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

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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

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

Search Results (806)

Search Parameters:
Keywords = subunit vaccine

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 3088 KiB  
Article
A Recombinant Shigella flexneri Strain Expressing ETEC Heat-Labile Enterotoxin B Subunit Shows Promise for Vaccine Development via OMVs
by Josune Salvador-Erro, Yadira Pastor and Carlos Gamazo
Int. J. Mol. Sci. 2024, 25(23), 12535; https://doi.org/10.3390/ijms252312535 - 22 Nov 2024
Viewed by 283
Abstract
Diarrheal diseases caused by Shigella and enterotoxigenic Escherichia coli (ETEC) are significant health burdens, especially in resource-limited regions with high child mortality. In response to the lack of licensed vaccines and rising antibiotic resistance for these pathogens, this study developed a recombinant Shigella [...] Read more.
Diarrheal diseases caused by Shigella and enterotoxigenic Escherichia coli (ETEC) are significant health burdens, especially in resource-limited regions with high child mortality. In response to the lack of licensed vaccines and rising antibiotic resistance for these pathogens, this study developed a recombinant Shigella flexneri strain with the novel incorporation of the eltb gene for the heat-labile enterotoxin B (LTB) subunit of ETEC directly into Shigella’s genome, enhancing stability and consistent production. This approach combines the immunogenic potential of LTB with the antigen delivery properties of S. flexneri outer membrane vesicles (OMVs), aiming to provide cross-protection against both bacterial pathogens in a stable, non-replicating vaccine platform. We confirmed successful expression through GM1-capture ELISA, achieving levels comparable to ETEC. Additionally, proteomic analysis verified that the isolated vesicles from the recombinant strains contain the LTB protein and the main outer membrane proteins and virulence factors from Shigella, including OmpA, OmpC, IcsA, SepA, and Ipa proteins, and increased expression of Slp and OmpX. Thus, our newly designed S. flexneri OMVs, engineered to carry ETEC’s LTB toxin, represent a promising strategy to be considered as a subunit vaccine candidate against S. flexneri and ETEC. Full article
Show Figures

Figure 1

Figure 1
<p>LTB detection in ETEC and different parental or transgenic <span class="html-italic">Shigella flexneri</span> strains using a GM1 capture-ELISA. (<b>A</b>) The presence of LTB was confirmed on ETEC and <span class="html-italic">S. flexneri::eltb</span> and <span class="html-italic">ΔtolR::eltb</span> strains. (<b>B</b>) In LTB-positive samples, no significant differences were observed between cell-associated and bacterial culture supernatant fractions. [ns, not significant (<span class="html-italic">p</span> ≥ 0.05)].</p>
Full article ">Figure 2
<p>Representative transmission electron microscopy (TEM) images of OMVs and HT-OMVs isolated from parental <span class="html-italic">Shigella flexneri</span>, <span class="html-italic">S. flexneri ΔtolR</span> mutant, and their respective recombinant containing <span class="html-italic">eltb</span> (scale bar = 200 nm).</p>
Full article ">Figure 3
<p>Proteomic analysis identified LTB abundance in the different OMV and HT products from ETEC and <span class="html-italic">Shigella flexneri</span> or mutant (<span class="html-italic">ΔtolR</span>), with and without the <span class="html-italic">eltb</span> insertion. Samples were analyzed in triplicates and the protein abundance was normalized to the total protein (*, <span class="html-italic">p</span> &lt; 0.05; ***, <span class="html-italic">p</span> &lt; 0.001; ns, non-significant; a.u.: arbitrary units).</p>
Full article ">Figure 4
<p>Differential expression of the main outer membrane proteins and virulence factors included in OMV and HT products from <span class="html-italic">Shigella flexneri</span> wild-type (<b>A</b>,<b>B</b>) or mutant <span class="html-italic">ΔtolR</span> (<b>C</b>,<b>D</b>) before (black) and after <span class="html-italic">eltb</span> mutation (white). Proteins were clustered in two groups based on their expression profile. Samples were analyzed in triplicates and the Log2 of the fold change is represented (*, <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, non-significant).</p>
Full article ">Figure 5
<p>Bacterial growth curves of <span class="html-italic">Shigella flexneri</span> wt, <span class="html-italic">S. flexneri::eltb, S. flexneri ΔtolR, and S. flexneri ΔtolR::eltb.</span> Bacterial suspensions were inoculated on Bioscreen C multi-well plates and incubated with continuous shaking at 37 °C. Absorbance values (O.D.<sub>580 nm</sub>) were automatically read at regular intervals of 10 min for 22 h. An arbitrary position of the stationary phase is indicated according to the growth curve kinetics.</p>
Full article ">Figure 6
<p>Biofilm formation assay results. (<b>A</b>) Comparison of biofilm formation among recombinant and parental <span class="html-italic">Shigella flexneri</span> strains, showing significant increases in biofilm production for the <span class="html-italic">S. flexneri::eltb</span> and <span class="html-italic">S. flexneri ΔtolR::eltb</span> strains (**, <span class="html-italic">p</span> &lt; 0.01) compared to the parental strains. (<b>B</b>) Evaluation of LTB involvement in biofilm formation, showing a significant decrease in biofilm formation capacity after the addition of GM1, the natural receptor of LTB (*, <span class="html-italic">p</span> &lt; 0.05), with no significant differences observed in the parental strain (ns).</p>
Full article ">Figure 7
<p>Effect of OMVs and HT-OMVs on the mitochondrial activity (MTT assay) on HeLa cells. Figures indicate the percentage (%) of mitochondrial activity after treatment for 2 or 24 h with different concentrations of OMVs and HT-OMVs (μg/mL) compared to untreated cells. Experiment was performed in triplicate. Error bars represent SEM.</p>
Full article ">
20 pages, 2059 KiB  
Review
Engineering Escherichia coli-Derived Nanoparticles for Vaccine Development
by Shubing Tang, Chen Zhao and Xianchao Zhu
Vaccines 2024, 12(11), 1287; https://doi.org/10.3390/vaccines12111287 - 18 Nov 2024
Viewed by 783
Abstract
The development of effective vaccines necessitates a delicate balance between maximizing immunogenicity and minimizing safety concerns. Subunit vaccines, while generally considered safe, often fail to elicit robust and durable immune responses. Nanotechnology presents a promising approach to address this dilemma, enabling subunit antigens [...] Read more.
The development of effective vaccines necessitates a delicate balance between maximizing immunogenicity and minimizing safety concerns. Subunit vaccines, while generally considered safe, often fail to elicit robust and durable immune responses. Nanotechnology presents a promising approach to address this dilemma, enabling subunit antigens to mimic critical aspects of native pathogens, such as nanoscale dimensions, geometry, and highly repetitive antigen display. Various expression systems, including Escherichia coli (E. coli), yeast, baculovirus/insect cells, and Chinese hamster ovary (CHO) cells, have been explored for the production of nanoparticle vaccines. Among these, E. coli stands out due to its cost-effectiveness, scalability, rapid production cycle, and high yields. However, the E. coli manufacturing platform faces challenges related to its unfavorable redox environment for disulfide bond formation, lack of post-translational modifications, and difficulties in achieving proper protein folding. This review focuses on molecular and protein engineering strategies to enhance protein solubility in E. coli and facilitate the in vitro reassembly of virus-like particles (VLPs). We also discuss approaches for antigen display on nanocarrier surfaces and methods to stabilize these carriers. These bioengineering approaches, in combination with advanced nanocarrier design, hold significant potential for developing highly effective and affordable E. coli-derived nanovaccines, paving the way for improved protection against a wide range of infectious diseases. Full article
Show Figures

Figure 1

Figure 1
<p>Commonly used nanocarriers to display antigens. (<b>A</b>) 3D structure of VLPs. (<b>B</b>) 3D structure of protein-based nanoparticles. T represents the triangulation of nanoparticles, and mer is the abbreviation of protomers.</p>
Full article ">Figure 2
<p>Nanoparticles offer several key advantages over conventional vaccines, leading to more potent and targeted immune responses. Unlike smaller proteins or particles (&lt;10 nm) that are quickly cleared from the bloodstream, nanoparticles (20–100 nm) efficiently target draining lymph nodes, the primary site of immune response initiation. Furthermore, nanoparticles are preferentially engulfed by antigen-presenting cells (APCs), particularly dendritic cells (DCs), maximizing antigen presentation and immune activation.</p>
Full article ">Figure 3
<p>Display of antigens using the genetic fusion approach. (<b>A</b>) Antigens can be inserted at the N-terminus, C-terminus or MIR site of HBc. (<b>B</b>) Two consecutive HBc are designed as nanocarriers with the insertion of a single antigen at the MIR site to reduce steric hindrance.</p>
Full article ">Figure 4
<p>Three chemoenzymatic methods to loading antigens onto HBc nanocarrier. (<b>A</b>) Soratase-mediated site-specific tagging is introduced into split HBc nanoscaffold. (<b>B</b>) Split intein-mediated <span class="html-italic">trans</span>-splicing is utilized to conjugate antigens onto split HBc nanoscaffold. (<b>C</b>) SpyCatcher/SpyTag mediated covalent conjugation is applied to couple antigens onto HBc nanoscaffold.</p>
Full article ">Figure 5
<p>Advanced nanotechnologies used in <span class="html-italic">E. coli</span>-derived vaccine development.</p>
Full article ">
28 pages, 1184 KiB  
Review
Immune Responses to Respiratory Syncytial Virus Vaccines: Advances and Challenges
by Gabriela Souza da Silva, Sofia Giacomet Borges, Bruna Bastos Pozzebon and Ana Paula Duarte de Souza
Microorganisms 2024, 12(11), 2305; https://doi.org/10.3390/microorganisms12112305 - 13 Nov 2024
Viewed by 812
Abstract
Respiratory Syncytial Virus (RSV) is a leading cause of acute respiratory infections, particularly in children and the elderly. This virus primarily infects ciliated epithelial cells and activates alveolar macrophages and dendritic cells, triggering an innate antiviral response that releases pro-inflammatory cytokines. However, immunity [...] Read more.
Respiratory Syncytial Virus (RSV) is a leading cause of acute respiratory infections, particularly in children and the elderly. This virus primarily infects ciliated epithelial cells and activates alveolar macrophages and dendritic cells, triggering an innate antiviral response that releases pro-inflammatory cytokines. However, immunity generated by infection is limited, often leading to reinfection throughout life. This review focuses on the immune response elicited by newly developed and approved vaccines against RSV. A comprehensive search of clinical studies on RSV vaccine candidates conducted between 2013 and 2024 was performed. There are three primary target groups for RSV vaccines: pediatric populations, infants through maternal immunization, and the elderly. Different vaccine approaches address these groups, including subunit, live attenuated or chimeric, vector-based, and mRNA vaccines. To date, subunit RSV vaccines and the mRNA vaccine have been approved using the pre-fusion conformation of the F protein, which has been shown to induce strong immune responses. Nevertheless, several other vaccine candidates face challenges, such as modest increases in antibody production, highlighting the need for further research. Despite the success of the approved vaccines for adults older than 60 years and pregnant women, there remains a critical need for vaccines that can protect children older than six months, who are still highly vulnerable to RSV infections. Full article
(This article belongs to the Special Issue Human Respiratory Syncytial Virus—Biology, Diagnosis and Prevention)
Show Figures

Figure 1

Figure 1
<p>RSV vaccine types in clinical trials and approved for use. There are different RSV vaccine types: subunit vaccines, recombinant vector vaccines, live attenuated vaccines, and mRNA vaccines. Three RSV proteins, F, G, and SH, were tested in the subunit vaccine approach. The F protein-based vaccines used two different F protein conformations, post-fusion and pre-fusion. The RSV vaccines approved for use in the elderly and pregnant are subunit and mRNA vaccines based on the pre-fusion conformation.</p>
Full article ">
14 pages, 6300 KiB  
Article
H9 Consensus Hemagglutinin Subunit Vaccine with Adjuvants Induces Robust Mucosal and Systemic Immune Responses in Mice by Intranasal Administration
by Liming Lin, Shunfan Zhu, Beibei Yang, Xin Zhang, Huimin Wu, Shixiang Wu, Li Wu, Jianhong Shu, Yulong He and Huapeng Feng
Microorganisms 2024, 12(11), 2294; https://doi.org/10.3390/microorganisms12112294 - 12 Nov 2024
Viewed by 585
Abstract
The H9N2 subtype avian influenza viruses mainly cause respiratory symptoms, reduce the egg production and fertility of poultry, and result in secondary infections, posing a great threat to the poultry industry and human health. Currently, all H9N2 avian influenza commercial vaccines are inactivated [...] Read more.
The H9N2 subtype avian influenza viruses mainly cause respiratory symptoms, reduce the egg production and fertility of poultry, and result in secondary infections, posing a great threat to the poultry industry and human health. Currently, all H9N2 avian influenza commercial vaccines are inactivated vaccines, which provide protection for immunized animals but cannot inhibit the spread of the virus and make it difficult to distinguish between the infected animals and vaccinated animals. In this study, a trimeric consensus H9 hemagglutinin (HA) subunit vaccine for the H9N2 subtype avian influenza virus based on a baculovirus expression system was first generated, and then the effects of three molecular adjuvants on the H9 HA subunit vaccine, Cholera toxin subunit B (CTB), flagellin, and granulocyte-macrophage colony-stimulating factor (GM-CSF) fused with H9 HA, and one synthetic compound, a polyinosinic–polycytidylic acid (PolyI:C) adjuvant, were evaluated in mice by intranasal administration. The results showed that these four adjuvants enhanced the immunogenicity of the H9 HA subunit vaccine for avian influenza viruses, and that GM-CSF and PolyI:C present better mucosal adjuvant activity for the H9 HA subunit vaccine. These results demonstrate that we have developed a potential universal H9 HA mucosal subunit vaccine with adjuvants in a baculovirus system that would be helpful for the prevention and control of H9N2 subtype avian influenza viruses. Full article
(This article belongs to the Topic Advances in Vaccines and Antimicrobial Therapy)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the structure of HA.</p>
Full article ">Figure 2
<p>Immunization schedule and sample collection time points.</p>
Full article ">Figure 3
<p>Double-digestion identification of recombinant transfer plasmids. (<b>A</b>) pFastBac-HA, M1: DL15000 DNA marker; (<b>B</b>) pFastBac-CTB-HA; (<b>C</b>) pFastBac-FliC-HA, M2: DL10000 DNA marker; (<b>D</b>) pFastBac-GM-CSF-HA.</p>
Full article ">Figure 4
<p>Identification of recombinant bacmids by PCR. (<b>A</b>) rBacmid-HA, M1: DL15000 DNA marker; (<b>B</b>) rBacmid-CTB-HA, M2: DL10000 DNA marker; (<b>C</b>) rBacmid-FliC-HA; (<b>D</b>) rBacmid-GM-CSF-HA.</p>
Full article ">Figure 5
<p>Observation of cytopathic effect in Sf9 cells transfected with recombinant bacmids. (<b>A</b>) rBV-HA; (<b>B</b>) rBV-CTB-HA; (<b>C</b>) rBV-FliC-HA; (<b>D</b>) rBV-GM-CSF-HA; (<b>E</b>) negative control. All images were magnified at 400×.</p>
Full article ">Figure 6
<p>Identification of trimer formation of recombinant proteins by Western blotting. (<b>A</b>) HA; (<b>B</b>) CTB-HA; (<b>C</b>) FliC-HA; (<b>D</b>) GM-CSF-HA.</p>
Full article ">Figure 7
<p>Identification of HA expression with the recombinant baculoviruses by indirect immunofluorescence assay. (<b>A</b>) rBV-HA; (<b>B</b>) rBV-CTB-HA; (<b>C</b>) rBV-FliC-HA; (<b>D</b>) rBV-GM-CSF-HA; (<b>E</b>) negative control. All images were magnified at 400×.</p>
Full article ">Figure 8
<p>Identification of the purified proteins by SDS-PAGE. (<b>A</b>) HA; (<b>B</b>) CTB-HA; (<b>C</b>) FliC-HA; (<b>D</b>) GM-CSF-HA.</p>
Full article ">Figure 9
<p>Identification of the purified HA proteins by Western blotting. (<b>A</b>) HA; (<b>B</b>) CTB-HA; (<b>C</b>) FliC-HA; (<b>D</b>) GM-CSF-HA.</p>
Full article ">Figure 10
<p>Specific IgA and IgG antibodies were induced in NW, BALF, and sera by HA and three molecular adjuvant-fused HAs through intranasal immunization. (<b>A</b>) IgA antibody titers in NLF and BALF, (<b>B</b>) IgA antibody titers in sera, (<b>C</b>) IgG antibody titers in NLF and BALF, * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001; Two-way ANOVA was used for significant analysis.</p>
Full article ">Figure 11
<p>Poly I:C significantly enhanced the induction of IgA and IgG antibodies by intranasal administration. (<b>A</b>) IgA antibody titers in NW and BALF; (<b>B</b>) IgA antibody titers in sera; (<b>C</b>) IgG antibody titers in sera.; **** <span class="html-italic">p</span> &lt; 0.0001; two-way ANOVA was used for significant analysis.</p>
Full article ">
57 pages, 990 KiB  
Review
Prokaryote- and Eukaryote-Based Expression Systems: Advances in Post-Pandemic Viral Antigen Production for Vaccines
by Nelli S. Khudainazarova, Dmitriy L. Granovskiy, Olga A. Kondakova, Ekaterina M. Ryabchevskaya, Angelina O. Kovalenko, Ekaterina A. Evtushenko, Marina V. Arkhipenko, Nikolai A. Nikitin and Olga V. Karpova
Int. J. Mol. Sci. 2024, 25(22), 11979; https://doi.org/10.3390/ijms252211979 - 7 Nov 2024
Viewed by 505
Abstract
This review addresses the ongoing global challenge posed by emerging and evolving viral diseases, underscoring the need for innovative vaccine development strategies. It focuses on the modern approaches to creating vaccines based on recombinant proteins produced in different expression systems, including bacteria, yeast, [...] Read more.
This review addresses the ongoing global challenge posed by emerging and evolving viral diseases, underscoring the need for innovative vaccine development strategies. It focuses on the modern approaches to creating vaccines based on recombinant proteins produced in different expression systems, including bacteria, yeast, plants, insects, and mammals. This review analyses the advantages, limitations, and applications of these expression systems for producing vaccine antigens, as well as strategies for designing safer, more effective, and potentially ‘universal’ antigens. The review discusses the development of vaccines for a range of viral diseases, excluding SARS-CoV-2, which has already been extensively studied. The authors present these findings with the aim of contributing to ongoing research and advancing the development of antiviral vaccines. Full article
Show Figures

Figure 1

Figure 1
<p>Types of the recombinant vaccines considered in the review. CP—Capsid Protein; AI—Antigen of Interest; Aux—Auxiliary additions to the antigen sequence; VLP—Virus-Like Particle; cVLP—Chimeric Virus-Like Particle.</p>
Full article ">
14 pages, 1892 KiB  
Article
Safety and Immunogenicity of Intranasal Razi Cov Pars as a COVID-19 Booster Vaccine in Adults: Promising Results from a Groundbreaking Clinical Trial
by Mohammad Hossein Fallah Mehrabadi, Monireh Hajimoradi, Ali Es-haghi, Saeed Kalantari, Mojtaba Noofeli, Ali Rezaei Mokarram, Seyed Hossein Razzaz, Maryam Taghdiri, Ladan Mokhberalsafa, Fariba Sadeghi, Vahideh Mohseni, Safdar Masoumi, Rezvan Golmoradi-Zadeh, Mohammad Hasan Rabiee, Masoud Solaymani-Dodaran and Seyed Reza Banihashemi
Vaccines 2024, 12(11), 1255; https://doi.org/10.3390/vaccines12111255 - 5 Nov 2024
Viewed by 831
Abstract
Protective antibodies in the upper respiratory tract prevent the spread of COVID-19 in the community. Intranasal vaccines could raise the specific secretory IgA and IgG levels. This is a single-center, randomized, double-blind, placebo-controlled clinical trial to evaluate the safety and immunogenicity of Razi [...] Read more.
Protective antibodies in the upper respiratory tract prevent the spread of COVID-19 in the community. Intranasal vaccines could raise the specific secretory IgA and IgG levels. This is a single-center, randomized, double-blind, placebo-controlled clinical trial to evaluate the safety and immunogenicity of Razi Cov Pars (RCP) intranasal recombinant protein subunit COVID-19 vaccine as a booster in adults. We compared specific IgG and IgA levels in the intranasal RCP group (n = 97) versus placebo (n = 96) in serum, saliva, and nasal mucosal secretions on days 0 and 14 and reported their Geometric Mean Ratios (GMR) and 95% confidence intervals (CI). We showed significant increases in IgA and IgG anti-RBD in the nasal mucosa in the RCP group, but their increase was not detectable in the serum and saliva. Anti-spike IgA in the nasal mucosa also increased in the RCP group compared to the placebo. This increase against the COVID-19 variant Omicron was also similar to that of the Wuhan. We detected no serious adverse reactions or anaphylaxis and all adverse events resolved completely during the follow-up period and were similar in both groups. Intranasal RCP is safe, stimulates the respiratory mucosal immunity, and could be a booster on various COVID-19 vaccines and be effective against new virus variants. Full article
Show Figures

Figure 1

Figure 1
<p>Participant’s flow diagram.</p>
Full article ">Figure 2
<p>Frequency of solicited systemic adverse reactions by study groups during the first week following vaccination.</p>
Full article ">Figure 3
<p>Scatter plots of individual values and their geometric means for specific IgG and IgA antibody levels in the serum and nasal mucosal secretions of the study participants at the time of booster IN vaccination and two weeks after. (<b>A</b>): Serum Anti-RBD IgG levels; (<b>B</b>): Serum Anti-RBD IgA levels; (<b>C</b>): Nasal mucosal Anti-RBD IgG levels; (<b>D</b>): Nasal mucosal Anti-RBD IgA levels. ns, not significant, * <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>Scatter plots of individual values and their geometric means of nasal mucosal specific IgA anti-spike antibodies comparing the antibody response to Wuhan and Omicron variants in the study participants by study groups at the time of booster IN vaccination and two weeks after. ns, not significant, * <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 5
<p>Scatter plots of individual values and their geometric means comparing the serum anti-Spike specific IgG in a subpopulation of phase III study participants in RAZI and Sinopharm groups in response to IN RCP or placebo (adjuvant), respectively, at the time of IN booster vaccination and two weeks after. * <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 ">
17 pages, 3795 KiB  
Article
Cell-Free Screening, Production and Animal Testing of a STI-Related Chlamydial Major Outer Membrane Protein Supported in Nanolipoproteins
by Mariam Mohagheghi, Abisola Abisoye-Ogunniyan, Angela C. Evans, Alexander E. Peterson, Gregory A. Bude, Steven Hoang-Phou, Byron Dillon Vannest, Dominique Hall, Amy Rasley, Dina R. Weilhammer, Nicholas O. Fischer, Wei He, Beverly V. Robinson, Sukumar Pal, Anatoli Slepenkin, Luis de la Maza and Matthew A. Coleman
Vaccines 2024, 12(11), 1246; https://doi.org/10.3390/vaccines12111246 - 1 Nov 2024
Viewed by 642
Abstract
Background: Vaccine development against Chlamydia, a prevalent sexually transmitted infection (STI), is imperative due to its global public health impact. However, significant challenges arise in the production of effective subunit vaccines based on recombinant protein antigens, particularly with membrane proteins like the Major [...] Read more.
Background: Vaccine development against Chlamydia, a prevalent sexually transmitted infection (STI), is imperative due to its global public health impact. However, significant challenges arise in the production of effective subunit vaccines based on recombinant protein antigens, particularly with membrane proteins like the Major Outer Membrane Protein (MOMP). Methods: Cell-free protein synthesis (CFPS) technology is an attractive approach to address these challenges as a method of high-throughput membrane protein and protein complex production coupled with nanolipoprotein particles (NLPs). NLPs provide a supporting scaffold while allowing easy adjuvant addition during formulation. Over the last decade, we have been working toward the production and characterization of MOMP-NLP complexes for vaccine testing. Results: The work presented here highlights the expression and biophysical analyses, including transmission electron microscopy (TEM) and dynamic light scattering (DLS), which confirm the formation and functionality of MOMP-NLP complexes for use in animal studies. Moreover, immunization studies in preclinical models compare the past and present protective efficacy of MOMP-NLP formulations, particularly when co-adjuvanted with CpG and FSL1. Conclusion: Ex vivo assessments further highlight the immunomodulatory effects of MOMP-NLP vaccinations, emphasizing their potential to elicit robust immune responses. However, further research is warranted to optimize vaccine formulations further, validate efficacy against Chlamydia trachomatis, and better understand the underlying mechanisms of immune response. Full article
(This article belongs to the Special Issue A One-Health Perspective on Immunization Against Infectious Diseases)
Show Figures

Figure 1

Figure 1
<p>Nanolipoprotein particles are a tool for studying membrane-bound proteins. (<b>A</b>) Nanolipoprotein particles, NLPs (8–25 nm disc-shaped particles), are formed by the spontaneous assembly of phospholipids into a bilayer stabilized by an apolipoprotein scaffold protein. (<b>B</b>) Cell-free approach for NLP production without detergents or pre-purified proteins. (<b>C</b>) Cell-free approach for full-length membrane protein expression encapsulated in NLP.</p>
Full article ">Figure 2
<p>Cell-free screening allows for rapid, large-scale condition testing and optimization. (<b>A</b>) SDS—PAGE image of 25 µL MOMP protein reactions with various lipids and telodendrimer to identify a condition that optimizes solubility. (<b>B</b>) SDS page of 100 µL MOMP protein reactions with apolipoprotein scaffold and various lipid/telodendrimer combinations demonstrates the scalability of the cell-free techniques. M = Molecular weight marker.</p>
Full article ">Figure 3
<p>Cell-free production of MOMP-NLP protein complex for mouse studies. (<b>A</b>) Cell-free expression of MOMP-NLP showing components including MOMP DNA, Δ49ApoA1 DNA, and pre-prepared DMPC/telodendrimer lipid in a cell-free reaction chamber. (<b>B</b>) SDS-PAGE Sypro-stained image showing cell-free produced MOMP-NLP purified using Nickel bead gravity column (molecular weight (MW) marker—SeeBlue Plus2, Total protein (T), flow-through (FT)—MOMP protein not associated with an NLP, two of six washes (W)—to purify protein of interest, and seven elutions (E1–E7)—to recover MOMP-NLP. (<b>C</b>) Western blot micrograph showing cell-free produced MOMP using mAb40, a primary antibody against MOMP.</p>
Full article ">Figure 4
<p>Imaging and electrophysiology techniques confirm the formation of the MOMP-NLP complex. (<b>A</b>) Negative stain cryoEM of peak fraction of eluted, soluble MOMP-NLP complex (from <a href="#vaccines-12-01246-f003" class="html-fig">Figure 3</a>B) illustrates the circular disk-like shape of Cm MOMP-NLP and empty NLP. (<b>B</b>) Dynamic Light Scattering (DLS) measurement further confirms the formation of the MOMP-NLP complex. (<b>C</b>) The MOMP protein is active as a porin, as shown by electrophysiology. Single channel conductance assay of MOMP-NLPs in fixed bilayers at a fixed voltage, standard 200 mV. (<b>D</b>) Electrophysiology additionally confirms MOMP-porin is active; the top trace shows 10 picoamp jumps to open confirmation of porin. A flat line indicates no porin activity and baseline membrane bilayer reading value.</p>
Full article ">Figure 5
<p>Systemic and local disease burden following intranasal Cm challenge of mice vaccinated via IN/IM prime-boost regimen with recombinant MOMP-NLP and adjuvanted with CpG and FSL1. (<b>A</b>) Experimental schematic showing intranasal prime and intramuscular boost vaccinations, i.n. challenge and tissue harvest. (<b>B</b>) Log IFU of Cm recovered from mice lungs 10 days post-challenge. (<b>C</b>) Change in mice body weight and lung weight (g) at 10 days post i.n. challenge with Cm. (<b>D</b>) A summary chart shows vaccine antigens, adjuvants, and <span class="html-italic">p</span>-values.</p>
Full article ">Figure 6
<p>Systemic and local disease burden following intranasal Cm challenge of mice vaccinated via IM prime regimen with recombinant MOMP-NLP and adjuvanted with CpG and FSL1. (<b>A</b>) Experimental schematic showing intramuscular prime vaccinations, i.n. challenge and tissue harvest. (<b>B</b>) Log IFU of Cm recovered from mice lungs 10 days post-challenge. (<b>C</b>) Change in mice body weight and lung weight (g) at 10 days post i.n. challenge with Cm. (<b>D</b>) A summary chart shows vaccine antigens, adjuvants, and <span class="html-italic">p</span>-values.</p>
Full article ">Figure 7
<p>Gene and cytokine expression following prime-boost-boost mice vaccinations with recombinant MOMP-NLP adjuvanted with CpG and FSL1. (<b>A</b>) Experimental schematic showing an intramuscular prime and two intramuscular boost vaccinations followed by tissue harvest. (<b>B</b>) Overexpressed genes with greater than or equal to 2-fold change value of vaccinated mice relative to sham (PBS) vaccinated mice and respective activated pathways, ranked by <span class="html-italic">p</span>-value. (<b>C</b>) Illustrates the downregulated genes. For these experiments, cDNA samples were obtained from pooled spleen RNA samples of 5 different animals per group and <span class="html-italic">p</span>-value ranked pathways/processes generated by NCATS BioPlanet, Enrichr. (<b>D</b>) Secreted cytokines following stimulation of splenocytes 7–10 days after boost with MOMP.</p>
Full article ">Figure 8
<p>Several Cm rMOMP and MOMP-NLP vaccine formulations offer protection against Chlamydia infections. A chart summarizing rMOMP and MOMP-NLP formulations tested with various adjuvant combinations and several routes of vaccinations. Protection was determined by a 2-fold IFU log mean difference upon comparison to EB values. IFU log means between 1 and 2 log differences classified as “semi-protective”.</p>
Full article ">
15 pages, 3169 KiB  
Article
In Silico Development of a Multi-Epitope Subunit Vaccine against Bluetongue Virus in Ovis aries Using Immunoinformatics
by Priyansha Raj Sinha, Shubhada R. Hegde, Ruchika Mittal, Chikkamagaluru Chandrashekhar Jagat, Ullas Gowda, Rathna Chandrashekhar, Gayathri Muthaiah, Samer Shamshad, Mohammed Mudassar Chanda, Vishweshwar Ganji, Kalyani Putty and Divakar Hemadri
Pathogens 2024, 13(11), 944; https://doi.org/10.3390/pathogens13110944 - 29 Oct 2024
Viewed by 669
Abstract
The bluetongue virus (BTV), transmitted by biting midges, poses a significant threat to livestock globally. This orbivirus induces bluetongue disease, leading to substantial economic losses in the agricultural sector. The current control measures have limitations, necessitating the development of novel, efficient vaccines. In [...] Read more.
The bluetongue virus (BTV), transmitted by biting midges, poses a significant threat to livestock globally. This orbivirus induces bluetongue disease, leading to substantial economic losses in the agricultural sector. The current control measures have limitations, necessitating the development of novel, efficient vaccines. In this study, an immunoinformatics approach is employed to design a multi-epitope subunit vaccine for Ovis aries targeting six BTV serotypes. Focusing on the VP2 capsid protein, the vaccine incorporates B-cell, helper-T lymphocytes (HTL), and cytotoxic T-cell lymphocytes (CTL) epitopes. Molecular docking reveals stable interactions with TLR2 and TLR4 receptors, suggesting the stability of the complex, indicating the potential viability of the multi-epitope vaccine. The computational approach offers a rapid and tailored strategy for vaccine development, highlighting potential efficacy and safety against BTV outbreaks. This work contributes to understanding BTV and presents a promising avenue for effective control. Full article
(This article belongs to the Section Vaccines and Therapeutic Developments)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Multi-epitope subunit vaccine structure. Components include the adjuvant (orange), cytotoxic T lymphocyte (CTL) epitopes (grey), helper-T lymphocyte (HTL) epitopes (teal), B-cell epitopes (green), and histidine tag (red). (<b>B</b>) Vaccine design of consensus sequence.</p>
Full article ">Figure 2
<p>Tertiary structure of subunit vaccine of consensus sequence.</p>
Full article ">Figure 3
<p>(<b>A</b>) Ramachandran plot with 94.9% residues in favored region and (<b>B</b>) ProSA-server Z-score, of the consensus vaccine refined model.</p>
Full article ">Figure 4
<p>(<b>A</b>) Visualization of docked TLR2—consensus vaccine complex. (<b>B</b>) Enlarged view highlighting the interacting residues in TLR2 (green) and consensus vaccine (brown). Hydrogen bonds are shown as the black dashed line.</p>
Full article ">Figure 5
<p>(<b>A</b>) Visualization of docked TLR4—consensus vaccine complex. (<b>B</b>) Enlarged view highlighting the interacting residues in TLR4 (green) and consensus vaccine (blue). Hydrogen bonds are shown as the black dashed line.</p>
Full article ">Figure 6
<p>MD simulation results of consensus vaccine complex with TLR2 and TLR4 over 50 ns trajectory. (<b>A</b>) RMSD graph of TLR2-vaccine (purple) and TLR4-vaccine (yellow) complex. (<b>B</b>) RMSF graph of the TLR2 protein (magenta) and vaccine construct (green). (<b>C</b>) RMSF graph of the TLR4 protein (violet) and vaccine construct (cyan).</p>
Full article ">
16 pages, 3296 KiB  
Article
Evaluation of the Use of Sub-Immunodominant Antigens of Babesia bovis with Flagellin C Adjuvant in Subunit Vaccine Development
by Manuel J. Rojas, Reginaldo G. Bastos, Jinna A. Navas, Heba F. Alzan, Jacob M. Laughery, Paul A. Lacy, Massaro W. Ueti and Carlos E. Suarez
Vaccines 2024, 12(11), 1215; https://doi.org/10.3390/vaccines12111215 - 25 Oct 2024
Viewed by 563
Abstract
Bovine babesiosis caused by the tick-borne apicomplexan parasite Babesia bovis remains a threat for cattle worldwide, and new vaccines are needed. We propose using immune-subdominant (ISD) antigens as alternative vaccine candidates. We first determined that RAP-1 NT and RRA are subdominant antigens using [...] Read more.
Bovine babesiosis caused by the tick-borne apicomplexan parasite Babesia bovis remains a threat for cattle worldwide, and new vaccines are needed. We propose using immune-subdominant (ISD) antigens as alternative vaccine candidates. We first determined that RAP-1 NT and RRA are subdominant antigens using recombinant antigens in ELISAs against sera from B. bovis-protected cattle. Protected animals demonstrated high antibody responses against the known immunodominant rRAP-1 CT antigen, but significantly lower levels against the rRAP-1 NT and rRRA antigens. Next, a group of cattle (n = 6) was vaccinated with rRRA and rRAP-1 NT using a FliC–Emulsigen mix as the adjuvant, and there was a control group (n = 6) with the adjuvant mix alone. All but one immunized animal demonstrated elicitation of strong humoral immune responses against the two ISD antigens. Acute babesiosis occurred in both groups of cattle upon a challenge with the virulent B. bovis, but a significant delay in the average rate of decrease in hematocrit in the vaccinated group, and an early monocyte response, was found in half of the vaccinated animals. In conclusion, we confirmed the immune subdominance of rRRA and rRAP-1 NT and the ability of FliC to increase immunogenicity of ISD antigens and generate useful information toward developing future subunit vaccines against B. bovis. Full article
(This article belongs to the Special Issue Vaccines against Arthropods and Arthropod-Borne Pathogens)
Show Figures

Figure 1

Figure 1
<p><b>Sub-immunodominance of Rap-1 NT and RRA proteins.</b> Recombinant Rap-1 CT, Rap-1 NT, and RRA proteins were exposed to immune sera from cattle vaccinated with attenuated <span class="html-italic">Babesia bovis</span> and challenged with a virulent <span class="html-italic">B. bovis</span> strain. The binding of antibodies IgM (top panels (<b>a</b>,<b>b</b>)) and IgG (bottom panels (<b>c</b>,<b>d</b>)) to the recombinant proteins was evaluated by iELISA. After averaging the Optical Density (OD) values of sera from the calves and the adult cows, rRap-1 CT showed to be an immunodominant antigen, while Rap-1 NT and RRA were immune-subdominant antigens.</p>
Full article ">Figure 2
<p><b>Western blot anti-recombinant proteins RAP-1 CT, RAP-1 NT, RRA, and FliC.</b> Pre-immune and immune bovine sera (calf 1735) [1:10] Goat anti-bovine IgG 1:2500). See the corresponding bands inside the red boxes.</p>
Full article ">Figure 3
<p><b>IgM and IgG reactivity against recombinant RAP-1 NT and RRA.</b> (<b>A</b>) Upper panel: iELISA analysis of IgM antibody levels against rRAP-1 NT in cattle (n = 6) immunized with rRAP-1 NT and RRA using a FliC adjuvant. DPI: days post-immunization with the recombinant antigen mix. DPC represents days post-challenge with the <span class="html-italic">B. bovis</span> strain Vir-S74-T3Bo. Lower panel: iELISA analysis of IgM antibody levels against rRRA in cattle (n = 6) immunized with rRAP-1 NT and RRA using a FliC adjuvant. DPI: days post-immunization with the recombinant antigen mix. DPC represents days post-challenge with the <span class="html-italic">B. bovis</span> strain Vir-S74-T3Bo. (<b>B</b>) Upper panel: iELISA analysis of IgG antibody levels against rRAP-1 NT in cattle (n = 6) immunized with rRAP-1 NT and RRA using a FliC adjuvant. DPI: days post-immunization with the recombinant antigen mix. DPC represents days post-challenge with the <span class="html-italic">B. bovis</span> strain Vir-S74-T3Bo. Lower panel: iELISA analysis of IgG antibody levels against rRRA in cattle (n = 6) immunized with rRAP-1 NT and RRA using a FliC adjuvant. DPI: days post-immunization with the recombinant antigen mix. DPC represents days post-challenge with the <span class="html-italic">B. bovis</span> strain Vir-S74-T3Bo. Each point represents the average of two replicas, and bars represent the standard deviation. The IDs of the individual animals are as follows: C1 (blue), C2 (red), C3 (grey), C4 (yellow), C5 (cyan), C6 (green), C7 (dotted blue), C8 (dotted orange), C9 (dotted cyan), C10 (dotted yellow), C11 (dotted grey), and C12 (dotted green).</p>
Full article ">Figure 4
<p><b>Immunoblot analysis.</b> Membranes were incubated with pre-immune and immune bovine sera from the selected cow C1, immunized against rRRA and rRAP-1 NT proteins. Lane 1 represents a lysate from non-infected bovine erythrocytes; Lane 2 represents an Mo7-infected bovine erythrocyte lysate from a 50% parasitemia culture; Lane 3 represents an Mo7-infected bovine erythrocyte lysate from a 65% parasitemia culture; Lane 4 represents lysates derived from the cultured <span class="html-italic">B. bovis</span> virulent T2Bo strain used in the challenge; Lane 5 represents recombinant RRA; and Lane 6 represents recombinant RAP-1 NT proteins used in the immunization mix. Red boxes show the corresponding bands. SM: Size marker.</p>
Full article ">Figure 5
<p><b>Body Temperature.</b> Averaged body temperature for the six vaccinated cows and the six mock-vaccinated cows during the days post-immunization (DPI) with the rRRA and rRap-1 NT antigen mix and the days post-challenge (DPC) with a virulent <span class="html-italic">B. bovis</span> strain. Orange and blue arrows indicate the onset of critical temps (&gt;103 °F).</p>
Full article ">Figure 6
<p><b>Hematocrit.</b> Averaged hematocrit values for the six vaccinated cows and for the six mock-vaccinated cows during days post-challenge (DPC) with a virulent <span class="html-italic">B. bovis</span> strain (<b>A</b>). Slope lines for the averaged hematocrit from the six vaccinated (blue line) and six mock-vaccinated (orange line) cows (<b>B</b>).</p>
Full article ">Figure 7
<p><b>Parasite load.</b> Estimation of parasite loads using qPCR: Quantification Cycle (Cq) values were obtained using a quantitative PCR (qPCR) analysis, performed on total DNA in blood from the six vaccinated and the six mock-vaccinated cows during days post-challenge (DPC). (<b>A</b>) Graphical representation. The 1/Cq value was plotted, since the Cq values represent values that are inverse to parasite load. (<b>B</b>) Table describing the qPCR numerical data. Estimation of the average 1/Cq values obtained from the qPCR analysis on samples from vaccinated and mock-vaccinated cows. This analysis was performed on days 1, 3, 5, and 6 post-challenge. Significative differences for day 1 (<span class="html-italic">p</span> &lt; 0.095) were determined using the <span class="html-italic">t</span>-test.</p>
Full article ">Figure 8
<p><b>Monocytes.</b> Blood cell count in mock-vaccinated and rRRA–rRAP-1 NT-vaccinated cows. Monocytes: The analysis was performed during the vaccination phase and after the challenge showed an increase in absolute number of monocytes in 3 out of 6 vaccinated animals. Comparison between the monocyte count average from the mock-vaccinated cows and each one of the six vaccinated animals showed an increase in these values for cows C1, C3, and C6 (<span class="html-italic">p</span> &lt; 0.05). Challenge day (red arrow).</p>
Full article ">Figure 9
<p><b>Neutrophils.</b> Blood cells count in mock-vaccinated and rRRA–rRAP-1 NT-vaccinated cows. Neutrophils: The analysis was performed during the vaccination phase and after the challenge showed an increase in absolute number of monocytes in 3 out of 6 vaccinated animals. Comparison between the neutrophil count average from the mock-vaccinated cows and each one of the six vaccinated animals showed an increase in these values for cows C3 and C6 (<span class="html-italic">p</span> &lt; 0.05). Challenge day (red arrow).</p>
Full article ">
21 pages, 5581 KiB  
Article
Reverse Vaccinology Integrated with Biophysics Techniques for Designing a Peptide-Based Subunit Vaccine for Bourbon Virus
by Taghreed N. Almanaa
Bioengineering 2024, 11(11), 1056; https://doi.org/10.3390/bioengineering11111056 - 23 Oct 2024
Viewed by 681
Abstract
Despite the seriousness of the disease carried by ticks, little is known about the Bourbon virus. Only three US states have recorded human cases of Bourbon virus (BRBV) infection; in all cases, a tick bite was connected with the onset of the illness. [...] Read more.
Despite the seriousness of the disease carried by ticks, little is known about the Bourbon virus. Only three US states have recorded human cases of Bourbon virus (BRBV) infection; in all cases, a tick bite was connected with the onset of the illness. The Bourbon virus (BRBV) belongs to the Orthomyxoviridae family and Thogotovirus genus, originating in the states of the US, i.e., Kansas, Oklahoma and Missouri. The growing rates of BRBV infections in various parts of the US highlight the necessity for a thorough analysis of the virus’s transmission mechanisms, vector types and reservoir hosts. Currently, there are no vaccines or efficient antiviral therapies to stop these infections. It is imperative to produce a vaccination that is both affordable and thermodynamically stable to reduce the likelihood of future pandemics. Various computational techniques and reverse vaccinology methodologies were employed to identify specific B- and T-cell epitopes. After thorough examination, the linker proteins connected the B- and T-cell epitopes, resulting in this painstakingly constructed vaccine candidate. Furthermore, 3D modeling directed the vaccine construct toward molecular docking to determine its binding affinity and interaction with TLR-4. Human beta-defensin was used as an adjuvant and linked to the N-terminus to boost immunogenicity. Furthermore, the C-IMMSIM simulation resulted in high immunogenic activities, with activation of high interferon, interleukins and immunoglobulin. The results of the in silico cloning process for E. coli indicated that the vaccine construct will try its utmost to express itself in the host, with a codon adaptation CAI value of 0.94. A net binding free energy of −677.7 kcal/mol obtained during docking showed that the vaccine has a high binding affinity for immunological receptors. Further validation was achieved via molecular dynamic simulations, inferring the confirmational changes during certain time intervals, but the vaccine remained intact to the binding site for a 100 ns interval. The thermostability determined using an RMSF score predicted certain changes in the mechanistic insights of the loop region with carbon alpha deviations, but no major changes were observed during the simulations. Thus, the results obtained highlight a major concern for researchers to further validate the vaccine’s efficacy using in vitro and in vivo approaches. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Flow chart of the current study depicting the protocol for predicting the multi-epitope vaccine construct.</p>
Full article ">Figure 2
<p>There are two distinct ways to view the final MEVC. (<b>A</b>,<b>B</b>) illustrate the arrangement of epitopes throughout the vaccine construct; (<b>C</b>) depicts the 3D model, with each component clearly colored, highlighting the vaccine folding and loop regions at N- and C-terminals.</p>
Full article ">Figure 3
<p>(<b>A</b>) depicts the Ramachandran plot with a confidence score; (<b>B</b>) shows the X-ray- and NMR-based prediction of the modeled vaccine construct; and (<b>C</b>) presents the sequence positioning in a 3D vaccine construct.</p>
Full article ">Figure 4
<p>A secondary structure diagram illustrating the presence of alpha helices (46.93%), beta strands (2.9%) and coils (51%) in the vaccine design with numerous epitopes.</p>
Full article ">Figure 5
<p>(<b>A</b>) depicts the binding pose of epitopes against nucleoprotein HTLs; (<b>B</b>) presents the binding pose of polymerase subunit PA HTLs in complex with epitopes.</p>
Full article ">Figure 6
<p>(<b>A</b>) shows the graphical insights of binding free energies for epitopes against nucleoprotein HTLs; (<b>B</b>) illustrates the binding free energies’ calculation of polymerase subunit PA HTLs in complex with the predicted epitopes.</p>
Full article ">Figure 7
<p>The red line, surrounded by a black circle, symbolizes the MEV’s restriction cloned into the pET28a(+) transcription vector in silico.</p>
Full article ">Figure 8
<p>MEBV in silico cloning and immune simulation investigations. (<b>A</b>) The amount of interleukin and interferon produced in milliliters per nanogram in response to MEBV. (<b>B</b>) The immunoglobulin response measured in microliters in response to the MEBV antigen.</p>
Full article ">Figure 9
<p>(<b>A</b>) TLR-4 receptor and vaccine with docked pose. The vaccine construct is shown in cyan and is also encoded by (C)<b>,</b> whereas the TLR-4 receptor networks A, B, C and D are colored green, red, yellow and pink, respectively. (<b>B</b>) shows the receptor’s interaction residues with vaccine construct encoded by (C).</p>
Full article ">Figure 10
<p>(<b>A</b>) An examination of the vaccine–TLR4 complexes and vaccine RMSD graphs at 100 ns time intervals. (<b>B</b>) The vaccine–TLR complexes’ RMSF plot. (<b>C</b>) The vaccine–TLR complexes’ H-bonds plot.</p>
Full article ">
12 pages, 1627 KiB  
Article
Comparative Analysis of the Neutralizing Capacity of Monovalent and Bivalent Formulations of Betuvax-CoV-2, a Subunit Recombinant COVID-19 Vaccine, Against Various Strains of SARS-CoV-2
by Anna V. Vakhrusheva, Ekaterina A. Romanovskaya-Romanko, Marina A. Stukova, Maria M. Sukhova, Ksenia S. Kuznetsova, Aleksandr V. Kudriavtsev, Maria E. Frolova, Taras V. Ivanishin, Igor V. Krasilnikov and Artur A. Isaev
Vaccines 2024, 12(10), 1200; https://doi.org/10.3390/vaccines12101200 - 21 Oct 2024
Viewed by 704
Abstract
SARS-CoV-2, the causal agent of the COVID-19 pandemic, is characterized by rapid evolution, which poses a significant public health challenge. Effective vaccines that provide robust protection, elicit strong immune responses, exhibit favorable safety profiles, and enable cost-effective large-scale production are crucial. The RBD-Fc-based [...] Read more.
SARS-CoV-2, the causal agent of the COVID-19 pandemic, is characterized by rapid evolution, which poses a significant public health challenge. Effective vaccines that provide robust protection, elicit strong immune responses, exhibit favorable safety profiles, and enable cost-effective large-scale production are crucial. The RBD-Fc-based Betuvax-CoV-2 vaccine has previously demonstrated a favorable safety profile and induced a significant anti-SARS-CoV-2 humoral immune response in clinical trials. Due to the rapid evolution and emergence of new SARS-CoV-2 strains, the relevance of bivalent vaccine formulations has increased. Methods: This study compared the neutralizing capacity of monovalent and bivalent vaccine formulations against different SARS-CoV-2 strains detected with a SARS-CoV-2 microneutralization assay (MNT). Findings: The monovalent Wuhan-based vaccine generated neutralizing antibodies against the Wuhan and Omicron BA.2 variants but not the distinct Omicron BQ.1 strain. Conversely, the monovalent BA.2-based vaccine induced neutralizing antibodies against both Omicron strains but not Wuhan. While the bivalent Wuhan and BA.2-based vaccine was effective against strains containing the same antigens, it was insufficient to neutralize the distinctive BQ.1 strain at a small dosage. Interpretation: These findings suggest that the vaccine composition should closely match the circulating SARS-CoV-2 strain to elicit the optimal neutralizing antibody response and include the appropriate dosage. Moreover, this study did not find additional advantages of using the bivalent form over the monovalent form for the vaccination against a single prevailing SARS-CoV-2 strain. Full article
(This article belongs to the Special Issue SARS-CoV-2 Infections; Treatment and Development of Vaccine)
Show Figures

Figure 1

Figure 1
<p>Betuvax-CoV-2 vaccine. Schematic of the RBD-Fc antigen construction (<b>A</b>); graphic representation of Betuvax-CoV-2 betulin-based nanoparticle with absorbed antigens (<b>B</b>); and transmission electron microscopy of the Betuvax-CoV-2 vaccine, the bar size in 100 nm (<b>C</b>).</p>
Full article ">Figure 2
<p>Scheme of BALB/c mice immunization.</p>
Full article ">Figure 3
<p>The level of IgG-specific antibodies to the RBD-Fc protein of the SARS-CoV-2 coronavirus variant Wuhan (<b>A</b>), BA.2 (<b>B</b>), and BQ.1 (<b>C</b>) in BALB/c mice after single and double immunization with samples of the candidate vaccine or placebo. The GMT ± standard deviation group is presented. For statistical processing, a two-way ANOVA test with Sidak’s post hoc test was used (**** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> &lt; 0.001, ns—non-significant).</p>
Full article ">Figure 4
<p>The level of neutralizing antibodies to the SARS-CoV-2 strains Wuhan (<b>A</b>), BA.2 (<b>B</b>), and BQ.1 (<b>C</b>) in BALB/c mice after double immunization with samples of the candidate vaccine. The GMT ± standard deviation group is presented. For statistical processing, a one-way ANOVA test with Dannet’s test was used (**** <span class="html-italic">p</span> &lt; 0.0001, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
36 pages, 3132 KiB  
Review
The Ambivalence of Post COVID-19 Vaccination Responses in Humans
by Radha Gopalaswamy, Vivekanandhan Aravindhan and Selvakumar Subbian
Biomolecules 2024, 14(10), 1320; https://doi.org/10.3390/biom14101320 - 17 Oct 2024
Viewed by 1608
Abstract
The Coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has prompted a massive global vaccination campaign, leading to the rapid development and deployment of several vaccines. Various COVID-19 vaccines are under different phases of clinical trials and include [...] Read more.
The Coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has prompted a massive global vaccination campaign, leading to the rapid development and deployment of several vaccines. Various COVID-19 vaccines are under different phases of clinical trials and include the whole virus or its parts like DNA, mRNA, or protein subunits administered directly or through vectors. Beginning in 2020, a few mRNA (Pfizer-BioNTech BNT162b2 and Moderna mRNA-1273) and adenovirus-based (AstraZeneca ChAdOx1-S and the Janssen Ad26.COV2.S) vaccines were recommended by WHO for emergency use before the completion of the phase 3 and 4 trials. These vaccines were mostly administered in two or three doses at a defined frequency between the two doses. While these vaccines, mainly based on viral nucleic acids or protein conferred protection against the progression of SARS-CoV-2 infection into severe COVID-19, and prevented death due to the disease, their use has also been accompanied by a plethora of side effects. Common side effects include localized reactions such as pain at the injection site, as well as systemic reactions like fever, fatigue, and headache. These symptoms are generally mild to moderate and resolve within a few days. However, rare but more serious side effects have been reported, including allergic reactions such as anaphylaxis and, in some cases, myocarditis or pericarditis, particularly in younger males. Ongoing surveillance and research efforts continue to refine the understanding of these adverse effects, providing critical insights into the risk-benefit profile of COVID-19 vaccines. Nonetheless, the overall safety profile supports the continued use of these vaccines in combating the pandemic, with regulatory agencies and health organizations emphasizing the importance of vaccination in preventing COVID-19’s severe outcomes. In this review, we describe different types of COVID-19 vaccines and summarize various adverse effects due to autoimmune and inflammatory response(s) manifesting predominantly as cardiac, hematological, neurological, and psychological dysfunctions. The incidence, clinical presentation, risk factors, diagnosis, and management of different adverse effects and possible mechanisms contributing to these effects are discussed. The review highlights the potential ambivalence of human response post-COVID-19 vaccination and necessitates the need to mitigate the adverse side effects. Full article
Show Figures

Figure 1

Figure 1
<p>Summary of host response to COVID-19 vaccines. The COVID-19 vaccination-induced host responses can be broadly divided into immediate or delayed hypersensitivity. While the former response elicits allergic reactions and anaphylaxis, the latter response results in mild, moderate, or severe adverse events. The immediate hypersensitivity response is caused either by a classical, IgE-mediated activation of mast cells and basophils or an alternative non-classical pathway involving IgG and other antibodies activating neutrophils and basophils. Autoimmunity due to COVID-19 vaccination can be caused by molecular mimicry, bystander activation of immune cells, viral epitope spreading, or adjuvant-mediated immune response. The overall magnitude and durability of immune response as well as adverse effects mediated by COVID-19 vaccination are determined by several factors, including the age, sex, genetic makeup, immune status, and underlying health conditions of the host as well as the nature of the vaccine used. Image created in Biorender.</p>
Full article ">Figure 2
<p>Effects of COVID-19 vaccination-induced immunity. Following vaccination, the immune response against COVID-19 is mediated mainly by the development of Abs against SARS-CoV-2 proteins. The magnitude of immune response developed and its impact on the host protection is determined by the nature of Ab response elicited. An effective neutralizing Ab response neutralizes the virus, controls the infecting viral load and protects the vaccinated host against severe disease and/or death due to infection. However, a sub-optimal non-neutralizing Ab response leads to poor neutralization of the virus and ineffective control of viral load in the organs and may also contribute to Ab-mediated adverse effects (AE), which may enhance the disease manifestations. Image created in Biorender.</p>
Full article ">Figure 3
<p>Key pathways of COVID-19 vaccine-induced adverse immune reactions. The COVID-19 vaccine is comprised of the SARS-CoV-2 S protein (either as mRNA or protein) combined with an adjuvant such as polyethylene glycol (PEG). In the classical pathway, internalization of the viral and adjuvant-derived antigens (Ag) in the vaccine by antigen-presenting cells (APC) results in the presentation of antigenic epitopes to the T helper (Th) cells, which produces cytokines and activates Ag-specific B cells to produce various antibodies, such as IgG, IgE, IgM, etc. The Ag-specific IgE Abs binds to the FcεR1 and activates basophils and mast cells to produce histamine, which leads to allergy and/or anaphylaxis reactions. In the non-classical pathway, the antigens were taken up directly by the MRGPRX2 receptor on mast cells, which results in the induction of histamine and allergic responses. In addition, the immune complex formation by the Ag-specific and/or anti-idiotypic IgG, IgE, IgM Abs activates the C3a and C5a complement components, which ultimately results in complement activation-related pseudo-allergic reaction (CARPA). Finally, in the alternative/additional pathway, the antigen–IgG complex is taken up by neutrophils through FcγRs, which activates these polymorphonuclear cells to produce reactive oxygen species (ROS), proteases such as neutrophil-elastases (NE), Protease-3 (PR3), cathepsin G (CatG), and the formation of neutrophil extracellular traps (NETosis). The combined action of these pathways may contribute to the overall allergy and anaphylactic response due to COVID-19 vaccination. Image created in Biorender.</p>
Full article ">Figure 4
<p>Various mechanisms of adverse immune activation by COVID-19 vaccines. The viral S protein, either as mRNA or recombinant, adenovector-DNA, is endocytosed through Toll-like receptors (TLR) present on antigen-presenting cells (APCs). These endosomes trigger intracellular signaling pathways that result in the activation of Interferon regulatory factor-7 (IRF-7) and nuclear factor k B (NFkB) networks. Activated IRF7 and NFkB upregulate the production of proinflammatory cytokines IL-6 and TNFα. Alternatively, the viral components can escape from the endosome and trigger the cGAS signaling pathway, which activates STING/IRF3 network that ultimately results in the upregulation of proinflammatory type I interferons (IFN) response. Finally, the viral nucleic acids are translated into peptides and presented by the APC to activate T cells through the T cell receptor (TcR). Activation of naïve T cells results in the production of cytokines. Exposure to IL-4 skews the naïve T cells to an anti-inflammatory, Th2-type T cells that produce IL-3, IL-5, and IL-9, all of which can activate mast cells to elicit an allergic/anaphylactic reaction. In contrast, exposure to IL-12 and IFNγ polarizes the naïve T cells into Th1-type cells, which contributes to the proinflammatory response. Apart from the viral-derived molecules, vaccine adjuvants, such as CpG, can be recognized by TLR on the APC, with further activation of the NFkB pathway, leading to the production of inflammatory response. The viral nucleic acids also form a complex with platelet factor-4 (PF4) produced by the blood platelets. This complex activates Ag-specific B cells to produce anti-DNA/PF4 complex IgG, which binds with the FCγRIIa receptor on the platelets and activates these cells to form aggregates, leading to vaccine-induced thrombotic thrombocytopenia (VITT). Thus, both APCs and platelets play divergent roles in mounting immune dysregulation upon exposure to viral antigens and/or adjuvants. Image created in Biorender.</p>
Full article ">
22 pages, 3581 KiB  
Article
Immunopeptidomics of Salmonella enterica Serovar Typhimurium-Infected Pig Macrophages Genotyped for Class II Molecules
by Carmen Celis-Giraldo, Carlos F. Suárez, William Agudelo, Nieves Ibarrola, Rosa Degano, Jaime Díaz, Raúl Manzano-Román and Manuel A. Patarroyo
Biology 2024, 13(10), 832; https://doi.org/10.3390/biology13100832 - 16 Oct 2024
Viewed by 1129
Abstract
Salmonellosis is a zoonotic infection that has a major impact on human health; consuming contaminated pork products is the main source of such infection. Vaccination responses to classic vaccines have been unsatisfactory; that is why peptide subunit-based vaccines represent an excellent alternative. Immunopeptidomics [...] Read more.
Salmonellosis is a zoonotic infection that has a major impact on human health; consuming contaminated pork products is the main source of such infection. Vaccination responses to classic vaccines have been unsatisfactory; that is why peptide subunit-based vaccines represent an excellent alternative. Immunopeptidomics was used in this study as a novel approach for identifying antigens coupled to major histocompatibility complex class II molecules. Three homozygous individuals having three different haplotypes (Lr-0.23, Lr-0.12, and Lr-0.21) were thus selected as donors; peripheral blood macrophages were then obtained and stimulated with Salmonella typhimurium (MOI 1:40). Although similarities were observed regarding peptide length distribution, elution patterns varied between individuals; in total, 1990 unique peptides were identified as follows: 372 for Pig 1 (Lr-0.23), 438 for Pig 2 (Lr.0.12) and 1180 for Pig 3 (Lr.0.21). Thirty-one S. typhimurium unique peptides were identified; most of the identified peptides belonged to outer membrane protein A and chaperonin GroEL. Notably, 87% of the identified bacterial peptides were predicted in silico to be elution ligands. These results encourage further in vivo studies to assess the immunogenicity of the identified peptides, as well as their usefulness as possible protective vaccine candidates. Full article
(This article belongs to the Section Infection Biology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Comparative peptide length distribution: (<b>a</b>) frequency peptide; (<b>b</b>) peptide percentage.</p>
Full article ">Figure 2
<p>GibbsCluster 2.0 results for the dataset for peptides obtained per pig in this study. Sequences: the number of peptides used for each analysis. Group: the number of peptides selected by the server for creating elution motifs. Trash cluster: the percentage of peptides removed as being outliers, considering a 2 threshold.</p>
Full article ">Figure 3
<p>Distribution of eluted ligand (EL) prediction, considering total data percentages: strong EL (&lt;1%Rank), weak EL (&lt;5%Rank), and no EL (≥5%Rank).</p>
Full article ">Figure 4
<p>Comparison of the logos obtained, considering only peptides predicted to be eluted ligands (EL &lt; 5%Rank). All the results were considered for creating the MHC II (<b>a</b>), SLA-DR (<b>b</b>), and SLA-DQ (<b>c</b>) logos for each pig.</p>
Full article ">Figure 4 Cont.
<p>Comparison of the logos obtained, considering only peptides predicted to be eluted ligands (EL &lt; 5%Rank). All the results were considered for creating the MHC II (<b>a</b>), SLA-DR (<b>b</b>), and SLA-DQ (<b>c</b>) logos for each pig.</p>
Full article ">Figure 5
<p>A MixMHC2pred motif was predicted from the haplotype sequences for the pigs used in this study.</p>
Full article ">Figure 6
<p>A NetMHCIIpan-4.0 motif was predicted from the haplotype sequences from the pigs used in this study.</p>
Full article ">Figure 7
<p>Distribution of Salmonella enterica gene, serovar Typhimurium enrichment identified in this study.</p>
Full article ">
15 pages, 4592 KiB  
Article
Inserting CTL Epitopes of the Viral Nucleoprotein to Improve Immunogenicity and Protective Efficacy of Recombinant Protein against Influenza A Virus
by Marina Shuklina, Liudmila Stepanova, Olga Ozhereleva, Anna Kovaleva, Inna Vidyaeva, Alexandr Korotkov and Liudmila Tsybalova
Biology 2024, 13(10), 801; https://doi.org/10.3390/biology13100801 - 7 Oct 2024
Viewed by 1102
Abstract
Conserved influenza virus proteins, such as the hemagglutinin stem domain (HA2), nucleoprotein (NP), and matrix protein (M), are the main targets in the development of universal influenza vaccines. Previously, we constructed a recombinant vaccine protein Flg-HA2-2-4M2ehs containing the extracellular domain of the M2 [...] Read more.
Conserved influenza virus proteins, such as the hemagglutinin stem domain (HA2), nucleoprotein (NP), and matrix protein (M), are the main targets in the development of universal influenza vaccines. Previously, we constructed a recombinant vaccine protein Flg-HA2-2-4M2ehs containing the extracellular domain of the M2 protein (M2e) and the aa76–130 sequence of the second HA subunit as target antigens. It demonstrated immunogenicity and broad protection against influenza A viruses after intranasal and parenteral administration. This study shows that CD8+ epitopes of NP, inserted into a flagellin-fused protein carrying M2e and HA2, affect the post-vaccination immune humoral response to virus antigens without reducing protection. No differences were found between the two proteins in their ability to stimulate the formation of follicular Th in the spleen, which may contribute to a long-lasting antigen-specific humoral response. The data obtained on Balb/c mice suggest that the insertion of CTL NP epitopes into the flagellin-fused protein carrying M2e and HA2 reduces the antibody response to M2e and A/H3N2. In C57Bl6 mice, this stimulates the formation of NP-specific CD8+ Tem and virus-specific mono- and multifunctional CD4+ and CD8+ Tem in the spleen and completely protects mice from influenza virus subtypes A/H1N1pdm09 and A/H3N2. Full article
(This article belongs to the Section Immunology)
Show Figures

Figure 1

Figure 1
<p>Schematic structure (<b>A</b>) and structural 3D models (<b>B</b>) of monomeric recombinant fusion proteins Flg-HA2-2-4M2ehs and Flg-HA2-2-NP335-NP255-4M2ehs: orange—M2es, pink—M2eh, blue—the HA2-2 fragment, yellow—the NP fragments, and green—flagellin. SDS-PAGE Coomassie brilliant blue staining and Western blot (<b>C</b>) of the recombinant fusion proteins Flg-HA2-2-NP335-NP255-4M2ehs (lane 1), Flg-HA2-2-4M2ehs (lane 2), and Flagellin (lane 3) are also shown.</p>
Full article ">Figure 2
<p>Design of the experiment.</p>
Full article ">Figure 3
<p>Antibody response in serum (log2 titers). BALB/c mice (<span class="html-italic">n</span> = 5/group) were immunized s.c. with 10 μg of Flg-HA2-2-4M2ehs or Flg-HA2-2-NP335-NP255-4M2ehs on days 0, 14, and 28. The control group of mice received PBS. Two weeks after the second boost, M2e-specific IgG (<b>A</b>,<b>B</b>), anti-M2e IgG subclasses (<b>C</b>), IgA (<b>D</b>), and virus-specific IgG (<b>E</b>,<b>F</b>) titers were evaluated by ELISA. The data are presented as Tukey plots. Statistically significant differences between groups are indicated as *—<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>The populations of CD4+CD44+CXCR5+ Tfh cells in lymph nodes (<b>A</b>) and spleens (<b>B</b>) on day 7 after the third immunization with Flg-HA2-2-4M2ehs or Flg-HA2-2-NP335-NP255-4M2ehs. The data are presented as Tukey plots. <span class="html-italic">p</span> values indicate statistically significant differences between groups: **—<span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>M2e-specific (<b>A</b>,<b>B</b>) and NP-peptide-specific (<b>C</b>,<b>D</b>) CD8+ and CD4+ effector memory T cells in spleens and the cytokine profile after subcutaneous immunization of C57Bl6 mice with the recombinant proteins Flg-HA2-2-4M2ehs and Flg-HA2-2-NP335-NP255-4M2ehs. The data are presented as Tukey plots. Significant differences from the control are shown as *—<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 6
<p>H3N2-specific CD8+ (<b>A</b>) and CD4+ (<b>B</b>) effector memory T cells and the cytokine profile in spleens after subcutaneous immunization of C57Bl6 mice with the recombinant proteins Flg-HA2-2-4M2ehs and Flg-HA2-2-NP335-NP255-4M2ehs. Expression of the degranulation marker CD107a+ (<b>C</b>) and expression of CD107a+ and IFN-γ (<b>D</b>) on A/H3N2-specific CD4+ and CD8+ effector memory T cells in the spleen of C57Bl6 mice are also presented. The data are presented as Tukey plots. Significant differences between experimental groups are shown as * —<span class="html-italic">p</span> &lt; 0.05; **—<span class="html-italic">p</span> &lt; 0.01; and ****—<span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 7
<p>Efficacy of immunization. Balb/c mice (<span class="html-italic">n</span> = 10/group) were immunized with fusion proteins Flg-HA2–2-4M2ehs and Flg-HA2-2-NP335-NP255-4M2ehs. Two weeks after the second boost, mice were challenged with 10 LD50 of (<b>A</b>) A/California/07/09 (H1N1pdm09) or (<b>B</b>) A/Aichi/2/68 (H3N2). Body weight (left) and survival rates (right) were monitored daily for 14 days. The <span class="html-italic">p</span> values (Mantel-Cox test) indicating statistically significant differences between immunized and control groups are presented as *—<span class="html-italic">p</span> &lt; 0.05; ***—<span class="html-italic">p</span> &lt; 0.001; and ****—<span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
19 pages, 2968 KiB  
Review
Self-Assembling Peptides for Vaccine Adjuvant Discovery
by Jingyi Fan, Istvan Toth and Rachel J. Stephenson
Immuno 2024, 4(4), 325-343; https://doi.org/10.3390/immuno4040021 - 1 Oct 2024
Viewed by 884
Abstract
Vaccination is credited as a significant medical achievement contributing to the decline in morbidity and mortality of infectious diseases. Traditional vaccines composed of inactivated and live-attenuated whole pathogens confer the induction of potent and long-term immune responses; however, traditional vaccines pose a high [...] Read more.
Vaccination is credited as a significant medical achievement contributing to the decline in morbidity and mortality of infectious diseases. Traditional vaccines composed of inactivated and live-attenuated whole pathogens confer the induction of potent and long-term immune responses; however, traditional vaccines pose a high risk of eliciting autoimmune and allergic responses as well as inflammations. New modern vaccines, such as subunit vaccines, employ minimum pathogenic components (such as carbohydrates, proteins, or peptides), overcome the drawbacks of traditional vaccines and stimulate effective immunity against infections. However, the low immunogenicity of subunit vaccines requires effective immune stimulants (adjuvants), which are an indispensable factor in vaccine development. Although there are several approved adjuvants in human vaccines, the challenges of matching and designing appropriate adjuvants for specific vaccines, along with managing the side effects and toxicity of existing adjuvants in humans, are driving the development of new adjuvants. Self-assembling peptides are a promising biomaterial rapidly emerging in the fields of biomedicine, vaccination and material science. Here, peptides self-assemble into ordered supramolecular structures, forming different building blocks in nanoparticle size, including fibrils, tapes, nanotubes, micelles, hydrogels or nanocages, with great biostability, biocompatibility, low toxicity and effectiveness at controlled release. Self-assembling peptides are effective immunostimulatory agents used in vaccine development to enhance and prolong immune responses. This review describes the predominant structures of self-assembling peptides and summarises their recent applications as vaccine adjuvants. Challenges and future perspectives on self-assembled peptides as vaccine adjuvants are also highlighted. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic representations of supramolecular self-assembling peptides.</p>
Full article ">Figure 2
<p>(<b>a</b>) Ball and stick model of an α-helical secondary structure (side view), and (<b>b</b>) heptad wheel representation of a coiled-coil α-helical structure.</p>
Full article ">Figure 3
<p>Self-assembling nanocages from coiled-coil peptides [<a href="#B27-immuno-04-00021" class="html-bibr">27</a>].</p>
Full article ">Figure 4
<p>(<b>a</b>) Ball and stick model of a β-sheet (side view), and (<b>b</b>) β-sheets secondary structure indicating both antiparallel and parallel β-sheets.</p>
Full article ">Figure 5
<p>Schematic structure of peptide amphiphiles. Region 1 is a hydrophobic domain composed of hydrophobic amino acids/alkyl/aromatic groups. Region 2 is a β-sheet or α-helix segment for the interfacial curvature (β-sheet or α-helix) of self-assembled structures by hydrogen bonds in the aqueous. Region 3 composed of charged amino acids (including arginine (Arg), glutamic acid (Glu), and lysine (Lys)) increase solubility of amphiphilic peptides, and Region 4 confers a functional peptide epitope specific to the disease target [<a href="#B47-immuno-04-00021" class="html-bibr">47</a>].</p>
Full article ">Figure 6
<p>Schematic structure of a cyclic peptide self-assembling into a nanotube.</p>
Full article ">Figure 7
<p>Coil29 conjugated to CD8<sup>+</sup> T cell epitope. PADRE is a T helper peptide and SIINFEKL is a T cell OVA peptide.</p>
Full article ">Figure 8
<p>Structures of (<b>a</b>) Q11 and (<b>b</b>) Q11-PAS.</p>
Full article ">Figure 9
<p>Examples of three classes of peptide amphiphiles (<b>a</b>) Amphiphilic peptide containing 15 leucine (Leu) residues as typical hydrophobic domain. (<b>b</b>) Lipopeptide is composed of two long hydrophobic lipid alkyl chains as a lipidated peptide. (<b>c</b>) Supramolecular peptide amphiphile conjugates composed of 1-adamantaneacetic acid coupled to the short peptide GFFY(K) <sub>n</sub> (n = 2,3) with formulation of nanofibers in phosphate buffered saline (PBS).</p>
Full article ">Figure 10
<p>Structures of example cyclic peptides, (<b>a</b>) cyclic decapeptide and (<b>b</b>) cycl (D-Trp-Tyr) [<a href="#B91-immuno-04-00021" class="html-bibr">91</a>].</p>
Full article ">
Back to TopTop