Host Innate Immune Responses against SARS-CoV-2

A special issue of Vaccines (ISSN 2076-393X). This special issue belongs to the section "COVID-19 Vaccines and Vaccination".

Deadline for manuscript submissions: closed (31 March 2022) | Viewed by 16246

Special Issue Editor

Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
Interests: host-pathogen interaction; inflammasome; cGAS STING; HPV cancers; RIG-I

Special Issue Information

Dear Colleagues,

The novel coronavirus (CoV) disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has rapidly evolved into pandemic scale since it first appeared in Wuhan, China. Most often, the morbidity and mortality associated with SAR-CoV-2 infection are due to the collateral damage caused by exaggerated and unabated immune response (“cytokine storm”) mounted by the host to protect against the infection. It is now apparent that the induction of a “cytokine storm” is the root cause of severity sequelae, such as acute respiratory distress syndrome (ARDS) and multi-organ dysfunction in late-stage COVID-19 patients. The induction of a cytokine storm is the root cause of pathogenic inflammation in SARS-CoV-2-infected patients. For those whom were critically ill or have died from SARS-CoV-2 infection, severe clinical manifestations did not develop in the early stages of the disease. In fact, these patients deteriorated suddenly in the later stages of the disease or in the process of recovery, rapidly succumbing to ARDS/acute lung injury and multiple-organ failure, resulting in death within a short time. Cytokine storm is considered to be one of the major causes of ARDS and multiple-organ failure and plays an important role in the process of disease aggravation. This Special Issue of Vaccines will cover all of these topics relevant to understanding host innate immune responses against SARS-CoV-2.

Dr. Adi Idris
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Vaccines is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • SARS-CoV-2
  • inflammasome
  • interferon
  • toll-like receptors

Published Papers (3 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

7 pages, 1403 KiB  
Communication
A Peptide-Based Assay Discriminates Individual Antibody Response to the COVID-19 Pfizer/BioNTech mRNA Vaccine
by Immacolata Polvere, Serena Voccola, Alfredina Parrella, Gaetano Cardinale, Lucrezia Zerillo, Romualdo Varricchio, Jessica Raffaella Madera, Romania Stilo, Pasquale Vito and Tiziana Zotti
Vaccines 2021, 9(9), 987; https://doi.org/10.3390/vaccines9090987 - 3 Sep 2021
Cited by 1 | Viewed by 2051
Abstract
The coronavirus disease 2019 (COVID-19) mRNA vaccine developed by Pfizer/BioNTech has been shown to be capable of developing an excellent antibody response against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein, with good production of neutralizing antibodies. Herein, we analyzed differences [...] Read more.
The coronavirus disease 2019 (COVID-19) mRNA vaccine developed by Pfizer/BioNTech has been shown to be capable of developing an excellent antibody response against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein, with good production of neutralizing antibodies. Herein, we analyzed differences in the antibody response elicited by inoculation of the Pfizer/BioNTech vaccine through a peptide-based enzyme-linked immunosorbent assay (ELISA) that utilizes synthetic peptides derived from the spike protein in the immuno-adsorbent phase. Immunoreactivity against synthetic peptides was measured at different time points from vaccination and was also correlated with the SARS-CoV-2 neutralizing capacity. Our results indicate that all vaccinated subjects except one show reactive antibodies to at least one peptide at both 30 and 60 days after injection of the first dose. Only one of the 19 analyzed subjects showed no antibody response toward any of the selected peptides, consistently with a lower neutralizing capacity. More importantly, our data showed that the antibody response elicited by inoculation of the two doses of the Pfizer vaccine appears to be qualitatively individual, both in the type of recognized peptides and in the temporal persistence of the antibody response. Together with previous published data, our findings suggest that for effective pandemic control, it is important to constantly monitor the antibody protection in the population, and the assay described here could be a valid tool for this purpose. Full article
(This article belongs to the Special Issue Host Innate Immune Responses against SARS-CoV-2)
Show Figures

Figure 1

Review

Jump to: Research

28 pages, 1187 KiB  
Review
Surviving the Storm: Cytokine Biosignature in SARS-CoV-2 Severity Prediction
by Rahnuma Ahmad and Mainul Haque
Vaccines 2022, 10(4), 614; https://doi.org/10.3390/vaccines10040614 - 14 Apr 2022
Cited by 10 | Viewed by 3235
Abstract
A significant part of the world population has been affected by the devastating SARS-CoV-2 infection. It has deleterious effects on mental and physical health and global economic conditions. Evidence suggests that the pathogenesis of SARS-CoV-2 infection may result in immunopathology such as neutrophilia, [...] Read more.
A significant part of the world population has been affected by the devastating SARS-CoV-2 infection. It has deleterious effects on mental and physical health and global economic conditions. Evidence suggests that the pathogenesis of SARS-CoV-2 infection may result in immunopathology such as neutrophilia, lymphopenia, decreased response of type I interferon, monocyte, and macrophage dysregulation. Even though most individuals infected with the SARS-CoV-2 virus suffer mild symptoms similar to flu, severe illness develops in some cases, including dysfunction of multiple organs. Excessive production of different inflammatory cytokines leads to a cytokine storm in COVID-19 infection. The large quantities of inflammatory cytokines trigger several inflammation pathways through tissue cell and immune cell receptors. Such mechanisms eventually lead to complications such as acute respiratory distress syndrome, intravascular coagulation, capillary leak syndrome, failure of multiple organs, and, in severe cases, death. Thus, to devise an effective management plan for SARS-CoV-2 infection, it is necessary to comprehend the start and pathways of signaling for the SARS-CoV-2 infection-induced cytokine storm. This article discusses the current findings of SARS-CoV-2 related to immunopathology, the different paths of signaling and other cytokines that result in a cytokine storm, and biomarkers that can act as early signs of warning for severe illness. A detailed understanding of the cytokine storm may aid in the development of effective means for controlling the disease’s immunopathology. In addition, noting the biomarkers and pathophysiology of severe SARS-CoV-2 infection as early warning signs can help prevent severe complications. Full article
(This article belongs to the Special Issue Host Innate Immune Responses against SARS-CoV-2)
Show Figures

Figure 1

15 pages, 567 KiB  
Review
mRNA COVID-19 Vaccines and Long-Lived Plasma Cells: A Complicated Relationship
by Girolamo Giannotta and Nicola Giannotta
Vaccines 2021, 9(12), 1503; https://doi.org/10.3390/vaccines9121503 - 20 Dec 2021
Cited by 17 | Viewed by 10167
Abstract
mRNA COVID-19 vaccines have hegemonized the world market, and their administration to the population promises to stop the pandemic. However, the waning of the humoral immune response, which does not seem to last so many months after the completion of the vaccination program, [...] Read more.
mRNA COVID-19 vaccines have hegemonized the world market, and their administration to the population promises to stop the pandemic. However, the waning of the humoral immune response, which does not seem to last so many months after the completion of the vaccination program, has led us to study the molecular immunological mechanisms of waning immunity in the case of mRNA COVID-19 vaccines. We consulted the published scientific literature and from the few articles we found, we were convinced that there is an immunological memory problem after vaccination. Although mRNA vaccines have been demonstrated to induce antigen-specific memory B cells (MBCs) in the human population, there is no evidence that these vaccines induce the production of long-lived plasma cells (LLPCs), in a SARS-CoV-2 virus naïve population. This obstacle, in our point of view, is caused by the presence, in almost all subjects, of a cellular T and B cross-reactive memory produced during past exposures to the common cold coronaviruses. Due to this interference, it is difficult for a vaccination with the Spike protein alone, without adjuvants capable of prolonging the late phase of the generation of the immunological memory, to be able to determine the production of protective LLPCs. This would explain the possibility of previously and completely vaccinated subjects to become infected, already 4–6 months after the completion of the vaccination cycle. Full article
(This article belongs to the Special Issue Host Innate Immune Responses against SARS-CoV-2)
Show Figures

Figure 1

Back to TopTop