[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (2,409)

Search Parameters:
Keywords = spike protein

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 3213 KiB  
Article
Intranasal Prime–Boost with Spike Vectors Generates Antibody and T-Cell Responses at the Site of SARS-CoV-2 Infection
by Muriel Metko, Jason Tonne, Alexa Veliz Rios, Jill Thompson, Haley Mudrick, David Masopust, Rosa Maria Diaz, Michael A. Barry and Richard G. Vile
Vaccines 2024, 12(10), 1191; https://doi.org/10.3390/vaccines12101191 - 18 Oct 2024
Abstract
Background: Long-lived, re-activatable immunity to SARS-CoV-2 and its emerging variants will rely on T cells recognizing conserved regions of viral proteins across strains. Heterologous prime–boost regimens can elicit elevated levels of circulating CD8+ T cells that provide a reservoir of first responders upon [...] Read more.
Background: Long-lived, re-activatable immunity to SARS-CoV-2 and its emerging variants will rely on T cells recognizing conserved regions of viral proteins across strains. Heterologous prime–boost regimens can elicit elevated levels of circulating CD8+ T cells that provide a reservoir of first responders upon viral infection. Although most vaccines are currently delivered intramuscularly (IM), the initial site of infection is the nasal cavity. Methods: Here, we tested the hypothesis that a heterologous prime and boost vaccine regimen delivered intranasally (IN) will generate improved immune responses locally at the site of virus infection compared to intramuscular vaccine/booster regimens. Results: In a transgenic human ACE2 murine model, both a Spike-expressing single-cycle adenovirus (SC-Ad) and an IFNß safety-enhanced replication-competent Vesicular Stomatitis Virus (VSV) platform generated anti-Spike antibody and T-cell responses that diminished with age. Although SC-Ad-Spike boosted a prime with VSV-Spike-mIFNß, SC-Ad-Spike alone induced maximal levels of IgG, IgA, and CD8+ T-cell responses. Conclusions: There were significant differences in T-cell responses in spleens compared to lungs, and the intranasal boost was significantly superior to the intramuscular boost in generating sentinel immune effectors at the site of the virus encounter in the lungs. These data show that serious consideration should be given to intranasal boosting with anti-SARS-CoV-2 vaccines. Full article
(This article belongs to the Section COVID-19 Vaccines and Vaccination)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Safety and immunogenicity of intranasal delivery of viral vaccines expressing SARS-CoV-2 Spike. (<b>A</b>) Schematic of Adenoviral and VSV-vectors expressing Spike. (<b>B</b>) Detection of SARS-CoV-2 Spike protein by Western Blot 24 h post-infection of 293T ACE2 cells with different viral vaccines. (<b>C</b>) IFNβ detection by ELISA of 293T ACE2 cells infected with VSV-based vaccines. <span class="html-italic">p</span> &lt; 0.0001 (****), ns = statistically non-significant, One-way ANOVA. (<b>D</b>) Survival curve of transgenic k18-hACE2 mice infected intranasally with different doses of VSV-based vaccines. <span class="html-italic">p</span> = 0.031 (*), Log–rank test with Grehan–Breslow–Wilcoxon test. (<b>E</b>) LegendPlexCytokine brain profile of mice experiencing toxicity 5–6 days after intranasal inoculation of VSV-Spike-GFPL.IL-12p70 <span class="html-italic">p</span> = 0.0333 (*), IL-1β 1 × 10<sup>5</sup> <span class="html-italic">p</span> = 0.0016 (**), IL-1β 5 × 10<sup>5</sup> <span class="html-italic">p</span> = 0.0068 (**), GMCSF <span class="html-italic">p</span> = 0.0184 (*), <span class="html-italic">p</span> &lt; 0.0001 (****), One-way ANOVA.</p>
Full article ">Figure 2
<p>Intranasal, but not intramuscular, delivery of viruses induces T-cell responses in the lung airways. (<b>A</b>) K18-hACE2 mice were vaccinated intranasally (IN) or intramuscularly (IM) with VSV-Spike-mIFNß (dose of 2 × 10<sup>8</sup> pfu) or SC-Ad-Spike (dose of 10<sup>10</sup> vp). Spleens, bronchoalveolar fluid (BAL), and blood were collected 14 days post-vaccination. (<b>B</b>) Serum was used at 1:1000 dilution to test for anti-SARS-CoV-2 Spike IgG antibodies by ELISA. Similar amounts of antibody were produced IN and IM for both vaccines. <span class="html-italic">p</span> = ns, one-way ANOVA. (<b>C</b>) Representative flow plots of BAL samples showing anti-Spike-tetramer+ gate and VSV-Nucleocapsid tetramer+ gate. <span class="html-italic">N</span> = 3 mice per group. (<b>D</b>) Quantification of BAL flow cytometry results. Frequency (%) of CD8+ T cells. Frequency (%) and count of CD8+ Spike tet+ T cells, and CD8+ VSV-Nuc tet+ T cells within lymphocytes detected by flow cytometry. <span class="html-italic">p</span> = ns, Unpaired <span class="html-italic">T</span>-test. (<b>E</b>) Quantification of splenocyte flow cytometry results. Frequency (%) of CD8+ T cells. Frequency (%) and count of CD8+ Spike tet+ T cells, and CD8+ VSV-Nuc tet+ T cells within lymphocytes detected by flow cytometry. <span class="html-italic">p</span> = 0.0096 (**), <span class="html-italic">p</span> = 0.0111 (*), Unpaired <span class="html-italic">T</span>-test.</p>
Full article ">Figure 3
<p>SC-Ad-Spike induces anti-Spike CD8+ T-cell responses in the lungs independent of prime and boost regimens. (<b>A</b>) K18-hACE2 mice were vaccinated intranasally (IN) with VSV-Spike- mIFNß (dose of 2 × 10<sup>8</sup> pfu) or SC-Ad-Spike (dose of 10<sup>10</sup> vp). Blood collected on day 21 prior to intranasal (IN) boost vaccination. Spleens, bronchoalveolar fluid (BAL), and blood were collected 3 weeks post boost. (<b>B</b>) Serum used at 1:1000 dilution to test for anti-SARS-CoV-2 Spike IgG antibodies by ELISA. Similar amounts of antibody were produced for all prime and boost combinations. <span class="html-italic">p</span> = ns (<b>C</b>) BAL fluid used neat to test for anti-SARS-CoV-2 Spike IgG antibodies by ELISA. <span class="html-italic">p</span> = ns. (<b>D</b>) BAL fluid used neat to test for anti-SARS-CoV-2 Spike IgA antibodies by ELISA. <span class="html-italic">p</span> = 0.0005 (***), <span class="html-italic">p</span> = 0.0024 (**) (<b>E</b>) Representative flow plots of BAL samples showing anti-Spike-tetramer+ gate and VSV-Nucleocapsid tetramer+ gate. <span class="html-italic">N</span> = 3 mice per group. (<b>F</b>) Quantification of BAL flow cytometry results. Frequency (%) of CD8+ T cells. Frequency (%) and count of CD8+ Spike tet+ T cells, and CD8+ VSV-Nuc tet+ T cells within lymphocytes detected by flow cytometry. <span class="html-italic">p</span> &lt; 0.0001 (****), <span class="html-italic">p</span> = 0.0006 (***), <span class="html-italic">p</span> = 0.0068 (**), <span class="html-italic">p</span> = 0.012 (*), one-way ANOVA. (<b>G</b>) Quantification of splenocyte flow cytometry results. Frequency (%) of CD8+ T cells. Frequency (%) and count of CD8+ Spike tet+ T cells, and CD8+ VSV-Nuc tet+ T cells within lymphocytes detected by flow cytometry. <span class="html-italic">p</span> &lt; 0.0001 (****), <span class="html-italic">p</span> = 0.0006 (***), <span class="html-italic">p</span> = 0.0068 (**), <span class="html-italic">p</span> = 0.0246 (*), one-way ANOVA.</p>
Full article ">Figure 4
<p>Anti-Spike Ab and T-cell response decrease with age. (<b>A</b>) Old (263 week) k18-hACE2 mice were vaccinated intranasally (IN) with VSV-Spike- mIFNß (dose of 2 × 10<sup>8</sup> pfu) or SC-Ad-Spike (dose of 10<sup>10</sup> vp). Blood collected on day 21 prior to intranasal (IN) boost vaccination. Spleens, bronchoalveolar fluid (BAL), and blood were collected 3 weeks post boost. (<b>B</b>) Serum used at 1:1000 dilution to test for anti-SARS-CoV-2 Spike IgG antibodies by ELISA. Similar amounts of antibody were produced for all prime and boost combinations. <span class="html-italic">p</span> = ns (<b>C</b>) Quantification of BAL flow cytometry results. Frequency (%) of CD8+ T cells. Frequency (%) and count of CD8+ Spike tet+ T cells, and CD8+ VSV-Nuc tet+ T cells within lymphocytes detected by flow cytometry. <span class="html-italic">p</span> = 0.0009 (***), <span class="html-italic">p</span> = 0.0438 (*), <span class="html-italic">p</span> = ns, one-way ANOVA. (<b>D</b>) Quantification of splenocyte flow cytometry results. Frequency (%) of CD8+ T cells. Frequency (%) and count of CD8+ Spike tet+ T cells, and CD8+ VSV-Nuc tet+ T cells within lymphocytes detected by flow cytometry. <span class="html-italic">p</span> = 0.0302 (*), <span class="html-italic">p</span> = ns, one-way ANOVA.</p>
Full article ">Figure 5
<p>Intranasal boosting significantly enhances IgA and CD8+ T-cell responses compared to intramuscular boosting. (<b>A</b>) K18-hACE2 mice were vaccinated intramuscularly (IM) with Sc-Ad-Spike (dose of 10<sup>10</sup> vp). Six weeks later, blood was collected prior to intranasal (IN) boost vaccination with VSV-Spike- mIFNß or SC-Ad-Spike or PBS. Spleens, bronchoalveolar fluid (BAL), and blood were collected 3 weeks post boost. (<b>B</b>) Serum used at 1:1000 dilution to test for anti-SARS-CoV-2 Spike IgG antibodies by ELISA. <span class="html-italic">p</span> &lt; 0.0001 (****), <span class="html-italic">p</span> = ns, one-way ANOVA. (<b>C</b>) BAL fluid used neat to test for anti-SARS-CoV-2 Spike IgG antibodies by ELISA. <span class="html-italic">p</span> = ns, one-way ANOVA. (<b>D</b>) BAL fluid used neat to test for anti-SARS-CoV-2 Spike IgA antibodies by ELISA. <span class="html-italic">p</span> &lt; 0.0001 (****), <span class="html-italic">p</span> = 0.0067 (**), <span class="html-italic">p</span> = 0.0292 (*), <span class="html-italic">p</span> = ns, one-way ANOVA. (<b>E</b>) Quantification of BAL flow cytometry results. Frequency (%) of CD8+ T cells. Frequency (%) and count of CD8+ Spike tet+ T cells, and CD8+ VSV-Nuc tet+ T cells within lymphocytes detected by flow cytometry. <span class="html-italic">p</span> &lt; 0.0001 (****), <span class="html-italic">p</span> = 0.0094 (**), <span class="html-italic">p</span> = 0.0447 (*), <span class="html-italic">p</span> = ns, one-way ANOVA. (<b>F</b>) Quantification of splenocyte flow cytometry results. Frequency (%) of CD8+ T cells. Frequency (%) and count of CD8+ Spike tet+ T cells, and CD8+ VSV-Nuc tet+ T cells within lymphocytes detected by flow cytometry. <span class="html-italic">p</span> &lt; 0.0001 (****), <span class="html-italic">p</span> = 0.0006 (***), <span class="html-italic">p</span> = 0.0087 (**), <span class="html-italic">p</span> = 0.0123 (*), <span class="html-italic">p</span> = ns, one-way ANOVA. (<b>G</b>) K18-hACE2 mice were vaccinated IN or IM with SC-Ad-Spike or VSV-Spike-mIFNβ. Blood collected on day 21 prior to intranasal boost vaccination with PBS, VSV-Spike-mIFNβ, or Sc-Ad-Spike. Spleens, bronchoalveolar fluid (BAL), and blood were collected 3 weeks post boost. BAL fluid used undiluted to test for anti-SARS-CoV-2 Spike IgG antibodies by ELISA. <span class="html-italic">p</span> = ns, one-way ANOVA. (<b>H</b>) BAL fluid used neat to test for anti-SARS-CoV-2 Spike IgA antibodies by ELISA. <span class="html-italic">p</span> &lt; 0.0001 (****), <span class="html-italic">p</span> = ns, one-way ANOVA. (<b>I</b>) Quantification of BAL flow cytometry results. Frequency (%) of CD8+ T cells. Frequency (%) and count of CD8+ Spike tet+ T cells. <span class="html-italic">p</span> &lt; 0.0001 (****), <span class="html-italic">p</span> = 0.0006 (***), <span class="html-italic">p</span> = 0.0044 (**), <span class="html-italic">p</span> = ns, one-way ANOVA.</p>
Full article ">
32 pages, 5551 KiB  
Review
Unveiling the Interplay—Vitamin D and ACE-2 Molecular Interactions in Mitigating Complications and Deaths from SARS-CoV-2
by Sunil J. Wimalawansa
Biology 2024, 13(10), 831; https://doi.org/10.3390/biology13100831 - 16 Oct 2024
Viewed by 434
Abstract
The interaction of the SARS-CoV-2 spike protein with membrane-bound angiotensin-converting enzyme-2 (ACE-2) receptors in epithelial cells facilitates viral entry into human cells. Despite this, ACE-2 exerts significant protective effects against coronaviruses by neutralizing viruses in circulation and mitigating inflammation. While SARS-CoV-2 reduces ACE-2 [...] Read more.
The interaction of the SARS-CoV-2 spike protein with membrane-bound angiotensin-converting enzyme-2 (ACE-2) receptors in epithelial cells facilitates viral entry into human cells. Despite this, ACE-2 exerts significant protective effects against coronaviruses by neutralizing viruses in circulation and mitigating inflammation. While SARS-CoV-2 reduces ACE-2 expression, vitamin D increases it, counteracting the virus’s harmful effects. Vitamin D’s beneficial actions are mediated through complex molecular mechanisms involving innate and adaptive immune systems. Meanwhile, vitamin D status [25(OH)D concentration] is inversely correlated with severity, complications, and mortality rates from COVID-19. This study explores mechanisms through which vitamin D inhibits SARS-CoV-2 replication, including the suppression of transcription enzymes, reduced inflammation and oxidative stress, and increased expression of neutralizing antibodies and antimicrobial peptides. Both hypovitaminosis D and SARS-CoV-2 elevate renin levels, the rate-limiting step in the renin-angiotensin-aldosterone system (RAS); it increases ACE-1 but reduces ACE-2 expression. This imbalance leads to elevated levels of the pro-inflammatory, pro-coagulatory, and vasoconstricting peptide angiotensin-II (Ang-II), leading to widespread inflammation. It also causes increased membrane permeability, allowing fluid and viruses to infiltrate soft tissues, lungs, and the vascular system. In contrast, sufficient vitamin D levels suppress renin expression, reducing RAS activity, lowering ACE-1, and increasing ACE-2 levels. ACE-2 cleaves Ang-II to generate Ang(1–7), a vasodilatory, anti-inflammatory, and anti-thrombotic peptide that mitigates oxidative stress and counteracts the harmful effects of SARS-CoV-2. Excess ACE-2 molecules spill into the bloodstream as soluble receptors, neutralizing and facilitating the destruction of the virus. These combined mechanisms reduce viral replication, load, and spread. Hence, vitamin D facilitates rapid recovery and minimizes transmission to others. Overall, vitamin D enhances the immune response and counteracts the pathological effects of SARS-CoV-2. Additionally, data suggests that widely used anti-hypertensive agents—angiotensin receptor blockers and ACE inhibitors—may lessen the adverse impacts of SARS-CoV-2, although they are less potent than vitamin D. Full article
(This article belongs to the Special Issue SARS-CoV-2 and Immunology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Infections and immune-related broader functions of vitamin D (calcitriol, 1,25(OH)<sub>2</sub>D). The figure illustrates muti-system-wide functions of vitamin D related through the modulation of innate and adaptive immune systems, resulting in lowering complications from infections and chronic disease burdens [⇧ = increased; ⇩ = reduced; RAS: renin-angiotensin-system; CVS: cardiovascular system] (after Wimalawansa, Nutrients, 2022) [<a href="#B51-biology-13-00831" class="html-bibr">51</a>].</p>
Full article ">Figure 2
<p>Pathological and physiological responses of the renin-angiotensin system. Peach and green boxes illustrate the renin-angiotensin system’s regulatory and counter-regulatory physiologic pathways. When excess angiotensin-II (Ang-II) is synthesized, as in the case of hypovitaminosis D and SARS-CoV-2 infection, this leads to the over-activation of the AT1 receptors (AT1-R) with pathological manifestations, as indicated in the peach colored boxes [⇧ = increased; ⇩ = reduced; ARDS = acute respiratory distress syndrome; RAS, renin-angiotensin system; ACE, angiotensin-converting enzyme; ACE-2, angiotensin-converting enzyme 2; Ang 1–7, angiotensin 1–7; Ang-I, angiotensin-I; Ang-II, angiotensin-II; AT1R, type 1 angiotensin-II receptor; MasR, MAS proto-oncogene receptor. PHT, pulmonary hypertension].</p>
Full article ">Figure 3
<p>This diagram outlines the status of the renin-angiotensin axis (RAS) axis: (<b>A</b>) physiological status, (<b>B</b>) pathological/activated status in the presence of vitamin D deficiency, and (<b>C</b>) following SARS-CoV-2 infection. RAS axis homeostasis is disrupted by hypovitaminosis D. SARS-CoV-2 or other coronal viral infections markedly activate the RAS, leading to pathologically elevated levels of angiotensin -II and the suppression of ACE-2. This hyperactivation of the RAS leads to increased complications and mortality (⇧ = increased; ⇩ = reduced; ACE: angiotensin-converting enzyme; ARB: angiotensin receptor blockers; AT1R: type 1 angiotensin-II receptor; ARDS: acute respiratory distress syndrome).</p>
Full article ">Figure 4
<p>Vit D strengthens innate and adaptive immune systems. This summary outlines the correlation between vitamin D, angiotensin-converting enzyme-2 (ACE-2), angiotensin-converting enzyme inhibitors (ACEi), and angiotensin II receptor blockers (ARBs) concerning severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and their impact on COVID-19 morbidity and mortality ([↑ = increased; ↓ = reduced; RAS: renin-angiotensin-system; CVS: cardiovascular system; ACE: angiotensin-converting enzyme; ARB: angiotensin receptor blockers; AT1R: type 1 angiotensin-II receptor; ARDS: acute respiratory distress syndrome; HTN: hypertension).</p>
Full article ">
13 pages, 829 KiB  
Review
Peptide-Based Inhibitors of Protein–Protein Interactions (PPIs): A Case Study on the Interaction Between SARS-CoV-2 Spike Protein and Human Angiotensin-Converting Enzyme 2 (hACE2)
by Aizhan Rakhmetullina, Piotr Zielenkiewicz and Norbert Odolczyk
Biomedicines 2024, 12(10), 2361; https://doi.org/10.3390/biomedicines12102361 - 16 Oct 2024
Viewed by 338
Abstract
Protein–protein interactions (PPIs) are fundamental to many critical biological processes and are crucial in mediating essential cellular functions across diverse organisms, including bacteria, parasites, and viruses. A notable example is the interaction between the SARS-CoV-2 spike (S) protein and the human angiotensin-converting enzyme [...] Read more.
Protein–protein interactions (PPIs) are fundamental to many critical biological processes and are crucial in mediating essential cellular functions across diverse organisms, including bacteria, parasites, and viruses. A notable example is the interaction between the SARS-CoV-2 spike (S) protein and the human angiotensin-converting enzyme 2 (hACE2), which initiates a series of events leading to viral replication. Interrupting this interaction offers a promising strategy for blocking or significantly reducing infection, highlighting its potential as a target for anti-SARS-CoV-2 therapies. This review focuses on the hACE2 and SARS-CoV-2 spike protein interaction, exemplifying the latest advancements in peptide-based strategies for developing PPI inhibitors. We discuss various approaches for creating peptide-based inhibitors that target this critical interaction, aiming to provide potential treatments for COVID-19. Full article
Show Figures

Figure 1

Figure 1
<p>The complex structure of <span class="html-italic">h</span>ACE2 and SARS-CoV-2 spike proteins (PDB id: 6m0j)— computer-generated, cartoon representation. (<b>a</b>) The interaction interface between <span class="html-italic">h</span>ACE2 (blue) and the spike (orange) is shown as the solvent accessible surface area and highlighted by magenta and cyan colors, respectively; (<b>b</b>,<b>c</b>) depicted amino acid residues forming the interface for a particular protein are shown as sticks in this representation; (<b>d</b>) schematic diagram of interactions between proteins. Residues are colored according to the type: positive (H, K, R); negative (D, E); S, T, N, Q = neutral; A, V, L, I, M = aliphatic; F, Y, W = aromatic; G = Gly. Type of contacts: hydrogen bonds (blue line); salt bridges (red line); nonbonded contacts (gray dash line). Protein visualization was prepared by the PyMOL Molecular Graphics System, Version 3.0.0 Schrödinger, LLC.</p>
Full article ">
13 pages, 3257 KiB  
Article
SERS-Driven Ceftriaxone Detection in Blood Plasma: A Protein Precipitation Approach
by Aradhana Dwivedi, Oleg Ryabchykov, Chen Liu, Edoardo Farnesi, Michael Stenbæk Schmidt, Thomas Bocklitz, Jürgen Popp and Dana Cialla-May
Chemosensors 2024, 12(10), 213; https://doi.org/10.3390/chemosensors12100213 (registering DOI) - 16 Oct 2024
Viewed by 253
Abstract
Accurate detection of antibiotics in biological samples is essential for clinical diagnoses and therapeutic drug monitoring. This research examines how proteins and other substances in blood plasma affect the detection of the antibiotic ceftriaxone using surface-enhanced Raman spectroscopy (SERS). We detected ceftriaxone spiked [...] Read more.
Accurate detection of antibiotics in biological samples is essential for clinical diagnoses and therapeutic drug monitoring. This research examines how proteins and other substances in blood plasma affect the detection of the antibiotic ceftriaxone using surface-enhanced Raman spectroscopy (SERS). We detected ceftriaxone spiked in blood plasma without sample preparation within the range of 1 mg/mL to 50 µg/mL. By employing a pretreatment approach involving methanol-based protein precipitation to eliminate interfering substances from a spiked blood plasma solution, we could detect ceftriaxone down to 20 µg/mL. The comparative analysis demonstrates that the protein precipitation step enhances the sensitivity of SERS-based detection of drugs in the matrix blood plasma. The insights derived from this study are highly beneficial and can prove advantageous in developing new antibiotic detection methods that are both sensitive and selective in complex biological matrices. These methods can have important implications for clinical treatments. Full article
(This article belongs to the Special Issue Surface-Enhanced Raman Spectroscopy for Bioanalytics)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Raman spectra of ceftriaxone in powder form (red curve), saturated aqueous solution (black curve), and SERS spectra in aqueous media (blue curve).</p>
Full article ">Figure 2
<p>SERS spectra of ceftriaxone spiked in the blood plasma, in the concentration range: (<b>a</b>) 1 mg/mL to 0 μg/mL, (<b>b</b>) within the range 100 μg/mL to 0 μg/mL.</p>
Full article ">Figure 3
<p>Scheme for precipitated blood plasma sample preparation.</p>
Full article ">Figure 4
<p>SERS spectra of ceftriaxone spiked in the precipitated blood plasma, in the concentration range: (<b>a</b>) 1 mg/mL to 0 μg/mL, (<b>b</b>) within the range 100 μg/mL to 0 μg/mL.</p>
Full article ">Figure 5
<p>PLS-DA classification model test for ceftriaxone detection in: (<b>a</b>) blood plasma, and (<b>b</b>) precipitated blood plasma. The results demonstrate that ceftriaxone can be detected in blood plasma starting from 50 μg/mL, while in precipitated blood plasma, it can be detected already at 20 μg/mL.</p>
Full article ">Figure 6
<p>Background SERS spectra of blood plasma (red curve) and precipitated blood plasma (black curve).</p>
Full article ">
8 pages, 928 KiB  
Brief Report
Modifications in Immune Response Patterns Induced by Kynurenine and One-Residue-Substituted T Cell Epitopes in SARS-CoV-2-Specific Human T Cells
by Mieko Tokano, Rie Takagi and Sho Matsushita
COVID 2024, 4(10), 1676-1683; https://doi.org/10.3390/covid4100116 - 15 Oct 2024
Viewed by 309
Abstract
Peptide p176-190, derived from the SARS-CoV-2 spike protein, is one of the major T cell epitopes that elicits the HLA-DR-restricted IL-8 response of human CD4+ T cells. Using PBMCs from a healthy individual primed with an S-protein-based SARS-CoV-2 vaccine, we established a [...] Read more.
Peptide p176-190, derived from the SARS-CoV-2 spike protein, is one of the major T cell epitopes that elicits the HLA-DR-restricted IL-8 response of human CD4+ T cells. Using PBMCs from a healthy individual primed with an S-protein-based SARS-CoV-2 vaccine, we established a CD4+ T cell line (TM45) and cloned T cells (TM45.2) specific for the peptide. We showed that (i) co-incubation with kynurenine leads to increased IL-8; (ii) T cells incubated in the absence of kynurenine recovered the original levels of cytokine production; and (iii) peptide p176-190 substituted at 176 Leucine for neutral hydrophilic serine completely abolished the cytokine responses of TM45.2 cells, thereby suggesting that 176 L is the first anchor residue for binding to HLA-DR. These observations collectively indicate that (i) enhanced IL-8 responses can be induced by kynurenine, which is produced under infectious conditions in COVID-19; (ii) the response is not a permanent change in the T cell phenotype; and (iii) IL-8 responses associated with harmful neutrophil extracellular traps can be abrogated by a single amino acid substitution of the viral antigens. These findings may shed light on a novel strategy for designing vaccines for viral infections that are accompanied by increased kynurenine production. Full article
Show Figures

Figure 1

Figure 1
<p>TM45 cells were incubated in the presence of irradiated (30 Gy) autologous PBMCs prepulsed with the peptide p176-190 and human rIL-2 (50 U/mL), with or without kynurenine. After 7 days, TM45 cells (1 × 10<sup>4</sup>/well) were cultured in 96-well flat-bottom culture plates in the presence of irradiated autologous PBMCs (1 × 10<sup>5</sup>/well) and 5 μM peptide p176-190 in R10H medium. Four days later, the culture supernatant fluid was collected for IL-8 (<b>A</b>) and GM-CSF (<b>B</b>) ELISAs (n = 6). Values obtained by subtracting the IL-8 concentration in the absence of peptide (Δpg/mL) are shown. * <span class="html-italic">p</span> &lt; 0.05, compared to ΔIL-8 with peptide p176-190 in the absence of kynurenine. (<b>C</b>) TM45 cells were incubated in the presence of irradiated (30 Gy) autologous PBMCs prepulsed with the peptide p176-190, human rIL-2 (50 U/mL), with or without kynurenine. After 7 days, TM45 cells (1 × 10<sup>4</sup>/well) were cultured in 96-well flat-bottom culture plates in the presence of irradiated autologous PBMCs (1 × 10<sup>5</sup>/well) and 5 μM peptide p176-190 using R10H medium. A portion of TM45 cells was maintained for an additional 7 days. Four days later, culture supernatant fluid was collected for an IL-8 ELISA (n = 6). Values obtained by subtracting the IL-8 concentration in the absence of the peptide (Δpg/mL) are shown. Data are expressed as the mean ± standard deviation (SD) and were compared using a one-way analysis of variance and Tukey’s post hoc test. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared to Day 0 or Day 14.</p>
Full article ">Figure 2
<p>(<b>A</b>) An overhead view of the HLA class II-peptide complex from the perspective of the TCR. Peptides rotate 130 degrees counterclockwise with each advancement of one residue. Yellow painted parts of the beta 1 domain indicate polymorphic residues. This is an original illustration. (<b>B</b>) A schematic diagram of the HLA class II-peptide complex viewed from the N-terminus of the binding peptide along the longitudinal axis (displaying only the first 4 residues). This is an original illustration. (<b>C</b>) TM45.2 cells were cultured in 96-well flat-bottom culture plates in the presence of irradiated autologous PBMCs and three types of peptides using R10H medium. “WT” represents the peptide p176-190. L176S refers to peptide p176-190 with a substitution of leucine at position 176 replaced by the neutral hydrophilic serine, while M177S indicates peptide p176-190 with a substitution of methionine at position 177 replaced by the neutral hydrophilic serine. Four days later, culture supernatant fluid was collected for an IFNγ ELISA (n = 6). Data are expressed as the mean ± SD and were compared by a one-way analysis of variance and Tukey’s post hoc test. ** <span class="html-italic">p</span> &lt; 0.01 compared to the culture without peptide.</p>
Full article ">
17 pages, 15851 KiB  
Article
Cross-Species Susceptibility of Emerging Variants of SARS-CoV-2 Spike
by Meng Li, Fei Lv, Zihao Li, Chenyu Zhao, Xiao Wang, Pingfen Zhu and Xuming Zhou
Genes 2024, 15(10), 1321; https://doi.org/10.3390/genes15101321 - 14 Oct 2024
Viewed by 332
Abstract
Background: The continuous evolution of SARS-CoV-2 and the emergence of novel variants with numerous mutations have heightened concerns surrounding the possibility of cross-species transmission and the establishment of natural animal reservoirs for the virus, but the host range of emerging SARS-CoV-2 variants has [...] Read more.
Background: The continuous evolution of SARS-CoV-2 and the emergence of novel variants with numerous mutations have heightened concerns surrounding the possibility of cross-species transmission and the establishment of natural animal reservoirs for the virus, but the host range of emerging SARS-CoV-2 variants has not been fully explored yet. Methods: We employed an in vitro model comprising VSV∆G* pseudotyped viruses bearing SARS-CoV-2 spike proteins to explore the plausible host range of SARS-CoV-2 emerging variants. Results: The overall host tropism of emerging SARS-CoV-2 variants are consistent with that of the SARS-CoV-2 wuhan-hu-1 strain with minor difference. Pseudotyped viruses bearing spike protein from RaTG13 and RmYN02 can enter cell cultures from a broad range of mammalian species, revealing that mink and hamsters may act as potential intermediate hosts. We further investigated 95 potential site-specific mutations in the SARS-CoV-2 spike protein that could impact viral infectivity across different species. The results showed that 13 of these mutations notably increased the transduction rates by more than two-fold when compared to the wild-type spike protein. Further examination of these 13 mutations within cell cultures from 31 different species revealed heightened sensitivity in cells derived from palm civets, minks, and Chinese horseshoe bats to the VSV∆G*-SARS2-S mutants. Specific mutations, such as L24F, R158G, and L212I, were seen to significantly enhance the capacity for SARS-CoV-2 of cross-species transmission. Conclusions: This study offers critical insights for the ongoing surveillance and monitoring efforts of SARS-CoV-2 evolution, emphasizing the need for the vigilant monitoring of specific mutations in both human and animal populations. Full article
(This article belongs to the Section Animal Genetics and Genomics)
Show Figures

Figure 1

Figure 1
<p>Schematic description of in vitro infection model. (<b>A</b>) A comparative analysis of S proteins from SARS-CoV-2 wuhan-hu-1, B.1.617.2 (Delta), XBB, XBB.1.5, XBB.1.16, EG.5.1, BA.2.75, BA.2.86, Bat-CoV RaTG13, and Bat-CoV RmYN02. The short lines with different colors represent amino acid variations: the blue lines represent amino acid substitutions; the white lines (or fragments) represent deletions or gaps; the red or green lines represent one of the substitutions at this position that enhances or decreases the receptor binding affinity. The orange arrows mean mutations at these sites enhance the capacity for cross-species transmission. (<b>B</b>) A schematic description of the in vitro infection model that comprised GFP-encoding VSV pseudotyped viruses and cell cultures derived from multiple animals. (<b>C</b>) The tested species were displayed as their position in the phylogenetic tree (TimeTree, <a href="https://timetree.org/home" target="_blank">https://timetree.org/home</a> (accessed on 9 October 2024)). Primary cell cultures, immortalized cells, and cell lines derived from forty-nine species were tested in this research. while the susceptibility towards SARS-CoV-2 infection was experimentally or naturally tested in 14 different species. High represents the species that are highly susceptible to SARS-CoV-2 infection; medium represents the species that can be occasionally infected by SARS-CoV-2; low represents the species that are rarely infected by SARS-CoV-2.</p>
Full article ">Figure 2
<p>VSV pseudotyped viruses bearing S proteins from different SARS-CoV-2 variants can transduce cell cultures from a broad range of mammalian species. The transduction rates (mean ± SD, n = 3) of cell cultures from human beings, hamsters, palm civets, Rock hyrax, and Asian particolored bat can be efficiently transduced by constructed SARS-CoV-2 pseudotyped viruses. The bars under the black lines represent that these cell cultures were minimally transduced. The pink and red dash lines represent the standard for defining moderate and efficient transduction, respectively.</p>
Full article ">Figure 3
<p>VSV pseudotyped viruses bearing S proteins from Bat-CoV RaTG13 and Bat-CoV RmYN02 can transduce cell cultures from a broad range of mammalian species. The pink and red dash lines represent the standard for defining moderate and efficient transduction, respectively. The bar was presented as mean ± SD (n = 3).</p>
Full article ">Figure 4
<p>The prediction and functional analysis of potential mutations in SARS-CoV-2 wuhan-hu-1 spike protein. (<b>A</b>) The statistics of the mutation rates of each site. The red line means the threshold for mutation rate equals 0.3%; the blue line means the threshold for mutation rate equals 0.1%. (<b>B</b>) 52 sites were chosen based on the mutation rates of each site of spike protein. The most frequent mutation is marked in blue; other mutations are marked in black, while potential mutation are marked in red. Among these sites, mutations at 373, 446, 452, 477, 478, and 496 enhanced the viral entry of SARS-CoV-2. The mutations at 95, 572, and 681 promote spike mediate viral entry of SARS-CoV-2 into primary cells derived from specific wild animals. These mutations are marked in one of the subunits of the spike protein that is marked in blue, and ACE2 proteins are marked in green. (<b>C</b>) The infection of Huh-7 cells by 95 pseudotyped viruses. The data were displayed as mean ± SD (n = 3). The red dot line indicates the two-fold threshold compared to transduction rates of the WT proteins, while the blue dot line indicates the 50% threshold compared to transduction rates of the WT proteins.</p>
Full article ">Figure 5
<p>Infection animal cell cultures by different pseudotyped viruses bearing mutated S protein. Tested species were displayed as their position in the phylogenetic tree (TimeTree, <a href="https://timetree.org/home" target="_blank">https://timetree.org/home</a> (accessed on 9 October 2024)). 34 cell cultures from 31 species were derived from the kidneys, heart, lungs, and other tissues. These species displayed variant susceptibility based on previous research. Cells were grouped by their distribution, their human–animal contacts, and their sensitivity to different pseudotyped viruses. High represents the species that are highly susceptible to SARS-CoV-2 infection; medium represents the species that can be occasionally infected by SARS-CoV-2; low represents the species are rarely infected by SARS-CoV-2, the asterisk (*) represents the species are assessed by in vitro infection analysis. +, ++, +++, and ++++ represent rarely-, occasionally-, medium-, and frequently human-animal contact.</p>
Full article ">
7 pages, 16715 KiB  
Case Report
Kaposi Sarcoma as a Possible Cutaneous Adverse Effect of ChAdOx1 nCov-19 Vaccine: A Case Report
by Yan-Han Li, Yu-Tzu Lin, Shu-Han Chuang and Hui-Ju Yang
Vaccines 2024, 12(10), 1168; https://doi.org/10.3390/vaccines12101168 - 14 Oct 2024
Viewed by 539
Abstract
The COVID-19 pandemic prompted the rapid development of vaccines, including the ChAdOx1 nCov-19 (AstraZeneca) vaccine. While effective, adverse effects have been reported, including cutaneous manifestations. Kaposi sarcoma (KS), a vascular tumor linked to Kaposi sarcoma herpesvirus/human herpesvirus 8 (HHV-8), has seen increased detection [...] Read more.
The COVID-19 pandemic prompted the rapid development of vaccines, including the ChAdOx1 nCov-19 (AstraZeneca) vaccine. While effective, adverse effects have been reported, including cutaneous manifestations. Kaposi sarcoma (KS), a vascular tumor linked to Kaposi sarcoma herpesvirus/human herpesvirus 8 (HHV-8), has seen increased detection during the pandemic. This study reports a case of classic cutaneous KS in a 79-year-old male following the first dose of the ChAdOx1 nCov-19 vaccine, without prior SARS-CoV-2 infection. The patient developed multiple reddish-blue papules on his legs and feet, confirmed as KS through histopathology. Treatment included radiotherapy and sequential chemotherapy with Doxorubicin. The potential reactivation of latent HHV-8 by the vaccine is explored through mechanisms involving the SARS-CoV-2 spike protein and adenovirus vector, which may induce immune responses and inflammatory pathways. Although establishing a direct causal link remains challenging, the case highlights the need for vigilance regarding KS reactivation post-vaccination. Further large-scale studies are warranted to elucidate the relationship between COVID-19 vaccines and latent virus reactivation, ensuring comprehensive safety assessments and informed public health decisions. Full article
Show Figures

Figure 1

Figure 1
<p>Clinical images of Kaposi sarcoma. (<b>A</b>) The patient presented with dark brown macules over the left foot (circled). (<b>B</b>) The patient presented with dark brown macules over the right foot (circled). (<b>C</b>,<b>D</b>) Two larger reddish erythematous papules on his left calf.</p>
Full article ">Figure 2
<p>Representative histopathological images of tumor. (<b>A</b>) Neoplastic cells infiltrating the dermis and subcutaneous fat (H&amp;E, ×40); strong and diffuse positive staining of neoplastic cells for CD31, ×200 (<b>B</b>), SMA, ×200 (<b>C</b>), and HHV-8, ×200 (<b>D</b>), for supporting the diagnosis of Kaposi sarcoma.</p>
Full article ">
15 pages, 2518 KiB  
Article
Molecular Characterization and Genomic Surveillance of SARS-CoV-2 Lineages in Central India
by Purna Dwivedi, Mukul Sharma, Afzal Ansari, Arup Ghosh, Subasa C. Bishwal, Suman Kumar Ray, Manish Katiyar, Subbiah Kombiah, Ashok Kumar, Lalit Sahare, Mahendra Ukey, Pradip V. Barde, Aparup Das and Pushpendra Singh
Viruses 2024, 16(10), 1608; https://doi.org/10.3390/v16101608 (registering DOI) - 14 Oct 2024
Viewed by 459
Abstract
Since the first reported case of COVID-19 in December 2019, several SARS-CoV-2 variants have evolved, and some of them have shown higher transmissibility, becoming the prevalent strains. Genomic epidemiological investigations into strains from different time points, including the early stages of the pandemic, [...] Read more.
Since the first reported case of COVID-19 in December 2019, several SARS-CoV-2 variants have evolved, and some of them have shown higher transmissibility, becoming the prevalent strains. Genomic epidemiological investigations into strains from different time points, including the early stages of the pandemic, are very crucial for understanding the evolution and transmission patterns. Using whole-genome sequences, our study describes the early landscape of SARS-CoV-2 variants in central India retrospectively (including the first known occurrence of SARS-CoV-2 in Madhya Pradesh). We performed amplicon-based whole-genome sequencing of randomly selected SARS-CoV-2 isolates (n = 38) collected between 2020 and 2022 at state level VRDL, ICMR-NIRTH, Jabalpur, from 11899 RT-qPCR-positive samples. We observed the presence of five lineages, namely B.1, B.1.1, B.1.36.8, B.1.195, and B.6, in 19 genomes from the first wave cases and variants of concern (VOCs) lineages, i.e., B.1.617.2 (Delta) and BA.2.10 (Omicron) in the second wave cases. There was a shift in mutational pattern in the spike protein coding region of SRAS-CoV-2 strains from the second wave in contrast to the first wave. In the first wave of infections, we observed variations in the ORF1Ab region, and with the emergence of Delta lineages, the D614G mutation associated with an increase in infectivity became a prominent change. We have identified five immune escape variants in the S gene, P681R, P681H, L452R, Q57H, and N501Y, in the isolates collected during the second wave. Furthermore, these genomes were compared with 2160 complete genome sequences reported from central India that encompass 109 different SARS-CoV-2 lineages. Among them, VOC lineages Delta (28.93%) and Omicron (56.11%) were circulating predominantly in this region. This study provides useful insights into the genetic diversity of SARS-CoV-2 strains over the initial course of the COVID-19 pandemic in central India. Full article
(This article belongs to the Special Issue Molecular Epidemiology of SARS-CoV-2, 3rd Edition)
Show Figures

Figure 1

Figure 1
<p>Phylogenetic analysis of the 38 SARS-CoV-2 genomes sequenced at ICMR-NIRTH with the reference genome (NC_045512) hCoV19/Wuhan/WH01/2019. Lineage distribution is depicted by different colors.</p>
Full article ">Figure 2
<p>Phylogenetic analysis of the 38 SARS-CoV-2 genomes sequenced at ICMR-NIRTH with the 2160 genomes obtained from GISAID. Classification of the genome sequences according to the Pangolin lineages, Nextstrain, and GISAID clade is shown in color. The labels corresponding to the ICMR-NIRTH genome sequences generated during this study are highlighted and have been marked with light yellow color.</p>
Full article ">Figure 3
<p>Global distribution of 5 lineages of SARS-CoV-2 from GISAID database until August 2022. (<b>A</b>) Month-wise distribution of lineages from worldwide submitted data. (<b>B</b>) Distribution of lineages across different continents of the world. (<b>C</b>) Comparison of lineages present in central Indian states with other Indian states and Asian countries.</p>
Full article ">
13 pages, 1200 KiB  
Article
Validation of the Enzyme-Linked ImmunoSpot Analytic Method for the Detection of Human IFN-γ from Peripheral Blood Mononuclear Cells in Response to the SARS-CoV-2 Spike Protein
by Laura E. Carreto-Binaghi, Milton Nieto-Ponce, Andrea Palencia-Reyes, Rodolfo L. Chávez-Domínguez, Jessica Blancas-Zaragoza, Pablo Franco-Mendoza, Montserrat A. García-Ramos, Claudia I. Hernández-Lázaro, Martha Torres and Claudia Carranza
Biomolecules 2024, 14(10), 1286; https://doi.org/10.3390/biom14101286 - 11 Oct 2024
Viewed by 603
Abstract
COVID-19 vaccine evaluations are mainly focused on antibody analyses, but there is growing interest in measuring the cellular immune responses from the researchers evaluating these vaccines. The cellular responses to several COVID-19 vaccines have been studied using the enzyme-linked immunospot (ELISPOT) assay for [...] Read more.
COVID-19 vaccine evaluations are mainly focused on antibody analyses, but there is growing interest in measuring the cellular immune responses from the researchers evaluating these vaccines. The cellular responses to several COVID-19 vaccines have been studied using the enzyme-linked immunospot (ELISPOT) assay for IFN-γ. However, the ELISPOT assay is no longer used only for research purpose and so the performance of this assay must be validated. Since the bioanalytical validation of ELISPOT-IFN-γ is essential for evaluating the method’s effectiveness and establishing confidence in a vaccine’s immunogenicity, the present work validates the ELISPOT-IFN-γ assay’s performance in determining the frequency of IFN-γ-producing cells after stimulation with the SARS-CoV-2 spike protein. The validation was performed in peripheral blood mononuclear cells from volunteers immunized with anti-COVID-19 vaccines. According to the findings, the LOD was 17 SFU and the LLOQ was 22 SFU, which makes the method highly sensitive and suitable for evaluating low levels of cellular responses. The procedure’s accuracy is confirmed by the correlation coefficients for the spike protein and anti-CD3+, being 0.98 and 0.95, respectively. The repeatability and intermediate precision tests were confirmed to be reliable by obtaining a coefficient of variation of ≤25%. The results obtained in this validation enable the assay to be employed for studying antigen-specific cells and evaluating cellular responses to vaccines. Full article
(This article belongs to the Section Biological Factors)
Show Figures

Figure 1

Figure 1
<p>Linearity of the ELISPOT-IFNγ assay. The linearity of the method was evaluated in PBMCs from a single donor following 18 h of incubation with medium, Spike S1 protein, or anti-CD3<sup>+</sup> antibody (positive control). (<b>a</b>) Scanned images of SFUs/well for PBMCs. The 95% confidence intervals (CIs) are shown in (<b>b</b>).</p>
Full article ">Figure 2
<p>IFN-γ LOD and LLOQ characterization. The PBMC of three donors were cultured in OPTmizer medium and analyzed 10 times. Using the average of the results of each volunteer, the LOD of 17 spots/well (dotted gray line) and LLOQ of 22 spots/well (dotted green line) were calculated. Data are shown as the mean of the SFUs of each donor.</p>
Full article ">Figure 3
<p>Repeatability assay of IFN-γ-producing PBMCs. The repeatability of the ELISPOT-IFNγ assay was evaluated by one analyst using PBMCs from three distinct donors. The PBMCs were exposed to Spike protein, PHA, or anti-CD3<sup>+</sup> activating antibody. Data are shown as the mean of the SFUs. The significant differences in the SFUs after PHA or CD3<sup>+</sup> exposure in each donor are indicated with asterisks (**** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">
14 pages, 3551 KiB  
Article
Genome-Wide Identification and Expression Analysis of SNAP Gene Family in Wheat
by Xiaohan Zhang, Yanan Yu, Yumeng Sun, Yan Bai, Yongjun Shu and Changhong Guo
Genes 2024, 15(10), 1311; https://doi.org/10.3390/genes15101311 - 11 Oct 2024
Viewed by 429
Abstract
Background/Objectives: The SNAP gene family is a class of proteins containing a SNAP domain, which plays a crucial role in the growth and development of plants. Methods: Bioinformatics methods were used to systematically analyze the gene structure, phylogenetic evolution, chromosomal distribution, [...] Read more.
Background/Objectives: The SNAP gene family is a class of proteins containing a SNAP domain, which plays a crucial role in the growth and development of plants. Methods: Bioinformatics methods were used to systematically analyze the gene structure, phylogenetic evolution, chromosomal distribution, physicochemical properties, conserved motifs, and cis-acting elements of the TaSNAP family members. Results: The TaSNAP family comprises members that encode proteins ranging between 120 and 276 amino acids, with isoelectric points spanning from 4.87 to 7.92. Phylogenetic analysis elucidated the categorization of the eight TaSNAP into three distinct subfamilies, wherein members of the same subfamily display marked similarities in their gene structures. Chromosomal mapping revealed the distribution of TaSNAP family members across chromosomes 2A, 2B, 2D, 7A, 7B, and 7D. Utilizing the Plant CARE tool, we identified ten elements linked to plant hormones and four associated with stress responses. Expression analysis via qRT-PCR was performed to assess the levels of the eight TaSNAP genes in various tissues and under diverse abiotic stress conditions. The results indicated heightened expression of most genes in roots compared to spikes. Notably, under ABA stress, the majority of genes exhibited upregulation, whereas certain genes were downregulated under PEG stress, implying a substantial role for SNAP protein in wheat growth and development. Conclusions: This study conducted a comprehensive bioinformatics analysis of each member of the wheat SNAP family, laying a crucial foundation for future functional investigations. Full article
(This article belongs to the Special Issue Genes and Genomics of Plants Under Abiotic Stresses)
Show Figures

Figure 1

Figure 1
<p>The unrooted phylogenetic tree of <span class="html-italic">SNAP</span> gene family in wheat and the phylogenetic tree of <span class="html-italic">SNAP</span> proteins in <span class="html-italic">Arabidopsis</span>, wheat, and rice. The NJ tree was constructed using the TaSNAP amino acid sequence with MEGA software (version 11.0.13) and 1000 bootstrap repetitions. Wheat SNAP proteins were divided into three groups (Group I marked with blue solid circles, Group II marked with yellow solid circles, and Group III marked with green solid circles; red solid circles do not contain wheat SNAP proteins, so they were not included in the grouping). The numbers at the nodes are bootstrap values, representing support levels.</p>
Full article ">Figure 2
<p>(<b>A</b>) presents the phylogenetic and gene structure analysis of <span class="html-italic">TaSNAP</span>, while (<b>B</b>) shows the protein structure analysis of <span class="html-italic">TaSNAP</span>. Through the results, the grouping and function of <span class="html-italic">TaSNAP</span> are analyzed. (<b>A</b>): Phylogenetic tree; gene structure; (<b>B</b>): protein structure; (<b>C</b>): conserved domains of B-box genes family in wheat.</p>
Full article ">Figure 2 Cont.
<p>(<b>A</b>) presents the phylogenetic and gene structure analysis of <span class="html-italic">TaSNAP</span>, while (<b>B</b>) shows the protein structure analysis of <span class="html-italic">TaSNAP</span>. Through the results, the grouping and function of <span class="html-italic">TaSNAP</span> are analyzed. (<b>A</b>): Phylogenetic tree; gene structure; (<b>B</b>): protein structure; (<b>C</b>): conserved domains of B-box genes family in wheat.</p>
Full article ">Figure 3
<p>The chromosome location of the <span class="html-italic">SNAP</span> gene family in wheat.</p>
Full article ">Figure 4
<p>Cis-acting element analysis of <span class="html-italic">TaSNAP</span>.</p>
Full article ">Figure 5
<p>Expression profile of <span class="html-italic">TaSNAP</span> gene family in different tissues of wheat. a: root; b: stem; c: leaf; d: spike; e: pollen.</p>
Full article ">Figure 6
<p>Expression profile of <span class="html-italic">SNAP</span> gene family under different treatments in wheat. Note: a: control; b: six hours of 0.2 mol L<sup>−1</sup> NaCl treatment; c: six hours of 100 μmol L<sup>−1</sup> ABA treatment; d: six hours of 20% PEG treatment.</p>
Full article ">
13 pages, 5104 KiB  
Article
Structure and Antigenicity of the Porcine Astrovirus 4 Capsid Spike
by Danielle J. Haley, Sarah Lanning, Kyle E. Henricson, Andre A. Mardirossian, Iyan Cirillo, Michael C. Rahe and Rebecca M. DuBois
Viruses 2024, 16(10), 1596; https://doi.org/10.3390/v16101596 - 11 Oct 2024
Viewed by 409
Abstract
Porcine astrovirus 4 (PoAstV4) has been recently associated with respiratory disease in pigs. In order to understand the scope of PoAstV4 infections and to support the development of a vaccine to combat PoAstV4 disease in pigs, we designed and produced a recombinant PoAstV4 [...] Read more.
Porcine astrovirus 4 (PoAstV4) has been recently associated with respiratory disease in pigs. In order to understand the scope of PoAstV4 infections and to support the development of a vaccine to combat PoAstV4 disease in pigs, we designed and produced a recombinant PoAstV4 capsid spike protein for use as an antigen in serological assays and for potential future use as a vaccine antigen. Structural prediction of the full-length PoAstV4 capsid protein guided the design of the recombinant PoAstV4 capsid spike domain expression plasmid. The recombinant PoAstV4 capsid spike was expressed in Escherichia coli, purified by affinity and size-exclusion chromatography, and its crystal structure was determined at 1.85 Å resolution, enabling structural comparisons to other animal and human astrovirus capsid spike structures. The recombinant PoAstV4 capsid spike protein was also used as an antigen for the successful development of a serological assay to detect PoAstV4 antibodies, demonstrating that the recombinant PoAstV4 capsid spike retains antigenic epitopes found on the native PoAstV4 capsid. These studies lay a foundation for seroprevalence studies and the development of a PoAstV4 vaccine for swine. Full article
Show Figures

Figure 1

Figure 1
<p>Predicting the boundaries of the PoAstV4 capsid spike domain: (<b>A</b>) Phylogenetic analysis of human and animal astroviruses was performed using MEGA X with a MUSCLE (EMBL-EBI) alignment of full-length ORF2 capsid protein sequences and the maximum-likelihood method and a JTT matrix-based model. The tree with the highest log likelihood (−43,729.83) is shown. The PoAstV4 and BoAstV capsid sequences are labeled with their accession number. The PoAstV4 sequences are boxed in cyan and PoAstV1, 2, 3, 5 sequences are boxed in gray. (<b>B</b>) Pairwise amino acid sequence identity between PoAstV4 spike and other astrovirus spikes, generated using MUSCLE (EMBL-EBI) alignment. (<b>C</b>) Top: AlphaFold2 prediction of the full-length PoAstV4 capsid, with the predicted spike domain colored in cyan, and the rest of the sequence in gray. Bottom: Design of the recombinant PoAstV4 spike expression construct showing the predicted residues for the PoAstV4 spike domain.</p>
Full article ">Figure 2
<p>Purification of recombinant PoAstV4 capsid spike protein: (<b>A</b>) SDS-PAGE of affinity purification elution fractions of recombinant PoAstV4 capsid spike protein. Lane 1, BioRad Precision Plus molecular weight markers; Lane 2, final wash; Lane 3–6, purified PoAstV4 spike elutions. (<b>B</b>) Size exclusion chromatography traces on a Superdex 200 column of PoAstV4 spike in cyan and BioRad gel filtration standards in gray.</p>
Full article ">Figure 3
<p>Structure of the porcine astrovirus 4 capsid spike protein: (<b>A</b>) PoAstV4 spike dimer, with individual protomers colored in magenta and cyan. (<b>B</b>) Electron density maps contoured at 1σ around residues 591–599. (<b>C</b>) Overlay of the AlphaFold2 predicted model (dark gray) with the experimentally determined crystal structure. (<b>D</b>) Overlay of the AlphaFold3 [<a href="#B27-viruses-16-01596" class="html-bibr">27</a>] predicted model (light gray) with the crystal structure. Figures were generated in PyMOL.</p>
Full article ">Figure 4
<p>Comparison of the PoAstV4 spike to other astrovirus spikes. Structures are shown as cartoons, with one protomer colored rainbow and the other protomer colored gray. PDB codes are noted. TM alignment scores between the PoAstV4 spike and the respective astrovirus spike are reported below its structure. Figures were generated in PyMOL.</p>
Full article ">Figure 5
<p>Anti-PoAstV4 spike IgG ELISA: (<b>A</b>) ELISA data showing a dose-dependent response towards recombinant PoAstV4 spike antigen in two presumed seropositive (+) pig serum samples. Low-no reactivity was observed in the presumed seronegative (−) pig serum samples from two CDCD piglets. Each sample was measured in triplicate, with the average reported, and error bars represent the standard deviation. (<b>B</b>) A matching negative control ELISA using an ELISA plate not coated with an antigen showed low or no reactivity (&lt;0.197 absorbance at 450 nm), supporting the specificity of the ELISA.</p>
Full article ">
20 pages, 4410 KiB  
Article
Implementation of an Immunoassay Based on the MVA-T7pol-Expression System for Rapid Identification of Immunogenic SARS-CoV-2 Antigens: A Proof-of-Concept Study
by Satendra Kumar, Liangliang Nan, Georgia Kalodimou, Sylvia Jany, Astrid Freudenstein, Christine Brandmüller, Katharina Müller, Philipp Girl, Rosina Ehmann, Wolfgang Guggemos, Michael Seilmaier, Clemens-Martin Wendtner, Asisa Volz, Gerd Sutter, Robert Fux and Alina Tscherne
Int. J. Mol. Sci. 2024, 25(20), 10898; https://doi.org/10.3390/ijms252010898 - 10 Oct 2024
Viewed by 408
Abstract
The emergence of hitherto unknown viral pathogens presents a great challenge for researchers to develop effective therapeutics and vaccines within a short time to avoid an uncontrolled global spread, as seen during the coronavirus disease 2019 (COVID-19) pandemic. Therefore, rapid and simple methods [...] Read more.
The emergence of hitherto unknown viral pathogens presents a great challenge for researchers to develop effective therapeutics and vaccines within a short time to avoid an uncontrolled global spread, as seen during the coronavirus disease 2019 (COVID-19) pandemic. Therefore, rapid and simple methods to identify immunogenic antigens as potential therapeutical targets are urgently needed for a better pandemic preparedness. To address this problem, we chose the well-characterized Modified Vaccinia virus Ankara (MVA)-T7pol expression system to establish a workflow to identify immunogens when a new pathogen emerges, generate candidate vaccines, and test their immunogenicity in an animal model. By using this system, we detected severe acute respiratory syndrome (SARS) coronavirus 2 (SARS-CoV-2) nucleoprotein (N)-, and spike (S)-specific antibodies in COVID-19 patient sera, which is in line with the current literature and our observations from previous immunogenicity studies. Furthermore, we detected antibodies directed against the SARS-CoV-2-membrane (M) and -ORF3a proteins in COVID-19 patient sera and aimed to generate recombinant MVA candidate vaccines expressing either the M or ORF3a protein. When testing our candidate vaccines in a prime-boost immunization regimen in humanized HLA-A2.1-/HLA-DR1-transgenic H-2 class I-/class II-knockout mice, we were able to demonstrate M- and ORF3a-specific cellular and humoral immune responses. Hence, the established workflow using the MVA-T7pol expression system represents a rapid and efficient tool to identify potential immunogenic antigens and provides a basis for future development of candidate vaccines. Full article
(This article belongs to the Special Issue Viral Infection and Virology Methods)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the MVA-T7pol expression system. The T7 polymerase gene was placed under the control of the vaccinia virus early/late promoter p7.5 and was inserted into MVA deletion site II, as descripted previously [<a href="#B29-ijms-25-10898" class="html-bibr">29</a>]. The SARS-CoV-2 gene sequences of N<sub>HA</sub>, E<sub>HA</sub>, M<sub>HA</sub>, ORF3a<sub>HA</sub>, ORF6<sub>HA</sub>, ORF7a<sub>HA</sub>, and ORF8<sub>HA</sub> were inserted into the vector plasmid pOS6 [<a href="#B29-ijms-25-10898" class="html-bibr">29</a>] or pTM3 [<a href="#B34-ijms-25-10898" class="html-bibr">34</a>], and expression was placed under transcriptional control of the T7 promoter. The T7-RNA polymerase, which is expressed by recombinant MVA-T7pol during its replication cycle, allows for a transient expression of the SARS-CoV-2 antigens in the cytoplasm of infected cells that are co-transfected with the plasmids pOS6-N<sub>HA</sub>, pOS6-ORF3a<sub>HA</sub>, pTM3-M<sub>HA</sub>, pOS6-ORF8<sub>HA</sub>, pOS6-ORF7a<sub>HA</sub>, pOS6-E<sub>HA</sub>, or pOS-ORF6<sub>HA</sub>. Of note, the target SARS-CoV-2 gene sequences are not inserted into the MVA-T7pol genome. I–VI: major deletion sites of MVA-T7pol. Created with BioRender.com.</p>
Full article ">Figure 2
<p>Identification of SARS-CoV-2 proteins expressed by the MVA-T7pol system. To detect the targeted SARS-CoV-2 proteins, CEF cells were infected with recombinant MVA-T7pol at an MOI of 10 and transfected with the vector plasmids pOS6 [<a href="#B29-ijms-25-10898" class="html-bibr">29</a>] or pTM3 [<a href="#B34-ijms-25-10898" class="html-bibr">34</a>] containing the encoding sequences of the targeted SARS-CoV-2 proteins that were placed under the T7 promoter. To express SARS-CoV-2 spike protein, CEF cells were infected with recombinant MVA-S<sub>HA</sub> at an MOI of 10. Proteins were separated by SDS-PAGE and analyzed with an antibody directed against the HA-tag (<b>a</b>) or by using human serum (<b>b</b>,<b>c</b>). Non-infected cells (Mock) and cells infected with MVA-T7pol served as controls. Lane 1: N<sub>HA</sub> (47 kDa); lane 2: E<sub>HA</sub> (10 kDa); lane 3: M<sub>HA</sub> (26 kDa); lane 4: S<sub>HA</sub> (190 kDa + 90 kDa) [<a href="#B24-ijms-25-10898" class="html-bibr">24</a>]; lane 5: ORF3a<sub>HA</sub> (32 kDa); lane 6: ORF6<sub>HA</sub> (8 kDa); lane 7: ORF7a<sub>HA</sub> (14 kDa); lane 8: ORF8<sub>HA</sub> (15 kDa); lane 9: MVA-T7pol; lane 10: non-infected cells (Mock). Red arrow: N protein; black arrow: S protein; blue arrow: ORF3a protein; green arrow: M protein.</p>
Full article ">Figure 3
<p>Virological characterization of MVA-SARS-CoV-2-M (MVA-M) and MVA-SARS-CoV-2-ORF3a (MVA-ORF3a). (<b>a</b>,<b>b</b>) Schematic diagram of the MVA genome with the major deletion sites I to VI. (<b>a</b>) The encoding sequence of the full-length SARS-CoV-2 membrane protein (M) was inserted into the vector plasmid pLW73 [<a href="#B45-ijms-25-10898" class="html-bibr">45</a>] (pLW73-M). Expression of SARS-CoV-2-M was controlled by the VACV-specific promoter PmH5 [<a href="#B46-ijms-25-10898" class="html-bibr">46</a>] and was inserted via homologous recombination between MVA DNA sequences (flank-1, flank-2) adjacent to the intergenomic region between the open reading frames (ORF) of the essential viral genes, <span class="html-italic">MVA069R</span> and <span class="html-italic">MVA070L</span>, and copies cloned in the MVA vector plasmid pLW73-M. Repetitive sequences served to remove the marker gene GFP by intergenomic homologous recombination (marker gene deletion) to generate MVA-M. (<b>b</b>) The deletion III site was targeted to insert the gene sequence encoding SARS-CoV-2-ORF3a under the transcriptional control of VACV promotor PmH5 [<a href="#B46-ijms-25-10898" class="html-bibr">46</a>]. Repetitive sequences served to remove the marker gene mCherry by intragenomic homologous recombination (marker gene deletion) to generate MVA-ORF3a. (<b>c</b>,<b>d</b>) Genetic integrity of MVA-M and MVA-ORF3a. PCR analysis of genomic viral DNA confirmed stable insertion of the SARS-CoV-2-M sequence into the intergenomic region between <span class="html-italic">069R</span> and <span class="html-italic">070L</span> of the MVA genome and SARS-CoV-2-ORF3a sequence inserted into the deletion III of the MVA genome. (<b>e</b>,<b>f</b>) Multiple-step growth analysis of recombinant MVA-M, MVA-ORF3a, and non-recombinant MVA (MVA). Recombinant viruses and non-recombinant MVA (MVA) amplified in CEF cells but failed to efficiently grow in human HaCat cells.</p>
Full article ">Figure 4
<p>Synthesis of membrane (M) and ORF3a proteins in MVA-M- and ORF3a-infected cells. (<b>a</b>,<b>b</b>) CEF cells were infected at an MOI of 5, and cell lysates were collected at 0, 4, 8, 24, and 48 h post infection (hpi). Polypeptides in the cell lysates were separated by SDS-PAGE and analyzed with antibodies against the M and ORF3a proteins. (<b>c</b>,<b>d</b>) Vero cells were infected at an MOI of 0.5 with MVA-M or MVA-ORF3a and fixed with paraformaldehyde after 16 hpi. Permeabilized cells were probed with antibodies against the M and ORF3a proteins. Polyclonal goat anti-mouse secondary antibody was used for M-specific fluorescent staining (green), and polyclonal goat anti-rabbit secondary antibody was used for ORF3a-specific fluorescent staining (green). Cell nuclei were counterstained with DAPI (blue). Scale bar: 50 μm.</p>
Full article ">Figure 5
<p>Activation of SARS-CoV-2-M- and SARS-CoV-2-ORF3a-specific CD8 T cells after vaccination with MVA-M and MVA-ORF3a. Groups of <span class="html-italic">HLA-A2.1-/HLA-DR1-transgenic H-2 class I-/class II-knockout</span> mice (n = 6–10) were immunized with 10<sup>7</sup> PFU of MVA-M or MVA-ORF3a via the i.m. route. Mice immunized with non-recombinant MVA (MVA) served as controls. Splenocytes were collected and prepared at day 35 after prime immunization (14 days after booster immunization). Splenocytes were either stimulated with SARS-CoV-2-ORF3a- or SARS-CoV-2-M-specific peptides and were measured by IFN-γ ELISPOT assay (<b>a</b>,<b>e</b>,<b>f</b>) and intracellular cytokines staining (ICS) plus FACS analysis (<b>b</b>–<b>d</b>). (<b>a</b>,<b>e</b>,<b>f</b>) IFN-γ spot-forming colonies (SFC) measured by ELISPOT assay. (<b>b</b>,<b>c</b>) IFN-γ producing CD8 T cells measured by FACS analysis. Graphs show the mean frequency and absolute number of IFN-γ+ CD8 T cells. (<b>d</b>) Cytokine profile of ORF3a<sub>82-90</sub> specific CD8 T cells. Graph shows the mean frequency of IFN-γ<sup>−</sup>TNF-α<sup>+</sup>, IFN-γ<sup>+</sup>TNF-α<sup>+</sup>, and IFN-γ<sup>+</sup>TNF-α<sup>−</sup> cells within the cytokine-positive CD8 T-cell compartment. Bars represent the mean + standard error of the mean (SEM). Differences between group were analyzed by unpaired, two-tailed <span class="html-italic">t</span>-test: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; ns, not significant.</p>
Full article ">Figure 6
<p>Antigen-specific humoral immunity induced by MVA-ORF3a and MVA-M. (<b>a</b>,<b>b</b>) Groups of <span class="html-italic">HLA-A2.1-/HLA-DR1-transgenic H-2 class I-/class II-</span>knockout mice were immunized with 10<sup>7</sup> PFU of MVA-M and MVA-ORF3a via the i.m. route. Mice immunized with non-recombinant MVA (MVA) served as controls. Serum samples were collected 18 days after the prime immunization (prime) and 14 days after the booster immunization (prime-boost). Sera were analyzed for (<b>a</b>) ORF3a- and (<b>b</b>) M-specific IgG by ELISA. Dashed lines represent the limits of detection (LOD). Differences between group were analyzed by unpaired, two-tailed <span class="html-italic">t</span>-test: ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; ns, not significant.</p>
Full article ">
20 pages, 1957 KiB  
Review
Advancements in the Development of Anti-SARS-CoV-2 Therapeutics
by Junjie Huang, Qianqian Ma, Zhengding Su and Xiyao Cheng
Int. J. Mol. Sci. 2024, 25(19), 10820; https://doi.org/10.3390/ijms251910820 - 9 Oct 2024
Viewed by 941
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the virus that causes COVID-19, and so far, it has occurred five noteworthy variants of concern (VOC). SARS-CoV-2 invades cells by contacting its Spike (S) protein to its receptor on the host cell, angiotensin-converting enzyme [...] Read more.
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the virus that causes COVID-19, and so far, it has occurred five noteworthy variants of concern (VOC). SARS-CoV-2 invades cells by contacting its Spike (S) protein to its receptor on the host cell, angiotensin-converting enzyme 2 (ACE2). However, the high frequency of mutations in the S protein has limited the effectiveness of existing drugs against SARS-CoV-2 variants, particularly the Omicron variant. Therefore, it is critical to develop drugs that have highly effective antiviral activity against both SARS-CoV-2 and its variants in the future. This review provides an overview of the mechanism of SARS-CoV-2 infection and the current progress on anti-SARS-CoV-2 drugs. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Structural features of SARS-CoV-2. (<b>A</b>). Virus structure of SARS-CoV-2; (<b>B</b>). Structure of S-protein.</p>
Full article ">Figure 2
<p>Structure of ACE2 (helix-1 in cyan, PBD ID:6M0J).</p>
Full article ">Figure 3
<p>Structure of S protein binding ACE2. From left to right, the number of RBD in the upward conformation increases and binds more ACE2. The S protein is stained green, RBD is stained purple, and ACE2 is stained red. From left to right, PDB ID:7VRW, 7DX7, 7DX8, and 7DX9.</p>
Full article ">
16 pages, 3108 KiB  
Article
An Analysis of Combined Molecular Weight and Hydrophobicity Similarity between the Amino Acid Sequences of Spike Protein Receptor Binding Domains of Betacoronaviruses and Functionally Similar Sequences from Other Virus Families
by Jamie D. Dixson, Lavanya Vumma and Rajeev K. Azad
Microorganisms 2024, 12(10), 2021; https://doi.org/10.3390/microorganisms12102021 - 5 Oct 2024
Viewed by 614
Abstract
Recently, we proposed a new method, based on protein profiles derived from physicochemical dynamic time warping (PCDTW), to functionally/structurally classify coronavirus spike protein receptor binding domains (RBD). Our method, as used herein, uses waveforms derived from two physicochemical properties of amino acids (molecular [...] Read more.
Recently, we proposed a new method, based on protein profiles derived from physicochemical dynamic time warping (PCDTW), to functionally/structurally classify coronavirus spike protein receptor binding domains (RBD). Our method, as used herein, uses waveforms derived from two physicochemical properties of amino acids (molecular weight and hydrophobicity (MWHP)) and is designed to reach into the twilight zone of homology, and therefore, has the potential to reveal structural/functional relationships and potentially homologous relationships over greater evolutionary time spans than standard primary sequence alignment-based techniques. One potential application of our method is inferring deep evolutionary relationships such as those between the RBD of the spike protein of betacoronaviruses and functionally similar proteins found in other families of viruses, a task that is extremely difficult, if not impossible, using standard multiple alignment-based techniques. Here, we applied PCDTW to compare members of four divergent families of viruses to betacoronaviruses in terms of MWHP physicochemical similarity of their RBDs. We hypothesized that some members of the families Arteriviridae, Astroviridae, Reoviridae (both from the genera rotavirus and orthoreovirus considered separately), and Toroviridae would show greater physicochemical similarity to betacoronaviruses in protein regions similar to the RBD of the betacoronavirus spike protein than they do to other members of their respective taxonomic groups. This was confirmed to varying degrees in each of our analyses. Three arteriviruses (the glycoprotein-2 sequences) clustered more closely with ACE2-binding betacoronaviruses than to other arteriviruses, and a clade of 33 toroviruses was found embedded within a clade of non-ACE2-binding betacoronaviruses, indicating potentially shared structure/function of RBDs between betacoronaviruses and members of other virus clades. Full article
Show Figures

Figure 1

Figure 1
<p>Dendrogram of betacoronavirus spike protein RBD sequences and arterivirus GP2 sequences constructed using MWHP PCDTW with Euclidean distance and UPGMA hierarchical clustering. ACE2-binding betacoronavirus sequences are highlighted in red, and non-ACE2-binding betacoronavirus sequences are highlighted in blue. In most cases, the host organism is encoded into the taxa label. In some cases, the host was labeled as “UnkArtV.” In those cases, the Uniprot record should be consulted for additional information concerning the host organism. The three arterivirus sequences that cluster near the ACE2-binding betacoronaviruses have blue text labels and are from Oliver’s Shrew. All black text labels not in a colored box represent Arterivirus GP2 sequences that did not cluster closely with the betacoronavirus sequences.</p>
Full article ">Figure 2
<p>Dendrogram of betacoronavirus spike protein RBD sequences, arterivirus GP2 sequences and torovirus sequences constructed using MWHP PCDTW with Euclidean distance and UPGMA hierarchical clustering. ACE2-binding betacoronavirus sequences are highlighted in red, and non-ACE2-binding betacoronavirus sequences are highlighted in blue. Three Arterivirus GP2 sequences that are unique within this study, in that they cluster very closely with ACE2-binding betacoronavirus sequence, are labeled with blue text. This combined dendrogram underscores the similarity of the three arterivirus GP2 sequences to those of the ACE2-binding betacoronavirus sequences.</p>
Full article ">Figure 3
<p>(<b>A</b>) Pairwise global alignment identity matrix showing identities for all pairwise comparisons of 51 betacoronavirus spike protein RBD sequences and three arterivirus GP2 sequences. (<b>B</b>) Pairwise distance matrix showing MWHP PCDTW distances which have been scaled to 100 for all pairwise comparisons of 51 betacoronavirus spike protein RBD sequences and three arterivirus GP2 (ArtVGP2) sequences. The Pearson Correlation Coefficient for the values in A and B is 0.44 with a <span class="html-italic">p</span>-Value of 4.98 × 10<sup>−140</sup>.</p>
Full article ">Figure 4
<p>Scatterplot of pairwise global alignment identity percentages and MWHP PCDTW distances scaled to 100 for all comparisons made between arterivirus GP2 sequences and betacoronavirus sequences underlying the dendrogram shown in <a href="#microorganisms-12-02021-f001" class="html-fig">Figure 1</a>. The line shown is a polynomial regression line. This shows that, in general terms, the two signals are not different ways of measuring the same signal. In other words, there is additional information in the MWHP PCDTW signal that is not in the identities.</p>
Full article ">Figure 5
<p>Superimposition of the SARS-CoV-2 RBD (6MOJ) and a synthetic construct homology model which has 0% identity to 6MOJ. The synthetic model exhibits an extremely low pruned RMSD value of 0.21 (~96% of residues considered) and also a very low TM-Score indicating that the two structures are nearly identical [<a href="#B15-microorganisms-12-02021" class="html-bibr">15</a>,<a href="#B18-microorganisms-12-02021" class="html-bibr">18</a>].</p>
Full article ">
19 pages, 21418 KiB  
Article
Genetic Transformation of Triticum dicoccum and Triticum aestivum with Genes of Jasmonate Biosynthesis Pathway Affects Growth and Productivity Characteristics
by Dmitry N. Miroshnichenko, Alexey V. Pigolev, Alexander S. Pushin, Valeria V. Alekseeva, Vlada I. Degtyaryova, Evgeny A. Degtyaryov, Irina V. Pronina, Andrej Frolov, Sergey V. Dolgov and Tatyana V. Savchenko
Plants 2024, 13(19), 2781; https://doi.org/10.3390/plants13192781 - 4 Oct 2024
Viewed by 462
Abstract
The transformation protocol based on the dual selection approach (fluorescent protein and herbicide resistance) has been applied here to produce transgenic plants of two cereal species, emmer wheat and bread wheat, with the goal of activating the synthesis of the stress hormone jasmonates [...] Read more.
The transformation protocol based on the dual selection approach (fluorescent protein and herbicide resistance) has been applied here to produce transgenic plants of two cereal species, emmer wheat and bread wheat, with the goal of activating the synthesis of the stress hormone jasmonates by overexpressing ALLENE OXIDE SYNTHASE from Arabidopsis thaliana (AtAOS) and bread wheat (TaAOS) and OXOPHYTODIENOATE REDUCTASE 3 from A. thaliana (AtOPR3) under the strong constitutive promoter (ZmUbi1), either individually or both genes simultaneously. The delivery of the expression cassette encoding AOS was found to affect morphogenesis in both wheat species negatively. The effect of transgene expression on the accumulation of individual jasmonates in hexaploid and tetraploid wheat was observed. Among the introduced genes, overexpression of TaAOS was the most successful in increasing stress-inducible phytohormone levels in transgenic plants, resulting in higher accumulations of JA and JA-Ile in emmer wheat and 12-OPDA in bread wheat. In general, overexpression of AOS, alone or together with AtOPR3, negatively affected leaf lamina length and grain numbers per spike in both wheat species. Double (AtAOS + AtOPR3) transgenic wheat plants were characterized by significantly reduced plant height and seed numbers, especially in emmer wheat, where several primary plants failed to produce seeds. Full article
Show Figures

Figure 1

Figure 1
<p>Emmer wheat plants transformed with <span class="html-italic">AtAOS</span> and <span class="html-italic">AtOPR3</span> genes around the flowering stage as grown in the greenhouse; note developmental differences between the primary T0 plants, RAB4, which is silenced for expression of introduced genes and the RAB2a, RAB5a, and RAB5b plants with a high level of constitutive expression of both the <span class="html-italic">AtAOS</span> and <span class="html-italic">AtOPR3</span> genes.</p>
Full article ">Figure 2
<p>Relative expression levels of <span class="html-italic">AtAOS</span> and <span class="html-italic">AtOPR3</span> in leaves of transgenic emmer (<b>a</b>) and bread (<b>b</b>) wheat lines; T4 homozygous plants, with the exceptions of RAB2 and RAR5, where leaf extracts of T0 plants are analyzed; data are means of at least three biological replicates ± SE; (<b>a</b>,<b>b</b>) expression levels in plants of ‘double’ transgenic lines carrying <span class="html-italic">AtAOS</span> and <span class="html-italic">AtOPR3</span> genes; (<b>c</b>) expression levels of <span class="html-italic">AtAOS</span> gene in transgenic lines of bread wheat Sar-60, for normalization, the relative expression level detected in SAB1 plants (panel (<b>b</b>)) is used.</p>
Full article ">Figure 3
<p>Production of transgenic wheat plants constitutively overexpressing <span class="html-italic">TaAOS</span> gene. (<b>a</b>) Transient <span class="html-italic">RFP</span> gene expression; morphogenic explant 24 h after the delivery of pANIC-<span class="html-italic">TaAOS</span> plasmid to Runo cells; (<b>b</b>) aging and necrosis of Runo wheat tissue with <span class="html-italic">RFP</span> expression; 45 days of in vitro culture; (<b>c</b>) early stage of transgenic somatic embryo formation of emmer wheat Runo, 60 days after bombardment with decreased concentration of herbicide; (<b>d</b>) formation of the RFP-positive single embryo-like structure of Sar-60 surrounded by leafy structures with RFP fluorescence on the medium with decreased herbicide concentration, 80 days after bombardment; (<b>e</b>) segregation of introduced expression cassette in T1 embryos germinated in vitro; 5 days of culture; transgenic line SD3 (<b>f</b>) RFP fluorescence in T2 kernels of homozygous sub-line RD1 in comparison with non-transgenic kernels of emmer wheat Runo. Bright field images are shown on the left side and fluorescent images are shown on the right side.</p>
Full article ">Figure 4
<p>Expression levels of <span class="html-italic">TaAOS</span> gene in leaves of transgenic wheat lines; (<b>a</b>) emmer wheat (cv. Runo) transgenic lines; (<b>b</b>) bread wheat (Sar-60) transgenic lines; data are means of at least five biological replicates ± SE; stars above the graphs indicate statistically significant differences with non-transgenic wheat (* <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, **** <span class="html-italic">p</span> ≤ 0.001).</p>
Full article ">Figure 5
<p>Analysis of leaf length of non-transgenic emmer wheat (cv. Runo) and transgenic plants with overexpression of <span class="html-italic">AtAOS</span> and <span class="html-italic">AtOPR3</span> (RAB1) or <span class="html-italic">TaAOS</span> (RD1 and RD4). Values represent the lengths of 1st, 2nd, 3rd, and 4th leaves measured in 22–25 plants (transgenic lines) or 38 plants (non-transgenic (Runo)) (average ± sd). Stars indicate statistically significant differences calculated according Dunnett’s multiple comparison test: (“****”, <span class="html-italic">p</span> &lt; 0.001), (NS, non-significant).</p>
Full article ">Figure 6
<p>Analysis of leaf length of non-transgenic bread emmer wheat Sar-60 and transgenic lines with overexpression of <span class="html-italic">AtAOS</span> (SA7), <span class="html-italic">TaAOS</span> (SD2, SD3), or <span class="html-italic">AtAOS</span> and <span class="html-italic">AtOPR3</span> simultaneously (SAB1, SAB3). Values represent the lengths of 1st, 2nd, 3rd, and 4th leaves measured in 22–25 plants (average ± sd). Stars indicate statistically significant differences calculated according to Dunnett’s multiple comparisons test (“*”, <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; 0.005), (“****”, <span class="html-italic">p</span> &lt; 0.001), (NS, non-significant).</p>
Full article ">Figure 7
<p>The morphology of transgenic bread wheat lines of plants transformed with <span class="html-italic">AtAOS</span> (SA7), <span class="html-italic">TaAOS</span> (SD2, SD3) and with both <span class="html-italic">AtAOS</span> and <span class="html-italic">AtOPR3</span> (SAB1, SAB3) genes. (<b>a</b>,<b>b</b>) plants are in boot developmental stage; (<b>c</b>,<b>d</b>) plants are in early ripening developmental stage.</p>
Full article ">Figure 8
<p>Average plant height and the productivity of transgenic wheat plants transformed with <span class="html-italic">AtAOS</span> (SA7), <span class="html-italic">TaAOS</span> (RD1, RD4, SD2, SD3), and <span class="html-italic">AtAOS</span> and <span class="html-italic">AtOPR3</span> simultaneously (RAB1, SAB1, SAB3). (<b>a</b>,<b>b</b>), average plant height; (<b>c</b>,<b>d</b>), mean number of seeds per spike; stars indicate statistically significant differences with corresponding non-transgenic wheat cultivar calculated according to Dunnett’s multiple comparisons test (“*”, <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), (ns, not significant).</p>
Full article ">Figure 9
<p>Schematic representation of the pANIC-<span class="html-italic">TaAOS</span> expression cassette used for emmer wheat and bread wheat transformation. <span class="html-italic">OsAct1</span>, rice <span class="html-italic">Actin 1</span> promoter; <span class="html-italic">BAR</span>, BASTA resistance gene (phosphinothricin acetyl transferase); 35ST, CaMV 35S terminator; PvUbi1, <span class="html-italic">Ubiquitin 1</span> promoter from <span class="html-italic">Panicum virgatum</span>; pporRFP, Red Fluorescent Protein gene from <span class="html-italic">Porites porites</span>; <span class="html-italic">NosT</span>, <span class="html-italic">Nopaline Synthase</span> terminator; <span class="html-italic">ZmUbi1</span>, maize <span class="html-italic">Ubiquitin 1</span> promoter; OCS T, octopine synthase terminator sequence; attB1 and attB2—site-specific recombination sequences; <span class="html-italic">Amp<sup>R</sup></span>, ampicillin resistance gene; <span class="html-italic">Kan<sup>R</sup></span>, kanamycin resistance gene. Arrows indicate promoters; regions controlling the expression of <span class="html-italic">TaAOS</span> gene are highlighted in green color.</p>
Full article ">
Back to TopTop