub>2</sub>, Et<sub>3</sub>N, dry DMF, MW, 60 °C, 25 min, 90%; (iv) TBAF, THF, r.t., 3 h, quant; (v) alkyne, NiCl<sub>2</sub>·6H<sub>2</sub>O, CuI, TMEDA, O<sub>2</sub>, THF, r.t., 24 h, 31–67%; (vi) HCl 37%, MeOH, r.t., 24 h, 51–88%; (vii) NH<sub>3</sub>/MeOH (7N), MeOH, r.t., 24 h, 71–100%; (viii) (a) <b>18</b>, t-BuMgCl (1.7 M in THF), THF, 0 °C to r.t., 20 h; (b) HCl 37%, THF, r.t., 6 h, 28–84%.</p>
Full article ">
17 pages, 1408 KiB  
Article
Ab Initio Study of Electron Capture in Collisions of Protons with CO2 Molecules
by Luis Méndez and Ismanuel Rabadán
Molecules 2025, 30(1), 74; https://doi.org/10.3390/molecules30010074 - 28 Dec 2024
Viewed by 244
Abstract
Ab initio calculations of cross sections for electron capture by protons in collisions with CO2 are carried out at energies between 100 eV/u and 50 keV/u, employing a semiclassical method within the Franck–Condon framework. The scattering wave function is expanded in a [...] Read more.
Ab initio calculations of cross sections for electron capture by protons in collisions with CO2 are carried out at energies between 100 eV/u and 50 keV/u, employing a semiclassical method within the Franck–Condon framework. The scattering wave function is expanded in a set of ab initio electronic wave functions of the HCO2+ supermolecule. The calculation is performed on several trajectory orientations to obtain orientation-averaged total cross sections. A two-state model with an exponential interaction between the entrance and the lowest charge transfer channel is proposed to describe the main aspects of the charge transfer process and to estimate the precision of the molecular expansion. The symmetry of the HOMO πg of CO2 is relevant to choose the signs of the molecular functions and to set up the orientation average of the cross sections. Very good agreement is found with the experimental charge transfer cross sections. Full article
Show Figures

Figure 1

Figure 1
<p>Potential energy curves (top panels) and dynamical couplings (bottom panels) along trajectories (<b>a</b>) t<sub>‖</sub>, (<b>b</b>) t<sub>⊥0</sub>, and (<b>c</b>) t<sub>⊥90</sub>, with <math display="inline"><semantics> <mrow> <mi>b</mi> <mo>=</mo> <mn>4.0</mn> <mo> </mo> <mrow> <mi>bohr</mi> </mrow> </mrow> </semantics></math>. The dynamical couplings shown in the bottom panels are coupling <math display="inline"><semantics> <msub> <mi>M</mi> <mn>12</mn> </msub> </semantics></math>, black lines; <math display="inline"><semantics> <msub> <mi>M</mi> <mn>13</mn> </msub> </semantics></math>, red lines; and <math display="inline"><semantics> <msub> <mi>M</mi> <mn>23</mn> </msub> </semantics></math>, green lines. The adiabatic states are numbered in increasing energy order, as shown in the top panels. The two green lines in the energies of panel (<b>c</b>) correspond to the two molecular states dissociating in H(1s) + CO<sub>2</sub><sup>+</sup> (<math display="inline"><semantics> <mrow> <msup> <mi>A</mi> <mn>2</mn> </msup> <msub> <mi mathvariant="normal">Π</mi> <mi>u</mi> </msub> </mrow> </semantics></math>).</p>
Full article ">Figure 2
<p>Color maps of the dynamical couplings, <math display="inline"><semantics> <mfenced separators="" open="&#x2329;" close="&#x232A;"> <msub> <mi>χ</mi> <mn>1</mn> </msub> <mfenced separators="" open="|" close="|"> <mi mathvariant="normal">d</mi> <mo>/</mo> <mi mathvariant="normal">d</mi> <mi>Z</mi> </mfenced> <msub> <mi>χ</mi> <mn>2</mn> </msub> </mfenced> </semantics></math>, between the first two electronic states of (H + CO<sub>2</sub>)<sup>+</sup> system for trajectories parallel (top) and perpendicular (bottom) to the CO<sub>2</sub> molecular axis. Comparison between the Demkov model (Equation (<a href="#FD8-molecules-30-00074" class="html-disp-formula">8</a>)) with <math display="inline"><semantics> <mrow> <msub> <mi>E</mi> <mn>0</mn> </msub> <mo>=</mo> <mn>0.006</mn> </mrow> </semantics></math> hartree and the ab initio data.</p>
Full article ">Figure 3
<p>Energy difference and coupling used in the 2-state model of <a href="#sec2dot2-molecules-30-00074" class="html-sec">Section 2.2</a> with <math display="inline"><semantics> <mrow> <msub> <mi>E</mi> <mn>0</mn> </msub> <mo>=</mo> <mo>+</mo> <mn>0.004</mn> </mrow> </semantics></math> hartree (black lines) and <math display="inline"><semantics> <mrow> <msub> <mi>E</mi> <mn>0</mn> </msub> <mo>=</mo> <mo>−</mo> <mn>0.006</mn> </mrow> </semantics></math> hartree (red lines) to estimate the error produced by the swapping of asymptotic energies, while keeping the same interaction, on the CT cross-section.</p>
Full article ">Figure 4
<p>Charge transfer cross sections obtained with the two-state models and the ab initio results for trajectories parallel to the CO<sub>2</sub> axis (t<sub>‖</sub>). The full lines are the results from the models with <math display="inline"><semantics> <mrow> <msub> <mi>E</mi> <mn>0</mn> </msub> <mo>=</mo> <mo>+</mo> <mn>0.004</mn> </mrow> </semantics></math> hartree (black line) and <math display="inline"><semantics> <mrow> <msub> <mi>E</mi> <mn>0</mn> </msub> <mo>=</mo> <mo>−</mo> <mn>0.006</mn> </mrow> </semantics></math> hartree of (red line). The dashed lines are the ab initio results for production of CO<sub>2</sub><sup>+</sup>(<math display="inline"><semantics> <mrow> <msup> <mi>X</mi> <mn>2</mn> </msup> <msub> <mi mathvariant="normal">Π</mi> <mi>g</mi> </msub> </mrow> </semantics></math>) (long-dashed green) and for production of CO<sub>2</sub><sup>+</sup>(<math display="inline"><semantics> <mrow> <msup> <mi>A</mi> <mn>2</mn> </msup> <msub> <mi mathvariant="normal">Π</mi> <mi>u</mi> </msub> </mrow> </semantics></math>) (short-dashed orange). Symbols are experimental data: • [<a href="#B5-molecules-30-00074" class="html-bibr">5</a>]; ▴ [<a href="#B6-molecules-30-00074" class="html-bibr">6</a>]; ⧫ [<a href="#B7-molecules-30-00074" class="html-bibr">7</a>]; ▾ [<a href="#B8-molecules-30-00074" class="html-bibr">8</a>]; ◂ [<a href="#B10-molecules-30-00074" class="html-bibr">10</a>].</p>
Full article ">Figure 5
<p>Color maps of the charge transfer probability in proton collisions with CO<sub>2</sub> as a function of the impact parameter <span class="html-italic">b</span> and the impact energy <span class="html-italic">E</span> with the 2-state model of <a href="#sec2dot2-molecules-30-00074" class="html-sec">Section 2.2</a> with <math display="inline"><semantics> <mrow> <msub> <mi>E</mi> <mn>0</mn> </msub> <mo>=</mo> <mo>−</mo> <mn>0.006</mn> </mrow> </semantics></math> hartree (top) and <math display="inline"><semantics> <mrow> <msub> <mi>E</mi> <mn>0</mn> </msub> <mo>=</mo> <mo>+</mo> <mn>0.004</mn> </mrow> </semantics></math> hartree (bottom).</p>
Full article ">Figure 6
<p>Color maps showing the probability of electron capture as function of the position of the impact parameter vector, <math display="inline"><semantics> <mi mathvariant="bold-italic">b</mi> </semantics></math> in the Cartesian plane, with <span class="html-italic"><b>v</b></span> perpendicular to this plane. Each panel corresponds to a different impact energy, as labeled. The position of the molecule in the coordinate system is also shown.</p>
Full article ">Figure 7
<p>Orientation-averaged CT cross-sections in proton collisions with CO<sub>2</sub> as functions of the impact energy. The lines are the present calculations of the total OAXS, obtained by averaging over different number of trajectory orientations: 3t, (<a href="#FD20-molecules-30-00074" class="html-disp-formula">20</a>); 4t, (<a href="#FD21-molecules-30-00074" class="html-disp-formula">21</a>); 5t, (<a href="#FD22-molecules-30-00074" class="html-disp-formula">22</a>); and 6t, (<a href="#FD23-molecules-30-00074" class="html-disp-formula">23</a>), as indicated in the figure. Experimental results: • [<a href="#B5-molecules-30-00074" class="html-bibr">5</a>]; ▴ [<a href="#B6-molecules-30-00074" class="html-bibr">6</a>]; ⧫ [<a href="#B7-molecules-30-00074" class="html-bibr">7</a>]; ▾ [<a href="#B8-molecules-30-00074" class="html-bibr">8</a>]; ◂ [<a href="#B10-molecules-30-00074" class="html-bibr">10</a>].</p>
Full article ">Figure 8
<p>Orientation-averaged charge-transfer cross section in proton collisions with CO<sub>2</sub> as a function of the impact energy. Lines are the present calculations: solid line, total charge transfer; red-dashed line, charge transfer to CO<sub>2</sub><sup>+</sup>(X); and green dashed-dotted line, charge transfer to CO<sub>2</sub><sup>+</sup>(A). Symbols are experimental data: • [<a href="#B5-molecules-30-00074" class="html-bibr">5</a>]; ▴ [<a href="#B6-molecules-30-00074" class="html-bibr">6</a>]; ⧫ [<a href="#B7-molecules-30-00074" class="html-bibr">7</a>]; ▾ [<a href="#B8-molecules-30-00074" class="html-bibr">8</a>]; ◂ [<a href="#B10-molecules-30-00074" class="html-bibr">10</a>].</p>
Full article ">Figure 9
<p>Scheme with the two families of projectile trajectories employed to study H<sup>+</sup> + CO<sub>2</sub> collisions: on the left, the t<sub>⊥</sub> family with <math display="inline"><semantics> <mi mathvariant="bold-italic">v</mi> </semantics></math> perpendicular to the molecular axis, <math display="inline"><semantics> <mi mathvariant="bold-italic">ρ</mi> </semantics></math>; on the right, the t<sub>‖</sub> family, with <math display="inline"><semantics> <mi mathvariant="bold-italic">v</mi> </semantics></math> parallel to <math display="inline"><semantics> <mi mathvariant="bold-italic">ρ</mi> </semantics></math>. The t<sub>⊥</sub> family includes subfamilies with different values of the angle <math display="inline"><semantics> <mi>α</mi> </semantics></math> between the impact parameter vector, <span class="html-italic"><b>b</b></span>, and <math display="inline"><semantics> <mi mathvariant="bold-italic">ρ</mi> </semantics></math> being <math display="inline"><semantics> <mrow> <mi>α</mi> <mo>=</mo> <mn>0</mn> </mrow> </semantics></math>, <math display="inline"><semantics> <mn>30</mn> <mo>°</mo> </semantics></math>, <math display="inline"><semantics> <mn>45</mn> <mo>°</mo> </semantics></math>, <math display="inline"><semantics> <mn>60</mn> <mo>°</mo> </semantics></math>, and <math display="inline"><semantics> <mn>90</mn> <mo>°</mo> </semantics></math>.</p>
Full article ">Figure A1
<p>Potential energy curves (middle diagram) of the first two electronic states of (H-CO<sub>2</sub>)<sup>+</sup> along a projectile trajectory perpendicular to the CO<sub>2</sub> internuclear axis (top left) with impact parameter <math display="inline"><semantics> <mrow> <mi>b</mi> <mo>=</mo> <mn>8</mn> </mrow> </semantics></math> bohr, and the non-adiabatic coupling between the two states (bottom) with the molecular orbitals depicted at five stages of the collisions, drawn in color squares and marked next to the energies and coupling lines at approximately their corresponding value of <span class="html-italic">Z</span>. At the first four stages, the electronic density for the two electronic states is shown in the top-right diagram. The two molecular orbitals depicted resemble a <math display="inline"><semantics> <msub> <mi>π</mi> <mi>g</mi> </msub> </semantics></math> on the CO<sub>2</sub> molecule and the <math display="inline"><semantics> <mrow> <mn>1</mn> <mi>s</mi> </mrow> </semantics></math> on the projectile.</p>
Full article ">
24 pages, 3828 KiB  
Article
Development of Primer Panels for Whole-Genome Amplification and Sequencing of Human Seasonal Coronaviruses: hCoV-OC43, hCoV-HKU1, hCoV-229E, and hCoV-NL63
by Tamila Musaeva, Artem Fadeev, Maria Pisareva, Veronika Eder, Andrey Ksenafontov, Margarita Korzhanova, Valery Tsvetkov, Alexander Perederiy, Irina Kiseleva, Daria Danilenko, Dmitry Lioznov and Andrey Komissarov
Viruses 2025, 17(1), 13; https://doi.org/10.3390/v17010013 - 25 Dec 2024
Viewed by 410
Abstract
Human seasonal coronaviruses (hCoVs) are a group of viruses that affect the upper respiratory tract. While seasonal patterns and the annual variability of predominant hCoV species are well-documented, their genetic and species diversity in St. Petersburg and across Russia remains largely unexplored. In [...] Read more.
Human seasonal coronaviruses (hCoVs) are a group of viruses that affect the upper respiratory tract. While seasonal patterns and the annual variability of predominant hCoV species are well-documented, their genetic and species diversity in St. Petersburg and across Russia remains largely unexplored. In this study, we developed a two-pool, long-amplicon (900–1100 bp) PCR primer panel for the whole-genome sequencing of four seasonal hCoV species. The panel was validated using nasopharyngeal swab samples collected within the Global Influenza Hospital Surveillance Network (GIHSN) project. Over a period of six epidemiological seasons from 2017 to 2023, we retrospectively analyzed 14,704 nasopharyngeal swabs collected from patients hospitalized in St. Petersburg clinics. Of these samples, 5010 (34.07%) tested positive for respiratory viruses, with 424 (2.88% of all samples) identified as seasonal human coronaviruses. The assessment of species diversity showed that predominant hCoV species alternate between seasons. Whole-genome sequences for 85 seasonal human coronaviruses (hCoVs) with >70% genome coverage were obtained, including 23 hCoV-OC43, 6 hCoV-HKU1, 39 hCoV-229E, and 17 hCoV-NL63. These represent the first near-complete genomes of seasonal hCoVs from the Russian Federation, addressing a significant gap in the genomic epidemiology of these viruses. A detailed phylogenetic analysis of the sequenced genomes was conducted, highlighting the emergence of hCoV-229E subclades 7b.1 and 7b.2, which carry numerous substitutions in the Spike protein. Additionally, we sequenced a historical hCoV-229E isolate collected in the USSR in 1979, the oldest sequenced 229E virus from Eurasia, and demonstrated that it belongs to Genotype 2. The newly developed PCR-based sequencing protocol for seasonal hCoVs is straightforward and well-suited for genomic surveillance, providing a valuable tool to enhance our understanding of the genetic diversity of human seasonal coronaviruses. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Percentage of specimens testing positive for different respiratory viruses using RT-PCR (excluding mixed infections) over six consecutive epidemic seasons. (<b>b</b>) Assessment of the species diversity of seasonal coronaviruses in St. Petersburg in 2017–2023 (normalized Shannon index; from 0 to 1, where 1 is the maximum possible diversity); for details see <a href="#app1-viruses-17-00013" class="html-app">Supplementary Table S1a–d</a>.</p>
Full article ">Figure 2
<p>Average coverage plots for different hCoV species based on sequencing results. The number of specimens used for the calculations for each hCoV type is indicated in the figure.</p>
Full article ">Figure 3
<p>Scatterplots of percentage min 20× vs. Ct values for different hCoVs based on the sequencing results. The color gradient represents the log(Fold80) value. hCoV-229E: <span class="html-italic">n</span>=30, hCoV-HKU1: <span class="html-italic">n</span> = 8, hCoV-NL63: <span class="html-italic">n</span> = 14, hCoV-OC43: <span class="html-italic">n</span> = 22.</p>
Full article ">Figure 4
<p>Scatterplots of the percentage of mapped reads vs. the Ct value for different hCoVs, based on the sequencing results. The color gradient represents the log(Fold80) value. hCoV-229E: <span class="html-italic">n</span> = 30, hCoV-HKU1: <span class="html-italic">n</span> = 8, hCoV-NL63: <span class="html-italic">n</span> = 14, hCoV-OC43: <span class="html-italic">n</span> = 22.</p>
Full article ">Figure 5
<p>Phylogenetic trees of seasonal human coronaviruses circulating in St. Petersburg from 2017 to 2023. (<b>a</b>) hCoV-HKU1, (<b>b</b>) hCoV-OC43, (<b>c</b>) hCoV-229E, and (<b>d</b>) hCoV-NL63. Green diamonds indicate strains sequenced in this study.</p>
Full article ">Figure 5 Cont.
<p>Phylogenetic trees of seasonal human coronaviruses circulating in St. Petersburg from 2017 to 2023. (<b>a</b>) hCoV-HKU1, (<b>b</b>) hCoV-OC43, (<b>c</b>) hCoV-229E, and (<b>d</b>) hCoV-NL63. Green diamonds indicate strains sequenced in this study.</p>
Full article ">Figure 5 Cont.
<p>Phylogenetic trees of seasonal human coronaviruses circulating in St. Petersburg from 2017 to 2023. (<b>a</b>) hCoV-HKU1, (<b>b</b>) hCoV-OC43, (<b>c</b>) hCoV-229E, and (<b>d</b>) hCoV-NL63. Green diamonds indicate strains sequenced in this study.</p>
Full article ">Figure 5 Cont.
<p>Phylogenetic trees of seasonal human coronaviruses circulating in St. Petersburg from 2017 to 2023. (<b>a</b>) hCoV-HKU1, (<b>b</b>) hCoV-OC43, (<b>c</b>) hCoV-229E, and (<b>d</b>) hCoV-NL63. Green diamonds indicate strains sequenced in this study.</p>
Full article ">
18 pages, 3296 KiB  
Article
miR-24-3p Is Antiviral Against SARS-CoV-2 by Downregulating Critical Host Entry Factors
by Parrish Evers, Spencer M. Uguccioni, Nadine Ahmed, Magen E. Francis, Alyson A. Kelvin and John P. Pezacki
Viruses 2024, 16(12), 1844; https://doi.org/10.3390/v16121844 - 28 Nov 2024
Viewed by 897
Abstract
Despite all the progress in treating SARS-CoV-2, escape mutants to current therapies remain a constant concern. Promising alternative treatments for current and future coronaviruses are those that limit escape mutants by inhibiting multiple pathogenic targets, analogous to the current strategies for treating HCV [...] Read more.
Despite all the progress in treating SARS-CoV-2, escape mutants to current therapies remain a constant concern. Promising alternative treatments for current and future coronaviruses are those that limit escape mutants by inhibiting multiple pathogenic targets, analogous to the current strategies for treating HCV and HIV. With increasing popularity and ease of manufacturing of RNA technologies for vaccines and drugs, therapeutic microRNAs represent a promising option. In the present work, miR-24-3p was identified to inhibit SARS-CoV-2 entry, replication, and production; furthermore, this inhibition was retained against common mutations improving SARS-CoV-2 fitness. To determine the mechanism of action, bioinformatic tools were employed, identifying numerous potential effectors promoting infection targeted by miR-24-3p. Of these targets, several key host proteins for priming and facilitating SARS-CoV-2 entry were identified: furin, NRP1, NRP2, and SREBP2. With further experimental analysis, we show that miR-24-3p directly downregulates these viral entry factors to impede infection when producing virions and when infecting the target cell. Furthermore, we compare the findings with coronavirus, HCoV-229E, which relies on different factors strengthening the miR-24-3p mechanism. Taken together, the following work suggests that miR-24-3p could be an avenue to treat current coronaviruses and those likely to emerge. Full article
(This article belongs to the Special Issue Viruses, MicroRNAs and Host Interactions)
Show Figures

Figure 1

Figure 1
<p>miR-24-3p inhibits SARS-CoV-2 replication and virion production. (<b>A</b>) Calu3 Cells were reverse transfected with miR-24-3p or con-miR for 24 h prior to infection with SARS-CoV-2, MOI of 0.01, for 24 h, 48 h, or 72 h. RT qPCR (technical triplicate) was then performed to quantify relative vRNA in samples. (<b>B</b>) Production of SARS-CoV-2 was assessed by collecting an aliquot of the supernatant at each time point. (<b>C</b>) Subgenomic cellular RNA of SARS-CoV-2 was assessed by lysing cells at each time point. <span class="html-italic">n</span> = 3. Error bars represent SEM. <span class="html-italic">p</span> &lt; 0.01 **, 0.001 ***, 0.0001 ****.</p>
Full article ">Figure 2
<p>Pathway Gene Ontology (GO) of highly expressed miR-24-3p targets. Panther GO was performed on miRDB predicted targets that are highly expressed (RPKM ≥ 20) in the cell lines used in the present study: Huh7, Calu-3, and A549. Most genes belong to unclassified categories; however, these were removed for clarity. A total of 75–102 targets were highly expressed in each cell line.</p>
Full article ">Figure 3
<p>miR-24-3p downregulates Furin and SREBP2 mRNA. Calu-3 cells were reverse transfected with 100 nM miR-24-3p or con-miR for 72 h before lysis for RT-qPCR. Treatment with miR-24-3p post-transcriptionally represses (<b>A</b>) furin and (<b>B</b>) SREBP2 at the mRNA level. <span class="html-italic">n</span> = 3. Error bars represent SEM. <span class="html-italic">p</span> &lt; 0.01 **, 0.001 ***.</p>
Full article ">Figure 4
<p>miR-24-3p decreases the entry of SARS-CoV-2 in an S pseudovirus model. (<b>A</b>) Scheme depicting the generation of SARS-CoV-2 S pseudotyped virus. (<b>B</b>) Validation of pseudovirus components via western blot of HEK293T producing cell lysates and extracellular supernatant. (<b>C</b>) Pseudovirus entry assay quantified by luciferase microplate reader (technical triplicate). Assay was performed after 24 h reverse transfection of miR-24-3p followed by 48 h pseudovirus infection and lysis using a passive lysis buffer in either (<b>C</b>) Huh7 or (<b>D</b>) Calu-3 cells. Both con-miR and miR-24-3p values were normalized to the average con-mR value. <span class="html-italic">n</span> = 3. Error bars represent SEM. <span class="html-italic">p</span> &lt; 0.001 ***.</p>
Full article ">Figure 5
<p>miR-24-3p maintains effectiveness against common SARS-CoV-2 S mutations. Pseudovirus entry assay quantified by luciferase microplate reader in technical triplicate. The assay was performed after 24 h reverse transfection of miR-24-3p or con-miR followed by 48 h pseudovirus S infection with D614G or N501Y S mutants in either (<b>A</b>) Huh7 or (<b>B</b>) ACE2 stably expressing A549 cell line. Lysis was performed using a passive lysis buffer. Both con-miR and miR-24-3p values were normalized to the average con-miR value. <span class="html-italic">n</span> = 3. Error bars represent SEM. <span class="html-italic">p</span> &lt; 0.05 *, 0.001 ***.</p>
Full article ">Figure 6
<p>miR-24-3p downregulates Furin, SREBP2, and NRP2 impairing production and entry of SARS-CoV-2. (<b>A</b>) Scheme depicting experimental workflow for assessing target abundance and pseudovirus S production during miRNA pre-treatment. Briefly, HEK293T cells were pre-treated with miR-24-3p or con-miR 24 h before transfection with the plasmids to produce pseudovirions. After 48 h, the pseudovirus was collected and the pseudovirus produced during miR-24-3p treatment or con-miR treatment were then used to infect healthy untreated Huh7 cells. A luciferase assay was then performed on these Huh7 cells to quantify the amount of pseudovirus produced. (<b>B</b>) Western blot analysis of lysates from control or miR-24-3p-treated HEK293Ts producing S-pseudovirus. Several essential proviral targets and the viral S protein were probed for. (<b>C</b>) Western blot analysis of supernatant from control or miR-24-3p-treated HEK293Ts cells producing S-pseudovirus. (<b>D</b>) S-pseudovirus entry assay performed on non-treated Huh7 cells following production in HEK293T cells reverse transfected with miR-24-3p or con-miR. (<b>E</b>) S-pseudovirus entry assay from (<b>C</b>) normalized to total S production from (<b>D</b>). <span class="html-italic">n</span> = 2. Error bars represent SEM. For the luciferase data, both con-miR and miR-24-3p values were normalized to the average con-mR value. <span class="html-italic">p</span> &lt; 0.01 **, 0.0001 ****.</p>
Full article ">Figure 7
<p>Diagram illustrating the targets and effects of miR-24-3p during SARS-CoV-2 infection.</p>
Full article ">
14 pages, 428 KiB  
Article
Exploring 4,7-Disubstituted Pyrimido[4,5-d]pyrimidines as Antiviral and Anticancer Agents
by Eleftheria A. Georgiou, Konstantinos Paraskevas, Christina Koutra, Leentje Persoons, Dominique Schols, Steven De Jonghe and Ioannis K. Kostakis
Molecules 2024, 29(23), 5549; https://doi.org/10.3390/molecules29235549 - 25 Nov 2024
Viewed by 850
Abstract
Thirteen new 4,7-disubstituted pyrimido[4,5-d]pyrimidines were synthesized via a straightforward methodology starting from thiourea. The anti-proliferative activity of these compounds was evaluated across a diverse panel of eight cancer cell lines, with derivatives 7d and 7h showing efficacy against several hematological cancer [...] Read more.
Thirteen new 4,7-disubstituted pyrimido[4,5-d]pyrimidines were synthesized via a straightforward methodology starting from thiourea. The anti-proliferative activity of these compounds was evaluated across a diverse panel of eight cancer cell lines, with derivatives 7d and 7h showing efficacy against several hematological cancer types. Furthermore, all compounds were assessed for their antiviral potency against a panel of viruses. Compounds featuring a cyclopropylamino group and an aminoindane moiety exhibited remarkable efficacy against human coronavirus 229E (HCoV-229E). These findings highlight the pyrimidino[4,5-d]pyrimidine scaffold as an interesting framework for the design of novel antiviral agents against HCoVs, with compounds 7a, 7b, and 7f emerging as strong candidates for further investigation. Full article
(This article belongs to the Special Issue Design, Synthesis and Biological Evaluation of Heterocyclic Compounds)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Structures of known kinase inhibitors.</p>
Full article ">Scheme 1
<p>Reagents and conditions: (a) dimethyl sulfate, H<sub>2</sub>O, 80 °C; (b) 2-(ethoxymethylene)malononitrile, triethylamine, ethanol abs., rt; (c) <span class="html-italic">m</span>CPBA, CH<sub>2</sub>Cl<sub>2</sub>, 25 °C; (d) appropriate amine, THF, 25 °C; (e) DMF-DMA, toluene, rt; (f) suitable aniline, CH<sub>3</sub>CO<sub>2</sub>H, reflux.</p>
Full article ">
11 pages, 947 KiB  
Article
The Effects of Different Respiratory Viruses on the Oxidative Stress Marker Levels in an In Vitro Model: A Pilot Study
by Barbara Bażanów, Katarzyna Michalczyk, Alina Kafel, Elżbieta Chełmecka, Bronisława Skrzep-Poloczek, Aleksandra Chwirot, Kamil Nikiel, Aleksander Olejnik, Alicja Suchocka, Michał Kukla, Bartosz Bogielski, Jerzy Jochem and Dominika Stygar
Int. J. Mol. Sci. 2024, 25(22), 12088; https://doi.org/10.3390/ijms252212088 - 11 Nov 2024
Viewed by 853
Abstract
Respiratory viruses are among the most common causes of human infections. Examining pathological processes linked to respiratory viral infections is essential for diagnosis, treatment strategies, and developing novel therapeutics. Alterations in oxidative stress levels and homeostasis are significant processes associated with respiratory viral [...] Read more.
Respiratory viruses are among the most common causes of human infections. Examining pathological processes linked to respiratory viral infections is essential for diagnosis, treatment strategies, and developing novel therapeutics. Alterations in oxidative stress levels and homeostasis are significant processes associated with respiratory viral infections. The study aimed to compare selected oxidative stress markers: total oxidative status (TOS), total antioxidant capacity (TAC), and the oxidative stress index (OSI) levels and glutathione peroxidase (GPx) and glutathione reductase (GR) activities in normal (MRC5 cell line) and tumor (A549 cell line) lung cells infected with human coronaviruses (HCoV) OC43 and 229E, human adenovirus type 5 (HAdV5), or human rhinovirus A (HRV A). We observed that a respiratory viral infection more significantly affected non-enzymatic oxidative stress markers in a lung adenocarcinoma model (A549 cells), while human lung fibroblasts (MRC-5 cell line) presented changes in enzymatic and non-enzymatic oxidative stress markers. We suggest that further detailed research is required to analyze this phenomenon. Full article
Show Figures

Figure 1

Figure 1
<p>Oxidative stress marker levels: (<b>a</b>) the total oxidative status (TOS), (<b>b</b>) total antioxidant capacity (TAC), (<b>c</b>) oxidative stress index (OSI), (<b>d</b>) glutathione peroxidase (GPx) activity, and (<b>e</b>) glutathione reductase (GR) activity in the cells infected with different respiratory viruses and in control cells of the human lung carcinoma cell line (A549). The results are presented as Me (Q<sub>1</sub>; Q<sub>3</sub>)—median (lower-upper quartile) or M ± SD—mean ± standard deviation. For better visualization in part (<b>b</b>,<b>e</b>), the logarithmic scale was used. Abbreviations: HAdV5—human adenovirus 5; HcoV-229E—human coronavirus 229E; HcoV-OC43—human coronavirus OC43; HRV A—human rhinovirus A; IU—international activity unit.</p>
Full article ">Figure 2
<p>Oxidative stress marker levels: (<b>a</b>) the total oxidative status (TOS), (<b>b</b>) total antioxidant capacity (TAC), (<b>c</b>) oxidative stress index (OSI), (<b>d</b>) glutathione peroxidase (GPx) activity, and (<b>e</b>) glutathione reductase (GR) activity in the cells infected with different respiratory viruses and in control cells of the human lung carcinoma cell line (MCR-5). The results are presented as Me (Q<sub>1</sub>; Q<sub>3</sub>)—median (lower-upper quartile) or M ± SD—mean ± standard deviation. Abbreviations: HAdV5—human adenovirus 5; HcoV-229E—human coronavirus 229E; HcoV-OC43—human coronavirus OC43; HRV A—human rhinovirus A; IU—international activity unit.</p>
Full article ">
17 pages, 3786 KiB  
Article
Dual Functionality of Papaya Leaf Extracts: Anti-Coronavirus Activity and Anti-Inflammation Mechanism
by Yujia Cao, Kah-Man Lai, Kuo-Chang Fu, Chien-Liang Kuo, Yee-Joo Tan, Liangli (Lucy) Yu and Dejian Huang
Foods 2024, 13(20), 3274; https://doi.org/10.3390/foods13203274 - 16 Oct 2024
Viewed by 1743
Abstract
Papaya leaves have been used as food and traditional herbs for the treatment of cancer, diabetes, asthma, and virus infections, but the active principle has not been understood. We hypothesized that the anti-inflammatory activity could be the predominant underlying principle. To test this, [...] Read more.
Papaya leaves have been used as food and traditional herbs for the treatment of cancer, diabetes, asthma, and virus infections, but the active principle has not been understood. We hypothesized that the anti-inflammatory activity could be the predominant underlying principle. To test this, we extracted papaya leaf juice with different organic solvents and found that the ethyl acetate (EA) fraction showed the most outstanding anti-inflammatory activity by suppressing the production of nitric oxide (NO, IC50 = 24.94 ± 2.4 μg/mL) and the expression of pro-inflammatory enzymes, such as inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX-2), and cytokines including interleukins (IL-1β and IL-6), and a tumor necrosis factor (TNF-α) in lipopolysaccharide (LPS)-induced RAW 264.7 cells. Transcriptomic analysis and Western blot results revealed its anti-inflammatory mechanisms were through the MAPK signaling pathway by inhibiting the phosphorylation of ERK1/2, JNKs, and p38 and the prevention of the cell surface expression of TLR4. Furthermore, we discovered that the EA fraction could inhibit the replication of alpha-coronavirus (HCoV-229E) and beta-coronavirus (HCoV-OC43 and SARS-CoV-2) and might be able to prevent cytokine storms caused by the coronavirus infection. From HPLC-QTOF-MS data, we found that the predominant phytochemicals that existed in the EA fraction were quercetin and kaempferol glycosides and carpaine. Counter-intuitively, further fractionation resulted in a loss of activity, suggesting that the synergistic effect of different components in the EA fraction contribute to the overall potent activity. Taken together, our results provide preliminary evidence for papaya leaf as a potential anti-inflammatory and anti-coronavirus agent, warranting further study for its use for human health promotion. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The inhibitory effects of papaya leaf juice extract (PLJE) on NO production in LPS-induced RAW 264.7 cells (<b>A</b>). Cytotoxicity of PLJE (<b>B</b>). Data points and bar represent arithmetic means ± SD. ns, not significant. * <span class="html-italic">p</span> &lt; 0.05, **** <span class="html-italic">p</span> &lt; 0.0001 compared to DMSO or control group.</p>
Full article ">Figure 2
<p>Suppressive effects of fractions extracted from PLJE on NO production in LPS-induced RAW 264.7 cells (<b>A</b>). Cell viability of RAW 264.7 cells treated with five fractions (<b>B</b>). IC<sub>50</sub> of EA fraction on NO production in LPS-induced RAW 264.7 cells (<b>C</b>). Data points and bar represent arithmetic means ± SD. ns, not significant. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 compared to DMSO group, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>####</sup> <span class="html-italic">p</span> &lt; 0.0001 compared within group.</p>
Full article ">Figure 3
<p>The inhibitory effects of EA fraction on inflammation-related protein expression in LPS-stimulated RAW 264.7 models (<b>A</b>). The expression levels of iNOS (<b>B</b>), COX-2 (<b>C</b>), and TLR4 (<b>D</b>) were determined by Western blot. The mRNA expression levels of IL-1β (<b>E</b>), IL-6 (<b>F</b>), and TNF-α (<b>G</b>) were tested by qRT-PCR. Data points and bar represent arithmetic means ± SD. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 compared to DMSO group.</p>
Full article ">Figure 4
<p>GO (<b>A</b>) and KEGG (<b>B</b>) enrichment scatter plots of EA (25 μg/mL) vs. DMSO group. GeneRatio is the ratio of the number of DEGs annotated to the GO or KEGG term to the total number of DEGs. The size of the dot represents the number of genes annotated to the terms.</p>
Full article ">Figure 5
<p>Effects of EA fraction on MAPK pathway in LPS-induced RAW 264.7 cell (<b>A</b>). Suppressive effects of EA fraction on the LPS-induced phosphorylation ratio of ERK1/2 (<b>B</b>), JNK (<b>C</b>), and p38 (<b>D</b>). Data points and bar represent arithmetic means ± SD. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 compared to DMSO group.</p>
Full article ">Figure 6
<p>Schematic diagram of potential contribution of papaya leaves in LPS-induced signaling pathways.</p>
Full article ">Figure 7
<p>Plaque reduction neutralization tests (PRNT) of EA fraction and nirmatrelvir against infectious HCoVs-OC43 (<b>A</b>), HCoV-229E (<b>B</b>), and SARS-CoV-2 (<b>C</b>). Data points and bar represent arithmetic shown which are the mean ± SD of at least two independent tests performed. ns, not significant. *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 compared between indicated groups.</p>
Full article ">
15 pages, 4617 KiB  
Article
Human Coronavirus 229E Uses Clathrin-Mediated Endocytosis as a Route of Entry in Huh-7 Cells
by Sabina Andreu, Inés Ripa, José Antonio López-Guerrero and Raquel Bello-Morales
Biomolecules 2024, 14(10), 1232; https://doi.org/10.3390/biom14101232 - 29 Sep 2024
Viewed by 996
Abstract
Human coronavirus 229E (HCoV-229E) is an endemic coronavirus responsible for approximately one-third of “common cold” cases. To infect target cells, HCoV-229E first binds to its receptor on the cell surface and then can follow different pathways, entering by direct fusion or by taking [...] Read more.
Human coronavirus 229E (HCoV-229E) is an endemic coronavirus responsible for approximately one-third of “common cold” cases. To infect target cells, HCoV-229E first binds to its receptor on the cell surface and then can follow different pathways, entering by direct fusion or by taking advantage of host cell mechanisms such as endocytosis. Based on the role of clathrin, the process can be classified into clathrin-dependent or -independent endocytosis. This study characterizes the role of clathrin-mediated endocytosis (CME) in HCoV-229E infection of the human hepatoma cell line Huh-7. Using specific CME inhibitory drugs, we demonstrated that blocking CME significantly reduces HCoV-229E infection. Additionally, CRISPR/Cas9-mediated knockout of the µ subunit of adaptor protein complex 2 (AP-2) further corroborated the role of CME, as KOs showed over a 50% reduction in viral infection. AP-2 plays an important role in clathrin recruitment and the maturation of clathrin-coated vesicles. Our study also confirmed that in Huh-7 cells, HCoV-229E requires endosomal acidification for successful entry, as viral entry decreased when treated with lysomotropic agents. Furthermore, the colocalization of HCoV-229E with early endosome antigen 1 (EEA-1), only present in early endosomes, suggested that the virus uses an endosomal route for entry. These findings highlight, for the first time, the role of CME in HCoV-229E infection and confirm previous data of the use of the endosomal route at a low pH in the experimental cell model Huh-7. Our results provide new insights into the mechanisms of entry of HCoV-229E and provide a new basis for the development of targeted antiviral therapies. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Viral Infections)
Show Figures

Figure 1

Figure 1
<p>Chemical inhibitors of CME block conjugate transferrin but not conjugate dextran internalization at non-cytotoxic doses. (<b>A</b>) Cell viability of Huh-7 cells treated with chlorpromazine, dynasore or pitstop 2 for 24 h. Viability was measured by a MTT assay and calculated as the percentage of cell viability compared to untreated cells; the columns represent the mean percentage of relative cellular viability ± S.D. (<span class="html-italic">n</span> = 4) after drug exposure. The dotted lines mark the area in which values are considered non cytotoxic. (<b>B</b>) The uptake of transferrin in Huh-7 cells is blocked by CME inhibitors. Cells were treated for 1 h with chlorpromazine, dynasore or pitstop 2. Then, they were maintained for 30 min on ice with Tf CF<sup>®</sup>543 (5 μg/mL). Finally, cells were incubated 5 min at 37 °C before fixation. (<b>C</b>) Quantification of Tf conjugate. ROIs from groups of 20 cells and three areas of each image were measured. Measurement of mean fluorescence intensity from the 555 channel in an ROI was performed. The mean percentage of fluorescence ± S.D. is shown. (<span class="html-italic">n</span> = 4); * <span class="html-italic">p</span> &lt; 0.05. (<b>D</b>) The uptake of dextran in the Huh-7 cells was not disrupted by the CME inhibitors. Cells were treated for 1 h with chlorpromazine, dynasore or pitstop 2. Then, they were maintained for 30 min on ice with CF<sup>®</sup>555 Labeled Dye Dextran 10,000 MW (5 μg/mL). Finally, the cells were incubated for 10 min at 37 °C before fixation. (<b>E</b>) Quantification of dextran conjugate. Same procedure as with Tf quantification. Scale bar = 20 µm. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>HCoV-229E infection is blocked by CME-inhibitory drugs. Huh-7 cells were treated with either 10 µM chlorpromazine, 100 µM dynasore or 50 µM pitstop 2 for 1 h and subsequently infected with HCoV-229E-GFP at an m.o.i. of 0.5. Cells were maintained in the presence of the drugs until they were collected at 20 h p.i. (<b>A</b>) Flow cytometry analysis: this graph shows the percentage of normalized infection at 20 h p.i. ± S.D. Triplicate experiments were performed (<span class="html-italic">n</span> = 4). (<b>B</b>) The fluorescence microscopy images show the GFP+ signal, which corresponds to viral infection. Cellular nuclei are stained with DAPI. Scale bar = 10 µm. (<b>C</b>) Infectious particles of the progeny virus were titrated in Huh-7 cells to determine the 50% tissue culture infective dose (TCID<sub>50</sub>)/mL. The graph shows the mean ± S.D. (<span class="html-italic">n</span> = 3) viral production. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Validation of the knockout of the AP2M1 gene in Huh-7 cells by CRISPR/Cas9. (<b>A</b>) AP2M1 expression was checked by immunoblotting. Western blot analysis of total cell lysates subjected to SDS-PAGE, showing AP2M1 for each subcloned cell. β-actin was chosen as the protein loading control. The black arrow points to the specific AP2M1 band. Values of immunoblot quantification are reported as mean ± S.D. (<span class="html-italic">n</span> = 3); * <span class="html-italic">p</span> &lt; 0.05. (<b>B</b>) Indel values of C5 and C7. Percentage of efficiency of CRISPR-Cas9 cut. (<b>C</b>) Sanger sequence view showing wild-type (control) and edited (C5, C7) sequences in the region around the guide sequence. The horizontal black underlined region represents the guide sequence. The horizontal red underline is the PAM site. The vertical black dotted line represents the actual cut site. Cutting and error-prone repair usually result in mixed sequencing bases after the cut. (<b>D</b>) Conjugate transferrin internalization is blocked in AP2M1-KO cells. The uptake of transferrin in KO AP2M1 Huh-7 cells C5 and C7 is blocked. Cells were maintained for 30 min on ice with Tf CF<sup>®</sup>543 (5 μg/mL). Finally, they were incubated 5 min at 37 °C before fixation. Nuclei were stained with DAPI. Scale bar = 20 µm. Original images can be found in <a href="#app1-biomolecules-14-01232" class="html-app">Supplementary Materials</a>.</p>
Full article ">Figure 4
<p>The knockout of AP2M1 in Huh-7 cells contributed to the decrease in HCoV-229E infection. KO cells C5 and C7 and non-KO cells (wild-type) were infected with HCoV-229E-GFP at an m.o.i. of 0.5. At 20 h p.i., samples were processed, and infection was analyzed by the following techniques: (<b>A</b>) Flow cytometry data show the mean of the percentage of normalized infection 20 h p.i. (% GFP+ cells) ± S.D. (<span class="html-italic">n</span> = 4). * <span class="html-italic">p</span> &lt; 0.05. The plots represent the histograms of GFP-positive (+) and GFP-negative (−) cells. (<b>B</b>) Infectious particles of the progeny virus for each condition were titrated in Huh-7 cells to determine the 50% tissue culture infective dose (TCID<sub>50</sub>)/mL. The graph shows the mean ± S.D. (<span class="html-italic">n</span> = 3) viral production. (<b>C</b>) Western blot analysis of total cell lysates subjected to SDS-PAGE, showing the viral GFP for each condition. β-actin was chosen as the protein loading control. Values of immunoblot quantification are reported as mean ± S.D. (<span class="html-italic">n</span> = 3); * <span class="html-italic">p</span> &lt; 0.05. Original images can be found in <a href="#app1-biomolecules-14-01232" class="html-app">Supplementary Materials</a>.</p>
Full article ">Figure 5
<p>Inhibition of endosomal acidification prevents the entry of HCoV-229E in the Huh-7 cell line. Huh-7 cells were treated with NH<sub>4</sub>Cl at different concentrations for 1 h and during infection with HCoV-229E-GFP at an m.o.i. of 3. (<b>A</b>) Fluorescence microscopy images showing GFP-positive cells at 20 h p.i. and nuclei stained with DAPI for each condition. Scale bar = 100 µm. (<b>B</b>) The flow cytometry data report the mean percentage of GFP+ cells, normalized to the non-treated control. * <span class="html-italic">p</span> &lt; 0.05 (<span class="html-italic">n</span> = 4). (<b>C</b>) Huh-7 cells were treated with 20 mM NH<sub>4</sub>Cl from 1 h before infection to 6 h p.i. (short times) or from 6 to 24 h p.i. (long times), with HCoV-229E-GFP at an m.o.i. of 3. The flow cytometry analysis reports the mean percentage of GFP+ cells at 24 h p.i., normalized to the non-treated control. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01 (<span class="html-italic">n</span> = 4).</p>
Full article ">Figure 6
<p>Quantitative confocal analysis of the colocalization of HCoV-229E and EEA-1 signal hot spots. (<b>A</b>) Cells were maintained with HCoV-229E for 15 min on ice and then incubated for 30 min at 33 °C. Then, they were fixed and labeled with the EEA-1 primary antibody and AlexaFluor-555-conjugated secondary antibody (red). The nuclei were stained with DAPI (blue). In the far-right panel, all three signal bandwidth images are combined, with colocalized red and green signals seen as yellow. Scale bar = 20 µm. (<b>B</b>) Two lineal sections were selected for each signal (yellow lines). The red EEA-1 and green viral GFP signal intensities in this line were plotted as a function of X–Y distance across the cell. Scale bar = 10 µm.</p>
Full article ">
16 pages, 2978 KiB  
Article
A Gain-of-Function Cleavage of TonEBP by Coronavirus NSP5 to Suppress IFN-β Expression
by Hyun Park, Sang Min Lee, Su Ji Jeong, Yeong Cheon Kweon, Go Woon Shin, Whi Young Kim, Whaseon Lee-Kwon, Chan Young Park and Hyug Moo Kwon
Cells 2024, 13(19), 1614; https://doi.org/10.3390/cells13191614 - 26 Sep 2024
Viewed by 811
Abstract
Human coronaviruses (HCoVs) modify host proteins to evade the antiviral defense and sustain viral expansion. Here, we report tonicity-responsive enhancer (TonE) binding protein (TonEBP) as a cellular target of HCoVs. TonEBP was cleaved into N-terminal and C-terminal fragments (TonEBP NT and TonEBP CT, [...] Read more.
Human coronaviruses (HCoVs) modify host proteins to evade the antiviral defense and sustain viral expansion. Here, we report tonicity-responsive enhancer (TonE) binding protein (TonEBP) as a cellular target of HCoVs. TonEBP was cleaved into N-terminal and C-terminal fragments (TonEBP NT and TonEBP CT, respectively) by NSP5 from all the HCoVs tested. This cleavage resulted in the loss of TonEBP’s ability to stimulate the TonE-driven transcription. On the other hand, TonEBP NT promoted viral expansion in association with the suppression of IFN-β expression. TonEBP NT competed away NF-κB binding to the PRD II domain on the IFN-β promoter. A TonEBP mutant resistant to the cleavage by NSP5 did not promote the viral expansion nor suppress the IFN-β expression. These results demonstrate that HCoVs use a common strategy of targeting TonEBP to suppress the host immune defense. Full article
(This article belongs to the Collection Cellular Immunology and COVID-19)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>TonEBP is cleaved by human coronavirus NSP5. (<b>A</b>) HEK293T cells were transfected with a TonE-driven Photinus luciferase plasmid. pRL-TK plasmid driving the expression of Renilla luciferase was co-transfected to normalize transfection efficiency. After 16 h, the cells were infected with HCoV-OC43 for 24 h at different MOIs indicated. The cells were then treated without (mock) or with 50 mM NaCl for 6 h before analyses of Photinus and Renilla luciferase activity to calculate TonE activity. (<b>B</b>) A549 cells were infected with HCoV-OC43 at an MOI of 20 for up to 24 h, as indicated. The cells were then analyzed by immunoblotting (IB) for TonEBP, OC43 N, and GAPDH. Full-length TonEBP and cleaved TonEBP are marked by solid and open arrowhead, respectively. (<b>C</b>) A549 cells were infected with HCoV-OC43 for 24 h at MOI up to 10, as indicated, before immunoblotting. (<b>D</b>) HEK293T cells were transfected for 16 h with the TonE-driven luciferase plasmid and pRL-TK, along with an empty vector (marked “−“) or a plasmid expressing one of NSPs (NSP1 to NSP16 except NSP11, as marked 1 to 16 except 11) from SARS-CoV-2. The cells were then treated without or with 50 mM NaCl and luciferase was measured. (<b>E</b>) Cells in isotonic conditions (mock) were transfected with one of the SARS-CoV-2 NSPs followed by immunoblotting for TonEBP, Flag, and GAPDH. (<b>F</b>) Cells were transfected with the plasmid expressing SARS-CoV-2 NSP5 for 4 h and treated with 0, 30, or 50 μM of GC376 for an additional 20 h. (<b>G</b>) Cells were transfected with a plasmid expressing either wild-type or catalytically inactive mutant C145A SARS-CoV-2 NSP5. (<b>H</b>) Cells were transfected with the TonE-driven luciferase plasmid and pRL-TK, along with the empty vector (-) or increasing amounts of the plasmid expressing SARS-CoV-2 NSP5 or C145A for 16 h. The transfected cells were treated without or with 50 mM NaCl for 6 h before analyses of luciferases. (<b>I</b>) Cells were co-transfected with plasmids expressing GFP-TonEBP (GFP is conjugated to the N-terminus of TonEBP) and either wild type or catalytically inactive mutant of NSP5 from HCoV-229E, HCoV-NL63, HCoV-OC43, bat CoV-HKU9 or SARS-CoV-2 (SCoV2). Full-length GFP-TonEBP and cleaved GFP-TonEBP are marked by filled and open arrowhead, as in (<b>E</b>–<b>G</b>) above. Data are shown as the mean ± SEM, <span class="html-italic">n</span> = 3; * <span class="html-italic">p</span> &lt; 0.033, ** <span class="html-italic">p</span> &lt; 0.002, and *** <span class="html-italic">p</span> &lt; 0.001 using <span class="html-italic">t</span>-tests (<b>A</b>,<b>D</b>,<b>H</b>). MOI, multiplicity of infection; GFP, green fluorescent protein; NSP, non-structural protein.</p>
Full article ">Figure 2
<p>SARS-CoV-2 NSP5 targets Q1127 of TonEBP. (<b>A</b>) HEK293T cells were co-transfected with the plasmid expressing either GFP-TonEBP (see above) or TonEBP-GFP (GFP is conjugated to the C-terminus of TonEBP) along with the empty vector or His-tagged SARS-CoV-2 NSP5-expressing vector, as indicated. At 48 h later, the cells were fixed and stained with DAPI. Fluorescence images for GFP, DAPI, and the two merged are shown. Scale bar: 100 µm. GFP fluorescence intensity was quantified as the mean ± SEM, <span class="html-italic">n</span> = 3 (bottom). (<b>B</b>) Cells were transfected for 24 h with constructs as in (<b>A</b>). The cells were then treated for 6 h with vehicle (DMSO) or 10 μM MG132 before immunoblotting. Full-length GFP-TonEBP and cleaved GFP-TonEBP are marked by solid and open arrowhead, respectively. Relative intensity of the cleaved GFP-TonEBP (open arrowhead) was quantified as the mean ± SEM, <span class="html-italic">n</span> = 3 (bottom). (<b>C</b>) Schematic of TonEBP primary sequence with putative cleavage sites marked with Qs predicted using NetCorona1.0 (with a score higher than 0.35). RHD, Rel-homology domain; TAD, transcription activation domain. (<b>D</b>) Cells were transfected with various combinations of plasmids expressing GFP-TonEBP (TonEBP), GFP-TonEBP with Q1127A mutation (Q1127A), and NSP5, as indicated, before immunoblotting. ns <span class="html-italic">p</span> &gt; 0.05, and *** <span class="html-italic">p</span> &lt; 0.001 using <span class="html-italic">t</span>-tests (<b>A</b>,<b>B</b>).</p>
Full article ">Figure 3
<p>Cleavage of TonEBP promotes expansion of HCoV-OC43. (<b>A</b>) U2OS stable cells expressing doxycycline (DOX)-inducible YFP, YFP-TonEBP wild type (WT) or YFP-TonEBP Q1127A were cultured without or with DOX, as indicated, and analyzed by immunoblotting. Anti-GFP antibody was used to detect YFP. (<b>B</b>) The cells in (<b>A</b>) were infected with HCoV-OC43 at an MOI of 0.001 and covered with 1% low-melting agarose. After 24 h, cell culture plates were fixed and stained with anti-HCoV-OC43 N protein antibody (red) and DAPI (blue). YFP is shown in yellow. Scale bar: 100 µm. (<b>C</b>) Number of cells in a single focus of N protein in TonEBP WT and TonEBP Q1127A relative to YFP was shown in box and whisker plots, <span class="html-italic">n</span> = 7–11. (<b>D</b>) The cells in (<b>A</b>) were infected with HCoV-OC43 as in (<b>B</b>) and cultured for 68 h. HCoV-OC43 was quantified from the culture media using RT-qPCR. Relative virus titer was normalized to the virus titer of the doxycycline-free condition of each cell line. Mean ± SEM, <span class="html-italic">n</span> = 3. (<b>E</b>) A single colony of cells stably expressing DOX-inducible YPF (YPF#1) and two colonies expressing U2OS stable cells expressing YFP-TonEBP NT were cultured without or with DOX. (<b>F</b>) The cells in (<b>E</b>) were cultured and infected with HCoV-OC43 as in (<b>B</b>). (<b>G</b>) Number of cells in a single focus of N protein in (<b>E</b>) was analyzed as in (<b>C</b>). Box and whisker plots are shown, <span class="html-italic">n</span> = 7–29. (<b>H</b>) The cells in (<b>E</b>) were infected with HCoV-OC43 as in (<b>F</b>) and cultured for 68 h. HCoV-OC43 was quantified from the culture media. Statistical analysis was conducted using one-way ANOVA combined with Bonferroni’s multiple comparisons test; ns <span class="html-italic">p</span> &gt; 0.05, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Cleaved TonEBP blocks viral induction of IFN-β. (<b>A</b>) HEK293T cells were transfected for 24 h with an <span class="html-italic">IFN-β</span> promoter luciferase reporter plasmid and pRL-TK in combination with a plasmid expressing MDA5, MAVS, TBK1, IRF3-5D, or p65 (see (<b>F</b>) below for the signaling pathway), along with various amounts of expression plasmid encoding TonEBP NT, as indicated, before analyses of luciferases. (<b>B</b>) The cells were transfected for 24 h with one of the plasmids expressing MDA5, MAVS, TBK1, IRF3-5D, or p65, without or with the expression plasmid for TonEBP NT, as indicated. The level of <span class="html-italic">IFN-β</span> mRNA was then determined using RT-qPCR. The transcript level of <span class="html-italic">IFN-β</span> was normalized to the expression of GAPDH. (<b>C</b>) Cells were transfected and analyzed as in (<b>A</b>) except that two expression plasmids for IRF3-5D and p65 were used instead of single plasmid for the signaling molecules. (<b>D</b>) Cells were transfected and analyzed as in (<b>B</b>) except that two expression plasmids for IRF3-5D and p65 were used instead of single plasmid for the signaling molecules. (<b>E</b>) Cells were transfected as in (<b>D</b>) for 48 h except for variable amounts of the plasmid for TonEBP NT, and IFN-β in the culture media were measured using enzyme-linked immunosorbent assay. (<b>F</b>) A model for the inhibitory role of cleaved TonEBP (TonEBP NT) in the MDA5–IFN-β signaling pathway. (<b>G</b>) Cells were co-transfected for 24 h with various combinations of four plasmids expressing TonEBP WT, TonEBP Q1127A, MAVS and SARS-CoV-2 NSP5, as indicated. The level of <span class="html-italic">IFN-β</span> mRNA was then determined using RT-qPCR. The transcript level of <span class="html-italic">IFN-β</span> was normalized to the expression of <span class="html-italic">GAPDH</span>. (<b>H</b>) Cells were co-transfected for 48 h with various combinations of four plasmids expressing TonEBP WT, TonEBP Q1127A, MAVS and SARS-CoV-2 NSP5, as indicated. IFN-β in the culture media were measured. Data are shown as the mean ± SEM, <span class="html-italic">n</span> = 3; ns <span class="html-italic">p</span> &gt; 0.05, * <span class="html-italic">p</span> &lt; 0.033, ** <span class="html-italic">p</span> &lt; 0.002, and *** <span class="html-italic">p</span> &lt; 0.001 using <span class="html-italic">t</span>-tests.</p>
Full article ">Figure 5
<p>Cleavage of TonEBP promotes binding to the PRD II domain of the <span class="html-italic">IFN-β</span> promoter. (<b>A</b>) HEK293T cells were co-transfected for 16 h with a plasmid expressing GFP-IRF3 without or with the plasmid expressing Flag-TonEBP NT. Cell lysates (Input) were immunoprecipitated (IP) with anti-Flag antibody. (<b>B</b>) Cells were transfected and analyzed as in (<b>A</b>) except that YPF-p65 was expressed instead of GFP-IRF3. (<b>C</b>) HEK293T cells were co-transfected for 24 h with various combinations of two plasmids expressing Flag-TonEBP NT and MAVS, as indicated. Chromatin immunoprecipitation was performed using normal rabbit IgG or anti-TonEBP antibody. The <span class="html-italic">IFN-β</span> promoter region indicated was quantified using qPCR. Mean ± SEM, <span class="html-italic">n</span> = 3. (<b>D</b>) Cells were co-transfected for 24 h with various combinations of three plasmids expressing Flag-TonEBP, Flag-TonEBP Q1127A and SARS-CoV-2 NSP5, as indicated. Chromatin immunoprecipitation was performed as in (<b>C</b>). (<b>E</b>) Schematic of TonEBP NT (1–1127) and TonEBP sNT (1–548). (<b>F</b>) Cells were transfected for 24 h with the <span class="html-italic">IFN-β</span> promoter luciferase reporter plasmid and pRL-TK plus one of the combinations of plasmids expressing p65, TonEBP NT, and TonEBP sNT (1–548), as indicated. Luciferases were measured to assess the activity of <span class="html-italic">IFN-β</span> promoter. (<b>G</b>) Nucleotide sequence of a portion of the human <span class="html-italic">IFN-β</span> promoter for binding of IRF3 and NF-κB (p65). Sequences of three probes are shown below. PRDIII, PRDI, and PRDII regions are marked by yellow, green, and orange colors, respectively. (<b>H</b>,<b>I</b>) The probes shown above were biotinylated and individually immobilized on streptavidin sensor chips for surface plasmon resonance assay. Various concentrations of p65 (<b>H</b>) or TonEBP sNT (1–548) (<b>I</b>) recombinant proteins were injected over the sensor surface. Binding affinity was determined by plotting the maximum response unit versus the concentration of injected proteins using GraphPad Prism 9.0. The values of K<sub>D</sub> are shown. (<b>J</b>) 100 nM each of Cy5.5-labeled <span class="html-italic">IFN-β</span> promoter probes were individually incubated without or with 500 nM TonEBP sNT (1–548) and resolved in an agarose gel. (<b>K</b>) Combinations of 0, 100, 200, or 500 nM TonEBP sNT (1–548) and p65 recombinant proteins (0 or 100 nM), as indicated, were each incubated for 25 min with 100 nM of Cy5.5-labeled IRF3/p65 probe and resolved in an agarose gel. The solid arrowhead denotes the complex of TonEBP sNT (1–548) with the probe and the open arrowhead denotes the complex of p65 with the probe. (<b>L</b>) A model for the IFN-β induction and its inhibition by TonEBP NT by replacing DNA bound p65. Data are shown as the mean ± SEM, <span class="html-italic">n</span> = 3; ns <span class="html-italic">p</span> &gt; 0.05, and *** <span class="html-italic">p</span> &lt; 0.001 using <span class="html-italic">t</span>-tests (<b>C</b>,<b>D</b>,<b>F</b>).</p>
Full article ">
12 pages, 1425 KiB  
Article
Comparison of Extraction Methods for the Detection of Avian Influenza Virus RNA in Cattle Milk
by Chantal J. Snoeck, Aurélie Sausy, Manon Bourg and Judith M. Hübschen
Viruses 2024, 16(9), 1442; https://doi.org/10.3390/v16091442 - 10 Sep 2024
Viewed by 1380
Abstract
Since early 2024, a multistate outbreak of highly pathogenic avian influenza H5N1 has been affecting dairy cattle in the USA. The influenza viral RNA concentrations in milk make it an ideal matrix for surveillance purposes. However, viral RNA detection in multi-component fluids such [...] Read more.
Since early 2024, a multistate outbreak of highly pathogenic avian influenza H5N1 has been affecting dairy cattle in the USA. The influenza viral RNA concentrations in milk make it an ideal matrix for surveillance purposes. However, viral RNA detection in multi-component fluids such as milk can be complex, and optimization of influenza detection methods is thus required. Raw bulk tank milk and mastitis milk samples were artificially contaminated with an avian influenza strain and subjected to five extraction methods. HCoV-229E and synthetic RNA were included as exogenous internal process controls. Given the high viral load usually observed in individual raw milk samples, four out of five tested methods would enable influenza detection in milk with normal texture, over a time window of at least 2 weeks post-onset of clinical signs. Nevertheless, sample dilution 1:3 in molecular transport medium prior to RNA extraction provided the best results for dilution of inhibitory substances and a good recovery rate of influenza RNA, that reached 12.5 ± 1.2% and 10.4 ± 3.8% in two independent experiments in bulk milk and 11.2 ± 3.6% and 10.0 ± 2.9% on two cohorts of mastitis milk samples. We have also shown compatibility of an influenza RT-qPCR system with synthetic RNA detection for simultaneous validation of the RNA extraction and RT-qPCR processes. Full article
(This article belongs to the Special Issue Advances in Animal Influenza Virus Research: Third Edition)
Show Figures

Figure 1

Figure 1
<p><b>Schematic illustration of the five methods compared for viral RNA extraction of IAV in cattle milk.</b> Method M1 corresponds to the manufacturer’s instructions with RNA extraction directly from 140 µL of medium or milk. For methods M2 or M3, milk samples were diluted 1:1 (method M2) or 1:3 (method M3) in molecular transport medium prior to RNA extraction from the diluted sample. For method M4, a swab was dipped into undiluted milk prior to being discharged in molecular transport medium. RNA was extracted from the molecular transport medium. Finally, for method M5, milk was diluted 1:3 in molecular transport medium prior to RNA extraction omitting adding lysis into AVL and directly loading on the silica-based membrane spin column.</p>
Full article ">Figure 2
<p><b>IAV and IPC detection in virus transport medium (VTM) and raw bulk tank milk.</b> Triplicate VTM and bulk milk samples were artificially contaminated with four ten-fold dilutions of IAV and extracted with methods M1 to M4. Constant quantities of HCoV-229E and commercial IPC were added at the lysis step. (<b>A</b>) Cq values obtained with IAV-1 RT-qPCR (y-axis) corresponding to final IAV concentrations ranging from 8.2 × 10<sup>3</sup> to 8.2 × 10<sup>6</sup> gc/mL of milk (x-axis). Mean values and standard deviations from three technical replicates are shown. (<b>B</b>,<b>C</b>) Cq values from RT-qPCR detecting both hCoV-229E (<b>B</b>) and commercial IPC (<b>C</b>) in a duplex format. The results of samples contaminated with four IAV dilutions in three technical replicates each are shown. In the absence of inhibition, Cq values for HCoV-229E or commercial IPC are expected to be similar across RNA extraction methods.</p>
Full article ">Figure 3
<p><b>Assessment of compatibility of IAV and commercial IPC detection in IAV-2 duplex RT-qPCR system.</b> (<b>A</b>) Tenfold dilutions of A/Cambodia/E0826360/2020 (H3N2) viral RNA were tested in technical triplicates in mastermix containing no IPC and no IPC detection primers and probe (IAV target, black symbols), and mastermix containing both a constant quantity of IPC RNA (1 × 10<sup>3</sup> copies) and IPC detection primers and probe (IAV target, red symbols). Detection of IPC in the VIC channel in duplex mastermix is shown with the green symbols (IPC target). Mean Cq values and standard deviations from three technical replicates are shown. (<b>B</b>) Cq values obtained with IAV-2 singleplex RT-qPCR for samples artificially contaminated with four ten-fold dilutions of IAV and extracted with methods M1 to M4 (same as for <a href="#viruses-16-01442-f002" class="html-fig">Figure 2</a>A). Mean Cq values and standard deviations from three technical replicates are shown. (<b>C</b>) Bland–Altman plot of viral load (in log<sub>10</sub> gc/µL) measured with IAV-2 RT-qPCR in singleplex or duplex. Average mean of all differences and 95% limits of agreement are shown with dotted lines. (<b>D</b>) Cq values of detection of commercial IPC with IAV-2 duplex RT-qPCR. The results of three technical replicates of samples contaminated with four IAV dilutions are shown. Lower Cq values are systematically obtained for commercial IPC with IAV-2 duplex compared to duplex detection with HCoV-229E (<a href="#viruses-16-01442-f002" class="html-fig">Figure 2</a>C), due to higher sensitivity of IAV-2 RT-qPCR chemistry for amplification of the commercial IPC.</p>
Full article ">Figure 4
<p><b>IAV and IPC detection in bulk milk and individual milk samples from cows with mastitis.</b> A single IAV concentration was seeded into VTM (technical replicates, <span class="html-italic">n</span> = 3), bulk milk (technical replicates, <span class="html-italic">n</span> = 3) and mastitis milk samples (biological replicates, <span class="html-italic">n</span> = 6), while a constant concentration of commercial IPC was added at the lysis step. IAV detection was performed with IAV-2 duplex RT-qPCR. (<b>A</b>) Recovery rates, calculated as the ratio of viral loads in a sample extracted with methods M1–M4 to the average viral load in VTM with method M1. Mean values and standard deviations of the replicates are shown. (<b>B</b>) Cq values (and mean and standard deviations) for commercial IPC detection with IAV-2 duplex RT-qPCR are shown. (<b>C</b>) Viral loads measured in individual mastitis samples extracted with methods M1–M4 are displayed, with samples with curdling highlighted in red.</p>
Full article ">Figure 5
<p><b>IAV and IPC detection in a second set of individual milk samples from cows with mastitis.</b> A single IAV concentration was seeded into VTM, bulk milk and individual mastitis milk samples (<span class="html-italic">n</span> = 12) while constant concentration of commercial IPC was added at the lysis step. IAV detection was performed with IAV-2 duplex RT-qPCR. (<b>A</b>) Recovery rates in bulk milk (three technical replicates) and mastitis milk samples (12 biological replicates) obtained with methods M1, M3 and M5 are shown. Mean values and standard deviations of the replicates are shown. (<b>B</b>) Cq values (and mean and standard deviations) for commercial IPC detection with IAV-2 duplex RT-qPCR are shown (<b>C</b>). Viral loads measured in individual mastitis samples extracted with methods M1, M3 and M5 are displayed, with samples with curdling highlighted in red.</p>
Full article ">
19 pages, 4491 KiB  
Article
Myrtus communis L. Essential Oil Exhibits Antiviral Activity against Coronaviruses
by Dar-Yin Li, Matthew G. Donadu, Taylor Shue, Georgios Dangas, Antonis Athanasiadis, Shuiyun Lan, Xin Wen, Basem Battah, Stefania Zanetti, Vittorio Mazzarello, Stefan G. Sarafianos, Marco Ferrari and Eleftherios Michailidis
Pharmaceuticals 2024, 17(9), 1189; https://doi.org/10.3390/ph17091189 - 10 Sep 2024
Viewed by 1212
Abstract
Human coronaviruses are a continuous threat to the human population and have limited antiviral treatments, and the recent COVID-19 pandemic sparked interest in finding new antiviral strategies, such as natural products, to combat emerging coronaviruses. Rapid efforts in the scientific community to identify [...] Read more.
Human coronaviruses are a continuous threat to the human population and have limited antiviral treatments, and the recent COVID-19 pandemic sparked interest in finding new antiviral strategies, such as natural products, to combat emerging coronaviruses. Rapid efforts in the scientific community to identify effective antiviral agents for coronaviruses remain a focus to minimize mortalities and global setbacks. In this study, an essential oil derived from Myrtus communis L. (MEO) is effective against HCoV-229E and HCoV-OC43 virus infections in comparison to two FDA-approved drugs, Remdesivir and Nirmatrelvir. Gas-chromatography and mass spectrometry were used to identify the chemical composition of MEO. Slight antioxidant activity was observed in MEO, indicating a role in oxidative stress. A dose–response curve measuring the EC50 indicates a high potency against HCoV-229E and HCoV-OC43 virus infections on Huh7.5 cells with low cytotoxicity using a PrestoBlue cell viability assay. Our findings demonstrate that MEO exhibits potent antiviral activity against HCoV-229E and HCoV-OC43 on Huh7.5 cells within a low-cytotoxicity range, but not on SARS-CoV-2. Artificial bacterial chromosome plasmids that expressed SARS-CoV-2 used for replicon—to determine viral replication and viral assembly/egress on HEK293T/17 cells—and virus-like particles on Huh7.5-AT cells—to determine viral entry and assembly/egress—showed no antiviral activity with MEO in comparison to Remdesivir. This study reveals the potential effectiveness of MEO as an alternative natural remedy to treat human coronaviruses and a potential antiviral agent for future coronavirus infections. Full article
Show Figures

Figure 1

Figure 1
<p>MEO inhibits HCoV-229E infection. (<b>A</b>) Experimental design of MEO experiment for dose–response curve. Huh7.5 cells were seeded in two 96-well plates with Remdesivir and Nirmatrelvir as antiviral controls. The experiment was performed in triplicate, and the starting concentration for MEO was 1:1000 (0.8469 mg/mL) and received a 1:2 serial dilution. The starting concentrations for Remdesivir and Nirmatrelvir were 250 nM and 25 µM, respectively, and were diluted with a 1:2 serial dilution. After HCoV-229E was added to the plates an hour after drug treatment, cells were fixed with 4% PFA one day post-infection. Immunofluorescent staining (IF) was performed to visualize infected cells using Cytation 7. (<b>B</b>) Dose–response curve showing that MEO has antiviral activity against HCoV-229E with EC<sub>50</sub> = 0.1204 mg/mL MEO concentration starting at 1:1000 (0.8469 mg/mL) with a 1:2 serial dilution and infected with 1:10 HCoV-229E virus. (<b>C</b>) Cytotoxicity assay measuring cell viability in MEO-treated Huh7.5 cells. A 1:10 (84.69 mg/mL) starting concentration for MEO was used with a 1:2 serial dilution. Huh7.5 cells treated with MEO were normalized to the untreated Huh7.5 cells.</p>
Full article ">Figure 2
<p>Immunofluorescent staining for HCoV-229E Spike protein. Huh7.5 cells without MEO treatment showed a mean fluorescence intensity of 22 with HCoV-229E. At a 1:2000 (0.42345 mg/mL) Myrtus concentration, the HCoV-229E viral infection has a mean fluorescence intensity of 7. Nuclei were stained with Hoechst, and HCoV-229E was stained with HCoV-229E spike protein and Alexa Fluor 488, labeled goat anti-mouse secondary antibody, and was imaged at a 1000 µm scale.</p>
Full article ">Figure 3
<p>MEO inhibits HCoV-OC43 infection. (<b>A</b>) Experimental layout of MEO with HCoV-OC43 infection to determine EC<sub>50</sub>. Two collagen-coated 96-wells plates were seeded with Huh7.5 cells to evaluate the dose–response curve of Remdesivir, Nirmatrelvir, and MEO. Each drug was conducted in triplicates with the starting concentration for MEO to be 1:1000 (0.8469 mg/mL) and serially diluted 1:2. The starting concentrations for Remdesivir and Nirmatrelvir were 7 µM and 25 µM, respectively, and were diluted with a 1:2 serial dilution. An hour after drug treatment, HCoV-OC43 was added, and cells were fixed with 4% PFA at 3 days post-infection. Immunofluorescent staining (IF) was performed to visualize infected cells using the Cytation 7. (<b>B</b>) A dose–response curve showed that MEO has antiviral activity against HCoV-OC43 with EC<sub>50</sub> = 1.405 mg/mL MEO concentration starting at a concentration of 1:1000 (0.8469 mg/mL) with a 1:2 serial dilution and infected with 1:20 HCoV-OC43E virus.</p>
Full article ">Figure 4
<p>Immunofluorescent staining for HCoV-OC43 spike protein. Huh7.5 cells without MEO treatment showed a mean fluorescence intensity of 12 with HCoV-OC43. At a 1:1000 (0.8469 mg/mL) MEO concentration, HCoV-OC43 viral infection has a mean fluorescence intensity of 4. Nuclei were stained with Hoechst and HCoV-OC43 was stained with anti-coronavirus antibody, OC-43 strain, clone 541-8F, and Alexa Fluor 488, labeled goat anti-mouse secondary antibody, and was imaged at a 1000 µm scale.</p>
Full article ">Figure 5
<p>MEO does not inhibit SARS-CoV-2 viral replication based on SARS-CoV-2 replicon. (<b>A</b>) Normalized SARS-CoV-2 replicon transfection to determine if MEO inhibits viral replication. Using GFP reporter in the SARS-CoV-2 replicon to calculate the percent of transfected cells, there was no difference in the number of transfected cells compared to normalized and untreated wells. NLuc activity from the SARS-CoV-2 replicon plasmid was used to quantify the amount of viral replication under MEO-treated conditions. Titration of MEO with SARS-CoV-2 replicon transfected cells showed no difference in viral replication via NLuc activity. Cell viability is not affected by SARS-CoV-2 replicon, nor by MEO cytotoxicity at the tested MEO concentrations. The relative luminescence graph validates that viral replicon is not inhibited by MEO. (<b>B</b>) Remdesivir substantially inhibited SARS-CoV-2 viral replication and normalized transfection level similar to the relative luminescence units, as untreated wells received about an eight-fold increase compared to 7.5 µM. At 15 µM of Remdesivir, cell viability dropped below 50%, exhibiting Remdesivir cytotoxicity.</p>
Full article ">Figure 6
<p>No inhibition against SARS-CoV-2 VLPs for MEO. (<b>A</b>) Immunofluorescence images showed no difference in VLP inhibition for untreated and MEO-treated wells at 1:200 (4.2345 mg/mL) concentration. Remdesivir inhibited SARS-CoV-2 VLP in a dose-dependent manner, which acted as positive control. To determine the percentage of cells infected with SARS-CoV-2 VLPs, the cells were counterstained with Hoechst and imaged for GFP reporter signal from the replicon plasmids and Hoechst dye using Cytation 7. All images are in 1000 µm scale. (<b>B</b>) The VLP transduction assay with MEO treatment showed similar infection levels as untreated wells. At 1:100 (8.469 mg/mL) concentration of MEO, cells were not viable, and viral replication remained consistent. (<b>C</b>) Remdesivir showed potent inhibition against SARS-CoV-2 VLP as an inhibitor of SARS-CoV-2 viral replication.</p>
Full article ">Figure 7
<p>MEO treatment has little impact SARS-CoV-2 Omicron BA.1 VLP formation/release. SARS-CoV-2 BA.1 VLPs were produced in HEK293T/17 cells that were pre-treated with either a 1:1000 (0.8469 mg/mL) or 1:500 (1.6938 mg/mL) concentration of MEO in DMEM (10% FBS + 1% NEAA) to assess the effect of MEO on the formation of infectious VLPs. VLPs were harvested via centrifugation and concentrated 20× using 100,000 MW Amicon filter units. VLPs were titrated on Huh7.5-AT cells at a starting dilution of 1:5 and continued with a 1:2 dilution. At one day post-transduction, the cells were counterstained with 1:5000 Hoechst dye and imaged using Cytation 7 for the number of GFP+ and total cells. Then, the cell culture supernatant was measured for NLuc activity. (<b>A</b>) Hoechst staining shows that there was a slight decrease in cell viability for VLPs formed in the presence of MEO treatment in a dose-dependent manner (<span class="html-italic">p</span> &lt; 0.0001). (<b>B</b>) Analysis of the GFP+ cells representing cells successfully transduced with VLP and undergoing replication of the replicon plasmid showed minor differences between MEO-treated and untreated VLPs, but only at the highest dilutions of VLP delivery (<span class="html-italic">p</span> = 0.01). (<b>C</b>) NLuc activity shows no difference between MEO VLPs and untreated VLPs. Overall, MEO treatment during VLP production has no effect on nascent VLP particles.</p>
Full article ">
17 pages, 3316 KiB  
Article
ESCRT Protein VPS4A Is Required for the Formation of Replication Centers and Replication of Human Coronavirus 229E (HCoV-229E)
by Rinki Kumar, Rebecca Kaddis Maldonado, Neil D. Christensen, Maria C. Bewley, John M. Flanagan, Nicholas J. Buchkovich and Leslie J. Parent
COVID 2024, 4(9), 1338-1354; https://doi.org/10.3390/covid4090095 - 24 Aug 2024
Viewed by 1014
Abstract
Human coronavirus 229E (HCoV-229E) is an alpha coronavirus that infects humans and bats. In common with all positive-strand RNA viruses, 229E infection causes rearrangements of the host’s intracellular membranes to form replication organelles, a highly conserved and vital step in the viral replication [...] Read more.
Human coronavirus 229E (HCoV-229E) is an alpha coronavirus that infects humans and bats. In common with all positive-strand RNA viruses, 229E infection causes rearrangements of the host’s intracellular membranes to form replication organelles, a highly conserved and vital step in the viral replication cycle. Here, we investigated the role of the ESCRT protein VPS4A in 229E infection. We found that functional VPS4A was required for the formation of replication organelles and localizing viral RNA to these structures in host cells to facilitate viral genome replication. We validated this effect using small molecule inhibitors to VPS4A, significantly reducing virus replication. We also showed that other ESCRTS, like CHMP4B, were required for the virus replication step, whereas VPS37A was involved in the post-replication stages. The absence of a functional VPS4A prevented the remodeling of membranes to form viral replication centers and, therefore, exposed the viral RNA, triggering an inflammatory immune response as indicated by elevated levels of IL-6. Interestingly, we observed the role of VPS4A to be similar for the OC43 coronavirus, indicating it could be conserved across all four coronavirus genera, including SARS-CoV-2. Understanding more about the replication of coronaviruses is imperative to finding more effective ways to control them. Full article
Show Figures

Figure 1

Figure 1
<p>Functional VPS4A is necessary for coronavirus (CoV) propagation. MRC-5 cells were transduced and selected with lentiviruses to express GFP-VPS4A<sup>E228Q</sup> or GFP-vector under a dox-inducible promoter. Cells were infected at 0.05 MOI with 229E virus and then incubated in the presence or absence of doxycycline (dox) (100 ng/mL). Supernatants were harvested daily for 3 days, and media was replenished daily with or without dox. (<b>A</b>,<b>B</b>) 229E virus titers were assessed using TCID<sub>50</sub> assay for the GFP-vector and GFP-VPS4A<sup>E228Q</sup>. Graphs show results from three independent experiments carried out in triplicate. Results are shown as mean ± SD. *** <span class="html-italic">p</span> &lt; 0.005, ns = not significant. (<b>C</b>) Transduced and selected MRC-5 cells were infected with OC43 at an MOI of 0.05 in the presence or absence of doxycycline. Supernatants were harvested at 24, 48, and 72 hpi. Virus titers were assessed using TCID<sub>50</sub> assay. Graphs show results from two independent experiments performed in triplicate. Results are shown as mean ± SD. **** <span class="html-italic">p</span> &lt; 0.0001. (<b>D</b>) Normal MRC-5 cells were infected with 229E virus at an MOI of 0.05 and treated with either DMSO, DBeQ, or #4107 (inhibitors targeting VPS4A). Supernatants were collected at 48 hpi, and virus titers were assessed using TCID<sub>50</sub> assay. Graphs show results from two independent experiments performed in triplicate. Results are shown as mean ± SD. ** <span class="html-italic">p</span> &lt; 0.005.</p>
Full article ">Figure 2
<p>VPS4A plays a significant role in the formation of replication centers. Transduced and selected MRC–5 cells were plated in 6 cm Petri dishes (compatible with sample processing for electron microscopy). Cells were infected with 229E at an MOI of 0.05 in the presence or absence of doxycycline, fixed at ~36 hpi, and processed. Copper grids were imaged. (<b>A</b>). 229E virus particles were visualized in the intercellular space in control cells containing GFP–VPS4A<sup>E228Q</sup> in the absence of dox (–dox), while no virus particles were observed in the presence of dox (+dox). (<b>B</b>). Representative images of GFP–VPS4A<sup>E228Q</sup> cells in the absence and presence of dox are shown. Scale bar, 600 nm. Arrows indicate replication centers. (<b>C</b>). GFP–VPS4A<sup>E228Q</sup> cells were infected with 229E in the presence or absence of doxycycline (dox) and harvested at 24, 48, and 72 hpi for RNA isolation. Relative genome levels using the 229E M gene were analyzed using One-Step RT–qPCR, and fold changes are shown. Three independent experiments were performed in triplicate. Results are shown as mean ± SD. **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>GFP–VPS4A<sup>E228Q</sup> mutation causes a decrease in 229E genome replication. (<b>A</b>) Cells containing a dox-inducible gfp gene were subjected to infection for 48 h with 229E. In the absence of dox-induction, GFP (green) is slightly leaky but is greatly upregulated in the presence of dox. Single-molecule RNA FISH (smFISH) with probes targeting the 229E genome (red) were used to indicate 229E replication. (<b>B</b>) Sum fluorescence intensity was measured for the RNA channel. There was no statistically significant change in 229E expression. (<b>C</b>) Cells containing a dox-inducible GFP-VPS4A<sup>E228Q</sup> dominant-negative mutant gene were subjected to infection for 48 h with 229E. GFP-VPS4A<sup>E228Q</sup> (green) is only expressed in the presence of dox. SmFISH was used to label 229E genomes (red). (<b>D</b>) Dox-induced GFP–VPS4A<sup>E228Q</sup> expression significantly decreased 229E genomic RNA levels, as indicated by a change in the sum fluorescence intensity of the RNA channel. **** <span class="html-italic">p</span> &lt; 0.0001, ns = not significant. Nucleus blue, DAPI, white outline. Scale bar = 10 µm. (<b>E</b>) MRC–5 cells transduced and selected for GFP–VPS4A<sup>E228Q</sup> expression were plated on coverslips and infected with 229E in the presence or absence of dox for 48 h. Cells were fixed and stained for viral nucleocapsid protein (green), dsRNA (red), GFP (shown as white), and DAPI (blue).</p>
Full article ">Figure 4
<p>229E induces an inflammatory immune response in the presence of GFP-VPS4A<sup>E228Q</sup>. MRC-5 cells transduced and selected with GFP-VPS4A<sup>E228Q</sup> were infected at 0.05 MOI with 229E virus and then incubated in the presence or absence of doxycycline (dox) (100 ng/mL). Cells were harvested for RNA isolation at 24, 48, and 72 hpi. Following cDNA synthesis, the mRNA levels of (<b>A</b>) ISG15, (<b>B</b>) MX1, (<b>C</b>) IL-6, and (<b>D</b>) IFN-β were analyzed using RT-qPCR and normalized to GAPDH. Three independent experiments were performed in triplicate. Relative fold changes are shown as mean ± SD.</p>
Full article ">Figure 5
<p>Effect of other ESCRTs on 229E replication. (<b>A</b>) MRC–5 cells transduced with GFP-CHMP4B were infected at an MOI of 0.05 with 229E in the absence or presence of dox. Supernatants were collected at 24, 48, and 72 hpi. Virus titers were assessed using TCID<sub>50</sub> assay. Graphs show results from two independent experiments conducted in triplicate. (<b>B</b>) MRC–5 cells transduced with lentivirus expressing either sh–NTC or sh–VPS37A and selected using puromycin. Knockdown was assessed on mock lysates by western blot and probed for VPS37A using antibodies against VPS37A and p115 (loading control). (<b>C</b>) The NTC and sh-VPS37A KD cells were infected with 229E at an MOI of 0.05. Supernatants were collected at 24, 48, and 72 hpi. Virus titers were assessed using TCID<sub>50</sub> assay. Graphs show results from two independent experiments conducted in triplicate. ** <span class="html-italic">p</span> = 0.009 (48 h) and ** <span class="html-italic">p</span> = 0.0052 (72 h). (<b>D</b>) GFP-vector, GFP-VPS4A<sup>E228Q</sup>, GFP-CHMP4B, NTC, and VPS37A KD cells were infected with 229E at an MOI of 0.05 for 48 h. Dox treatment was conducted as previously described for all except the NTC and VPS37A KD cells. Cells were harvested at 48 hpi for RNA isolation. Relative genome levels using the 229E M gene were analyzed using One-Step RT-qPCR, and fold changes are shown. Three independent experiments were performed in triplicate. Results are shown as mean ± SD. ns = not significant, * <span class="html-italic">p</span> = 0.024 (CHMP4B), ** <span class="html-italic">p</span> = 0.0052 (VPS37A), and **** <span class="html-italic">p</span> &lt; 0.0001 (GFP-VPS4A<sup>E228Q</sup>), respectively.</p>
Full article ">
19 pages, 2061 KiB  
Article
Development of an Enantioselective Method by Liquid Chromatography to Monitor 3,4-Methylenedioxypyrovalerone in Culture Media from Ecotoxicity Assays
by Ariana Pérez-Pereira, Virgínia M. F. Gonçalves, Ana R. L. Ribeiro, Carla Fernandes, João S. Carrola, Cláudia Ribeiro and Maria E. Tiritan
Separations 2024, 11(8), 248; https://doi.org/10.3390/separations11080248 - 16 Aug 2024
Viewed by 883
Abstract
3,4-Methylenedioxypyrovalerone (MDPV) is an emerging, chiral, new psychoactive substance belonging to the synthetic cathinones group that has been frequently detected in wastewater effluents and aquatic environments. However, the knowledge of its enantioselective occurrence and toxicity toward aquatic organisms is scarce. The aim of [...] Read more.
3,4-Methylenedioxypyrovalerone (MDPV) is an emerging, chiral, new psychoactive substance belonging to the synthetic cathinones group that has been frequently detected in wastewater effluents and aquatic environments. However, the knowledge of its enantioselective occurrence and toxicity toward aquatic organisms is scarce. The aim of this work was to develop an enantioselective liquid chromatography (LC) method to monitor the enantiomers of MDPV in environmental and ecotoxicological assays. For that, different chiral columns and mobile phases in both normal and reversed elution modes were attempted. The optimized conditions were achieved using a Daicel® 3 μm—CHIRALPAK® IF-3 column with 5 mM of ammonium bicarbonate (NH4HCO3, pH 8.8) in ultra-pure water (UPW) and acetonitrile (ACN) (10:90, v/v) as a mobile phase, at a flow rate of 0.3 mL min−1. This condition was applied to monitor the racemate and the single enantiomers of MDPV in culture medium collected from ecotoxicity experiments. Racemization was observed for MDPV enantiomers (in individual exposure). The enantiomeric ratio (e.r.) of (S)-MDPV changed from an initial e.r. of 96.4/3.6 to 78.0/22.0 and for the (R)-enantiomer, the e.r changed from 15.6/84.4 to 28.3/71.7). These data highlight the importance of enantioselective monitoring of culture media in toxicity assays that involve chiral substances, since racemization can occur and lead to inaccuracy in the toxicity evaluation. Nevertheless, it is also important to stress that racemization may occur during storage conditions or sample procedures. Therefore, the enantioselective methodology is of utmost importance to warrant the quality of the results in enantioselective ecotoxicological studies. Full article
(This article belongs to the Collection Feature Paper Collection in Section 'Environmental Separations')
Show Figures

Figure 1

Figure 1
<p>Chromatograms of MDPV enantioseparation in the analytical Lux<sup>®</sup> 3 μm—Cellulose-2 (150 × 4.6 mm I.D.) column by LC-UV/Vis under the normal-elution mode. LC conditions: mobile phase, 0.1% of DEA in Hex/EtOH (99:1, <span class="html-italic">v</span>/<span class="html-italic">v</span>); flow rate of 0.7 mL min<sup>−1</sup>; injection volume of 40 μL; λ<sub>max</sub> at 315 nm; RT. (<b>a</b>) (<span class="html-italic">R</span>,<span class="html-italic">S</span>)-MDPV at 0.10 μg mL<sup>−1</sup> in Hex/EtOH (99:1, <span class="html-italic">v</span>/<span class="html-italic">v</span>); (<b>b</b>) (<span class="html-italic">S</span>)-MDPV at 0.10 μg mL<sup>−1</sup> in Hex/EtOH (99:1, <span class="html-italic">v</span>/<span class="html-italic">v</span>). * Stereogenic center.</p>
Full article ">Figure 2
<p>Chromatograms of MDPV enantioseparation in the analytical Lux<sup>®</sup> 3 μm—Cellulose-2 (150 × 4.6 mm I.D.) column by LC-UV/Vis under the reversed-elution mode. LC conditions: mobile phase, 5 mM of NH<sub>4</sub>OAc in UPW (pH 8.5) and ACN; flow rate of 1.0 mL min<sup>−1</sup>; injection volume of 40 μL; λ<sub>max</sub> at 315 nm; RT; (<b>a</b>) (<span class="html-italic">R</span>,<span class="html-italic">S</span>)-MDPV at 0.50 μg mL<sup>−1</sup> in UPW; mobile phase (70:30, <span class="html-italic">v</span>/<span class="html-italic">v</span>); (<b>b</b>) (<span class="html-italic">R</span>,<span class="html-italic">S</span>)-MDPV at 1.0 μg mL<sup>−1</sup> in UPW; mobile phase (74:26, <span class="html-italic">v</span>/<span class="html-italic">v</span>). * Stereogenic center.</p>
Full article ">Figure 3
<p>Chromatogram of the enantioseparation of 1.0 μg mL<sup>−1</sup> (<span class="html-italic">R</span>,<span class="html-italic">S</span>)-MDPV (in UPW) in the analytical Lux<sup>®</sup> 3 μm—Cellulose-2 (150 × 2.0 mm I.D.) column by LC-UV/Vis under the reversed-elution mode. LC conditions: mobile phase, 20 mM of NH<sub>4</sub>OAc in UPW and ACN (70:30, <span class="html-italic">v</span>/<span class="html-italic">v</span>) pH 8.5; flow rate at 0.3 mL min<sup>−1</sup>; injection volume of 5 μL; λ<sub>max</sub> at 315 nm; column oven temperature set at 25 °C. * Stereogenic center.</p>
Full article ">Figure 4
<p>Chromatograms of MDPV enantioseparation in the analytical amylose <span class="html-italic">tris</span>-(3-chloro-4-methylphenylcarbamate) column commercialized as the Daicel<sup>®</sup> 3 μm—CHIRALPAK<sup>®</sup> IF-3 (150 × 2.1 mm I.D.) column by LC-UV/Vis under the reversed-elution mode. LC conditions: mobile phase, 5 mM of NH<sub>4</sub>HCO<sub>3</sub> in UPW (pH 8.8) with ACN (10:90, <span class="html-italic">v</span>/<span class="html-italic">v</span>); flow rate of 0.3 mL min<sup>−1</sup>; injection volume of 5 μL; λ<sub>max</sub> set at 315 nm; column oven temperature, 30 °C. (<b>a</b>) (<span class="html-italic">R</span>,<span class="html-italic">S</span>)-MDPV at 1.0 μg mL<sup>−1</sup> in UPW; (<b>b</b>) (<span class="html-italic">S</span>)-MDPV at 1.0 μg mL<sup>−1</sup> in UPW.</p>
Full article ">Figure 5
<p>Total ion chromatogram (black) and selected reaction monitoring (<span class="html-italic">m</span>/<span class="html-italic">z</span> 276.00 &gt; 126.15 (pink); <span class="html-italic">m</span>/<span class="html-italic">z</span> 276.00 &gt; 135.05 (blue) of (<span class="html-italic">R</span>,<span class="html-italic">S</span>)-MDPV enantioseparation (1.0 μg mL<sup>−1</sup> in UPW) in the analytical Daicel<sup>®</sup> 3 μm—CHIRALPAK<sup>®</sup> IF-3 (150 × 2.1 mm I.D.) column by LC-MS/MS under the reversed-elution mode. LC conditions: mobile phase, 5 mM NH<sub>4</sub>HCO<sub>3</sub> in UPW (pH 8.8) with ACN (10:90, <span class="html-italic">v</span>/<span class="html-italic">v</span>); flow rate of 0.3 mL min<sup>−1</sup>; injection volume of 5 μL; column oven temperature set at 30 °C.</p>
Full article ">Figure 6
<p>Total ion chromatograms (black) and selected reaction monitoring (<span class="html-italic">m</span>/<span class="html-italic">z</span> 276.00 &gt; 126.15 (pink); <span class="html-italic">m</span>/<span class="html-italic">z</span> 276.00 &gt; 135.05 (blue)) of MDPV enantiomers in the analytical Daicel<sup>®</sup> 3 μm—CHIRALPAK<sup>®</sup> IF-3 (150 × 2.1 mm I.D.) column by LC-MS/MS under the reversed-elution mode in the isocratic mode. LC conditions: 5 mM of NH<sub>4</sub>HCO<sub>3</sub> in UPW (pH 8.8) with ACN (10:90, <span class="html-italic">v</span>/<span class="html-italic">v</span>) as the mobile phase; flow rate of 0.3 mL min<sup>−1</sup>; injection volume of 5 μL; column oven temperature set at 30 °C. (<b>a</b>) (<span class="html-italic">R</span>,<span class="html-italic">S</span>)-MDPV sample at 1.0 μg L<sup>−1</sup> (UPW)—Sample Collection 2; (<b>b</b>) (<span class="html-italic">S</span>)-MDPV sample at 1.0 μg L<sup>−1</sup> (UPW)—Sample Collection 2; (<b>c</b>) (<span class="html-italic">R</span>)-MDPV sample at 1.0 μg L<sup>−1</sup> (UPW)—Sample Collection 2.</p>
Full article ">Figure 7
<p>Bar chart of e.r. values of racemic and enantiopure MDPV standards and MDPV spiked in the culture medium samples of <span class="html-italic">D. magna</span> enantioselective ecotoxicity assays, analyzed by the MDPV enantioselective method previously optimized in the analytical Daicel<sup>®</sup> 3 μm—CHIRALPAK<sup>®</sup> IF-3 (150 × 2.1 mm I.D.) column by LC-MS/MS under the reversed-elution mode in the isocratic mode. LC conditions: mobile phase, 5 mM of NH<sub>4</sub>HCO<sub>3</sub> in UPW (pH 8.8) with ACN (10:90, <span class="html-italic">v</span>/<span class="html-italic">v</span>); flow rate of 0.3 mL min<sup>−1</sup>; injection volume of 5 μL; column oven temperature set at 30 °C. (a) MDPV standards at 1.0 μg L<sup>−1</sup> (UPW); (b) MDPV samples at 1.0 μg L<sup>−1</sup> (UPW); (c) MDPV samples at 1.0 μg L<sup>−1</sup> (UPW); (C1) Collection n° 1; (C2) Collection n° 2; (C3) Collection n° 3.</p>
Full article ">
16 pages, 3402 KiB  
Article
Antiviral Properties of Moringa oleifera Leaf Extracts against Respiratory Viruses
by Rosa Giugliano, Valeria Ferraro, Annalisa Chianese, Roberta Della Marca, Carla Zannella, Francesca Galdiero, Teresa M. A. Fasciana, Anna Giammanco, Antonio Salerno, Joseph Cannillo, Natalie Paola Rotondo, Giovanni Lentini, Maria Maddalena Cavalluzzi, Anna De Filippis and Massimiliano Galdiero
Viruses 2024, 16(8), 1199; https://doi.org/10.3390/v16081199 - 25 Jul 2024
Viewed by 1501
Abstract
Moringa oleifera (M. oleifera) is a plant widely used for its beneficial properties both in medical and non-medical fields. Because they produce bioactive metabolites, plants are a major resource for drug discovery. In this study, two different cultivars of leaves of [...] Read more.
Moringa oleifera (M. oleifera) is a plant widely used for its beneficial properties both in medical and non-medical fields. Because they produce bioactive metabolites, plants are a major resource for drug discovery. In this study, two different cultivars of leaves of M. oleifera (Salento and Barletta) were obtained by maceration or microwave-assisted extraction (MAE). We demonstrated that extracts obtained by MAE exhibited a lower cytotoxic profile compared to those obtained by maceration at concentrations ranged from 25 to 400 µg/mL, on both Vero CCL-81 and Vero/SLAM cells. We examined their antiviral properties against two viruses, i.e., the human coronavirus 229E (HCoV-229E) and measles virus (MeV), which are both responsible for respiratory infections. The extracts were able to inhibit the infection of both viruses and strongly prevented their attack and entry into the cells in a range of concentrations from 50 to 12 µg/mL. Particularly active was the variety of Salento that registered a 50% inhibitory concentration (IC50) at 21 µg/mL for HCoV-229E and at 6 µg/mL for MeV. We identified the presence of several compounds through high performance liquid chromatography (HPLC); in particular, chlorogenic and neochlorogenic acids, quercetin 3-O-β-d-glucopyranoside (QGP), and glucomoringin (GM) were mainly observed. In the end, M. oleifera can be considered a promising candidate for combating viral infections with a very strong action in the early stages of viral life cycle, probably by destructuring the viral particles blocking the virus–cell fusion. Full article
(This article belongs to the Special Issue Recent Advances in Antiviral Natural Products 2023)
Show Figures

Figure 1

Figure 1
<p>Cytotoxicity of <span class="html-italic">M. oleifera</span> extracts obtained by MAE (<b>A</b>,<b>B</b>) and maceration (<b>C</b>,<b>D</b>) on Vero CCL-81 and Vero/hSLAM cell lines. Positive control (CTRL+) was represented by untreated cells, negative control (CTRL−) by cells treated with DMSO and PBS was used as a vehicle. Two-way ANOVA was utilized for statistical analyses. Dunnett’s test was utilized for multiple comparisons. **** <span class="html-italic">p</span> &lt; 0.0001, * <span class="html-italic">p</span> &lt; 0.021, ns: not statistically significant.</p>
Full article ">Figure 2
<p>Antiviral activity of <span class="html-italic">M. oleifera</span> extracts against HCoV-229E. Co-treatment test (<b>A</b>) and virus pre-treatment (<b>B</b>) of MwS extracts; co-treatment test (<b>C</b>) and virus pre-treatment test (<b>D</b>) of MwB extracts. Algal extract at 50 µg/mL was used as a positive control (CTRL+), while infected and untreated cells represented negative control (CTRL−). Two-way ANOVA was used for statistical analysis. The Dunnett test was used for multiple comparisons. **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> ≤ 0.0002, * <span class="html-italic">p</span> &lt; 0.0332, ns: not statistically significant.</p>
Full article ">Figure 3
<p>Antiviral activity of <span class="html-italic">M. oleifera</span> extracts against HCoV-229E. Co-treatment test (<b>A</b>) and virus pre-treatment (<b>B</b>) of MaS extracts; co-treatment (<b>C</b>) and pre-treatment test (<b>D</b>) of MaB extracts. Algal extract at 50 µg/mL was used as a positive control (CTRL+), while infected and untreated cells represented negative control (CTRL−). Two-way ANOVA was used for statistical analysis. The Dunnett test was used for multiple comparisons. **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> ≤ 0.0002, ** <span class="html-italic">p</span> &lt; 0.0021, * <span class="html-italic">p</span> &lt; 0.0332, ns: not statistically significant.</p>
Full article ">Figure 4
<p>Antiviral activity of <span class="html-italic">M. oleifera</span> extracts against MeV. Co-treatment test (<b>A</b>) and virus pre-treatment (<b>B</b>) of MwS extracts; co-treatment test (<b>C</b>) and virus pre-treatment test of MwB extracts (<b>D</b>). The peptide AR-23 at 25 µg/mL was used as a positive control (CTRL+), while infected and untreated cells represented negative control (CTRL−). Two-way ANOVA was used for statistical analysis. The Dunnett test was used for multiple comparisons. **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> ≤ 0.0002, ** <span class="html-italic">p</span> &lt; 0.0021, * <span class="html-italic">p</span> &lt; 0.0332, ns: not statistically significant.</p>
Full article ">Figure 5
<p>Antiviral activity of <span class="html-italic">M. oleifera</span> extracts against MeV. Co-treatment test (<b>A</b>) and virus pre-treatment (<b>B</b>) of MaS extracts; co-treatment (<b>C</b>) and pre-treatment test (<b>D</b>) of MaB extracts. The peptide AR-23 at 25 µg/mL was used as a positive control (CTRL+), while infected and untreated cells represented negative control (CTRL−). Two-way ANOVA was used for statistical analysis. The Dunnett test was used for multiple comparisons. **** <span class="html-italic">p</span> &lt; 0.0001, ** <span class="html-italic">p</span> &lt; 0.0021, ns: not statistically significant.</p>
Full article ">Figure 6
<p>Temperature shift assays. Attachment and entry tests against HCoV-229E (<b>A</b>–<b>C</b>) and MeV (<b>B</b>–<b>D</b>). Positive controls were algal extract at 50 µg/mL (HCoV-229E), and peptide AR-23 at 25 µg/mL (MeV). Infected and untreated cells represented negative control (CTRL−). Two-way ANOVA was used for statistical analysis. The Dunnett test was used for multiple comparisons. **** <span class="html-italic">p</span> &lt; 0.0001, ** <span class="html-italic">p</span> &lt; 0.0021, ns: not statistically significant.</p>
Full article ">Figure 7
<p>HPTLC profile of <span class="html-italic">M. oleifera</span> extracts obtained under microwave irradiation and four reference standards sprayed with NP/PEG reagent; MwB2 and MwB3: <span class="html-italic">M. oleifera</span> Barletta extracted with 50% EtOH and 70% EtOH, respectively; MwS2 andMwS3: <span class="html-italic">M. oleifera</span> Salento extracted with 50% EtOH and 70% EtOH, respectively; CA: chlorogenic acid; NCA: neochlorogenic acid; QPG: quercetin 3-<span class="html-italic">O</span>-β-<span class="html-small-caps">d</span>-glucopyranoside; GM: glucomoringin; solvent: ethyl acetate/formic acid/acetic acid/water (100:11:11:26); λ: 366 nm.</p>
Full article ">
22 pages, 5600 KiB  
Article
The Novel A-Type Proanthocyanidin-Rich Phytocomplex SP4™ Acts as a Broad-Spectrum Antiviral Agent against Human Respiratory Viruses
by Giulia Sibille, Giuseppe Mannino, Ilaria Frasson, Marta Pavan, Anna Luganini, Cristiano Salata, Massimo E. Maffei and Giorgio Gribaudo
Int. J. Mol. Sci. 2024, 25(13), 7370; https://doi.org/10.3390/ijms25137370 - 5 Jul 2024
Cited by 1 | Viewed by 1535
Abstract
The appearance of new respiratory virus infections in humans with epidemic or pandemic potential has underscored the urgent need for effective broad-spectrum antivirals (BSAs). Bioactive compounds derived from plants may provide a natural source of new BSA candidates. Here, we investigated the novel [...] Read more.
The appearance of new respiratory virus infections in humans with epidemic or pandemic potential has underscored the urgent need for effective broad-spectrum antivirals (BSAs). Bioactive compounds derived from plants may provide a natural source of new BSA candidates. Here, we investigated the novel phytocomplex formulation SP4™ as a candidate direct-acting BSA against major current human respiratory viruses, including coronaviruses and influenza viruses. SP4™ inhibited the in vitro replication of SARS-CoV-2, hCoV-OC43, hCoV-229E, Influenza A and B viruses, and respiratory syncytial virus in the low-microgram range. Using hCoV-OC43 as a representative respiratory virus, most of the antiviral activity of SP4™ was observed to stem primarily from its dimeric A-type proanthocyanidin (PAC-A) component. Further investigations of the mechanistic mode of action showed SP4™ and its PAC-A-rich fraction to prevent hCoV-OC43 from attaching to target cells and exert virucidal activity. This occurred through their interaction with the spike protein of hCoV-OC43 and SARS-CoV-2, thereby interfering with spike functions and leading to the loss of virion infectivity. Overall, these findings support the further development of SP4™ as a candidate BSA of a natural origin for the prevention of human respiratory virus infections. Full article
Show Figures

Figure 1

Figure 1
<p>SP4™ inhibits the replication of hCoV-OC43, hCoV-229E, SARS-CoV-2, IAV, IBV, and RSV-A. (<b>A</b>,<b>B</b>) Focus forming reduction assays (FFRAs) were performed in HCT-8 and MRC5 cells infected with coronaviruses hCoV-OC43 (<b>A</b>) or hCoV-229E (<b>B</b>), respectively, and treated with different concentrations of SP4™ 1 h before, during, and post infection. At 72 h p.i., the viral foci were microscopically counted and converted into viral titer (plaque forming unit (PFU)/mL). (<b>C</b>–<b>G</b>) Virus yield reduction assays (VRAs) were performed in Calu-3 cells infected with SARS-CoV-2 Wuhan (<b>C</b>) or the Omicron BA.1 variant (<b>D</b>) and in A549 cells infected with influenza A (IAV) (<b>E</b>), influenza B (IBV) (<b>F</b>), or respiratory syncytial (RSV-A) viruses (<b>G</b>). Infected cell monolayers were treated throughout the experiment with different concentrations of SP4™. At 48 h p.i. for SARS-CoV-2 and IVs and at 72 h p.i. for RSV-A, cell supernatants were harvested and titrated by the plaque assay, as described in the Material and Methods. The data shown are the means ± standard deviations (SDs) (error bars) of <span class="html-italic">n</span> = 3 independent experiments performed in triplicate.</p>
Full article ">Figure 2
<p>Anti-hCoV activity of the different fractions of SP4™. (<b>A</b>) FFRAs were performed in HCT-8 cells infected with hCoV-OC43 and exposed to different concentration of the primary fractions F1-F3. At 72 h p.i., the viral foci were microscopically counted and converted into viral titer (PFU/mL). (<b>B</b>,<b>C</b>) The secondary fractions obtained from the fractionation of F1 (F1A-C, (<b>B</b>)) or F3 (F3A-E, (<b>C</b>)) were analyzed by FFRAs in hCoV-OC43-infected HCT-8 cells treated with different concentrations of the samples. The results are shown as the means ± SD of <span class="html-italic">n</span> = 3 independent experiments performed in triplicate.</p>
Full article ">Figure 3
<p>Synthetic dimeric PAC-As inhibit hCoV replication. (<b>A</b>,<b>B</b>) FFRAs were carried out in HCT-8 cells infected with hCoV-OC43 and treated with different concentrations of synthetic PAC-As (<b>A</b>) or PAC-Bs (<b>B</b>), present throughout the experiment. At 72 h p.i., the viral foci were microscopically counted and converted into viral titer (PFU/mL). The reported data are the means ± SDs of <span class="html-italic">n</span> = 3 independent experiments performed in triplicate.</p>
Full article ">Figure 4
<p>SP4™ acts during an early stage of the hCoV replicative cycle. (<b>A</b>) Schematic summary of the time-of-drug-addition experiment. HCT-8 cell monolayers were exposed to different concentrations of SP4™ 1 h prior to hCoV-OC43 infection (from −1 to 0 h; pre-treatment; red line), or during hCoV-OC43 infection (from 0 to 2 h p.i.; co-treatment; blue line), or after hCoV-OC43-infection (from 2 to 72 h p.i.; post-treatment; green line). (<b>B</b>) At 72 h p.i., hCoV-OC43 replication was assessed by FFRA, and the viral foci were microscopically counted and plotted as PFU/mL. The data shown are the means ± SD (error bars) of three independent experiments performed in triplicate and analyzed by a two-way ANOVA followed by Dunnett’s multiple comparison test. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>SP4™ prevents hCoV-OC43 attachment and entry. (<b>A</b>) For the attachment assay, prechilled HCT-8 cell monolayers were infected with hCoV-OC43 (50 PFU/well) at 4 °C for 2 h in the presence of different concentrations of SP4™ or F3, ranging from 0.39 to 25 μg/mL. Following viral adsorption, the infected cell monolayers were washed three times with medium, overlaid with growth medium supplemented with 1% methylcellulose, and incubated at 33 °C. At 72 h p.i., viral foci were immunostained and microscopically counted. The results shown are the means ± SD from <span class="html-italic">n</span> = 3 independent experiments performed in triplicate. (<b>B</b>) For the entry assay, prechilled HCT-8 cells were infected with hCoV-OC43 (50 PFU/well) for 2 h at 4 °C to allow virion attachment to the cells. Thereafter, the cells were washed with medium and treated with different amounts of SP4™ or F3 for 2 h at 33 °C, prior to the inactivation of bound extracellular virus with acidic glycine buffer for 30 s at room temperature. After further washings, cell monolayers were incubated for 72 h at 33 °C, and the viral foci were detected as in the attachment assay. The results shown are the means ± SD of <span class="html-italic">n</span> = 3 independent experiments performed in triplicate.</p>
Full article ">Figure 6
<p>SP4™ abrogates the infectivity of hCoV-OC43 virions. hCoV-OC43 (10<sup>5</sup> PFU/mL) was incubated at either 4 °C (<b>A</b>) or at 33 °C (<b>B</b>) for different times in the absence (control virus (CV)) or presence of 15 μg/mL SP4™ or F3. After incubation, the samples were diluted to reduce the concentration of SP4™ or fraction 3 below the concentration at which hCoV-OC43 attachment is inhibited (0.1 μg/mL). The foci were microscopically counted at 72 h p.i., and the mean number of foci counts was expressed as PFU/mL. The results are representative of <span class="html-italic">n</span> = 3 independent experiments performed in triplicate. The data were analyzed by a two-way ANOVA, followed by Dunnett’s multiple comparison test. A statistical analysis was performed by comparing the treated samples with the CV control for each condition. * <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; and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 7
<p>SP4™ interacts with the purified spike (S) protein of hCoV-OC43. (<b>A</b>) SDS-PAGE and the immunoblot analysis of the recombinant S protein of hCoV-OC43. The purified recombinant S protein preparation was analyzed by 4–15% SDS-PAGE, and the gels were either stained with Coomassie blue (left panel) or analyzed by immunoblotting with an anti-S (S1 subunit) polyclonal antibody (pAb) (right panel). Left panel: Coomassie blue-stained gel of 3 μg of the S protein preparation fractionated in the 160 kilodaltons (kDa) S1 + S2 component, the 95 kDa S1 subunit, and the 60 kDa S2 subunit. Right panel: Immunoblot analysis of 500 ng of the S protein preparation in which the S1 + S2 and the S1 components had been stained with anti-S1 monoclonal antibody (mAb). (<b>B</b>–<b>E</b>) SP4™ and F3 interact with the recombinant S protein of hCoV-OC43. (<b>B</b>,<b>C</b>) The purified recombinant S protein preparation (3 μg) was incubated at 33 °C with DMSO as a control or increasing amounts of either SP4™ (<b>B</b>) or F3 (<b>C</b>) for 3 h. The mixtures were then analyzed by 4–15% SDS–PAGE. (<b>D</b>,<b>E</b>) The recombinant S protein preparation (3 μg) was incubated at 33 °C for 30, 60, 120, and 180 min with DMSO or 9 μg of SP4™ (<b>D</b>) or 3 μg of F3 (<b>E</b>). At the indicated timepoints, the mixtures were analyzed by 4–15% SDS–PAGE. The gels were then stained with Coomassie blue, and images were acquired by ChemiDoc. The molecular weight markers are indicated in kilodaltons.</p>
Full article ">Figure 8
<p>SP4™ targets the purified S protein of SARS-CoV-2. (<b>A</b>) SDS-PAGE and immunoblot analysis of recombinant SARS-CoV-2 (Omicron BA.1) S protein preparation. The purified S protein preparation (S1 + S2) was fractionated by 4–15% SDS-PAGE and either stained with Coomassie blue (left panel) or analyzed by immunoblotting with an anti-S1 mAb (right panel). Left panel: Coomassie blue-stained gel of 3 μg of the recombinant S preparation resolved as the 160 kDa S1 + S2 protein band. Right panel: Immunoblot analysis of 500 ng of the same sample as in the left panel. (<b>B</b>–<b>E</b>) SP4™ and F3 interact with the S protein of SARS-CoV-2. (<b>B</b>,<b>C</b>) The purified recombinant S protein preparation (1 μg) was incubated with DMSO as a control or increasing concentrations of SP4™ (<b>B</b>) or F3 (<b>C</b>) for 3 h at 37 °C; the mixtures were then analyzed by 4–15% SDS–PAGE. (<b>D</b>,<b>E</b>) Recombinant S protein aliquots (1 μg) were incubated at 37 °C for 30, 60, 120, and 180 min with DMSO or 3 μg of SP4™ (<b>D</b>) or 1 μg of fraction 3 (<b>E</b>). At the indicated incubation timepoint, the mixtures were analyzed by 4–15% SDS-PAGE. the Gels were then stained with Coomassie blue, and images were acquired by ChemiDoc. The molecular weight markers are indicated in kilodaltons.</p>
Full article ">
Back to TopTop