[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

Journals

Article Types

Countries / Regions

Search Results (63)

Search Parameters:
Keywords = MHCI/MHCII

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 7480 KiB  
Article
Design, Development and Immunogenicity Study of a Multi-Epitope Vaccine Prototype Against SARS-CoV-2
by Mariyana Atanasova, Ivan Dimitrov, Nikola Ralchev, Aleksandar Markovski, Iliyan Manoylov, Silviya Bradyanova, Nikolina Mihaylova, Andrey Tchorbanov and Irini Doytchinova
Pharmaceuticals 2024, 17(11), 1498; https://doi.org/10.3390/ph17111498 - 7 Nov 2024
Viewed by 802
Abstract
Objectives: SARS-CoV-2 caused the COVID-19 pandemic, which overwhelmed global healthcare systems. Over 776 million COVID-19 cases and more than 7 million deaths were reported by WHO in September 2024. COVID-19 vaccination is crucial for preventing infection and controlling the pandemic. Here, we describe [...] Read more.
Objectives: SARS-CoV-2 caused the COVID-19 pandemic, which overwhelmed global healthcare systems. Over 776 million COVID-19 cases and more than 7 million deaths were reported by WHO in September 2024. COVID-19 vaccination is crucial for preventing infection and controlling the pandemic. Here, we describe the design and development of a next-generation multi-epitope vaccine for SARS-CoV-2, consisting of T cell epitopes. Methods: Immunoinformatic methods were used to derive models for the selection of MHC binders specific for the mouse strain used in this study among a set of human SARS-CoV-2 T cell epitopes identified in convalescent patients with COVID-19. The immunogenicity of the vaccine prototype was tested on humanized-ACE2 transgenic B6.Cg-Tg(K18-ACE2)2Prlmn/J mice by in vitro, in vivo, and ex vivo immunoassays. Results: Eleven binders (two from the Envelope (E) protein; two from the Membrane (M) protein; three from the Spike (S) protein; and four from the Nucleocapsid (N) protein) were synthesized and included in a multi-epitope vaccine prototype. The animals were immunized with a mix of predicted MHC-I, MHC-II, or MHC-I/MHC-II peptide epitopes in Complete Freund’s Adjuvant, and boosted with peptides in Incomplete Freund’s Adjuvant. Immunization with SARS-CoV-2 epitopes remodeled the lymphocyte profile. A weak humoral response and the significant production of IL-4 and IFN-γ from T cells were found after the vaccination of the animals. Conclusions: The multi-epitope vaccine prototype presented in this study demonstrates immunogenicity in mice and shows potential for human vaccine construction. Full article
Show Figures

Figure 1

Figure 1
<p>SARS-CoV-2 structure and genome.</p>
Full article ">Figure 2
<p>Contributions of the amino acids at the anchor positions 2, 5, and 9 in the peptide binding to H2-Db, according to the PLS model. Peptides with Trp at p5 and with Gln, His, and Trp at p9 are absent in the training set.</p>
Full article ">Figure 3
<p>Contributions of the amino acids at the anchor positions 1, 3, and 5 in the peptide binding to H2-Kb, according to the PLS model.</p>
Full article ">Figure 4
<p>Localization of the epitopes from <a href="#pharmaceuticals-17-01498-t002" class="html-table">Table 2</a> on the structures of SARS-CoV-2 proteins. (<b>A</b>) E protein (PDB: 5 × 29): YSFVSEETG (PDB: 5 × 29 structure contains only the last three amino acids from this epitope) (blue) and TLIVNSVLLFLAF (red). (<b>B</b>) M protein (PDB: 8CTK): LSYYKLGAS (blue) and LSYFIASF (red). (<b>C</b>) N protein (PDB: 8FG2): AQFAPSASAF (red), WYFYYLGTGP (blue), AGLPYGAN (green), and LALLLLDRL (yellow). (<b>D</b>) S protein (PDB: 6XR8): QSYGFQPTNGV (red), IPFAMQMAYRFNGI (blue), and EFRVYSSANNCTFE (green). The proteins E, M, and N are dimers, and the opposite regions contain the same epitopes (not marked). The images were generated by PyMOL Molecular Graphics System, version 2.5.0.</p>
Full article ">Figure 5
<p>Scheme of treatment of transgenic mice expressing the human ACE2 receptor.</p>
Full article ">Figure 6
<p>Flow cytometry analysis for splenocyte phenotyping. (<b>A</b>) Isolated lymphocytes from inguinal lymph nodes from all experimental mice were analyzed with combinations of anti-mouse antibodies, as described in <a href="#sec4-pharmaceuticals-17-01498" class="html-sec">Section 4</a>. The extracted results from all experiments are presented graphically. Results are represented as mean ±SD (<span class="html-italic">n</span> = 5). Data were analyzed by the one-way ANOVA 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). (<b>B</b>) Representative data of 5 experiments are shown.</p>
Full article ">Figure 7
<p>The humoral immune response against viral proteins was analyzed after immunization of B6.Cg-Tg(K18-ACE2)2Prlmn/J mice with SARS-CoV-2 peptide epitopes. Anti-S1, S2, M, N, and E protein IgG antibody levels were determined using ELISA. All samples were triplicated and average values were used for analysis. Summarized results for the calculated fold change obtained from individual mouse sera (<span class="html-italic">n</span> = 5) are presented and mean ± SD values were calculated for each group, in comparison to controls; <span class="html-italic">p</span> values were calculated using the one-way ANOVA test (<span class="html-italic">p</span> &lt; 0.05) in comparison to each group. Representative data of four experiments are shown.</p>
Full article ">Figure 8
<p>Cytokine production in the animals after in vivo SARS-CoV-2 peptide epitope administration. IL-4 and IFN-γ ELISpot assays were used to count the number of IL-4- and IFN-γ-producing T cells from mice treated in vivo with different mixes of peptides (<b>left panels</b>) or ex vivo with additional peptide stimulation (<b>right panels</b>). All samples were triplicated and the data are presented as mean ± SD for each group; p values were calculated using the one-way ANOVA test (* <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01) in comparison to each group. Representative data of four independent experiments are shown.</p>
Full article ">
16 pages, 2154 KiB  
Article
Temporal Changes in Splenic Immune Cell Populations following Infection with a Very Virulent plus MDV in Commercial Meat-Type Chickens
by Nagwa Khaled, Raveendra R. Kulkarni, Tobias Käser and Isabel M. Gimeno
Viruses 2024, 16(7), 1092; https://doi.org/10.3390/v16071092 - 6 Jul 2024
Viewed by 1047
Abstract
Marek’s disease virus (MDV) can cause severe immunosuppression in chickens. Our previous study showed that infection with very virulent plus (vv+) MDV strains of one-day-old commercial meat-type chickens possessing maternal antibodies against MDV resulted in severe depletion of splenocytes at 28–30 days of [...] Read more.
Marek’s disease virus (MDV) can cause severe immunosuppression in chickens. Our previous study showed that infection with very virulent plus (vv+) MDV strains of one-day-old commercial meat-type chickens possessing maternal antibodies against MDV resulted in severe depletion of splenocytes at 28–30 days of age. In the present study, we have investigated the effect of vv+MDV strain 686 on splenic immunophenotypes at 6, 20, and 30 days post-infection (dpi). Both live and dead cells were analyzed, and the data were statistically compared to the uninfected control. The results revealed a decrease in the total live cell population starting on day 20, primarily affecting B cells, CD8β+, and gamma delta (γδ) T cells, while the frequencies of both live and dead CD3+ and CD4+ T cells were increased. The MHC-I expression of CD3+ and CD4+ T cells was higher at 20 and 30 dpi, while the expression of MHC-II on these cells was downregulated at 6 dpi but was upregulated at 30 dpi. Collectively, these results suggest that maternal antibodies seem to delay the negative effects of vv+MDV on the splenic lymphoid populations, albeit being non-protective. Our results emphasize the importance of MD vaccination in vv+MDV endemic areas. Full article
(This article belongs to the Special Issue Marek's Disease Virus)
Show Figures

Figure 1

Figure 1
<p>The effect of 686 infection on frequencies of live cell populations. After 686 infection, single cell suspensions from spleens of uninfected control and 686-infected groups were collected for flow cytometry analysis at three time points: 6 dpi (panel <b>A</b>), 20 dpi (panel <b>B</b>), and 30 dpi (panel <b>C</b>)). Cell viability dye, Live/Dead™ near-infrared (Invitrogen, Carlsbad, CA, USA), was used to exclude dead cells. Frequencies of live cells were analyzed at each time point. Using an unpaired <span class="html-italic">t</span>-test, each data point represents the mean percentage of cells from seven samples of the respective treatment, and the error bars represent the standard deviation. Asterisks above the group averages plotted in the graph indicate statistical significance between the groups; * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>The effect of 686 infection on frequencies of T cell subsets within splenic CD45+ live and dead cell populations. After 686 infection, single cell suspensions from spleens of uninfected control and 686-infected groups were collected at three time points (6, 20, and 30 dpi) for flow cytometry analysis. Cell viability dye, Live/Dead™ near-infrared (Invitrogen, Carlsbad, CA, USA), was used to exclude dead cells. Frequencies of CD3+, TCRαβ+, and TCRγδ+ cells were measured within all CD45+ splenocytes in both live (<b>A</b>–<b>C</b>) and dead (<b>D</b>–<b>F</b>) populations. Using an unpaired <span class="html-italic">t</span>-test, each data point represents the mean percentage of cells from seven samples of the respective treatment, and the error bars represent the standard deviation. Asterisks above the group averages plotted in the graph indicate statistical significance between the groups; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>The effect of 686 infection on frequencies of T cell subsets within splenic CD3+ live and dead cell populations. After 686 infection, single cell suspensions from spleens of uninfected control and 686-infected groups were collected at three time points (6, 20, and 30 dpi) for flow cytometry analysis. Cell viability dye, Live/Dead™ near-infrared (Invitrogen, Carlsbad, CA, USA), was used to exclude dead cells. Frequencies of CD4+, CD8α+, CD8β+, CD4+CD8α-, and CD8α+CD4- T cells were measured within CD3+ splenocytes in both live (<b>A</b>–<b>C</b>) and dead (<b>D</b>–<b>F</b>) populations. Using an unpaired <span class="html-italic">t</span>-test, each data point represents the mean percentage of positive cells from seven samples of the respective treatment, and the error bars represent the standard deviation. Asterisks above the group averages plotted in the graph indicate statistical significance between the groups; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>The effect of 686 infection on frequencies of different TCRγδ+ cell subsets within splenic live and dead cell populations. After 686 infection, single cell suspensions from spleens of uninfected control and 686-infected groups were collected at three time points (6, 20, and 30 dpi) for flow cytometry analysis. Cell viability dye, Live/Dead™ near-infrared (Invitrogen, Carlsbad, CA, USA), was used to exclude dead cells. Frequencies of TCRγδ+ CD4+ and TCRγδ+ CD8β+ cells were measured within TCRγδ+ splenocytes in both live (<b>A</b>–<b>C</b>) and dead (<b>D</b>–<b>F</b>) populations. Using an unpaired <span class="html-italic">t</span>-test, each data point represents the mean percentage of positive cells from seven samples of the respective treatment, and the error bars represent the standard deviation. Asterisks above the group averages plotted in the graph indicate statistical significance between the groups; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>Effect of 686 infection on frequencies of B cells and macrophages within splenic CD45+ live and dead cell populations. After 686 infection, single cell suspensions from spleens of uninfected control and 686-infected groups were collected at two time points (20 and 30 dpi) for flow cytometry analysis. Cell viability dye, Live/Dead™ near-infrared (Invitrogen, Carlsbad, CA, USA), was used to exclude dead cells. Frequencies of B cells and macrophages were measured within CD45+ splenocytes in both live (<b>A</b>,<b>C</b>) and dead (<b>B</b>,<b>D</b>) populations. Using an unpaired <span class="html-italic">t</span>-test, each data point represents the mean percentage of positive cells from seven samples of the respective treatment, and the error bars represent the standard deviation. Asterisks above the group averages plotted in the graph indicate statistical significance between the groups; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
22 pages, 5893 KiB  
Article
Immunoinformatics and Reverse Vaccinology Approach for the Identification of Potential Vaccine Candidates against Vandammella animalimors
by Ahmad Hasan, Wadi B. Alonazi, Muhammad Ibrahim and Li Bin
Microorganisms 2024, 12(7), 1270; https://doi.org/10.3390/microorganisms12071270 - 22 Jun 2024
Cited by 1 | Viewed by 1324
Abstract
Vandammella animalimorsus is a Gram-negative and non-motile bacterium typically transmitted to humans through direct contact with the saliva of infected animals, primarily through biting, scratches, or licks on fractured skin. The absence of a confirmed post-exposure treatment of V. animalimorsus bacterium highlights the [...] Read more.
Vandammella animalimorsus is a Gram-negative and non-motile bacterium typically transmitted to humans through direct contact with the saliva of infected animals, primarily through biting, scratches, or licks on fractured skin. The absence of a confirmed post-exposure treatment of V. animalimorsus bacterium highlights the imperative for developing an effective vaccine. We intended to determine potential vaccine candidates and paradigm a chimeric vaccine against V. animalimorsus by accessible public data analysis of the strain by utilizing reverse vaccinology. By subtractive genomics, five outer membranes were prioritized as potential vaccine candidates out of 2590 proteins. Based on the instability index and transmembrane helices, a multidrug transporter protein with locus ID A0A2A2AHJ4 was designated as a potential candidate for vaccine construct. Sixteen immunodominant epitopes were retrieved by utilizing the Immune Epitope Database. The epitope encodes the strong binding affinity, nonallergenic properties, non-toxicity, high antigenicity scores, and high solubility revealing the more appropriate vaccine construct. By utilizing appropriate linkers and adjuvants alongside a suitable adjuvant molecule, the epitopes were integrated into a chimeric vaccine to enhance immunogenicity, successfully eliciting both adaptive and innate immune responses. Moreover, the promising physicochemical features, the binding confirmation of the vaccine to the major innate immune receptor TLR-4, and molecular dynamics simulations of the designed vaccine have revealed the promising potential of the selected candidate. The integration of computational methods and omics data has demonstrated significant advantages in discovering novel vaccine targets and mitigating vaccine failure rates during clinical trials in recent years. Full article
(This article belongs to the Section Microbial Biotechnology)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of methodology.</p>
Full article ">Figure 2
<p>(<b>A</b>) BepiPred Linear Epitope, (<b>B</b>) Chou &amp; Fasman Beta-Turn Prediction, (<b>C</b>) Emini Surface Accessibility Prediction, (<b>D</b>) Karplus &amp; Schulz Flexibility Prediction, (<b>E</b>) Kolaskar &amp; Tongaonkar Antigenicity, (<b>F</b>) Parker Hydrophilicity Prediction.</p>
Full article ">Figure 3
<p>The final multi-epitope vaccine peptide is represented schematically as a 350-long amino acid sequence. The N-terminal and C-terminal ends of the peptide feature adjuvants (depicted in red), and each adjuvant is connected to the multi-epitope sequence through EAAAK linkers (highlighted in blue).</p>
Full article ">Figure 4
<p>Docked vaccine construct with sbk8. (<b>A</b>) Docked vaccine (red) and sbk8 (purple). (<b>B</b>) Interaction occurs between the vaccine model and the sbk8 protein.</p>
Full article ">Figure 5
<p>(<b>A</b>) Secondary structure predicted through PDB-sum. (<b>B</b>) Ramachandran plot depicting the validation of the structure along with the graph presenting Z-score. (<b>C</b>) I-TASSER (<b>D</b>) Graph of solubility.</p>
Full article ">Figure 6
<p>(<b>A</b>) World population coverage for MHC-I. (<b>B</b>) World population coverage for MHC-II. (<b>C</b>) Combined world population coverage for HLA allele recognized as T-cell epitope.</p>
Full article ">Figure 7
<p>Codon optimization of the vaccine construct Final Vaccine. Here, CAI of the optimized codon and average GC content were 0.94 and 56.09%, respectively.</p>
Full article ">Figure 8
<p>The optimized codons encoding the vaccine protein were in silico cloned into the pET28a (+) vector to facilitate expression in microbial systems. The DNA sequence was inserted into the multiple cloning site of the cloning vector. In the representation, the red portion represents the gene sequence of the designed vaccine construct, while the black portion represents the backbone of the vector. Colored arrows indicate the location and direction of gene expression, with green denoting the kanamycin resistance gene.</p>
Full article ">Figure 9
<p>Host immune system computational modeling against designed vaccination. (<b>A</b>) Graph illustrating the synthesis of immunoglobulins. (<b>B</b>) Graph illustrating interleukins and cytokines’ induction. (<b>C</b>) The tally of plasma B lymphocytes is categorized according to their isotypes (IgM, IgG1, and IgG2). (<b>D</b>) The population of B lymphocytes by entity-state.</p>
Full article ">Figure 10
<p>Molecular dynamics (MD) simulation of the docked MEV complex. (<b>A</b>) deformability; (<b>B</b>) B factor; (<b>C</b>) Coherence Index; (<b>D</b>) elastic network analysis; (<b>E</b>) eigenvalue.</p>
Full article ">
11 pages, 1610 KiB  
Article
Evaluation of Immune Protection of a Bivalent Inactivated Vaccine against Aeromonas salmonicida and Vibrio vulnificus in Turbot
by Yunji Xiu, Jingyuan Yi, Ruixin Feng, Jiaxue Song, Yunfei Pang, Peng Liu and Shun Zhou
Fishes 2024, 9(4), 131; https://doi.org/10.3390/fishes9040131 - 9 Apr 2024
Viewed by 1529
Abstract
The Aeromonas salmonicida is responsible for causing furunculosis in various fish species. Furunculosis is a ubiquitous disease that affects the aquaculture industry and causes the mass mortality of turbot. Vibrio vulnificus is a pathogen that causes skin ulcers and hemorrhagic septicemia in fish, [...] Read more.
The Aeromonas salmonicida is responsible for causing furunculosis in various fish species. Furunculosis is a ubiquitous disease that affects the aquaculture industry and causes the mass mortality of turbot. Vibrio vulnificus is a pathogen that causes skin ulcers and hemorrhagic septicemia in fish, resulting in significant mortality in aquaculture. In this study, we have established a bivalent inactivated vaccine against A. salmonicida and V. vulnificus with Montanide™ ISA 763 AVG as an adjuvant. This bivalent inactivated vaccine was used to immunize turbot by intraperitoneal injection, and the relevant immune indexes were detected. The results demonstrate that the bivalent inactivated vaccine exhibited a relative percent survival (RPS) of 77% following A. salmonicida and V. vulnificus intraperitoneal challenge. The vaccinated group exhibited higher levels of acid phosphatase activity and lysozyme activity compared to the control group. ELISA results showed a significant increase in serum antibody levels in immunized turbot, which was positively correlated with immunity. In the kidney tissue, related immune genes (TLR5, CD4, MHCI and MHCII) were up-regulated significantly, showing that the vaccine can induce cellular and humoral immune responses in turbot. In conclusion, the bivalent inactivated vaccine against A. salmonicida and V. vulnificus was immunogenic, efficiently preventing turbot from infection, which has the potential to be applied in aquaculture. Full article
(This article belongs to the Special Issue Fish Diseases Diagnostics and Prevention in Aquaculture)
Show Figures

Figure 1

Figure 1
<p>Cumulative mortality of vaccinated and control group after challenge with <span class="html-italic">A. salmonicida</span> and <span class="html-italic">V. vulnificus</span>. The <span class="html-italic">x</span>-axis represents the days post-vaccination and the <span class="html-italic">y</span>-axis represents the cumulative mortality rate.</p>
Full article ">Figure 2
<p>ACP activity in serum of turbot after vaccination. The <span class="html-italic">x</span>-axis represents the weeks post-vaccination and the <span class="html-italic">y</span>-axis represents the ACP activity. Data are presented as the means ± SE (N = 3). The asterisk (*) indicates the statistical significance (<span class="html-italic">p</span> &lt; 0.05) between the vaccinated and control groups.</p>
Full article ">Figure 3
<p>Lysozyme activity in serum of turbot after vaccination. The <span class="html-italic">x</span>-axis represents the weeks post-vaccination and the <span class="html-italic">y</span>-axis represents the LZM activity. Data are presented as the means ± SE (N = 3). The asterisk (*) indicates the statistical significance (<span class="html-italic">p</span> &lt; 0.05) between the vaccinated and control groups.</p>
Full article ">Figure 4
<p>Variation trend of serum antibody titer against <span class="html-italic">A. salmonicida</span> (<b>A</b>) and <span class="html-italic">V. vulnificus</span> (<b>B</b>). The <span class="html-italic">x</span>-axis represents the weeks post-vaccination and the <span class="html-italic">y</span>-axis represents the serum antibody titers. The asterisk (*) indicates the statistical significance (<span class="html-italic">p</span> &lt; 0.05) between the vaccinated and control groups.</p>
Full article ">Figure 5
<p>The expression levels of immune-related genes were analyzed by qRT-PCR. (<b>A</b>): <span class="html-italic">TLR5</span> relative expression. (<b>B</b>): <span class="html-italic">CD4</span> relative expression. (<b>C</b>): <span class="html-italic">MHCI</span> relative expression. (<b>D</b>): <span class="html-italic">MHCII</span> relative expression. The kidney was sampled at 0, 1, 2, 3 and 4 weeks post-vaccination. Each bar represents the mean of three biological replicates. The asterisk (*) indicates the statistical significance (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
19 pages, 4552 KiB  
Article
Differential Immune Response Patterns Induced by Anionic and Cationic Lipid Adjuvants in Intranasal Anti-Influenza Immunization
by Anirban Sengupta, Noha Al-Otaibi, Claudia Devito, Francisca Lottersberger and Jorma Hinkula
Vaccines 2024, 12(3), 320; https://doi.org/10.3390/vaccines12030320 - 18 Mar 2024
Viewed by 1938
Abstract
Currently, vaccine development against different respiratory diseases is at its peak. It is of utmost importance to find suitajble adjuvants that can increase the potency of the vaccine candidates. This study aimed to determine the systemic and splenic immune mechanisms in mice models [...] Read more.
Currently, vaccine development against different respiratory diseases is at its peak. It is of utmost importance to find suitajble adjuvants that can increase the potency of the vaccine candidates. This study aimed to determine the systemic and splenic immune mechanisms in mice models induced by anionic and cationic lipid adjuvants in the presence of the vaccine-candidate influenza antigen hemagglutinin (HA). In the presence of the HA antigen, the cationic adjuvant (N3) increased conventional dendritic cell 1 (cDC1) abundance with enhanced MHCI and CD80-CD86 costimulatory marker expression, and significantly higher CD8T and Th17 populations with enhanced interferon-gamma (IFNγ) expression in CD8T and CD4T populations. Conversely, the anionic adjuvant (L3) increased the cDC2 population percentage with significantly higher MHCII and DEC205 expression, along with an increase in the CD4T and regulatory T cell populations. The L3-treated group also exhibited higher percentages of activated B and plasma cell populations with significantly higher antigen-specific IgG and IgA titer and virus neutralization potential. While the anionic adjuvant induced significantly higher humoral responses than the cationic adjuvant, the latter influenced a significantly higher Th1/Th17 response. For customized vaccine development, it is beneficial to have alternative adjuvants that can generate differential immune responses with the same vaccine candidate antigen. This study will aid the selection of adjuvants based on their charges to improve specific immune response arms in the future development of vaccine formulation. Full article
(This article belongs to the Section Vaccines against Tropical and other Infectious Diseases)
Show Figures

Figure 1

Figure 1
<p>Analysis of population percentage and activation of conventional dendritic cell subsets: (<b>A</b>) FSC and SSC plots were used to display the live single-cell populations, and, from those, we gated out the B220 negative populations. They were then gated to determine MHCII+CD11c+ populations. Subsequently, they were gated to determine the CD8a+ and CD11b+ populations to obtain the B220−MHCII+CD11c+CD11b-CD8a+ cDC1 and B220−MHCII+CD11c+CD11b+CD8a− cDC2 population percentages. These two dendritic cell subsets were then individually assessed for CD80+CD86+, MHCI+, and DEC205+ cDC1 and cDC2. Unstained panels from the live single cells are shown in the upper row, which creates the reference gate for the stained samples below. (<b>B</b>) The bar diagram represents the population percentage of splenocytes for cDC1 and cDC2. (<b>C</b>) CD80+CD86+ cDC1 and cDC2 populations are represented in the bar diagram as the percentage of individual cDC1 and cDC2 populations. (<b>D</b>) Mean fluorescence intensity (MFI) of MHCI expression in cDC1 and cDC2 populations is represented here in the bar diagram. (<b>E</b>) DEC205 MFI expression within cDC1 and cDC2. The graphs are representative images derived from at least four independent experiments. Each round of experiments included five mice in each set: (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, ns = non-significant). We followed the same controls, antibodies, machine settings, and gating strategies as our recently published work [<a href="#B44-vaccines-12-00320" class="html-bibr">44</a>].</p>
Full article ">Figure 2
<p>Cytotoxic CD8T cell and CD4T cell population dynamics and activation: (<b>A</b>) CD3+ cells were gated out from the live single-cell splenocyte suspension and were visualized using FSC and SSC plots. In the CD3+ gated population, CD4+ and CD8+ T cells were gated. The FOXP3, RORγT, and IFNγ expressions in CD3+CD4+ cells were analyzed. The CD3+CD8+ T cells were studied for IFNγ+ and CD28+ positive cells and MFI. Unstained gating (the upper row) was considered when drawing the respective gates for the stained samples (lower panel row). (<b>B</b>) Population percentage of CD3+CD8+ T cells out of the total splenocytes studied. (<b>C</b>) The absolute number of CD8T cells is shown for each group, normalized to 10,000 splenocytes. (<b>D</b>) The MFI of the activation and expression of the costimulatory marker of CD28. (<b>E</b>) The population percentage of CD3+CD4+ T cells. The percentage of CD4T cells shown here is on all splenocytes. (<b>F</b>) The absolute number of CD8T cells is shown for each group as normalized to 10,000 splenocytes. The graphs are representative images of at least four independent experiments. Each round of experiments had five mice in each set: (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01). We followed the same controls, antibodies, machine settings, and gating strategies as our recently published work [<a href="#B44-vaccines-12-00320" class="html-bibr">44</a>].</p>
Full article ">Figure 3
<p>Treg and Th17 cell populations and IFNγ+ CD4T and IFNγ+ CD8T cell subsets: (<b>A</b>) The differentiation of CD4T cells into CD3+CD4+FoxP3+RORγT- regulatory T cells was significantly higher in the L3 groups whereas (<b>B</b>) the CD3+CD4+FoxP3-RORγT+ Th17 cell population is higher in N3 groups. (<b>C</b>) The ratio of Treg:Th17 is much higher in the L3 group, whereas a significantly lower ratio in the N3 groups was observed. (<b>D</b>) The MFI level of IFNγ+ expression within the T cell subsets is shown by the bar diagram. (<b>E</b>) The absolute numbers of IFNγ+ CD4T and IFNγ+ CD8T cells are provided in the bar diagram, normalized to 10,000 splenocytes. The graphs are representative images derived from at least four independent experiments. Each round of experiments had five mice in each set. (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01). We followed the same controls, antibodies, machine settings, and gating strategies as our recently published work [<a href="#B44-vaccines-12-00320" class="html-bibr">44</a>].</p>
Full article ">Figure 4
<p>Population percentage of immature/transitional, memory, activated B, and plasma cells: (<b>A</b>) The B220<sup>low</sup> and B220+ cell populations were gated out from live single splenocyte suspensions, keeping unstained samples as a reference for gating. CD19<sup>low/neg</sup> cell populations were gated out from B220<sup>low</sup> populations, and CD19+ cells were gated out from B220+ cells. (<b>B</b>) Within the B220+CD19+ cell populations, CD27+MHCII+ populations were gated, of which CD40+CD80+ populations were determined to obtain the B220/CD45R+CD19+CD27+MHCII+CD40+CD80+ memory B cell population. (<b>C</b>) IgM+IgD+ (thick black box) and IgM<sup>high</sup>+IgD<sup>low/neg</sup> populations (yellow box) were gated on B220+CD19+ populations. From these, B220/CD45R+CD19+IgM<sup>high</sup>+IgD<sup>low/neg</sup>+CD43− immature/transitional B cell and B220/CD45R+CD19+IgM+IgD+MHCII+CD138- activated B cell population were determined. (<b>D</b>) From the B220<sup>low</sup> CD19<sup>low/neg</sup> population, IgM−IgD− cells were determined to finally obtain B220<sup>low</sup> CD19<sup>low/neg</sup> IgM-IgD−CD138+ plasma cells. (<b>E</b>) The bar diagram represents the immature/transitional B and memory B cell population percentages. (<b>F</b>) The bar diagram represents the activated B and plasma cells. The percentages exhibited on the y-axis of 4E and 4F represent the percentage within a total of 10,000 splenocytes analyzed. Hence, to calculate the absolute cell number, readers can use the conversion where 1% on the y-axis = 100 cells. Data in the graphs are the representative images derived from at least four independent experiments (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01). We followed the same controls, antibodies, machine settings, and gating strategies as our recently published work [<a href="#B44-vaccines-12-00320" class="html-bibr">44</a>].</p>
Full article ">Figure 5
<p>Higher expression of serum anti-HA IgG with enhanced neutralization capacity was observed in the N3 group: (<b>A</b>) Anti-HA IgG expression within the serum samples was determined at multiple timepoints post-first and -second dose of immunization by ELISA. (<b>B</b>) The anti-HA IgA titer was determined from the serum samples from each group on a HA-coated plate. (<b>C</b>) A hemagglutination Inhibition assay (HIA) was performed to determine the antibody response in the serum samples. The geometric means of the individual groups of the highest serum dilution capable of neutralizing the virus are plotted here, in log10 dilution factors represented by the <span class="html-italic">y</span>-axis. Titer &gt; 40 was the cutoff for seroprotection titer and is marked on the graph. The graphs are representative images derived from at least four independent experiments. Each round of experiments had five mice in each set: (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">
16 pages, 4044 KiB  
Article
CXCL12 Neutralizing Antibody Promotes Hair Growth in Androgenic Alopecia and Alopecia Areata
by Mei Zheng, Min-Ho Kim, Sang-Gyu Park, Won-Serk Kim, Sang-Ho Oh and Jong-Hyuk Sung
Int. J. Mol. Sci. 2024, 25(3), 1705; https://doi.org/10.3390/ijms25031705 - 30 Jan 2024
Cited by 3 | Viewed by 3444
Abstract
We had previously investigated the expression and functional role of C-X-C Motif Chemokine Ligand 12 (CXCL12) during the hair cycle progression. CXCL12 was highly expressed in stromal cells such as dermal fibroblasts (DFs) and inhibition of CXCL12 increased hair growth. Therefore, we further [...] Read more.
We had previously investigated the expression and functional role of C-X-C Motif Chemokine Ligand 12 (CXCL12) during the hair cycle progression. CXCL12 was highly expressed in stromal cells such as dermal fibroblasts (DFs) and inhibition of CXCL12 increased hair growth. Therefore, we further investigated whether a CXCL12 neutralizing antibody (αCXCL12) is effective for androgenic alopecia (AGA) and alopecia areata (AA) and studied the underlying molecular mechanism for treating these diseases. In the AGA model, CXCL12 is highly expressed in DFs. Subcutaneous (s.c.) injection of αCXCL12 significantly induced hair growth in AGA mice, and treatment with αCXCL12 attenuated the androgen-induced hair damage in hair organ culture. Androgens increased the secretion of CXCL12 from DFs through the androgen receptor (AR). Secreted CXCL12 from DFs increased the expression of the AR and C-X-C Motif Chemokine Receptor 4 (CXCR4) in dermal papilla cells (DPCs), which induced hair loss in AGA. Likewise, CXCL12 expression is increased in AA mice, while s.c. injection of αCXCL12 significantly inhibited hair loss in AA mice and reduced the number of CD8+, MHC-I+, and MHC-II+ cells in the skin. In addition, injection of αCXCL12 also prevented the onset of AA and reduced the number of CD8+ cells. Interferon-γ (IFNγ) treatment increased the secretion of CXCL12 from DFs through the signal transducer and activator of transcription 3 (STAT3) pathway, and αCXCL12 treatment protected the hair follicle from IFNγ in hair organ culture. Collectively, these results indicate that CXCL12 is involved in the progression of AGA and AA and antibody therapy for CXCL12 is promising for hair loss treatment. Full article
(This article belongs to the Special Issue Feature Papers in Molecular Endocrinology and Metabolism)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Neutralization of CXCL12 promotes hair growth in a testosterone-induced AGA model. (<b>A</b>) The back skin of 7-week-old C<sub>3</sub>H male mice was shaved and subcutaneous (s.c.) injections of testosterone propionate (TP) were used to induce the AGA. (<b>B</b>) The expression of CXCL12 in the dorsal skin of control and TP-treated mice was detected using immunofluorescence. CXCL12<sup>+</sup> cells (red) are indicated by white arrows, and DAPI staining (blue) indicates cell nuclei. The scale bar is set at 100 μm. (<b>C</b>) Different doses of CXCL12 neutralizing antibody (αCXCL12, 5, and 20 μg) were subcutaneously injected twice a week for three weeks. A daily topical treatment of 0.05% dutasteride served as a positive control. αCXCL12 administration accelerated hair growth in TP-treated C<sub>3</sub>H mice and the hair weight was measured. ### <span class="html-italic">p</span> &lt; 0.001 vs. control, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 vs. TP-treated, n = 7. (<b>D</b>) αCXCL12 (10, 100, or 1000 ng/mL) treatment increased the length of mouse vibrissa follicles in the AGA mimic ex vivo model. ## <span class="html-italic">p</span> &lt; 0.01 vs. control, * <span class="html-italic">p</span> &lt; 0.05 vs. TP-treated, n = 8. + or -: treated with or without TP, αCXCL12. The asterisk and sharp symbols indicate statistical differences using Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 2
<p>Androgen treatment enhances CXCL12 expression in dermal fibroblasts. (<b>A</b>) Dermal fibroblasts (DFs) were treated with various concentrations of TP (1, 10, and 100 nM) and DHT (1, 10, and 100 nM) for 24 h, and the expression of CXCL12 was assessed using qRT-PCR. <span>$</span> <span class="html-italic">p</span> &lt; 0.05 vs. control. (<b>B</b>) After treating DFs with different concentrations of TP and DHT for 48 h, the culture medium was collected and the secreted CXCL12 levels were quantified using ELISA. <span>$</span> <span class="html-italic">p</span> &lt; 0.05, <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01 vs. control. The dollar sign (<span>$</span>) indicates differences in one-way ANOVA. (<b>C</b>) Immunostaining revealed that the translocation of the AR (green) in DFs increased after TP (100 nM) and DHT (100 nM) treatment for 1 h, as indicated by the white arrows. DAPI staining (blue) marks the cell nuclei. The scale bar is set at 50 μm. (<b>D</b>) After AR-CRISPR/Cas9 knockout (AR-KO) for 48 h, DFs were treated with TP and DHT for an additional 48 h to collect the culture medium for ELISA analysis. AR-KO significantly reduced the secretion of CXCL12 from DFs. Western blot analysis indicated differences in AR expression between the control and AR-KO groups. ### <span class="html-italic">p</span> &lt; 0.001 vs. control, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. TP or DHT treatment. The asterisk and sharp symbols indicate statistical differences using Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 3
<p>CXCL12 secreted from DFs induces AR and CXCR4 in DPCs. The effects of rCXCL12 on the expression of AR and CXCR4 in DPCs were observed using qRT-PCR (<b>A</b>,<b>B</b>) and Western blot analysis (<b>C</b>). rCXCL12 increased the mRNA and protein expression of the AR and CXCR4 in DPCs. <span>$</span> <span class="html-italic">p</span> &lt; 0.05, <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01 vs. Control. (<b>D</b>) DFs were treated with 100 nM TP or DHT for 48 h and the culture medium (CM) was collected. CM from DFs treated with TP and DHT (DFCM<sup>TP</sup> and DFCM<sup>DHT</sup>) significantly reduced hair length in human hair organ culture. <span>$</span> <span class="html-italic">p</span> &lt; 0.05 vs. DFCM, n = 10. The dollar sign (<span>$</span>) indicates differences in a one-way ANOVA.</p>
Full article ">Figure 4
<p>CXCL12 neutralization improves alopecia areata. (<b>A</b>) In this experimental design, skin-draining lymph node (SDLN) cells were isolated from AA-affected C<sub>3</sub>H/HeN female mice and intradermally injected into the dorsal skin of naïve mice to induce AA. Severe hair loss was observed after 12 weeks, and αCXCL12 (20 μg) was subcutaneously injected twice in a 2-week interval. (<b>B</b>) Skin sections from normal and AA mice were stained with an anti-CXCL12 antibody (green). The expression of CXCL12 increased in the AA model. The scale bar is set at 500 μm. (<b>C</b>) The administration of αCXCL12 in AA mice significantly reduced the areas of hair loss, which are marked in the images with dotted lines. The extent of hair loss in both the control and αCXCL12-treated mice was quantified using Image J (v1.53t). *** <span class="html-italic">p</span> &lt; 0.001 vs. control, n = 3. (<b>D</b>) Skin sections of AA and αCXCL12-treated mice were stained with anti-CD8, anti-MHC-I, or anti-MHC-II antibodies. The αCXCL12-treated group had reduced expression of these immune reaction markers (green; white arrows). DAPI staining (blue) indicates cell nuclei. The scale bar is set at 100 μm. An asterisk indicates a statistical difference using Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 5
<p>CXCL12 neutralization prevents alopecia areata onset. (<b>A</b>) In this experimental design, 10-week-old C<sub>3</sub>H female mice were treated with αCXCL12 (20 μg) through subcutaneous injections once a week for 14 weeks following AA-affected SDLN cell transplantation. (<b>B</b>) αCXCL12 treatment significantly delayed the onset of AA, which was marked with dotted lines. (<b>C</b>) The incidence of AA onset in SDLN-transplanted C<sub>3</sub>H mice. Control (n = 10) or αCXCL12 treated (n = 9). (<b>D</b>) Skin sections were stained with an anti-CD8 antibody, and αCXCL12 reduced the expression of CD8<sup>+</sup> cells (green; white arrows). DAPI staining (blue) indicates the cell nuclei. The scale bar is set at 100 μm. <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01 vs. control. The dollar sign (<span>$</span>) indicates differences in one-way ANOVA.</p>
Full article ">Figure 6
<p>Involvement of JAK/STAT signaling in IFNγ-mediated CXCL12 secretion. (<b>A</b>) Dermal fibroblasts (DFs) were treated with IFNγ at various concentrations (1, 10, 30, and 100 ng/mL), and the expression of CXCL12 was significantly increased. <span>$</span> <span class="html-italic">p</span> &lt; 0.05 vs. control. (<b>B</b>) The secreted CXCL12 level was measured using ELISA, and IFNγ significantly increased CXCL12 secretion. <span>$</span><span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.001 vs. control. The dollar sign (<span>$</span>) indicates differences in one-way ANOVA. (<b>C</b>) Immunostaining showed that p-STAT3 (green) in DFs was increased by IFNγ (100 ng/mL), as indicated by the white arrows. DAPI staining (blue) indicates the cell nuclei. The scale bar is set at 100 μm. (<b>D</b>) JAK inhibitor (Baricitinib) and STAT3 inhibitor (Stattic) treatment attenuated the IFNγ-induced CXCL12 secretion from DFs. ## <span class="html-italic">p</span> &lt; 0.01 vs. control, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 vs. IFNγ treated. (<b>E</b>) αCXCL12 treatment increased the length of mouse vibrissae follicles in the AA mimic ex vivo model. # <span class="html-italic">p</span> &lt; 0.05 vs. control * <span class="html-italic">p</span> &lt; 0.05 vs. IFNγ treated, n = 8. Asterisk and sharp indicate statistical differences using Student’s <span class="html-italic">t</span>-test.</p>
Full article ">
18 pages, 4048 KiB  
Article
Live-Attenuated Salmonella-Based Oral Vaccine Candidates Expressing PCV2d Cap and Rep by Novel Expression Plasmids as a Vaccination Strategy for Mucosal and Systemic Immune Responses against PCV2d
by Khristine Kaith Sison Lloren and John Hwa Lee
Vaccines 2023, 11(12), 1777; https://doi.org/10.3390/vaccines11121777 - 28 Nov 2023
Cited by 1 | Viewed by 1731
Abstract
Oral vaccines are highly envisaged for veterinary applications due to their convenience and ability to induce protective mucosal immunity as the first line of defense. The present investigation harnessed live-attenuated Salmonella Typhimurium to orally deliver novel expression vector systems containing the Cap and [...] Read more.
Oral vaccines are highly envisaged for veterinary applications due to their convenience and ability to induce protective mucosal immunity as the first line of defense. The present investigation harnessed live-attenuated Salmonella Typhimurium to orally deliver novel expression vector systems containing the Cap and Rep genes from porcine circovirus type 2 (PCV2), a significant swine pathogen. The antigen expression by the vaccine candidates JOL2885 and JOL2886, comprising eukaryotic pJHL204 and pro-eukaryotic expression pJHL270 plasmids, respectively, was confirmed by Western blot and IFA. We evaluated their immunogenicity and protective efficacy through oral vaccination in a mouse model. This approach elicited both mucosal and systemic immunity against PCV2d. Oral administration of the candidates induced PCV2-specific sIgA, serum IgG antibodies, and neutralizing antibodies, resulting in reduced viral loads in the livers and lungs of PCV2d-challenged mice. T-lymphocyte proliferation and flow-cytometry assays confirmed enhanced cellular immune responses after oral inoculation. The synchronized elicitation of both Th1 and Th2 responses was also confirmed by enhanced expression of TNF-α, IFN-γ, IL-4, MHC-I, and MHC-II. Our findings highlight the effectiveness and safety of the constructs with an engineered-attenuated S. Typhimurium, suggesting its potential application as an oral PCV2 vaccine candidate. Full article
(This article belongs to the Special Issue Advances in Oral Vaccine Development)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the vaccine design, construction, and expression confirmation of the target antigens. (<b>A</b>) Schematic diagram of the eukaryotic expression plasmid (pJHL204) and dual-expression plasmid (pJHL270) illustrating the plasmid map, key components, and strategy for vaccine construction. The Cap and Rep cloned from PCV2d were linked by a self-cleaving peptide, P2A, and were inserted into the MCS region under the CMV promoter for eukaryotic expression in pJHL204 and pJHL270. An optimized sequence of Cap was inserted into the MCS region under the Ptrc promoter for prokaryotic expression in pJHL270. (<b>B</b>) Eukaryotic expression validation of Cap and Rep by the vaccine strains JOL2885 (p204:Cap-Rep) and JOL2886 (p270:EuCap-Rep+ProCap) through immunofluorescence and Western blot detection of Cap and Rep in transfected RAW 264.7 cells using hyperimmune rabbit antisera raised against the target proteins. Green fluorescence indicates positive expression of the respective proteins, and cell nuclei were stained with DAPI as shown by blue fluorophore. (<b>C</b>) Western blot validation of Cap expression by the prokaryotic side of JOL2886 (p270:EuCap-Rep+ProCap).</p>
Full article ">Figure 2
<p>Schematic diagram of <span class="html-italic">Salmonella</span>-based vaccine delivery. (<b>A</b>) Graphical representation of live-attenuated <span class="html-italic">Salmonella</span> with genotype <span class="html-italic">∆lon</span>, <span class="html-italic">∆cpxr</span>, <span class="html-italic">∆sifA</span>, <span class="html-italic">∆asd</span> electroporated with the constructed plasmids for bacterial-based vaccine delivery. (<b>B</b>) Vaccine delivery via intramuscular or oral administration, invasion of macrophages and dendritic cells (Antigen-presenting cells), and further spread through the bloodstream and lymphatics into different organs for induction of immune responses. The figure was created using BioRender online tool (<a href="https://app.biorender.com/" target="_blank">https://app.biorender.com/</a> (accessed on 14 October 2023)).</p>
Full article ">Figure 3
<p>Induction of humoral and mucosal immune responses by JOL2885 (p204:Cap-Rep) and JOL2886 (p270:EuCap-Rep+ProCap) in BALB/c mice. Groups of mice were given two doses of 1 × 10<sup>7</sup> CFU intramuscularly at 2-week intervals, whereas other groups of mice were given four doses of 1 × 10<sup>8</sup> CFU orally at 1-week intervals. (<b>A</b>,<b>B</b>) IgG level was assessed in the serum of immunized mice at 14 and 42 days after primary immunization by ELISA with Cap or Rep purified proteins as capture antigens. (<b>C</b>) IgG1 and IgG2a subclass were assessed in the serum of immunized mice at 42 days post-immunization using Cap purified protein as capture antigen. (<b>D</b>) The secretory IgA (sIgA) level in the lung homogenate and intestinal wash was assessed by ELISA using Cap purified protein as capture antigen. (<b>E</b>) Neutralizing antibody titer (NAb) at day 42 after primary immunization was quantified by serum neutralization (SN) test and analyzed by end-point dilution reduction assay. The SEM is denoted by the error bars. The data were analyzed by two-way ANOVA. <sup>ns</sup> <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, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Cell-mediated immune responses by JOL2885 (p204:Cap-Rep) and JOL2886 (p270:EuCap-Rep+ProCap) in immunized BALB/c mice. Three weeks post-immunization, collected splenocytes were stimulated with purified Cap and Rep proteins for 48 h. (<b>A</b>) Percentages of CD4<sup>+</sup> and CD8<sup>+</sup> T cells presented in a bar diagram as analyzed by flow cytometry. (<b>B</b>) Bar diagram showing splenocyte proliferation index after stimulation with Cap and Rep proteins. (<b>C</b>) Changes in the cytokine expression profile of IFN-γ, TNF-α, and IL-4 in orally immunized mice were determined by qPCR. (<b>D</b>) Bar diagram showing the percentage of MHCI and MHCII molecules. The data were analyzed by two-way ANOVA. <sup>ns</sup> <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, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5
<p>Viral load in the organs of vaccinated mice after PCV2d challenge. Groups of mice were immunized with 2 doses of 1 × 10<sup>7</sup> CFU intramuscularly at 2-week intervals or with 4 doses of 1 × 10<sup>8</sup> CFU orally at 1-week intervals and then challenged with PCV2d intraperitoneally. (<b>A</b>) PCV2 Cap genomic copies in the liver and lungs of vaccinated mice 21 days after PCV2d challenge. (<b>B</b>) Representative images of immunohistochemical detection of PCV2 in liver, lungs, and spleen of PCV2d-challenged mice. Positive immunolabeling for PCV2 antigen was indicated by a dark brown signal (yellow arrow). All images were taken at 400× magnification. The bars represent the mean values and the SEM is denoted by the error bars. The data were analyzed by two-way ANOVA.</p>
Full article ">
11 pages, 2164 KiB  
Article
An Avirulent Largemouth Bass Birnavirus Vaccine Candidate Protects Largemouth Bass against Birnavirus Infection
by Xiaozhe Fu, Mingju Luo, Qiang Lin, Hongru Liang, Yinjie Niu, Xia Luo, Baofu Ma and Ningqiu Li
Vaccines 2023, 11(12), 1740; https://doi.org/10.3390/vaccines11121740 - 22 Nov 2023
Cited by 1 | Viewed by 1126
Abstract
Background: Largemouth bass birnavirus (LBBV) disease outbreaks in largemouth bass fingerlings lead to high mortality in China. Therefore, the development of immersion immunization strategies is paramount. Methods: An avirulent LBBV strain was screened using a fish challenge assay. The proliferation dynamics of the [...] Read more.
Background: Largemouth bass birnavirus (LBBV) disease outbreaks in largemouth bass fingerlings lead to high mortality in China. Therefore, the development of immersion immunization strategies is paramount. Methods: An avirulent LBBV strain was screened using a fish challenge assay. The proliferation dynamics of the avirulent strain were determined in vitro and in vivo. The efficacy of the avirulent vaccine was evaluated using immune gene expression, viral load, and a virus challenge, and the safety was also assessed using a reversion to virulence test. Results: An avirulent virus strain, designated as largemouth bass birnavirus Guangdong Sanshui (LBBV-GDSS-20180701), was selected from five fish birnavirus isolates. The proliferation peak titer was 109.01 TCID50/mL at 24 hpi in CPB cells and the peak viral load was 2.5 × 104 copies/mg at 4 dpi in the head kidneys and spleens of largemouth bass. The largemouth bass that were immersed within an avirulent vaccine or injected with an inactivated vaccine were protected from the virulent LBBV challenge with a relative percent survival (RPS) of 75% or 42.9%, respectively. The expression levels of IL-12, MHCI, MHCII, CD8, CD4, and IgM in the avirulent group were significantly upregulated at a partial time point compared to the inactivated vaccine group. Moreover, the viral load in the avirulent vaccine group was significantly lower than those in the inactivated vaccine group and control group using real-time PCR. Conclusions: LBBV-GDSS-20180701 is a potential live vaccine candidate against LBBV disease. Full article
Show Figures

Figure 1

Figure 1
<p>Cumulative mortality curves for different experimental groups injected with different LBBV strains. (<b>a</b>) At a dose of 5 × 10<sup>4</sup> TCID<sub>50</sub> per fish. (<b>b</b>) At a dose of 5 × 10<sup>6</sup> TCID<sub>50</sub> per fish.</p>
Full article ">Figure 2
<p>Proliferation dynamics of avirulent strain in CPB cells (<b>a</b>) and in largemouth bass (<b>b</b>).</p>
Full article ">Figure 3
<p>qRT-PCR analysis of the immune-related gene expressions in different vaccine groups. Total RNAs were extracted from the spleen tissues on 0 d, 1 d, 2 d, 3 d, 4 d, 5 d, 6 d, and 7 d for detection of IL-12, MHCI, MHCII, CD8 and CD4, and from the head kidney tissues on 0 d, 7 d, 14 d, and 21 d for IgM detection post vaccination for use in qRT-PCR. * Significant differences from the inactivated group. Data are presented as means ± SE (N = 3), <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Immune protection of different vaccines against LBBV challenge. (<b>a</b>) Cumulative mortality curves for different groups after challenge with virulent LBBV. (<b>b</b>) Relative percent survival of different vaccine groups.</p>
Full article ">Figure 5
<p>Detection of LBBV loads in largemouth bass from different groups. Fish from avirulent, inactivated, immersion control, and injection control groups at 1, 2, and 3 days post challenge were collected. Then, head kidneys and spleens were sampled immediately for LBBV load experiment. The viral copy number per gram of different tissues was determined by real-time PCR. <sup>a</sup> represents that avirulent vaccine group was significantly different from immersion control group (<span class="html-italic">p</span> &lt; 0.05). <sup>b</sup> represents avirulent vaccine group was significantly different from inactivated vaccine group. <sup>c</sup> represents inactivated vaccine group was significantly different from injection control group.</p>
Full article ">Figure 6
<p>RT-PCR detection of avirulent vaccine strain in largemouth bass after serial propagations. M: DNA marker (DL2000); −: negative control; 1, 2, 3, 4, and 5: samples from 5 serial passages in largemouth bass vaccinated with avirulent strain. The size of amplified fragment is 339 bp.</p>
Full article ">
21 pages, 4105 KiB  
Article
Immunoinformatic Execution and Design of an Anti-Epstein–Barr Virus Vaccine with Multiple Epitopes Triggering Innate and Adaptive Immune Responses
by Naveed Ahmed, Ali A. Rabaan, Ameen S. S. Alwashmi, Hawra Albayat, Mutaib M. Mashraqi, Ahmad A. Alshehri, Mohammed Garout, Wesam A. Abduljabbar, Nik Yusnoraini Yusof and Chan Yean Yean
Microorganisms 2023, 11(10), 2448; https://doi.org/10.3390/microorganisms11102448 - 29 Sep 2023
Cited by 2 | Viewed by 1500
Abstract
One of the most important breakthroughs in healthcare is the development of vaccines. The life cycle and its gene expression in the numerous virus-associated disorders must be considered when choosing the target vaccine antigen for Epstein–Barr virus (EBV). The vaccine candidate used in [...] Read more.
One of the most important breakthroughs in healthcare is the development of vaccines. The life cycle and its gene expression in the numerous virus-associated disorders must be considered when choosing the target vaccine antigen for Epstein–Barr virus (EBV). The vaccine candidate used in the current study will also be effective against all other herpesvirus strains, based on the conservancy study, which verified that the protein is present in all herpesviruses. From the screening, two B-cell epitopes, four MHC-I, and five MHC-II restricted epitopes were chosen for further study. The refined epitopes indicated 70.59% coverage of the population in Malaysia and 93.98% worldwide. After removing the one toxin (PADRE) from the original vaccine design, it was projected that the new vaccine would not be similar to the human host and would instead be antigenic, immunogenic, non-allergenic, and non-toxic. The vaccine construct was stable, thermostable, soluble, and hydrophilic. The immunological simulation projected that the vaccine candidate would be subject to a long-lasting active adaptive response and a short-lived active innate response. With IgM concentrations of up to 450 cells per mm3 and active B-cell concentrations of up to 400 cells per mm3, the B-cells remain active for a considerable time. The construct also discovered other conformational epitopes, improving its ability to stimulate an immune response. This suggests that, upon injection, the epitope will target the B-cell surface receptors and elicit a potent immune response. Furthermore, the discotope analysis confirmed that our conformational B-cell epitope was not displaced during the design. Lastly, the docking complex was stable and exhibited little deformability under heat pressure. These computational results are very encouraging for future testing of our proposed vaccine, which may potentially help in the management and prevention of EBV infections worldwide. Full article
(This article belongs to the Special Issue 10th Anniversary of Microorganisms: Past, Present and Future)
Show Figures

Figure 1

Figure 1
<p>Graphical representation of the immunogenic epitopes. (<b>A</b>) Hydrophilicity; (<b>B</b>) linear epitope prediction. The green residues in all these graphs represent the non-epitope residues, while the ones in yellow represent the potential epitopes. (<b>C</b>) Discotope prediction of the conformational and discontinuous epitopes of the protein. The residues in green represent discontinuous but conformational epitopes; (<b>D</b>) Ellipro illustration of the first B-cell epitope, residues 699–733, as conformational and surface accessible; (<b>E</b>) Discotope 2D validating the Ellipro prediction; (<b>F</b>) 3D representation of all the conformational epitopes of the protein. The residues on the plots encircled in red represent the two finalized epitopes.</p>
Full article ">Figure 2
<p>(<b>A</b>) Malaysian population coverage of the vaccine candidate; (<b>B</b>) world population coverage of the vaccine candidate; (<b>C</b>) the vaccine construct, where 1 represents EAAAK, 2 represents GSSS, and 3 represents HEYGAEALERAG linkers. Red line: minimum number of epitope hits / HLA combinations recognized by 90% of the population.</p>
Full article ">Figure 3
<p>Modeling and physicochemical analysis of the construct; (<b>A</b>) the original and refined models superimposed on each other to visualize the minor structural difference; (<b>B</b>) protein solubility compared to the average solubility of proteins in <span class="html-italic">E. coli</span> using Protein sol; (<b>C</b>) localization of the vaccine construct; (<b>D</b>) disorder plot of the construct; (<b>E</b>) cartoon of the predicted secondary structure of the vaccine candidate.</p>
Full article ">Figure 4
<p>(<b>A</b>) MEMSTAT of the protein translating the localization shown in <a href="#microorganisms-11-02448-f004" class="html-fig">Figure 4</a>; (<b>B</b>) vaccine candidate solubility predicted by PSIPRED.</p>
Full article ">Figure 5
<p>Immune simulation analysis. (<b>A</b>) B-cell population; (<b>B</b>) B-cell population per state; (<b>C</b>) TH (Helper-T) cell population; (<b>D</b>) TC (Cytotoxic-T) cell population.</p>
Full article ">Figure 6
<p>(<b>A</b>) The conformational B-cell epitope predicted earlier is conserved in the final vaccine construct; (<b>B</b>) the 3D structure of TLR-2 (protein code: O60603) obtained from UniProt KB; (<b>C</b>) the docked complex, where the purple-colored chains are the non-interacting chains and the ones in pink have been predicted to interact with the vaccine candidate, whereas the vaccine candidate is shown in yellow color; (<b>D</b>–<b>L</b>) some of the molecular interactions showing bond length and AA residue involved, where the blue residues belong to the vaccine candidate and the pink/green residues belong to TLR-2.</p>
Full article ">Figure 7
<p>The pET28 (+) plasmid with the insert.</p>
Full article ">
26 pages, 2639 KiB  
Review
Insight into Cancer Immunity: MHCs, Immune Cells and Commensal Microbiota
by Minting Wen, Yingjing Li, Xiaonan Qin, Bing Qin and Qiong Wang
Cells 2023, 12(14), 1882; https://doi.org/10.3390/cells12141882 - 18 Jul 2023
Cited by 4 | Viewed by 2332
Abstract
Cancer cells circumvent immune surveillance via diverse strategies. In accordance, a large number of complex studies of the immune system focusing on tumor cell recognition have revealed new insights and strategies developed, largely through major histocompatibility complexes (MHCs). As one of them, tumor-specific [...] Read more.
Cancer cells circumvent immune surveillance via diverse strategies. In accordance, a large number of complex studies of the immune system focusing on tumor cell recognition have revealed new insights and strategies developed, largely through major histocompatibility complexes (MHCs). As one of them, tumor-specific MHC-II expression (tsMHC-II) can facilitate immune surveillance to detect tumor antigens, and thereby has been used in immunotherapy, including superior cancer prognosis, clinical sensitivity to immune checkpoint inhibition (ICI) therapy and tumor-bearing rejection in mice. NK cells play a unique role in enhancing innate immune responses, accounting for part of the response including immunosurveillance and immunoregulation. NK cells are also capable of initiating the response of the adaptive immune system to cancer immunotherapy independent of cytotoxic T cells, clearly demonstrating a link between NK cell function and the efficacy of cancer immunotherapies. Eosinophils were shown to feature pleiotropic activities against a variety of solid tumor types, including direct interactions with tumor cells, and accessorily affect immunotherapeutic response through intricating cross-talk with lymphocytes. Additionally, microbial sequencing and reconstitution revealed that commensal microbiota might be involved in the modulation of cancer progression, including positive and negative regulatory bacteria. They may play functional roles in not only mucosal modulation, but also systemic immune responses. Here, we present a panorama of the cancer immune network mediated by MHCI/II molecules, immune cells and commensal microbiota and a discussion of prospective relevant intervening mechanisms involved in cancer immunotherapies. Full article
Show Figures

Figure 1

Figure 1
<p>Illustration of MHC-I deficiency and immune evasion.</p>
Full article ">Figure 2
<p>Illustration of tsMHC-II’s role in tumor immune surveillance.</p>
Full article ">Figure 3
<p>Summary of NK cells’ function in anti-cancer immunity.</p>
Full article ">Figure 4
<p>Summary of the interaction between γδ T cells and cancer cells.</p>
Full article ">Figure 5
<p>Illustration of eosinophils’ role in tumor microenvironment.</p>
Full article ">Figure 6
<p>Summary of the influence of commensal microbiota in cancer immunity.</p>
Full article ">
30 pages, 2906 KiB  
Article
High-Resolution Genotyping of Expressed Equine MHC Reveals a Highly Complex MHC Structure
by Deepali Vasoya, Thomas Tzelos, Lindert Benedictus, Anna Eleonora Karagianni, Scott Pirie, Celia Marr, Charlotta Oddsdóttir, Constanze Fintl and Timothy Connelley
Genes 2023, 14(7), 1422; https://doi.org/10.3390/genes14071422 - 10 Jul 2023
Cited by 1 | Viewed by 2676
Abstract
The Major Histocompatibility Complex (MHC) genes play a key role in a number of biological processes, most notably in immunological responses. The MHCI and MHCII genes incorporate a complex set of highly polymorphic and polygenic series of genes, which, due to the technical [...] Read more.
The Major Histocompatibility Complex (MHC) genes play a key role in a number of biological processes, most notably in immunological responses. The MHCI and MHCII genes incorporate a complex set of highly polymorphic and polygenic series of genes, which, due to the technical limitations of previously available technologies, have only been partially characterized in non-model but economically important species such as the horse. The advent of high-throughput sequencing platforms has provided new opportunities to develop methods to generate high-resolution sequencing data on a large scale and apply them to the analysis of complex gene sets such as the MHC. In this study, we developed and applied a MiSeq-based approach for the combined analysis of the expressed MHCI and MHCII repertoires in cohorts of Thoroughbred, Icelandic, and Norwegian Fjord Horses. The approach enabled us to generate comprehensive MHCI/II data for all of the individuals (n = 168) included in the study, identifying 152 and 117 novel MHCI and MHCII sequences, respectively. There was limited overlap in MHCI and MHCII haplotypes between the Thoroughbred and the Icelandic/Norwegian Fjord horses, showcasing the variation in MHC repertoire between genetically divergent breeds, and it can be inferred that there is much more MHC diversity in the global horse population. This study provided novel insights into the structure of the expressed equine MHC repertoire and highlighted unique features of the MHC in horses. Full article
(This article belongs to the Section Animal Genetics and Genomics)
Show Figures

Figure 1

Figure 1
<p><b>Schematic workflow of equine MHC genotyping using a MiSeq High-throughput sequencing approach.</b> An overview of the bespoke bioinformatics pipeline used to analyze the equine MHCI and MHCII data generated using a MiSeq platform, starting with the raw MiSeq data to a final set of MHC alleles and haplotypes.</p>
Full article ">Figure 2
<p><b>Read frequency of the MHCI alleles identified in the cohort of Thoroughbred horses.</b> For each allele (horizontal axis), the percentage of sequencing reads (vertical axis) at which it was identified in the For1/Rev2.2 and For 3.2/Rev1 amplicons in each individual is represented by a blue and red colored dot, as described in the legend. The percentage of sequencing reads is shown on a logarithmic scale. The 0.2% cut-off threshold is shown as a horizontal bar.</p>
Full article ">Figure 3
<p><b>Read frequency of the (i) DQA, (ii) DQB, and (ii) DRB loci in the Thoroughbred cohort.</b> The relative frequencies of reads representing alleles for DQA1/DQA2, DQB1/DQB2, and DRB1/DRB2/DRB3 loci are shown. For each gene, the horizontal central line in the box plot represents the median value, the box represents the inter-quartile range, and the vertical lines extending from the box represent 1.5x the inter-quartile range. Individual outliers are represented by dots.</p>
Full article ">Figure 4
<p>Association between expressed MHCI and MHCII haplotypes in the (<b>A</b>) Thoroughbred and (<b>B</b>) Icelandic Norwegian Fjord Horse cohorts. In the alluvial plot, the width of the lines linking MHCI and MHCII haplotypes represents the frequency at which these MHCI/MHCII are observed to co-segregate in the cohort.</p>
Full article ">Figure 5
<p><b>Read frequency of the MHCI alleles identified in the cohort of Icelandic and Norwegian Fjord Horses.</b> For each allele (horizontal axis), the percentage of sequencing reads (vertical axis) at which it was identified in the For1/Rev2.2 and For 3.2/Rev1 amplicons in each individual is represented by a blue and red colored dot as described in the legend. The percentage of sequencing reads is shown on a logarithmic scale. The 0.2% cut-off threshold is shown as a horizontal bar.</p>
Full article ">
11 pages, 1404 KiB  
Article
Evidence of the Autophagic Process during the Fish Immune Response of Skeletal Muscle Cells against Piscirickettsia salmonis
by Cristián A. Valenzuela, Marco Azúa, Claudio A. Álvarez, Paulina Schmitt, Nicolás Ojeda and Luis Mercado
Animals 2023, 13(5), 880; https://doi.org/10.3390/ani13050880 - 28 Feb 2023
Cited by 4 | Viewed by 2219
Abstract
Autophagy is a fundamental cellular process implicated in the health of the cell, acting as a cytoplasmatic quality control machinery by self-eating unfunctional organelles and protein aggregates. In mammals, autophagy can participate in the clearance of intracellular pathogens from the cell, and the [...] Read more.
Autophagy is a fundamental cellular process implicated in the health of the cell, acting as a cytoplasmatic quality control machinery by self-eating unfunctional organelles and protein aggregates. In mammals, autophagy can participate in the clearance of intracellular pathogens from the cell, and the activity of the toll-like receptors mediates its activation. However, in fish, the modulation of autophagy by these receptors in the muscle is unknown. This study describes and characterizes autophagic modulation during the immune response of fish muscle cells after a challenge with intracellular pathogen Piscirickettsia salmonis. For this, primary cultures of muscle cells were challenged with P. salmonis, and the expressions of immune markers il-1β, tnfα, il-8, hepcidin, tlr3, tlr9, mhc-I and mhc-II were analyzed through RT-qPCR. The expressions of several genes involved in autophagy (becn1, atg9, atg5, atg12, lc3, gabarap and atg4) were also evaluated with RT-qPCR to understand the autophagic modulation during an immune response. In addition, LC3-II protein content was measured via Western blot. The challenge of trout muscle cells with P. salmonis triggered a concomitant immune response to the activation of the autophagic process, suggesting a close relationship between these two processes. Full article
(This article belongs to the Section Aquatic Animals)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Expression of immune molecular markers in rainbow-trout muscle cells after <span class="html-italic">Piscirickettsia salmonis</span> challenge. Primary cultures of fish skeletal muscle were challenge with <span class="html-italic">P. salmonis</span> (MOI 10) for 6, 8, and 24 h. As control group, cells were treated with a vehicle (DMEM + 10% FBS). (<b>A</b>) Toll-like receptors 3 and 9. (<b>B</b>) Proinflammatory cytokines <span class="html-italic">il-1β</span> and <span class="html-italic">tnfα</span>. (<b>C</b>) <span class="html-italic">il-8</span> and the antimicrobial peptide <span class="html-italic">hepcidin</span>. (<b>D</b>) Major complex of histocompatibility I and II (<span class="html-italic">mhc-I</span> and <span class="html-italic">mhc-II</span>). qPCR analyses were normalized with the <span class="html-italic">ef-1α</span> and <span class="html-italic">β-actin</span> genes, and the results are presented as means ± SEMs of triplicates from three independent experiments, using fold change compared to the control values (n = 3). The difference in the transcript levels is denoted by asterisks (*) considering a <span class="html-italic">p</span>-value &lt; 0.05 (*), <span class="html-italic">p</span>-value &lt; 0,01 (**) and <span class="html-italic">p</span>-value &lt; 0.001 (****) significant.</p>
Full article ">Figure 2
<p>Effect of <span class="html-italic">P. salmonis</span> challenge on the expression of autophagic genes in rainbow-trout muscle cells. Primary cultures of fish skeletal muscle were infected with <span class="html-italic">P. salmonis</span> at MOI 10 for 6, 8, and 24 h. As the control, cells without bacteria were sampled at the beginning of the trial (time 0). (<b>A</b>) Autophagic genes <span class="html-italic">becn1</span> and <span class="html-italic">atg9</span> involved in the induction of this process. (<b>B</b>) Autophagosome formation genes <span class="html-italic">atg12</span> and <span class="html-italic">atg5</span>. (<b>C</b>) Gene <span class="html-italic">atg4,</span> involved in the elongation of the autophagosome. (<b>D</b>) Vesicle-completion-associated genes <span class="html-italic">gabarap</span> and <span class="html-italic">lc3</span>. The data are presented as means ± SEMs of triplicates from three independent experiments, using fold change compared to the values of the time 0 group (<span class="html-italic">n</span> = 3). qPCR analyses were normalized with the ef-1α and β-actin genes. The difference in transcript levels is denoted by asterisks (**) when the main effects were significant (<span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 3
<p>Effects of <span class="html-italic">P. salmonis</span> on the level of autophagy and Rab protein markers in muscle cells challenged. Fish skeletal muscle cells were cultured and challenge with <span class="html-italic">P. salmonis</span> at MOI 10 (resuspended in differentiation medium) for 6 h. (<b>A</b>) Immunofluorescence of LC3 (LC3-I and LC3-II). (<b>B</b>) Representative Western blot of LC3-II. (<b>C</b>) Rab small GTPases gene expression and, (<b>D</b>) Representative Western blot of Rab7a. The data are presented as the means ± SEMs of triplicates from three independent experiments, using fold change compared to the control values (untreated cells, <span class="html-italic">n</span> = 3). The measure of actin was used as load control for Western blots. qPCR analyses were normalized by the <span class="html-italic">ef-1α</span> and <span class="html-italic">β-actin</span> genes. The difference between the control group and treated groups is denoted by asterisks (*) when the main effects were significant (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
16 pages, 1043 KiB  
Review
Exploiting Autophagy-Dependent Neoantigen Presentation in Tumor Microenvironment
by Evangelos Koustas, Eleni-Myrto Trifylli, Panagiotis Sarantis, Nikolaos Papadopoulos, Konstantinos Papanikolopoulos, Georgios Aloizos, Christos Damaskos, Nikolaos Garmpis, Anna Garmpi, Dimitris Matthaios and Michalis V. Karamouzis
Genes 2023, 14(2), 474; https://doi.org/10.3390/genes14020474 - 13 Feb 2023
Cited by 9 | Viewed by 2463
Abstract
Autophagy constitutes a well-known homeostatic and catabolic process that is responsible for degradation and recycling of cellular components. It is a key regulatory mechanism for several cellular functions, whereas its dysregulation is associated with tumorigenesis, tumor–stroma interactions and resistance to cancer therapy. A [...] Read more.
Autophagy constitutes a well-known homeostatic and catabolic process that is responsible for degradation and recycling of cellular components. It is a key regulatory mechanism for several cellular functions, whereas its dysregulation is associated with tumorigenesis, tumor–stroma interactions and resistance to cancer therapy. A growing body of evidence has proven that autophagy affects the tumor microenvironment, while it is also considered a key factor for function of several immune cells, such as APCs, T-cells, and macrophages. Moreover, it is implicated in presentation of neo-antigens of tumor cells in both MHC-I and MHC-II in dendritic cells (DCs) in functional activity of immune cells by creating T-cell memory, as well as in cross-presentation of neo-antigens for MHC-I presentation and the internalization process. Currently, autophagy has a crucial role in immunotherapy. Emergence of cancer immunotherapy has already shown some remarkable results, having changed therapeutic strategy in clinical practice for several cancer types. Despite these promising long-term responses, several patients seem to lack the ability to respond to immune checkpoint inhibitors. Thus, autophagy through neo-antigen presentation is a potential target in order to strengthen or attenuate the effects of immunotherapy against different types of cancer. This review will shed light on the recent advances and future directions of autophagy-dependent neo-antigen presentation and consequently its role in immunotherapy for malignant tumors. Full article
(This article belongs to the Special Issue Cell Signalling and Inflammation in Cancer)
Show Figures

Figure 1

Figure 1
<p>A schematic presentation of the macroautophagy pathway.Autophagy includes five major steps: (i) induction, (ii) nucleation, (iii) elongation, (iv) fusion, and (v) degradation. After mTOR is deactivated and ULK1 activated, cargo starts to become engulfed inside the previously empty phagophore and later nucleated, which requires activation of classIII PI3K by the ULK1 complex. Later, the phagophore is elongated, which is mediated via formation of the classIII PI3K- Beclin1 complex, resulting in formation of the autophagosome. Subsequently, the autophagosome is matured by two conjugation reactions and later fused with lysosomes, which leads to its degradation [<a href="#B3-genes-14-00474" class="html-bibr">3</a>,<a href="#B4-genes-14-00474" class="html-bibr">4</a>,<a href="#B5-genes-14-00474" class="html-bibr">5</a>,<a href="#B6-genes-14-00474" class="html-bibr">6</a>,<a href="#B7-genes-14-00474" class="html-bibr">7</a>]. This figure was created with BioRender.com (agreement number PU24S95UJE).</p>
Full article ">Figure 2
<p>A schematic presentation of TME implications in tumor growth and progression.ECM is characterized by desmoplasia, which results from aggregation of collagen/elastin fibers and CAFs. Endothelial cells (ECs) are related to neoangiogenesis under the stimulatory effect of VEGF. The new vasculature lacks proper intercellular connections (“leaky vessels”), permitting extravasation of tumor cells and formation of distant metastasis. Adipocytes induce ECM alteration via release of MMPs, while neutrophils also secrete MMP-9 and VEGF, inducing ECM modification, neoangiogenesis, and local invasion. NK cells secrete cytokines or directly induce tumor cell destruction, while Tregs release several cytokines, such as IL-2, which deregulates NK cells, while TAMs secrete cytokines that enhance angiogenesis. CAFs are produced via conversion of tissue-fibroblasts and degrade E-cadherin in ECM via MMP-3, promoting tumor invasion [<a href="#B22-genes-14-00474" class="html-bibr">22</a>,<a href="#B23-genes-14-00474" class="html-bibr">23</a>,<a href="#B24-genes-14-00474" class="html-bibr">24</a>,<a href="#B25-genes-14-00474" class="html-bibr">25</a>,<a href="#B26-genes-14-00474" class="html-bibr">26</a>,<a href="#B27-genes-14-00474" class="html-bibr">27</a>]. This figure was created with BioRender.com (agreement number AD24S95KVO).</p>
Full article ">
11 pages, 2419 KiB  
Article
Designing Novel Multi-Epitope Vaccine Construct against Prevotella intermedia-Interpain A: An Immunoinformatics Approach
by Pradeep Kumar Yadalam, Raghavendra Vamsi Anegundi, Safa Munawar, Ramya Ramadoss, Santhiya Rengaraj, Sindhu Ramesh, Mohammed Aljeldah, Basim R. Al Shammari, Ahmad A. Alshehri, Ameen S. S. Alwashmi, Safaa A. Turkistani, Abdulsalam Alawfi, Amer Alshengeti, Mohammed Garout, Amal A. Sabour, Maha A. Alshiekheid, Fatimah S. Aljebaly and Ali A. Rabaan
Medicina 2023, 59(2), 302; https://doi.org/10.3390/medicina59020302 - 6 Feb 2023
Cited by 3 | Viewed by 2657
Abstract
Background and Objectives: Periodontitis is a chronic multifactorial inflammatory infectious disease marked by continuous degradation of teeth and surrounding parts. One of the most important periodontal pathogens is P. intermedia, and with its interpain A proteinase, it leads to an increase [...] Read more.
Background and Objectives: Periodontitis is a chronic multifactorial inflammatory infectious disease marked by continuous degradation of teeth and surrounding parts. One of the most important periodontal pathogens is P. intermedia, and with its interpain A proteinase, it leads to an increase in lethal infection. Materials and Methods: The current study was designed to create a multi-epitope vaccine using an immunoinformatics method that targets the interpain A of P. intermedia. For the development of vaccines, P. intermedia peptides InpA were found appropriate. To create a multi-epitope vaccination design, interpain A, B, and T-cell epitopes were found and assessed depending on the essential variables. The vaccine construct was evaluated based on its stability, antigenicity, and allergenicity. Results: The vaccine construct reached a more significant population and was able to bind to both the binding epitopes of major histocompatibility complex (MHC)-I and MHC-II. Through the C3 receptor complex route, P. intermedia InpA promotes an immunological subunit. Utilizing InpA-C3 and vaccination epitopes as the receptor and ligand, the molecular docking and dynamics were performed using the ClusPro 2.0 server. Conclusion: The developed vaccine had shown good antigenicity, solubility, and stability. Molecular docking indicated the vaccine’s 3D structure interacts strongly with the complement C3. The current study describes the design for vaccine, and steady interaction with the C3 immunological receptor to induce a good memory and an adaptive immune response against Interpain A of P. intermedia. Full article
(This article belongs to the Special Issue New Trends in Autoimmunity and Vaccines)
Show Figures

Figure 1

Figure 1
<p>The antigenic propensity of reference ID 3BBA. AC: Amino acids.</p>
Full article ">Figure 2
<p>Graphical representation of the BCL epitopes’ confirmation with the BepiPred server identified by ABCPred with a 16-mer length score of 0.5 or above.</p>
Full article ">Figure 3
<p>Population coverage of all the selected epitopes for both MHC-I and II classes. (<b>A</b>): Indian Asian class-I coverage. (<b>B</b>): Indian Asian class-II coverage. MHC: major histocompatibility complex.</p>
Full article ">Figure 4
<p>Docked complexes of the vaccine and the epitopes. (<b>A</b>): Predicted structure of modeled vaccine construct using a Swiss model. (<b>B</b>): Molecular docking of epitopes with 2a37 complement c3 receptor.</p>
Full article ">
26 pages, 3279 KiB  
Article
Immunoinformatics Approach to Design a Multi-Epitope Nanovaccine against Leishmania Parasite: Elicitation of Cellular Immune Responses
by Maritsa Margaroni, Maria Agallou, Evgenia Tsanaktsidou, Olga Kammona, Costas Kiparissides and Evdokia Karagouni
Vaccines 2023, 11(2), 304; https://doi.org/10.3390/vaccines11020304 - 30 Jan 2023
Cited by 10 | Viewed by 2883
Abstract
Leishmaniasis is a vector-borne disease caused by an intracellular parasite of the genus Leishmania with different clinical manifestations that affect millions of people worldwide, while the visceral form may be fatal if left untreated. Since the available chemotherapeutic agents are not satisfactory, vaccination [...] Read more.
Leishmaniasis is a vector-borne disease caused by an intracellular parasite of the genus Leishmania with different clinical manifestations that affect millions of people worldwide, while the visceral form may be fatal if left untreated. Since the available chemotherapeutic agents are not satisfactory, vaccination emerges as the most promising strategy for confronting leishmaniasis. In the present study, a reverse vaccinology approach was adopted to design a pipeline starting from proteome analysis of three different Leishmania species and ending with the selection of a pool of MHCI- and MHCII-binding epitopes. Epitopes from five parasite proteins were retrieved and fused to construct a multi-epitope chimeric protein, named LeishChim. Immunoinformatics analyses indicated that LeishChim was a stable, non-allergenic and immunogenic protein that could bind strongly onto MHCI and MHCII molecules, suggesting it as a potentially safe and effective vaccine candidate. Preclinical evaluation validated the in silico prediction, since the LeishChim protein, encapsulated simultaneously with monophosphoryl lipid A (MPLA) into poly(D,L-lactide-co-glycolide) (PLGA) nanoparticles, elicited specific cellular immune responses when administered to BALB/c mice. These were characterized by the development of memory CD4+ T cells, as well as IFNγ- and TNFα-producing CD4+ and CD8+ T cells, supporting the potential of LeishChim as a vaccine candidate. Full article
(This article belongs to the Special Issue Reverse Vaccinology and Vaccine Antigens)
Show Figures

Figure 1

Figure 1
<p>Vaccination schedule. BALB/c mice were vaccinated subcutaneously twice, with 15 days intervals, with (i) PLGA-LeishChim, (ii) PLGA-LeishChim-MPLA or (iii) soluble LeishChim and 15 days post booster vaccination, spleens were removed.</p>
Full article ">Figure 2
<p>Schematic representation of the LeishChim construct. (<b>a</b>) The construct is composed of sequences obtained from 5 different proteins which were placed in tandem. (<b>b</b>) Amino acid sequence of LeishChim. Different font colors correspond to each protein from which sequence was derived and numbers indicate which part of each protein was included in LeishChim. Illustrator for Biological sequences (IBS) version 1.0 used for protein illustration is available at <a href="http://ibs.biocuckoo.org/online.php#" target="_blank">http://ibs.biocuckoo.org/online.php#</a>, accessed on 5 December 2018 [<a href="#B52-vaccines-11-00304" class="html-bibr">52</a>].</p>
Full article ">Figure 3
<p>Secondary and tertiary structure of LeishChim. (<b>a</b>) PSIPRED was used to predict its secondary structure. (<b>b</b>) The tertiary structure was predicted with i-TASSER algorithm, and the best model had a C-score of −2.42.</p>
Full article ">Figure 4
<p>Refinement and validation of LeishChim’s tertiary structure. (<b>a</b>) Protein’s refined model via application of GalaxyRefine server. The new structure is pictured in grey color. (<b>b</b>) Validation of the initial and the refined tertiary structure of chimeric protein with PROS-A web server. (<b>c</b>) ERRAT analysis before and after the refinement. In the ERRAT plot, regions of the 3D model that can be rejected at 95% confidence level are shown in gray lines and regions, while those that can be rejected at 99% level confidence are depicted in black lines. (<b>d</b>) Ramachandran analysis of the initial and refined protein model via PROCHECK server application. The most favored areas are represented with darker color.</p>
Full article ">Figure 5
<p>MD simulation of the LeishChim and MHCI or MHCII complexes with Normal Mode Analysis (NMA). (<b>a</b>,<b>b</b>) NMA mobility of the LeishChim-MHCI/II complexes with affine model arrows. (<b>c</b>,<b>d</b>) Deformability plots. (<b>e</b>,<b>f</b>) B-factor plots. (<b>g</b>,<b>h</b>) Eigenvalue plots. (<b>i</b>,<b>j</b>) Normal mode variance plots. The purple bars indicate variance of individual modes, while the green indicate cumulative variance. (<b>k</b>,<b>l</b>) Covariance map. Red, white and blue colors correspond to correlated, uncorrelated and anti-correlated motions. (<b>m</b>,<b>n</b>) Elastic network. The darker gray colors correspond to stiffer spring.</p>
Full article ">Figure 6
<p>In silico simulation of immune response against LeishChim in a two-immunization protocol. (<b>a</b>) Immunoglobulins and antigen levels, (<b>b</b>) B cell population per state, (<b>c</b>) generation of B cell population, (<b>d</b>) T helper (T<sub>H</sub>) cell population per state, (<b>e</b>) generation of T<sub>H</sub> population, (<b>f</b>) cytotoxic T (T<sub>C</sub>) cell population per state and (<b>g</b>) production of cytokines. The inner graph in (<b>g</b>) indicates the Simpson index D of IL-2. Simpson Index D was inferred as measurement of diversity.</p>
Full article ">Figure 7
<p>Cellular immune responses in vaccinated BALB/c mice. BALB/c mice (<span class="html-italic">n</span> = 5/group) were vaccinated subcutaneously with PBS, LeishChim, PLGA-LeishChim or PLGA-LeishChim-MPLA twice with 15 days interval. (<b>a</b>) Fifteen days post booster vaccination, spleen cells were harvested and were stimulated in vitro with LeishChim (5 μg/mL) for 96 h in 5% CO<sub>2</sub> at 37 °C. LeishChim-specific proliferation was measured by assessing thymidine incorporation. Samples were run in triplicates. In parallel, spleen cells were stimulated with LeishChim (5 μg/mL) for 24 h or 48 h for the detection of (<b>b</b>) central memory (CD44<sup>+</sup>CD62L<sup>+</sup>) CD4<sup>+</sup> T cells and (<b>c</b>–<b>f</b>) IFNγ- or TNFα-producing CD8<sup>+</sup> and CD4<sup>+</sup> T cells with flow cytometry. The results are presented as the mean value ± SD of each group of mice. Significant differences between vaccinated and PBS control group are indicated by asterisks: * <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 or **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
Back to TopTop